ArticlePDF AvailableLiterature Review

Astroglia in dementia and Alzheimer's disease

Authors:

Abstract and Figures

Astrocytes, the most numerous cells in the brain, weave the canvas of the grey matter and act as the main element of the homoeostatic system of the brain. They shape the microarchitecture of the brain, form neuronal-glial-vascular units, regulate the blood-brain barrier, control microenvironment of the central nervous system and defend nervous system against multitude of insults. Here, we overview the pathological potential of astroglia in various forms of dementias, and hypothesise that both atrophy of astroglia and reactive hypertrophic astrogliosis may develop in parallel during neurodegenerative processes resulting in dementia. We also show that in the transgenic model of Alzheimer's disease, reactive hypertrophic astrocytes surround the neuritic plaques, whereas throughout the brain parenchyma astroglial cells undergo atrophy. Astroglial atrophy may account for early changes in synaptic plasticity and cognitive impairments, which develop before gross neurodegenerative alterations.
Content may be subject to copyright.
Review
Astroglia in dementia and Alzheimer’s disease
JJ Rodrı
´
guez*
,1,2
, M Olabarria
1
, A Chvatal
2
and A Verkhratsky*
,1,2
Astrocytes, the most numerous cells in the brain, weave the canvas of the grey matter and act as the main element of the
homoeostatic system of the brain. They shape the microarchitecture of the brain, form neuronal-glial-vascular units, regulate the
blood–brain barrier, control microenvirionment of the central nervous system and defend nervous system against multitude of
insults. Here, we overview the pathological potential of astroglia in various forms of dementias, and hypothesise that both
atrophy of astroglia and reactive hypertrophic astrogliosis may develop in parallel during neurodegenerative processes
resulting in dementia. We also show that in the transgenic model of Alzheimer’s disease, reactive hypertrophic astrocytes
surround the neuritic plaques, whereas throughout the brain parenchyma astroglial cells undergo atrophy. Astroglial atrophy
may account for early changes in synaptic plasticity and cognitive impairments, which develop before gross neurodegenerative
alterations.
Cell Death and Differentiation (2009) 16, 378–385; doi:10.1038/cdd.2008.172; published online 5 December 2008
Astrocytes, for the first time visualised by Otto Deiters
1
and
Christianised by Michael von Lenhossek
2
(for historical
overview, see also references
3,4
), are the main type of glia
in the central nervous system (CNS). The astrocytes, which
dwell in both grey and white matter, are the main element of
brain homoeostatic system, being responsible for all aspects
of metabolic support, nutrition, control of ion and neurotrans-
mitter environment, regulation of brain-blood barrier and
defence of the CNS.
5,6
In addition, astroglial cells are
endowed with numerous signalling cascades, which include
a wide array of plasmalemmal neurotransmitter receptors and
intracellular second messenger pathways.
7–10
These path-
ways in combination with trans-cellular communication routes
represented by gap junctions
11,12
and regulated gliotransmit-
ter release
13
support information processing within glial
networks, the mechanisms and functional relevance of which
remains essentially enigmatic.
The main type of astroglia in the grey matter, the
protoplasmic astrocytes, are characterised by complex
morphology with elaborated processes, which cover neuronal
membranes, residing within the astrocytic territories; the latter
being defined by the arborisation of a single astrocyte.
Importantly, a single-astroglial microanatomical domain is
essentially free from the processes of other astrocytes, giving
the grey matter in the brain and in the spinal cord a regular
parcellating structure.
14
A single astrocyte in the rodent brain
has a volume B66 000 mm
3
, and the membrane of this
astrocyte covers around 140 000 synapses lying within the
astroglial domain;
15
human astrocytes are considerably larger
and more complex and their processes wrap up to 2 millions of
synapses.
16
Protoplasmic astrocytes send processes to
neighbouring blood vessels, where these processes form
endfeet plastering the capillary wall and participating in
formation of blood–brain barrier. As a result, astroglial cells
integrate neurones and blood vessels into relatively indepen-
dent neuronal-glial-vascular units. Astroglial endfeet are
capable of releasing vasoactive substances that control local
blood flow and coordinate the latter with neuronal activity.
17–19
Individual astroglial domains are further integrated into an
internally continuous superstructure, because the peripheral
processes of astroglial cells are coupled through gap
junctions, which form a specific pathway for intercellular
communications binding astroglia into a functional syncytium.
This coupling most likely has anatomical specificity, providing
for specific organisation of glial networks: for example, in
somato-sensory cortex astroglial coupling is restricted to the
Received 25.7.08; revised 23.10.08; accepted 24.10.08; Edited by N Bazan; published online 05.12.08
1
Faculty of Life Sciences, The University of Manchester, Manchester, UK and
2
Institute of Experimental Medicine, ASCR, Videnska 1083, 142 20 Prague 4, Czech
Republic
*Corresponding authors: JJ Rodrı
´
guez, Faculty of Life Sciences, The University of Manchester, 1.124 Stopford Building, Oxford Road, Manchester M13 9PT, UK.
E-mail: Jose.Rodriguez-arellano@manchester.ac.uk; Tel: þ 44 161 275 7324; Fax: þ 44 161 275 3938
or A Verkhratsky, Faculty of Life Sciences, The University of Manchester, 1.124 Stopford Building, Oxford Road, Manchester M13 9PT, UK.
E-mail: alex.verkhratsky@manchester.ac.uk; Tel: þ 44 161 275 5414; Fax: þ 44 161 275 3938
Keywords: astrocyte; Glia/Alzheimer’s disease; dementia
Abbreviations: 3xTg-AD, triple-transgenic mice model of AD; AD, Alzheimer’s disease; APP, Amyloid precursor protein; APP/ApoE-KO, Double-transgenic mice
overexpressing a human mutated form of APP (APP
V717F
) in combination with ApoE gene knockout; APP
751SL
, Single-transgenic mice expressing APP mutation APP
Ser
751
to Leu; APP
751SL
/PS1
M146L
, Double-transgenic mice expressing APP
751SL
and PS1
M146L
; APP
SWE
, Single-transgenic mice expressing the ‘Swedish’ APP double-
point mutations Lys
670
to Asn and Met
671
to Leu; APP
SWE
/PS1
dE9
, Double-transgenic mice expressing APPswe and PS1
dE9
; CNS, central nervous system; FAD,
Familial form of Alzheimer’s disease; GFAP, glial fibrillary acidic protein; HAD, HIV-1-associated dementia; nAChR, nicotinic cholinoreceptor; nbm, nucleus basalis
magnocellularis; PDAPP, Single-transgenic mice expressing the ‘Swedish’ and ‘Indiana’ APP mutations Asp
664
to Ala (PDAPP(D664A); PS1, Preseniline 1; PS1
dE9
,
Single-transgenic mice (line S-9) expressing human PS1 carrying the exon 9-deleted variant associated with FAD; PS1
M146L
, Single-transgenic mice expressing PS1
mutation Met
146
to Leu; Tau
P301L
, Single-transgenic mice expressing the Pro
301
to Leu tau mutation (P301L)
Cell Death and Differentiation (2009) 16, 378–385
&
2009 Macmillan Publishers Limited All rights reserved 1350-9047/09
$
32.00
www.nature.com/cdd
barrel superstructure.
20
Gap junctions provide for inter-
cellular diffusion of variety of substances, including ions,
second messengers and metabolites such as glucose and
ATP.
21–23
This intercellular route underlies long-distance
communications within glial networks; one of the examples
of such communication is represented by glial calcium
waves;
21,24,25
yet propagating waves of other molecules can
also be relevant for intercellular signalling and information
processing.
26
Functional integration between neuronal and glial networks
is accomplished mainly through chemical transmission,
although there are incidental appearances of neuronal–glial
gap junctions.
27,28
Synaptic transmission in neuronal–glial
circuitry occurs either at the level of tripartite synapse (when
perisynaptic astroglial processes are directly exposed to
neurotransmitter released from the neuronal terminals,
29,30
)
or in neuronal–glial synapses.
31,32
The mechanisms of glia to
neurones signalling are more complex; at least in part they
may involve the exocytotic release of gliotransmitters.
13
Astroglia in neurological diseases
Progression and outcome of neurological diseases are
determined by the balance between destruction, neuroprotec-
tion and regeneration. In this context, glial cells are invariably
involved in every kind of neuropathology, which, to a very
large extent, is shaped by glial performance.
6,33
Astrocytes, in
accordance with their homoeostatic function, are deeply
involved in neural diseases. Astroglia forms the first line of
brain defence by controlling the volume and composition of
extracellular space. Astrocytes buffer an excess of extra-
cellular K
þ
, thus maintaining neuronal excitability,
34
control
extracellular levels of glutamate,
35,36
thereby limiting the
intrinsic excitotoxicity of the latter, regulate fluid movements
and provide the main antioxidant system in the brain.
37
At the
very same time astroglial cells can contribute to neuronal
damage, when, for example, severe insults compromise
astrocyte metabolism, which leads to a depolarisation and
reversal of glutamate uptake system, the latter underlying the
release of additional glutamate, thus exacerbating the brain
damage.
6
Acute and chronic brain insults trigger a specific glial
reaction, generally known as reactive astrogliosis, repre-
sented by a complex morphofunctional remodelling of astro-
cytes.
38
Reactive astrogliosis is a defensive brain reaction
which is aimed at (i) isolation of the damaged area from the
rest of the CNS tissue, (ii) reconstruction of the blood–brain
barrier and (iii) facilitation of the remodelling of brain circuits
in areas surrounding the lesioned region.
39,40
As a result,
astrogliosis comes in different guises. Astrocytes surrounding
the lesion undergo a robust hypertrophy and proliferation,
which ultimately ends up in complete substitution of previously
existing tissue architecture with a permanent glial scar;
this process is called anisomorphic (i.e., changing the
morphology) astrogliosis. In astrocytes more distal to the
damage, the reactive changes are much milder and, although
astroglial cells modify their appearance and undergo multiple
biochemical and immunological changes, they do not distort
the normal architecture of CNS tissue, but rather permit
growth of neurites and synaptogenesis, thus facilitating the
remodelling of neuronal networks. This type of astrocyte
reaction is defined as isomorphic (i.e., preserving morphology)
astrogliosis.
Astroglia and dementia
Dementia is the ultimate scourge of mankind, being generally
absent in every other animal species; and as such it may be
considered as the most appalling and horrible disease,
because it effectively robs human beings from their intelli-
gence and turns them into helpless entities. The causes of
dementia are many, ranging from traumatic injuries (mechan-
ical, chemical or as a result of irradiation) and viral infections,
which may predispose to dementia-related chronic neuro-
degenerative conditions, to the intrinsic neurodegenerative
processes associated with genetic factors, predispositions
and other yet unknown reasons. The pathological role of
astroglia in dementiae begun to be explored only very
recently; however, some generalisations can already be
drawn.
First and foremost, both astrogliosis and astroglial dystro-
phy are manifest in different types of dementia; furthermore,
both these processes may develop in parallel depending on
the disease form and/or stage. The frontotemporal dementias
(the clinical term covering several types of sporadic non-
Alzheimer cognitive disruptions, which include e.g., Pick’s
disease and frontotemporal lobar degeneration), is, for
example, associated with a very early and profound apoptotic
death and dystrophy of astrocytes.
41
The degree of astroglial
loss directly correlates with the severity of dementia.
Conversely, other study, using postmortem tissues from 21
frontotemporal dementia cases, found prominent astrogliosis
in the frontal and temporal cortices at the very early stages of
the disease, with astrocyte densities increasing by 4–5 times
of the control.
42
Astrogliosis also assumes the leading pathological role in
thalamic dementia (the rather rare form of specific dementia of
unknown aetiology). Numerous alterations of protoplasmic
astrocytes, manifested by highly localised proliferation of
perivascular and peryneuronal astroglial processes, believed
to be a primary pathological change, which can produce
dementia even without severe neuronal loss.
43
Similarly, in
HIV-1-associated dementia (HAD) significant astroglial hyper-
trophy and increase in GFAP (glial fibrillary acidic protein)
expression is observed in entorhinal cortex and hippocam-
pus.
44
Furthermore, there are indications that HIV-1 infected
reactive astrocytes, together with activated microglial cells,
may constitute a primary cause of neuronal damage, through
the release of various inflammatory and death factors.
45,46
At
the same time, however, HAD also leads to a prominent
apoptotic death of astrocytes especially in the patients with
rapidly progressing cognitive deficits.
47
Astroglia and Alzheimer’s disease
Alzheimer’s disease (AD), existing in both genetic (familial AD
or FAD) and sporadic forms,
48
is characterised by severe
neurodegeneration associated with an occurrence of specific
histopathological markers represented by focal extracellular
deposits of fibrillar b-amyloid (also called neuritic plaques) in
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
379
Cell Death and Differentiation
brain parenchima and in the wall of blood vessels and
intraneuronal accumulation of neurofibrillary tangles com-
posed from abnormal hyperphosphorylated tau filaments. The
progression of AD is rather stereotypic with initial neurode-
generative events appearing in the transentorhinal cortex,
which afterwards spread to the entorhinal cortex and
hippocampus. Subsequently, the spreading wave of neuro-
degeneration swallows the rest of the temporal, frontal and
parietal lobes.
49,50
At the advanced stage of the AD, the grey
matter undergoes severe damage manifested by a profound
loss of neurones and synaptic contacts.
The pathological potential of glial cells in progression of
dementia (as well as in other types of brain pathology) was
originally suggested by Alois Alzheimer himself in 1910.
51
Many studies of the AD-related pathological potential of
astroglia have focused on the investigations of the effects of
b-amyloid on astrocytes. The treatment of cultured glial cells
with aggregated b-amyloid or with amyloid plaques isolated
from human AD brains triggered reactive astrogliosis.
52
In mixed astroglial-neuronal cultures b-amyloid peptide
(Ab 1–42) and its toxic fragment (Ab 25–35) provoked
sporadic increases and/or oscillations in cytosolic calcium
[Ca
2 þ
]
i
, which lasted for many hours.
53
These [Ca
2 þ
]
i
responses were observed solely in astrocytes and never in
neurones and were generated by Ca
2 þ
influx from extra-
cellular space, most likely through transmembrane channels
formed by Ab proteins. Importantly, astroglial [Ca
2 þ
]
i
fluctua-
tions were somehow linked to neuronal death, which occurred
24 h after Ab treatment; inhibition of Ca
2 þ
influx had a clear
neuroprotective effect.
53
In the AD human tissue the main astroglial reaction found
hitherto is represented by prominent astrogliosis, mostly
observed in the cells surrounding amyloid plaques.
54
Im-
portantly, activated astrocytes are capable of accumulating
large amounts of Ab;
55
the later being taken up by astrocytes
in association with neuronal debris. In addition, reactive
astrocytes seem to accumulate large amounts of neuronal
subtype of nicotinic cholinoreceptor (a7nAChRs), which is
known to have an exceptionally high affinity to b-amyloid.
Astroglial b-amyloid deposits are clearly associated with
plaques, as astrocytes positioned away from the plaques
show no b-amyloid burden.
54,55
Processes of activated
astrocytes were also reported to participate in plaques
formation.
54,55
At the very same time astroglial changes during AD
progression remain virtually unexplored. The main reason
for this is purely experimental as for many years the suitable
animal models for AD were absent.
Animal models of AD
Alzheimer’s disease, as every other dementia, is a sole
prerogative of humans; no animal suffers from AD.
56
Hence,
substantial efforts were invested in producing relevant animal
models of AD (for a comprehensive review of literature, see
references
57,58
). Initial models of AD were simply normal aged
animals,
59,60
which showed cholinergic involution associated
(in monkeys) with b-amyloid deposition.
61
As the AD is
manifested by a loss of cholinergic neurones,
62
several
cholinergic models of the disease were created. Among
these, the most relevant were the rodent models with lesions
in the nucleus basalis magnocellularis (nbm) that is the
equivalent of the nucleus of Meynert in humans.
56,63,64
These
models offered the possibility to investigate the differences in
structure, function and behaviour of the cholinergic systems in
young and aged animals.
63,64
The majority of these models,
however, were created by using non-selective excitotoxic
toxins such as NMDA, ibotenic acid, quisqualic acid and
certain alkaloid substances.
56
More refined lesion models
used more selective and specific toxins that affected only
cholinergic neurones in the relevant areas, such as septum,
nbm and the diagonal band of Broca, but preserved
non-cholinergic neurones.
56,65
These models were produced
by using AF64A cholinotoxin, which binds to the high affinity
choline uptake system, and 192 IgG-saporin that binds
selectively and irreversibly to low affinity nerve growth factor
receptor interrupting cholinergic neuronal protein synthesis.
Both these interventions lead to selective impairment and
death of cholinergic neurones.
None of the models mentioned above, however, mimicked
the histopathology (plaques and tangles) and progression of
AD. Therefore several experimental transgenic animals that
replicate various neuropathological features of AD have been
developed (see Table 1 and reference
58
). Initially, simple
transgenic models harbouring single-mutated b-amyloid-
related proteins (amyloid precursor protein (APP) or
presenilines, PS) or mutated tau were created. The very first
APP transgenic animal showing an AD-like pathology was
developed in 1995 by Games et al.
66
This model was called
PDAPP (single-transgenic mice expressing the ‘Swedish’ and
‘Indiana’ APP mutations Asp
664
to Ala (PDAPP(D664A)) and
showed many pathological features of AD, including extra-
cellular b-amyloid deposits, neuritic dystrophy, astrogliosis
and memory impairments, yet the latter did not correlate with
the degree of b-amyloid deposition.
58,66
Subsequently, Hsiao
et al.
67
developed the transgenic mice carrying APP
swe
mutation (Tg2576 mice) using a prion protein vector. These
animals had abundant Ab plaques as well as a memory and
learning impairment from 9 months of age. Subsequently, the
double-APP mutation, resulting the APP23 mouse was
created. This model showed a 14% loss of hippocampal
CA1 pyramidal neurons.
Mutations in PS proteins account for the majority of cases of
FAD.
58
To address the role of genetic risk factors in APP
Table 1 Summary of the transgenic AD animal models and the observed
neuropathology
Transgenic
mouse model Neuropathology Reference
APP
SWE
Plaques
68
PDAPP Plaques
69
APP
751SL
Plaques
70
PS1
M146L
Plaques
70
APP
SWE
/PS1
dE9
Plaques
71
APP/apoE-ko Plaques
72
TgCRND8 bAPP6
95
Plaques
73
APP
751SL
/PS1
M146L
Plaques
70
THY-Tau22 Tangles
74
Tau
P301L
Tangles
68
3xTg-AD Plaques and tangles
75
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
380
Cell Death and Differentiation
processing and memory, double transgenic have been
created. Co-expression of PS1
dE9
with APP resulted in a
viable model that showed accelerated Ab deposition and
memory deficits without tangle formation.
71
Expression of
mutant APP on an ApoE knockout background did not affect
the age of onset of Ab plaques but instead of fibrillar Ab
mature plaques, only diffuse plaques were formed, suggest-
ing that ApoE affects fibrillogenesis and/or clearance of Ab.
76
The pathological tau animals were developed in parallel, the
first model being created in 1995;
77
these transgenic animals
showed somatodendritic localisation of hyperphosphorlation
of tau, but have not developed neurofibrillary tangle pathology
(NFT). After identification of the pathogenic mutations of tau in
FTDP-17, different transgenic models with clear neuronal
NFT were produced.
57,58
The Tau
P301L
(single-transgenic
mice expressing the Pro
301
to Leu tau mutation (P301L))
mutation is the most common associated mutation linked with
FTDP-17.
78
Transgenic mice overexpressing Tau
P301L
exhibit
neurofibrillary tangles without Ab pathology and/or neuronal
loss, except for the spinal cord (Table 1;
58,78
).
The triple-transgenic (3xTg-AD) AD animal model. One
of the most advanced AD animal models is represented by
the triple-transgenic mice developed in 2003 by Salvatore
Oddo and Frank LaFerla. These animals (the 3xTg-AD
mouse model) harbour the mutant genes for amyloid
precursor protein (APP
Swe
), for presenilin 1 PS1
M146V
and
for tau
P301L
.
75,79
These mice are recognised as relevant AD
model because they show temporal- and region-specific Ab
and tau pathology, which closely resembles that seen in the
human AD brain. As well as progressively developing
plaques and tangles, the 3xTg-AD animals also show clear
functional and cognitive impairments including LTP, spatial
memory and long-term memory deficits. These all are
Figure 1 Bar graphs showing the complexity (a) of glial cytoskeleton by measuring the GFAP area coverage versus volume ratio of within the molecular layer of the dorsal
dentate gyrus of both control and 3xTg-AD mice at different ages. (b and c) Bar graphs showing the coverage area (b) and volume (c) of GFAP at 12 months of age in control
and 3xTg-AD mice; *Po0.05 compared with age matching controls. (d and e) Fluorescence photomicrographs showing GFAP-positive cells in control (d) and 3xTg-AD mice
(e) at 12 months of age. There is an evident decrease in arborisation, surface area and volume in 3xTg-AD mice when compared with matching controls. All animal procedures
were performed according to the Animal Scientific Procedures Act of 1986 under the licence from the United Kingdom Home Office. The animals group were housed and kept
on daily 12 h light-dark cycles dark schedule. All mice were given ad libitum access to food and water
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
381
Cell Death and Differentiation
manifest in an age-related manner; most importantly
functional deficits precede the appearance of histological
markers.
75,79
Cognitive deficits in the 3xTg-AD model
correlate with the accumulation of intraneuronal Ab.
80,81
It has to be noted, however, that all animal models of AD
developed so far have some limitations, the most serious one
being the absence of significant neuronal loss. This may
reflect some intrinsic differences between human and animal
brain, shorter lifespan of experimental animals or influence of
other yet unknown factors. At the same time absence of
neuronal loss in animals undergoing severe amyloidosis may
also question the toxic potential of neuritic plaques.
Glial atrophy and astrogliosis in transgenic AD
animals. We characterised morphology of astroglia in the
hippocampus of male 3xTG-AD mice using single and dual
immunohistochemical labelling.
82
For this labelling, we used
the antibodies to GFAP an astroglia cytoskeleton-specific
protein which expression varies between different brain
regions and is upregulated upon reactive astrogliosis, or
combined GFAP immunostaining with antibodies against
b-amyloid. Morphology of GFAP-positive astrolglial profiles
was reconstructed from series of optical confocal planes
taken from 50 mm thick fixed brain slices. After the
reconstruction of individual astrocytes the following
parameters were determined: surface area, volume and
cellular complexity (area versus volume; for the detailed
description of imaging/analysis techniques, see
references
83,84
).
Morphological characterisation of GFAP-positive astroglial
cells was performed on animals of different ages, from 3 to 12
months. To our surprise we found consistent reduction in the
GFAP expression (determined as surface and volume of
GFAP-positive profiles), which progressed with age. In very
young animals (3 months old), which did not have any signs of
AD pathology, astroglial morphology was already somewhat
altered. There was a slight reduction in astrocyte complexity
(by 1.29%; Figure 1a) that was directly associated with a
decrease in the number of processes and overall decrease in
both surface (12.62%) and volume (14.25%; data not shown)
of GFAP-labelled structures. Similar reduction in GFAP-
labelled profiles was observed at all ages, although this
decrease becomes significant only at 12 months of age. In
12-month-old animals the complexity was reduced by 5.04%
(P ¼ 0.0105; Figure 1a, d and e), whereas GFAP-positive
surface decreased by 38.99% and volume by 42.93%
(P ¼ 0.0211, 0.0292, respectively; Figure 1). On a cellular
level, this decrease in complexity was reflected by reduced
number of main processes and their arborisation (Figure 1c).
Incidentally, the same decrease in astrocytes complexity
was found in the postmortem brains of dementia patients. In
this study,
85
astroglial cells from neocortical area 11 were
Golgi impregnated and their complexity was characterised by
a fractal dimension. Astrocytes from adult and healthy aged
participants had similar complexity, whereas astrocytes from
patients with dementia showed significant decrease in fractal
dimension, thus indicating the decreased complexity. Further-
more, the volume of the brain parenchima occupied by the
processes of single astrocyte was smaller in demented brains.
At the very same time confocal images revealed that the
specific population of astrocytes surrounding amyloid plaques
display the typical reactive characteristics (Figure 2), showing
thick processes and enlarged cell bodies. Some of these
astrocytes closely associated with amyloid plaques showed
b-amyloid accumulation, as revealed by specific antibodies
(Figure 2b). Conversely, astrocytes positioned away from the
plaques did not show b-amyloid burden, which is consistent
with previous observations.
54,55
The 3xTg-AD animals show other changes that could be
associated with glial malfunction. For example, we observed a
transient increase in the number of asymmetric excitatory
synapses in AD mice at 2 months of age, which subsequently
dropped to control levels at 6–9 months. Nonetheless, this
increase in excitatory synaptic contacts re-emerged again at
12 months of age when the brain parenchyma was infested
with plaques, and neurones showed tangles.
86
In addition, we
Figure 2 Confocal image showing GFAP-positive (green) reactive astrocytes
surrounding b-amyloid plaques (red; a). (b) Reactive astrocytes (green) and an
astrocyte showing cytoplasmic b-amyloid accumulation (indicated by arrows;
colocalisation, yellow) near a neuritic plaque (red)
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
382
Cell Death and Differentiation
also found significant decrease in the neurogenic capacity
of 3x-Tg-AD mice at 12 months of age,
87
which may reflect
degenerative changes in ‘stem-cell’-like astrocytes.
Astroglial theory of AD?
The pivotal role of astroglia in various types of brain pathology
is widely accepted.
6,88,89
Nevertheless, the pathological
potential of astrocytes in human dementias remains virtually
unexplored. From the scarce data available in the literature,
we may conclude that astrocytic reactions in various types of
dementia can be manifold, ranging from proliferation and
astrogliosis to apoptosis and dystrophy.
In AD, these two reactions develop in parallel and may
represent the underlying mechanism for loss in synaptic
connectivity and plasticity, which in turn determine cognitive
deficits. The reaction of astrocytes to the AD progression is
spatially distinct (Figure 3): astroglial cells surrounding the
plaques undergo gliosis, whereas astrocytes distant from the
amyloid deposits develop atrophy. The first signs of this
atrophy appear early in the genetic form of AD pathology in
mice; and these signs precede the formation of both plaques
and tangles. It is then conceivable to speculate that dystrophic
astrocytes fail to provide adequate support for synaptic
contacts, which may lead to their remodelling (e.g., distorted
balance between excitatory and inhibitory synapses), with
ensuing disruption of neuronal circuitry and indeed the
synaptic loss is observed in early stages of AD when brain
parenchyma remains relatively free from neuritic plaques.
90,91
Furthermore, reduced astroglial coverage may significantly
affect extracellular brain homoeostasis. In particular, dys-
trophic astrocytes may have reduced ability for glutamate
uptake, thus increasing the overall brain vulnerability to
glutamate excitotoxicity. In contrast, the reactive astroglial
cells may serve another function the function of neuronal
killers. Indeed, the main pillar of b-amyloid hypothesis of AD,
the toxicity of the neuritic plaques has been considerably
shaken by experiments on transgenic animals. The latter,
despite acquiring a heavy burden of amyloid plaques, show
very little (if any) neuronal death.
58
This discrepancy is
fundamental, as massive neuronal death assumes the main
role in the development of severe dementia in humans.
Naturally, the absence of neuronal extinction in the animal
model can be explained by a variety of reasons, yet it may also
hint that alternative mechanisms of the neurodegenerative
process are in operation. These mechanisms could be
Figure 3 Astroglial hypothesis of Alzheimer’s disease
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
383
Cell Death and Differentiation
associated with both astroglial atrophy (reduced glutamate
uptake with increased glutamate excitotoxicity) and astro-
gliosis, as reactive astrocytes and activated microglia may
secrete numerous death-promoting molecules.
The AD, as well as other types of dementia, are complex
processes which engulf all cellular elements in the brain. Can
astrocytes assume the central stage in these pathologies?
This question requires further detailed and insightful
investigation.
Acknowledgements
. We thank the National Institute of Health, INTAS,
Alzheimer Research Trust UK and European commission for the grant support. AC
research was supported by the Grant Agency of the Czech Republic (nos. 305/06/
1316, 305/06/1464, 305/08/1384 and 309/08/1381), by the Ministry of Education,
Youth and Sports of the Czech Republic (nos. 1M0538 and LC554) and by the
Academy of Sciences of the Czech Republic (no. AVOZ50390512).
1. Deiters O. Untersuchungen u
¨ber
Gehirn und Ru
¨ckenmark
des Menschen und der
Sa
¨
ugethiere. Braunschweig: Vieweg, 1865.
2. Lenhossek Mv. Der feinere Bau des Nervensystems im Lichte neuester Forschung.
Fischer’s Medicinische Buchhandlung H Kornfield: Berlin, 1893.
3. Kettenmann H, Ransom BR. The concept of neuroglia: a historical perspective. In:
Kettenmann H, Ransom BR (eds). Neuroglia. OUP: Oxford, 2005, pp 1–16.
4. Verkhratsky A. Patching the glia reveals the functional organisation of the brain. Pflugers
Arch 2006; 453: 411–420.
5. Verkhratsky A, Butt A. Glial Neurobiology A text book. John Wiley & Sons: Chichester,
2007.
6. Giaume C, Kirchhoff F, Matute C, Reichenbach A, Verkhratsky A. Glia: the fulcrum of brain
diseases. Cell Death Differ 2007; 14: 1324–1335.
7. Porter JT, McCarthy KD. Astrocytic neurotransmitter receptors in situ and in vivo. Prog
Neurobiol 1997; 51: 439–455.
8. Verkhratsky A, Steinhauser C. Ion channels in glial cells. Brain Res Brain Res Rev 2000;
32: 380–412.
9. Verkhratsky A, Kirchhoff F. Glutamate-mediated neuronal-glial transmission. J Anat 2007;
210: 651–660.
10. Verkhratsky A. Calcium ions and integration in neural circuits. Acta Physiol (Oxf) 2006; 187:
357–369.
11. Dermietzel R, Gao Y, Scemes E, Vieira D, Urban M, Kremer M et al. Connexin43
null mice reveal that astrocytes express multiple connexins. Brain Res Brain Res Rev 2000;
32: 45–56.
12. Giaume C, McCarthy KD. Control of gap-junctional communication in astrocytic networks.
Trends Neurosci 1996; 19: 319–325.
13. Volterra A, Meldolesi J. Astrocytes, from brain glue to communication elements: the
revolution continues. Nat Rev Neurosci 2005; 6: 626–640.
14. Nedergaard M, Ransom B, Goldman SA. New roles for astrocytes: redefining the functional
architecture of the brain. Trends Neurosci 2003; 26: 523–530.
15. Bushong EA, Martone ME, Jones YZ, Ellisman MH. Protoplasmic astrocytes in CA1
stratum radiatum occupy separate anatomical domains. J Neurosci 2002; 22: 183–192.
16. Oberheim NA, Wang X, Goldman S, Nedergaard M. Astrocytic complexity distinguishes the
human brain. Trends Neurosci 2006; 29: 547–553.
17. Mulligan SJ, MacVicar BA. Calcium transients in astrocyte endfeet cause cerebrovascular
constrictions. Nature 2004; 431: 195–199.
18. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan T et al. Neuron-to-
astrocyte signaling is central to the dynamic control of brain microcirculation. Nat Neurosci
2003; 6: 43–50.
19. Iadecola C, Nedergaard M. Glial regulation of the cerebral microvasculature. Nat Neurosci
2007; 10: 1369–1376.
20. Houades V, Koulakoff A, Ezan P, Seif I, Giaume C. Gap junction-mediated astrocytic
networks in the mouse barrel cortex. J Neurosci 2008; 28: 5207–5217.
21. Scemes E, Giaume C. Astrocyte calcium waves: what they are and what they do. Glia
2006; 54: 716–725.
22. Kang J, Kang N, Lovatt D, Torres A, Zhao Z, Lin J et al. Connexin 43 hemichannels are
permeable to ATP. J Neurosci 2008; 28: 4702–4711.
23. Tabernero A, Medina JM, Giaume C. Glucose metabolism and proliferation in glia: role of
astrocytic gap junctions. J Neurochem 2006; 99: 1049–1061.
24. Cornell-Bell AH, Finkbeiner SM, Cooper MS, Smith SJ. Glutamate induces calcium waves
in cultured astrocytes: long-range glial signaling. Science 1990; 247: 470–473.
25. Verkhratsky A, Orkand RK, Kettenmann H. Glial calcium: homeostasis and signaling
function. Physiol Rev 1998; 78: 99–141.
26. Verkhratsky A, Toescu EC. Neuronal-glial networks as substrate for CNS integration. J Cell
Mol Med 2006; 10: 826–836.
27. Nedergaard M. Direct signaling from astrocytes to neurons in cultures of mammalian brain
cells. Science 1994; 263: 1768–1771.
28. Pakhotin P, Verkhratsky A. Electrical synapses between Bergmann glial cells and Purkinje
neurones in rat cerebellar slices. Mol Cell Neurosci 2005; 28: 79–84.
29. Halassa MM, Fellin T, Haydon PG. The tripartite synapse: roles for gliotransmission in
health and disease. Trends Mol Med 2007; 13: 54–63.
30. Araque A, Parpura V, Sanzgiri RP, Haydon PG. Tripartite synapses: glia, the
unacknowledged partner. Trends Neurosci 1999; 22: 208–215.
31. Lin SC, Bergles DE. Synaptic signaling between neurons and glia. Glia 2004; 47: 290–298.
32. Jabs R, Pivneva T, Huttmann K, Wyczynski A, Nolte C, Kettenmann H et al. Synaptic
transmission onto hippocampal glial cells with hGFAP promoter activity. J Cell Sci 2005;
118: 3791–3803.
33. Panickar KS, Norenberg MD. Astrocytes in cerebral ischemic injury: morphological and
general considerations. Glia 2005; 50: 287–298.
34. Kofuji P, Newman EA. Potassium buffering in the central nervous system. Neuroscience
2004; 129: 1045–1056.
35. Danbolt NC. Glutamate uptake. Progr Neurobiol 2001; 65: 1–105.
36. Kirischuk S, Kettenmann H, Verkhratsky A. Membrane currents and cytoplasmic sodium
transients generated by glutamate transport in Bergmann glial cells. Pflugers Arch 2007;
454: 245–252.
37. Dringen R. Metabolism and functions of glutathione in brain. Prog Neurobiol 2000; 62:
649–671.
38. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia 2005; 50: 427–434.
39. Butt AM, Hamilton N, Hubbard P, Pugh M, Ibrahim M. Synantocytes: the fifth element.
J Anat 2005; 207: 695–706.
40. Pekny M, Wilhelmsson U, Bogestal YR, Pekna M. The role of astrocytes and complement
system in neural plasticity. Int Rev Neurobiol 2007; 82: 95–111.
41. Broe M, Kril J, Halliday GM. Astrocytic degeneration relates to the severity of disease in
frontotemporal dementia. Brain 2004; 127: 2214–2220.
42. Kersaitis C, Halliday GM, Kril JJ. Regional and cellular pathology in frontotemporal
dementia: relationship to stage of disease in cases with and without Pick bodies. Acta
Neuropathol 2004; 108: 515–523.
43. Potts R, Leech RW. Thalamic dementia: an example of primary astroglial dystrophy of
Seitelberger. Clin Neuropathol 2005; 24: 271–275.
44. Vanzani MC, Iacono RF, Caccuri RL, Troncoso AR, Berria MI. Regional
differences in astrocyte activation in HIV-associated dementia. Medicina (B Aires) 2006;
66: 108–112.
45. Deshpande M, Zheng J, Borgmann K, Persidsky R, Wu L, Schellpeper C et al. Role of
activated astrocytes in neuronal damage: potential links to HIV-1-associated dementia.
Neurotox Res 2005; 7: 183–192.
46. Nardacci R, Antinori A, Kroemer G, Piacentini M. Cell death mechanisms in HIV-associated
dementia: the involvement of syncytia. Cell Death Differ 2005; 12 (Suppl 1): 855–858.
47. Thompson KA, McArthur JC, Wesselingh SL. Correlation between neurological
progression and astrocyte apoptosis in HIV-associated dementia. Ann Neurol 2001; 49:
745–752.
48. Alzheimer A. ber eine eigenartige Erkrankung der Hirnrinde. Allg Z Psychiat Psych-Gericht
Med 1907; 64: 146–148.
49. Thompson PM, Hayashi KM, Dutton RA, Chiang MC, Leow AD, Sowell ER et al. Tracking
Alzheimer’s disease. Ann N Y Acad Sci 2007; 1097: 183–214.
50. Thompson PM, Hayashi KM, de Zubicaray G, Janke AL, Rose SE, Semple J et al.
Dynamics of gray matter loss in Alzheimer’s disease. J Neurosci 2003; 23: 994–1005.
51. Alzheimer A. Beitra
¨
ge zur Kenntnis der pathologischen Neuroglia und ihrer Beziehungen
zu den Abbauvorga
¨
ngen im Nervengewebe. In: Nissl F, Alzheimer A (eds). Histologische
und histopathologische Arbeiten u
¨
ber die Grosshirnrinde mit besonderer Beru
¨
cksichtigung
der pathologischen Anatomie der Geisteskrankheiten. Gustav Fischer: Jena, 1910,
pp 401–562.
52. DeWitt DA, Perry G, Cohen M, Doller C, Silver J. Astrocytes regulate microglial
phagocytosis of senile plaque cores of Alzheimer’s disease. Exp Neurol 1998; 149:
329–340.
53. Abramov AY, Canevari L, Duchen MR. Changes in intracellular calcium and glutathione
in astrocytes as the primary mechanism of amyloid neurotoxicity. J Neurosci 2003; 23:
5088–5095.
54. Nagele RG, Wegiel J, Venkataraman V, Imaki H, Wang KC. Contribution of glial cells
to the development of amyloid plaques in Alzheimer’s disease. Neurobiol Aging 2004; 25:
663–674.
55. Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang HY. Astrocytes accumulate A
beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res
2003; 971: 197–209.
56. Toledano A, Alvarez MI. Lesions and dysfunctions of the nucleus basalis as Alzheimer’s
disease models: general and critical overview and analysis of the long-term changes in
several excitotoxic models. Curr Alzheimer Res 2004; 189: 214.
57. Cassel JC, Mathis C, Majchrzak M, Moreau PH, Dalrymple-Alford JC. Coexisting
cholinergic and parahippocampal degeneration: a key to memory loss in dementia and a
challenge for transgenic models? Neurodegener Dis 2008; 5: 304–317.
58. Gotz J, Streffer JR, David D, Schild A, Hoerndli F, Pennanen L et al. Transgenic animal
models of Alzheimer’s disease and related disorders: histopathology, behavior and
therapy. Mol Psychiatry 2004; 9: 664–683.
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
384
Cell Death and Differentiation
59. Biegon A, Greenberger V, Segal M. Quantitative histochemistry of brain
acetylcholinesterase and learning rate in the aged rat. Neurobiol Aging 1986; 7: 215–217.
60. Bertoni-Freddari C, Giuli C, Pieri C, Paci D. Quantitative investigation of the morphological
plasticity of synaptic junctions in rat dentate gyrus during aging. Brain Res 1986; 366:
187–192.
61. Sani S, Traul D, Klink A, Niaraki N, Gonzalo-Ruiz A, Wu CK et al. Distribution, progression
and chemical composition of cortical amyloid-beta deposits in aged rhesus monkeys:
similarities to the human. Acta Neuropathol 2003; 105: 145–156.
62. Bartus RT, Dean III RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory
dysfunction. Science 1982; 217: 408–414.
63. Pepeu G, Marconcini Pepeu I. Dysfunction of the brain cholinergic system during aging and
after lesions of the nucleus basalis of Meynert. J Neural Transm Suppl 1994; 44: 189–194.
64. Wellman CL, Pelleymounter MA. Differential effects of nucleus basalis lesions in young
adult and aging rats. Neurobiol Aging 1999; 20: 381–393.
65. Wiley RG. Neural lesioning with ribosome-inactivating proteins: suicide transport and
immunolesioning. Trends Neurosci 1992; 15: 285–290.
66. Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C et al.
Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid
precursor protein. Nature 1995; 373: 523–527.
67. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S et al. Correlative
memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996;
274: 99–102.
68. Arendash GW, Lewis J, Leighty RE, McGowan E, Cracchiolo JR, Hutton M et al. Multi-
metric behavioral comparison of APPsw and P301L models for Alzheimer’s disease:
linkage of poorer cognitive performance to tau pathology in forebrain. Brain Res 2004;
1012: 29–41.
69. Galvan V, Zhang J, Gorostiza OF, Banwait S, Huang W, Ataie M et al. Long-term
prevention of Alzheimer’s disease-like behavioral deficits in PDAPP mice carrying a
mutation in Asp664. Behav Brain Res 2008; 191: 246–255.
70. Gruart A, Lopez-Ramos JC, Munoz MD, Delgado-Garcia JM. Aged wild-type and APP,
PS1, and APP+PS1 mice present similar deficits in associative learning and synaptic
plasticity independent of amyloid load. Neurobiol Dis 2008; 30: 439–450.
71. Savonenko A, Xu GM, Melnikova T, Morton JL, Gonzales V, Wong MP et al. Episodic-like
memory deficits in the APPswe/PS1dE9 mouse model of Alzheimer’s disease:
relationships to beta-amyloid deposition and neurotransmitter abnormalities. Neurobiol
Dis 2005; 18: 602–617.
72. Dodart JC, Mathis C, Saura J, Bales KR, Paul SM, Ungerer A. Neuroanatomical
abnormalities in behaviorally characterized APP(V717F) transgenic mice. Neurobiol Dis
2000; 7: 71–85.
73. Janus C, Pearson J, McLaurin J, Mathews PM, Jiang Y, Schmidt SD et al. A beta peptide
immunization reduces behavioural impairment and plaques in a model of Alzheimer’s
disease. Nature 2000; 408: 979–982.
74. Schindowski K, Bretteville A, Leroy K, Begard S, Brion JP, Hamdane M et al. Alzheimer’s
disease-like tau neuropathology leads to memory deficits and loss of functional synapses in
a novel mutated tau transgenic mouse without any motor deficits. Am J Pathol 2006; 169:
599–616.
75. Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R et al.
Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular
Abeta and synaptic dysfunction. Neuron 2003; 39: 409–421.
76. Brendza RP, Bales KR, Paul SM, Holtzman DM. Role of apoE/Abeta interactions
in Alzheimer’s disease: insights from transgenic mouse models. Mol Psychiatry 2002; 7:
132–135.
77. Gotz J, Probst A, Spillantini MG, Schafer T, Jakes R, Burki K et al. Somatodendritic
localization and hyperphosphorylation of tau protein in transgenic mice expressing the
longest human brain tau isoform. EMBO J 1995; 14: 1304–1313.
78. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M et al.
Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing
mutant (P301L) tau protein. Nat Genet 2000; 25: 402–405.
79. Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM. Amyloid deposition precedes
tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol Aging 2003;
24: 1063–1070.
80. McKee AC, Carreras I, Hossain L, Ryu H, Klein WL, Oddo S et al. Ibuprofen reduces Abeta,
hyperphosphorylated tau and memory deficits in Alzheimer mice. Brain Res 2008; 1207:
225–236.
81. Carroll JC, Rosario ER, Chang L, Stanczyk FZ, Oddo S, LaFerla FM et al. Progesterone
and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice. J Neurosci
2007; 27: 13357–13365.
82. Rodriguez JJ, Mackie K, Pickel VM. Ultrastructural localization of the CB1 cannabinoid
receptor in mu-opioid receptor patches of the rat Caudate putamen nucleus. J Neurosci
2001; 21: 823–833.
83. Chvatal A, Anderova M, Kirchhoff F. Three-dimensional confocal morphometry a new
approach for studying dynamic changes in cell morphology in brain slices. JAnat2007;
210: 671–683.
84. Chvatal A, Anderova M, Hock M, Prajerova I, Neprasova H, Chvatal V et al. Three-
dimensional confocal morphometry reveals structural changes in astrocyte morphology
in situ. J Neurosci Res 2007; 85: 260–271.
85. Senitz D, Reichenbach A, Smith Jr TG. Surface complexity of human neocortical
astrocytic cells: changes with development, aging, and dementia. J Hirnforsch 1995; 36:
531–537.
86. Le
´
veille
´
F, Ortega JB, Rodrı
´
guez JJ, Meadows RS, Verkhratsky A, Nicole O et al. Increase
in excitatory synapses density and functional modification of NMDA receptors-mediated
responses induced by b-amyloid oligomers. Unpublished observations.
87. Rodrı
´
guez JJ, Jones VC, Tabuchi M, Allan SM, Knight EM, LaFerla FM et al. Impaired adult
neurogenesis in the dentate gyrus of a triple transgenic mouse model of Alzheimer’s
disease. PLOS One 2008; 3: e2935.
88. Tian GF, Azmi H, Takano T, Xu Q, Peng W, Lin J et al. An astrocytic basis of epilepsy.
Nat Med 2005; 11: 973–981.
89. Nedergaard M, Dirnagl U. Role of glial cells in cerebral ischemia. Glia 2005; 50: 281–286.
90. Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ. Synaptic alterations in CA1 in
mild Alzheimer disease and mild cognitive impairment. Neurology 2007; 68: 1501–1508.
91. Terry RD. Cell death or synaptic loss in Alzheimer disease. J Neuropathol Exp Neurol
2000; 59: 1118–1119.
Astroglia in Alzheimer disease
JJ Rodrı
´
guez et al
385
Cell Death and Differentiation
... Enlarged astrocytes in AD have been observed to surround and engulf Aβ plaques, the characteristic pathological hallmark of the disease. By phagocytosing Aβ, these astrocytes may help in reducing the burden of toxic protein aggregates in the brain, thereby exerting a neuroprotective effect [51,52]. Moreover, enlarged astrocytes may exert a neuroprotective role by modulating neuroinflammation. ...
Article
Full-text available
Despite decades of rigorous research and numerous clinical trials, Alzheimer’s disease (AD) stands as a notable healthcare challenge of this century, with effective therapeutic solutions remaining elusive. Recently, the endocannabinoid system (ECS) has emerged as an essential therapeutic target due to its regulatory role in different physiological processes, such as neuroprotection, modulation of inflammation, and synaptic plasticity. This aligns with previous research showing that cannabinoid receptor ligands have the potential to trigger the functional structure of neuronal and brain networks, potentially impacting memory processing. Therefore, our study aims to assess the effects of prolonged, intermittent exposure (over 90 days) to JWH-133 (0.2 mg/kg) and an EU-GMP certified Cannabis sativa L. (Cannabixir® Medium Flos, 2.5 mg/kg) on recognition memory, as well as their influence on brain metabolism and modulation of the expanded endocannabinoid system in APP/PS1 mice. Chronic therapy with cannabinoid receptor ligands resulted in reduced anxiety-like behavior and partially reversed the cognitive deficits. Additionally, a reduction was observed in both the number and size of Aβ plaque deposits, along with decreased cerebral glucose metabolism, as well as a decline in the expression of mTOR and CB2 receptors. Furthermore, the study revealed enlarged astrocytes and enhanced expression of M1 mAChR in mice subjected to cannabinoid treatment. Our findings highlight the pivotal involvement of the extended endocannabinoid system in cognitive decline and pathological aspects associated with AD, presenting essential preclinical evidence to support the continued exploration and assessment of cannabinoid receptor ligands for AD treatment.
... Within astrocytes, glucose is metabolized glycolytically to lactate, which is then exported as the neuron's primary source of energy; this process is stimulated by glutamate. Astrocytes have a key role in maintaining the functionality of synapses and neuronal circuits, and store glucose in the form of glycogen (Rodriguez et al., 2009). Astrocyte-to-neuron signaling in intact tissue contributes to synaptic plasticity (Fiacco et al., 2004). ...
Article
Full-text available
Neurons have a high energy need, requiring a continuous supply of glucose from the blood. Tight regulation of glucose metabolism in response to stimuli is essential for brain physiology. Glucose metabolism and cerebral blood flow are closely coordinated during neuronal activity to maintain proper brain function. Glucose uptake across the blood-brain barrier is facilitated by a carrier protein: the GLUT-1 transporter. The first way the body meets urgent demand for glucose is to increase the blood flow through vasodilatory responses generated by nitric oxide. If that is insufficient, the second way is to increase the density of GLUT-1 through the translocation of this transporter from intracellular stores. The third pathway is to increase GLUT-1 synthesis by stimulating SLC2A1 (GLUT-1 gene) transcription. A tandem of two key molecules, free estradiol and DHA, is involved in this critical regulation. Their relationship is synergistic and reciprocal: free estradiol with genomic and non-genomic actions via ERα, and DHA via the PPARα-RXRα and PPARɣ-RXRα heterodimers. We highlight several original mechanisms linking two main principles (neuronal stimulation and brain energy metabolism) with the fundamental roles played by DHA and free estradiol. In particular, it has been shown that from a certain level of chronic DHA deficiency, a permanent imbalance sets in with disturbances in glucose intake and brain metabolism. This DHA deficiency is an aggravating factor in some neuropathologies.
... (i) Astrogliosis and astroglial degeneration in AD Our knowledge about modifications of neuroglia and their roles in the progression of AD remains, at best, fragmentary (for review of modern theories and references, see Heneka et al., 2010;Nagele et al., 2004;Rodriguez et al., 2009;. It is generally accepted that the main glial reaction to AD is profound astrogliosis and, indeed, many studies of post-mortem tissues from AD patients found generalised astrogliosis, manifested by cellular hypertrophy and an increase in expression of GFAP and S100b. ...
... It is also observed that microglia and astrocytes are increased in areas near the Aβ plaques in AD [67]. The reactive astrocytes around the Aβ plaques induce inflammatory reactions and alter Ca 2+ signaling [68,69]. A loss of astrocytic function and reactivity contribute to AD [70]. ...
Article
Full-text available
Oxidative stress with a depletion of glutathione is a key factor in the initiation and progression of Alzheimer's disease (AD). N-Acetylcysteine (NAC), a glutathione precursor, provides neuroprotective effects in AD animal models. Its amide form, N-Acetylcysteine amide (NACA), has an extended bioavailability compared to NAC. This study evaluates the neuroprotective effects of NACA against Aβ1-42 peptide-induced AD-like pathology in rats. Male Wistar rats (2.5 months old) were divided into five groups: Normal Control (NC), Sham (SH), Aβ, Aβ + NACA and NACA + Aβ + NACA (n = 8 in all groups). AD-like pathology was induced by the intracerebroven-tricular infusion of Aβ1-42 peptide into the lateral ventricle. NACA (75 mg/kg) was administered either as a restorative (i.e., injection of NACA for 7 consecutive days after inducing AD-like pathology (Aβ + N group)), or as prophylactic (for 7 days before and 7 days after inducing the pathology (N + Aβ + N group)). Learning and memory, neurogenesis, expression of AD pathology markers, antioxidant parameters, neuroprotection, astrogliosis and microgliosis were studied in the hippocam-pus and the prefrontal cortex. All data were analyzed with a one-way ANOVA test followed by Bonferroni's multiple comparison test. NACA treatment reversed the cognitive deficits and reduced oxidative stress in the hippocampus and prefrontal cortex. Western blot analysis for Tau, Synapto-physin and Aβ, as well as a histopathological evaluation through immunostaining for neurogenesis, the expression of neurofibrillary tangles, β-amyloid peptide, synaptophysin, neuronal morphology and gliosis, showed a neuroprotective effect of NACA. In conclusion, this study demonstrates the neuroprotective effects of NACA against β-amyloid induced AD-like pathology.
... Previous studies have demonstrated that aberrant activity and dysfunction of glial cells in AD are associated with cognitive decline. Pathological examinations of AD brains reveal an abundance of activated microglia and astrocytes surrounding senile plaques [30,31]. Enhanced microglial phagocytosis and Aβ clearance mediated by microglia have been shown to alleviate cognitive impairment in AD mouse models [32]. ...
Article
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that impacts a substantial number of individuals globally. Despite its widespread prevalence, there is currently no cure for AD. It is widely acknowledged that normal synaptic function holds a key role in memory, cognitive abilities, and the interneuronal transfer of information. As AD advances, symptoms including synaptic impairment, decreased synaptic density, and cognitive decline become increasingly noticeable. The importance of glial cells in the formation of synapses, the growth of neurons, brain maturation, and safeguarding the microenvironment of the central nervous system is well recognized. However, during AD progression, overactive glial cells can cause synaptic dysfunction, neuronal death, and abnormal neuroinflammation. Both neuroinflammation and synaptic dysfunction are present in the early stages of AD. Therefore, focusing on the changes in glia-synapse communication could provide insights into the mechanisms behind AD. In this review, we aim to provide a summary of the role of various glial cells, including microglia, astrocytes, oligodendrocytes, and oligodendrocyte precursor cells, in regulating synaptic dysfunction. This may offer a new perspective on investigating the underlying mechanisms of AD.
Preprint
Full-text available
Neuroinflammation is a central process in the pathogenesis of several neurodegenerative diseases such as Alzheimer’s disease (AD), and there are active efforts to target pathways involved in neuroinflammation for molecular biomarker discovery and therapeutic development in neurodegenerative diseases. It was also proposed that there may be an infectious etiology in AD that is associated with viruses such as herpes simplex virus (HSV-1) and influenza A virus (IAV), leading to neuroinflammation-induced AD pathogenesis or disease progression. We sought to develop high-throughput, quantitative molecular biomarker assays using dissociated cells from human cerebral organoids (dcOrgs), that can used for screening compounds to reverse AD-associated neuroinflammation. We found that HSV-1 infection, but not IAV infection, in dcOrgs led to increased intracellular Aβ42 and phosphorylated Tau-Thr212 (pTau-212) expression, lower ratios of secreted Aβ42/40, as well as neuronal loss, and increased proportions of astrocytes and microglia, which are hallmarks of AD. Among the glia cell-type markers, Iba1 (microglia) and GFAP (astrocyte) expression were most strongly correlated with HSV-1 expression, which further supported that these biomarkers are perturbed by glia-mediated neuroinflammation. By performing large-scale RNA sequencing, we observed that differentially expressed transcripts in HSV-1 infected dcOrgs were specifically enriched for AD-associated GWAS genes, but not for genes associated with other common neurodegenerative, neuropsychiatric or autoimmune diseases. Immediate treatment of HSV-1 infected dcOrgs with anti-herpetic drug acyclovir (ACV) rescued most of the cellular and transcriptomic biomarkers in a dosage-dependent manner, indicating that it is possible to use our high-throughput platform to identify compounds or target genes that can reverse these neuroinflammation-induced biomarkers associated with AD.
Article
Full-text available
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Chapter
Brain aging is a complicated and diversified process that includes an ongoing decline in cognitive performance as well as various cellular and molecular changes in the brain. During an individual's life, glial cells such as oligodendrocytes, microglia, and astrocytes are crucial for maintaining healthy brain function. Astrocytes support neurons but may become less efficient with age, contributing to cognitive decline. Microglia, the brain's immune cells, can turn overactive with age, causing inflammation which can lead to neuronal damage. Oligodendrocytes, responsible for myelin sheath maintenance, also undergo changes, affecting neural communication. With advancing age, changes in glial cell function may lead to neuroinflammation, oxidative damage, and abnormalities in neural circuitry, all of which contribute to age related cognitive decline and neurodegenerative disorders. This chapter delves into the intricate dynamics of brain aging, focusing on the profound influence of age-related changes in glial cells.
Article
Full-text available
Cerebral venous abnormalities, distinct from traditional arterial diseases, have been linked to brain atrophy in a previous community‐based cohort study, specifically in relation to the reduction of deep medullary veins (r‐DMVs). To better understand the properties and biological functions of serum extracellular vesicles (EVs) in cerebral venous disease‐associated brain atrophy, EVs are extracted from the serum of both participants with r‐DMV and normal controls and analyzed their proteomic profiles using Tandem Mass Tag label quantitation analysis. Phenotypic experiments showed that EVs from individuals with r‐DMVs are able to disrupt the normal functions of neurons, endothelial cells, and smooth muscle cells, and induce A1 reactive astrocytes. Additionally, this study provided a comprehensive characterization of the proteomic profile of DMV EVs and found that the collagen hydroxyproline is upregulated, while complement C3 is downregulated in the r‐DMV group, suggesting that r‐DMV may not be a simple pathological phenomenon and highlighting the potential involvement of EVs in the progression of brain atrophy in r‐DMVs which has implications for the development of future therapeutic strategies.
Article
Full-text available
Protoplasmic astrocytes are increasingly thought to interact extensively with neuronal elements in the brain and to influence their activity. Recent reports have also begun to suggest that physiologically, and perhaps functionally, diverse forms of these cells may be present in the CNS. Our current understanding of astrocyte form and distribution is based predominately on studies that used the astrocytic marker glial fibrillary acidic protein (GFAP) and on studies using metal-impregnation techniques. The prevalent opinion, based on studies using these methods, is that astrocytic processes overlap extensively and primarily share the underlying neuropil. However, both of these techniques have serious shortcomings for visualizing the interactions among these structurally complex cells. In the present study, intracellular injection combined with immunohistochemistry for GFAP show that GFAP delineates only ∼15% of the total volume of the astrocyte. As a result, GFAP-based images have led to incorrect conclusions regarding the interaction of processes of neighboring astrocytes. To investigate these interactions in detail, groups of adjacent protoplasmic astrocytes in the CA1 stratum radiatum were injected with fluorescent intracellular tracers of distinctive emissive wavelengths and analyzed using three-dimensional (3D) confocal analysis and electron microscopy. Our findings show that protoplasmic astrocytes establish primarily exclusive territories. The knowledge of how the complex morphology of protoplasmic astrocytes affects their 3D relationships with other astrocytes, oligodendroglia, neurons, and vasculature of the brain should have important implications for our understanding of nervous system function.
Article
Full-text available
ALZHEIMER'S disease (AD) is the most common cause of progressive intellectual failure in aged humans. AD brains contain numerous amyloid plaques surrounded by dystrophic neurites, and show profound synaptic loss, neurofibrillary tangle formation and gliosis. The amyloid plaques are composed of amyloid beta-peptide (A beta), a 40-42-amino-acid fragment of the beta-amyloid precursor protein (APP)(1). A primary pathogenic role for APP/A beta is suggested by missense mutations in APP that are tightly linked to autosomal dominant forms of AD(2,3). A major obstacle to elucidating and treating AD has been the lack of an animal model. Animals transgenic for APP have previously failed to show extensive AD-type neuropathology(4-10), but we now report the production of transgenic mice that express high levels of human mutant APP (with valine at residue 717 substituted by phenylalanine) and which progressively develop many of the pathological hallmarks of AD, including numerous extracellular thioflavin S-positive AP deposits, neuritic plaques, synaptic loss, astrocytosis and microgliosis. These mice support a primary role for APP/A beta in the genesis of AD and could provide a preclinical model for testing therapeutic drugs.
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.
Chapter
This chapter provides a historical perspective that highlights the early period of glial research. Topics covered include Virchow's invention of the term neuroglia in 1856, Heinrich Müller' picture of a glial cell, stellate cells in white and gray matter, and Camillo Golgi's description of cells with characteristic features of astrocytes and oligodendrocytes.
Article
Alzheimer's disease (AD) is a devastating neurodegenerative disease that affects more than 15 million people worldwide. Within the next generation, these numbers will more than double. To assist in the elucidation of pathogenic mechanisms of AD and related disorders, such as frontotemporal dementia (FTDP-17), genetically modified mice, flies, fish and worms were developed, which reproduce aspects of the human histopathology, such as beta-amyloid-containing plaques and tau-containing neurofibrillary tangles (NFT). In mice, the tau pathology caused selective behavioral impairment, depending on the distribution of the tau aggregates in the brain. Beta-amyloid induced an increase in the numbers of NFT, whereas the opposite was not observed in mice. In beta-amyloid-producing transgenic mice, memory impairment was associated with increased levels of beta-amyloid. Active and passive beta-amyloid-directed immunization caused the removal of beta-amyloid plaques and restored memory functions. These findings have since been translated to human therapy. This review aims to discuss the suitability and limitations of the various animal models and their contribution to an understanding of the pathophysiology of AD and related disorders.
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
HIV-1-associated dementia (HAD) is an important complication of HIV-1 infection. Reactive astrogliosis is a key pathological feature in HAD brains and in other central nervous system (CNS) diseases. Activated astroglia may play a critical role in CNS inflammatory diseases such as HAD. In order to test the hypothesis that activated astrocytes cause neuronal injury, we stimulated primary human fetal astrocytes with HAD-relevant pro-inflammatory cytokine IL-1β. IL-1β-activated astrocytes induced apoptosis and significant changes in metabolic activity in primary human neurons. An FITC-conjugated pan-caspase inhibitor peptide FITC-VAD-FMK was used for confirming caspase activation in neurons. IL-1β activation enhanced the expression of death protein FasL in astrocytes, suggesting that FasL is one of the potential factors responsible for neurotoxicity observed in HAD and other CNS diseases involving glial inflammation. Our data presented here add to the developing picture of role of activated glia in HAD pathogenesis.
Book
Developmental function – producing new neural cellsDevelopmental function – neuronal guidanceRegulation of synaptogenesis and control of synaptic maintenance and eliminationStructural function – creation of the functional microarchitecture of the brainVascular function – creation of glial–vascular interface (blood–brain barrier) and glia–neurone–vascular unitsRegulation of brain microcirculationIon homeostasis in the extracellular spaceRegulation of extracellular glutamate concentrationWater homeostasis and regulation of the extracellular space volumeNeuronal metabolic supportAstroglia regulate synaptic transmissionIntegration in neuronal–glial networksAstrocytes as cellular substrate of memory and consciousness?