Astrocytes are key homeostatic and defensive cells of the central nervous system (CNS). They undertake numerous functions during development and in adulthood to support and protect the brain through finely regulated communication with other cellular elements of the nervous tissue. In Alzheimer’s disease (AD), astrocytes undergo heterogeneous morphological, molecular and functional alterations represented by reactive remodelling, asthenia and loss of function. Reactive astrocytes closely associate with amyloid β (Aβ) plaques and neurofibrillary tangles in advanced AD. The specific contribution of astrocytes to AD could potentially evolve along the disease process and includes alterations in their signalling, interactions with pathological protein aggregates, metabolic and synaptic impairments. In this review, we focus on the purinergic receptor, P2X7R, and discuss the evidence that P2X7R activation contributes to altered astrocyte functions in AD. Expression of P2X7R is increased in AD brain relative to non-demented controls, and animal studies have shown that P2X7R antagonism improves cognitive and synaptic impairments in models of amyloidosis and tauopathy. While P2X7R activation can induce inflammatory signalling pathways, particularly in microglia, we focus here specifically on the contributions of astrocytic P2X7R to synaptic changes and protein aggregate clearance in AD, highlighting cell-specific roles of this purinoceptor activation that could be targeted to slow disease progression.

Neuropathological changes in Alzheimer’s disease (AD) include the progressive deposition of senile plaques and neurofibrillary tangles (NFTs), alongside extensive and complex glial alterations, vascular changes, synapse and neuron loss, leading to cognitive impairment and dementia [1].

Astrocytes are a subpopulation of glial cells derived from neuroepithelial progenitors that account for 20–40% of total glial cells in humans, depending on the brain region [2,3]. Single-cell transcriptomics revealed considerable molecular heterogeneity among astrocyte populations in rodent brain [4] as well as a complex stratified architecture across cerebral layers [5] that is likely more diverse in human brain, which in addition contain several forms of astrocytes absent in other mammals [6].

Astrocytes perform critical functions in the developing and adult CNS [7]. For example, during development, astrocytes remodel neuronal circuits, participating in the formation and pruning of synapses [8,9]. Astrocytes are functionally integrated with synapses, with all astrocytic compartments found to abut synapses in adult mouse hippocampus [10]. A particularly high density of pre-synaptic terminals and/or dendritic spines contact astrocytic branches and leaflets [10,11] which may result from these being amongst the most dynamic of astrocytic structures in response to neuronal signals [10]. Astrocytes, together with other cellular and non-cellular elements, form multipartite synapses that regulate various aspects of synaptic function [12]. Astrocytes control synaptogenesis, synaptic maturation, synaptic maintenance and synaptic extinction through the release of multiple specific regulatory molecules including thrombospondins, hevins, glypicans, norrin and many more [13]. Astrocytes modulate synaptic activity by several mechanisms of which K+ buffering, glutamate clearance by astrocyte specific excitatory amino acid transporter (EAAT)1/2 receptors and supplying glutamine by the glutamate-(GABA)-glutamine shuttle are of particular relevance [14]. Astrocytes release small molecules such as adenosine-triphosphate (ATP) which is rapidly converted to adenosine by ectonucleotidases to supress excitatory transmission by acting on presynaptic adenosine A1 receptors (A1R) [15,16]. Astrocytes also participate in the maintenance of ion homeostasis allowing the rapid uptake of K+ from the extracellular space during neuronal activity [17]. Thus, astrocytes are actively engaged in the regulation of synaptic transmission, synaptic plasticity and maintenance of neuronal circuitry in the CNS, as previously reviewed [11].

In addition, astrocytic end-feet are a parenchymal component of the blood–brain barrier (BBB). They contribute to the regulation of blood flow in response to neuronal activity, along with perivascular neuronal terminals, endothelial cells and pericytes, in a process known as neurovascular coupling [18]. Astrocytes take up glucose, the main energy source of the brain, and store it as glycogen, or may supply it to neurons in the form of lactate [19,20], although this is debated. Furthermore, end-feet-specific expression of aquaporin-4 (AQP4) maintains the proper function of glymphatic system which facilitates the elimination of toxic solutes from the interstitial fluid [21].

Astrocytes in AD

Brain injury associated with trauma, stroke, neuroinfection or immune attack triggers specific and stereotypical defensive responses of astrocytes known as reactive astrogliosis in which astrocytes proliferate, form a glial scar and promote the recruitment of immune cells. In neurodegenerative diseases including AD, astrocytes undergo morphological, molecular and functional changes commonly known as astrocytic reactivity [22]. In contrast to the bona fide stereotypical reactive astrogliosis that is triggered by brain lesions associated with a breach of the BBB, astrocytic reactivity in chronic neurological diseases is highly dynamic and heterogeneous [22].

Glial fibrillary acidic protein (GFAP) is a cytoskeletal protein in astrocytes that is increased to allow cytoskeletal rearrangement in response to many physiological and disease stimuli [22–25]. In human AD brain, astrocytes expressing different isoforms and/or truncated forms of GFAP cluster around Aβ plaques [26,27]. This is similar in mice where, without altering their spatial distribution, astrocytes extend their processes towards amyloid deposits [28]. Astrocytes with GFAP-immunoreactivity are also associated with ghost neurofibrillary tangles (NFTs) in advanced stages of AD [29], a feature believed to result from astrocyte processes having penetrated extracellular ghost tangles or tangle-bearing neurons in advanced AD and becoming separated from the soma [30–32]. Moreover, increased numbers of GFAP-immunoreactive astrocytes in the superior temporal sulcus are among the molecular features that distinguish between AD cases with dementia and those showing resilience to cognitive decline [33].

Levels of GFAP in brain tissue are high in prodromal AD [34] and an increase in the levels of GFAP in cerebrospinal fluid (CSF) and plasma of AD patients is also detected in early stages, that peaks upon symptom onset [35]. Recent work using specific positron electron tomography (PET) ligands demonstrated an association between Aβ, but not tau, burden and CSF GFAP levels [36]. These authors further showed that the strength of tau-PET signals is more closely associated with increases in CSF levels of a putative marker of astrocyte reactivity YKL-40 [37] that is expressed only in a subset of astrocytes [38] and also by several other cell types [39]. Others have reported that CSF YKL-40 levels are distinct from grey matter loss associated with phosphorylated tau [40].

However, characterisation of GFAP immunostaining is insufficient to make conclusions about functional changes in astrocytes. Indeed, astrocytes show considerable transcriptomic heterogeneity in the diseased brain [41], even amongst cases with the same diagnosis [42]. Recent multi-transcriptomic analysis of human astrocytes in AD relative to control brain revealed increases in genes related to specialized astrocyte-neuron contacts at perisynaptic astrocyte leaflets that influence the function of adjacent synapses, alongside downregulation of endolysosomal and mitochondrial genes in astrocytes, that for mitochondrial genes were found to decline as disease severity increased [42]. The decrease in endolysosomal and mitochondria-related genes, but not the upregulation of synapse-related genes, was mirrored in transgenic APP/PS1 mice [42].

Astrocytes, like microglia, possess a myriad of cell surface receptors through which they recognise a variety of stimuli [43]. These include molecules released upon cell damage including adenosine triphosphate (ATP) which is rapidly converted to adenosine by ectonucleotidases, heat-shock proteins, and disease-specific pathological species of Aβ and tau [44–47]. The binding of these pathogenic molecules to astrocytic receptors is a key step for the initiation of signal transduction cascades that increase the transcription of target genes [48,49]. Below, we discuss how sensing through one of these receptors, the purinergic P2X7 receptor, may contribute to alterations in synaptic and endolysosome-related functions of astrocytes in AD (Figure 1).

Potential roles of astrocytic P2X7R in synaptic function and protein clearance in AD

Figure 1
Potential roles of astrocytic P2X7R in synaptic function and protein clearance in AD

In AD brain, high levels of ATP could activate P2X7R in astrocytes and contribute to defects in neurotransmission. The opening of P2X7R channels allows calcium influx which modulates the release of glutamate to the synaptic cleft, where it could bind to NMDA-Rs at the post-synapse. P2X7R could also participate in the regulation of inhibitory synapses, by modulating the release of GABA from astrocytes. In addition to their intimate association with synapses, astrocytes also play important roles in the maintenance of protein homeostasis through the internalisation and degradation of Aß and tau aggregates. In AD, astrocytic P2X7R could alter protein clearance pathways via HSPB1-mediated autophagy or the regulation of HSPG expression, which might influence astrocyte uptake and clearance of tau species.

Figure 1
Potential roles of astrocytic P2X7R in synaptic function and protein clearance in AD

In AD brain, high levels of ATP could activate P2X7R in astrocytes and contribute to defects in neurotransmission. The opening of P2X7R channels allows calcium influx which modulates the release of glutamate to the synaptic cleft, where it could bind to NMDA-Rs at the post-synapse. P2X7R could also participate in the regulation of inhibitory synapses, by modulating the release of GABA from astrocytes. In addition to their intimate association with synapses, astrocytes also play important roles in the maintenance of protein homeostasis through the internalisation and degradation of Aß and tau aggregates. In AD, astrocytic P2X7R could alter protein clearance pathways via HSPB1-mediated autophagy or the regulation of HSPG expression, which might influence astrocyte uptake and clearance of tau species.

Close modal

The purinoreceptor family includes highly conserved receptors that bind adenine (P0 receptors), adenosine (P1 receptors) and ADP/ATP (P2 receptors) [50–53]. P2 receptors are further classified into G-protein coupled (P2Y) and ion-gated (P2X) receptors [54]. To date, seven P2X (PX1-7R) and eight P2Y (P2Y1R, P2Y2R, P2Y4R, P2Y6R, P2Y11-14R) subtypes have been identified, including P2Y receptors with the ability to sense pyrimidines (e.g., uridine diphosphate [UDP], UDP-glucose, UDP-galactose) in addition to purines [55].

The P2X7R protein consists of a short intracellular N-terminal domain, two transmembrane α-helixes, an extracellular loop enriched in N-glycosylation sites and a long cytoplasmatic C-terminal domain [56,57]. P2X7R is typically found in a resting/closed or apo-state conformation, with a narrow cavity through which ATP must access to reach the active binding pocket. When ATP binds to P2X7R, conformational rearrangements lead to the opening of an ion-permeable channel that allows the influx (i.e., Na+, Ca2+) and efflux (i.e., K+) of small cations, and upon channel dilatation into a larger pore, the entry of large hydrophilic molecules at a slower rate [58]. In contrast to other P2X subtypes, P2X7R does not undergo desensitisation after activation of the receptor due to permanent stabilization provided by a palmitoylated cysteine rich region in the cytoplasmatic domain [59]. This feature of P2X7R activation dynamics likely contributes to the hyperpolarised astrocyte membrane potential that is important for astrocyte physiology and functions [60].

The human P2X7R gene is highly polymorphic with more than 150 non-synonymous SNPs, the majority of which lead to amino acid substitutions in the extracellular loop or the cytoplasmic C-terminal tail [61], affecting agonist binding affinity [62], trafficking to membranes [63], ion channel activity [64] and permeability of the pore [65]. In humans, there are seven splice variants of P2X7R, two of which are predominantly expressed in the CNS and immune tissues including the full-length (P2X7R A) and a C-terminally truncated form with an early stop codon (P2X7R B) [66,67]. The latter gives rise to a receptor deficient in the formation of a large permeable pore that retains ion channel properties [67].

Opening of the P2X7R channel is stimulated by ATP concentrations in the higher micromolar to millimolar range, in contrast to the lower ATP concentrations required for opening of other P2X family members, with ATP4− being a true agonist [68]. ATP can be sensed by P2X7Rs in rodent or human microglia which show the highest levels of P2X7R expression in the brain [69–72] and oligodendrocytes [73,74]. Whilst the neuronal localization of P2X7R remains controversial [75,76], P2X7R expression in astrocytes [71,72,77–80] has been confirmed by the presence of agonist-induced currents and Ca2+ responses [78,81–86]. Activation of P2X7R in astrocytes triggers various cellular signals including those that stimulate nitrous oxide and superoxide radicals production [87,88], Akt and mitogen-activated protein kinase (MAPK) signalling [89–91], secretion of cytokines and other mediators of inflammation [88,89,92–94], as well as the release of gliotransmitters [81,95,96].

P2X7R in AD

While there is limited evidence that single nucleotide polymorphisms that alter P2X7R activity influence the risk of AD [97], converging studies show enhanced levels of P2X7R mRNA and P2X7R protein in AD post-mortem brains in comparison with non-demented controls suggesting an involvement of this purinoreceptor in AD [71,98,99]. This increase is similarly observed in several transgenic mouse models of familial AD that overexpress mutant forms of APP including Tg2576 [80], APP/PS1 [100] and J20 [99] mice as well as in tauopathy models carrying mutations in MAPT [63]. Higher levels of P2X7R were observed at 12 months relative to 3-month-old APP/PS1 mice suggesting that P2X7R expression increases with disease progression [100]. P2X7R protein increases are particularly apparent surrounding Aβ plaques in AD brain [71,99,101], and this plaque-associated up-regulation is recapitulated in transgenic rodent models of amyloidogenesis [80,99,100]. In addition to changes in P2X7R expression, pharmacological antagonism or genetic deletion of P2X7R protects against disease in mouse models harbouring Aß [62,92] or tau pathology [63,91,93] indicating that P2X7R-mediated functions contribute to the disease process.

P2X7R contributions to astrocyte driven synaptic changes in AD

Astrocytes are implicated in the deterioration of synaptic transmission in AD, affecting both excitatory (glutamatergic) and inhibitory γ-aminobutyric acid (GABA)-ergic synapses [14]. Aβ induces calcium dysregulation in astrocytes which can affect their ability to modulate neurotransmission [102,103]. For example, astrocytes can induce neuronal hyperactivity through the transient receptor potential cation channel, subfamily A, member 1 (TRPA1) in response to Aβ, which potentiates the release of excitatory glutamate in APP/PS1 mice [104]. Reports describing an increase in the release of the N-methyl-D-aspartate receptor (NMDA-R) co-factor D-serine from these astrocytes are now questioned, since astrocytes do not produce D-serine but rather L-serine which can be shuttled to neurons to drive neuronal production of D-serine [105]. Compromised glucose metabolism is observed in prodromal stages of AD which correlates with disease progression [14,106]. Accordingly, reduced aerobic glycolysis is also observed in prodromal AD [107], one consequence of which is decreased L-serine synthesis by astrocytes [108]. This disrupts NMDA-R-mediated synaptic plasticity and cognitive function in AD mice, which can be recovered upon dietary L-serine supplementation [109].

In transgenic mice expressing AD-causing mutant forms of APP and PSEN1 (APP/PS1), astrocytes surrounding Aβ plaques have lower levels of EAAT2, which leads to an extra-synaptic accumulation of glutamate, neuronal hyperactivity [110] potentially mediated by neuronal NMDA receptors [111], and possibly neurotoxicity. However, whether or not this is true in human disease is uncertain since analysis of postmortem brain from AD cases with significant amyloid and tau pathology showed higher levels of astrocytic EAAT2 in comparison with non-demented cases carrying AD pathology, pointing towards a mechanism of astrocytic resilience against neuropathological changes in AD [112]. Astrocytic P2X7R could be activated by ATP, or potentially indirectly, by Aβ [113], in the vicinity of senile plaques to contribute to excess glutamate levels. Stimulation of P2X7R in hippocampal, spinal cord and substantia gelatinosa astrocytes using the potent broad agonist 2′(3′)-O-(4-benzoylbenzoyl)adenosine 5′-triphosphate (BzATP) induces glutamate release and stimulation of neighbouring NMDA-Rs through a Ca2+-dependent mechanism [114,115]. The opening of P2X7R pores could also mobilise other transmitter-containing vesicles following Ca2+ entry, but the precise molecular pathways that mediate these events remain obscure [75]. Astrocytes can also influence neuronal inhibition by increasing GABA release at the synaptic cleft in mice expressing five familial mutations in APP and PSEN1 (5xFAD mice, [116]), and some GABA release from astrocytes is regulated by P2X7Rs, at least in stratum radiatum astrocytes proximal to interneurons in the hippocampus [117].

P2X7R and astrocytic protein clearance pathways – implications for AD

Recent analysis showed that endolysosomal pathway components, fundamental for the uptake, processing, degradation and disposal of proteins and cellular debris, are down-regulated in AD astrocytes [42]. Indeed astrocytes, in addition to microglia, play central roles in the clearance of protein aggregates and other debris in degenerating AD brain [118]. By surrounding Aβ plaques, glia erect a physical and functional barrier to isolate and potentially clear proteinaceous aggregates from the affected neuropil [119].

Aß oligomers are observed within astrocytes in post-mortem AD brain [120] and mature healthy astrocytes engulf and degrade Aβ species in vitro and ex vivo [121,122]. Inhibition of reactive astrogliosis either increases [123] or reduces [124] levels of Aβ in APP/PS1 mice. The astrocyte-mediated internalisation of Aβ occurs in a ApoE-dependent manner, since ApoE deficient astrocytes are not capable of removing amyloid [125], with efficient Aβ uptake and clearance from astrocytes dependent on transcription factor EB (TFEB)-mediated lysosomal degradation [126]. Similarly, there is evidence that the Aβ sensor low density lipoprotein receptor-related protein 1 (LRP1) is critical for astrocytic uptake and degradation of Aß [47]. Astrocytes can also upregulate the expression of extracellular proteolytic enzymes that target Aβ including insulin degrading enzyme [127], released via an unconventional autophagy-dependent secretory pathway, and endothelin-converting enzyme-2 [128], they are efficient in autophagy and can potentially limit the accumulation of Aβ species in AD [118].

Astrocytes can also internalize modified forms of tau protein. In a tauopathy mouse model in which tau was expressed specifically in entorhinal cortex neurons, tau aggregates that spread trans-synaptically to the dentate gyrus were detected in astrocytes [129]. These data indicate that astrocytes internalize and may contribute to tau propagation. Indeed, extracellular forms of fibrillar tau are taken up by astrocytes [46], including at synapses for redirection into lysosomal degradation pathways to regulate tau spread [130]. Data also implicates heparin sulphate proteoglycans (HSPGs) and LRP1 in tau uptake by astrocytes, with the efficiency of the uptake varying depending on disease-associated tau modifications [131–134].

While the direct contributions of astrocytic P2X7R to these processes have not been investigated in detail, several independent studies demonstrated that pharmacological blockade or genetic deletion of P2X7R is beneficial in mouse models of AD, reporting reduced amyloid plaque number and abundance of soluble Aβ species in mouse models of amyloidosis [71,135]. In tauopathy mouse models, decreases in tau phosphorylation at certain epitopes [72,101] or a reduction in the abundance of misfolded tau forms [136] were reported. Although some alterations in microglial morphology and functions including phagocytosis, migration and cytokine release were observed upon P2X7R inhibition [72,101], no consistent changes were detected between the different mouse models [71,136]. No alterations in protein degradation pathways were described although P2X7R induction in microglia is known to impair lysosomal function, increasing levels of the autophagosome membrane-associated form of microtubule-associated protein 1 light chain 3 (LC3)-II in a Ca2+-dependent manner, up-regulating the formation of autophagosomes and autophagolysosomes, and increasing the release of autophagosomes [137,138]. We suggest that further exploration of the potential contribution of astrocytic P2X7R to these processes is warranted since P2X7R activation also regulates autophagy in astrocytes [139,140]. Astrocytes are damaged in status epilepticus, and they form vacuoles containing lysosome-associated membrane protein (LAMP)-1 [141]. P2X7R antagonism was found to decrease astrocyte damage in the molecular layer of the dentate gyrus and frontoparietal cortex under these conditions [142] which could be caused by a prolonged induction of the molecular chaperone small heat-shock protein (HSP)B1, a HSP that facilitates the folding and removal of aberrant proteins, and, in turn, promotes astroglial autophagy [139]. Others have shown similar effects in P2X7R knockout mice, where P2X7R signalling to focal adhesion kinase (FAK) was found to regulate HSPB25 and fine tune autophagy [140]. Since, as we discuss above, astrocytes efficiently internalise Aβ and modified forms of tau in disease, these data may suggest that P2X7R-induced regulation of autophagy in astrocytes is important for limiting proteinaceous spread in AD and tauopathies. Finally, tau internalization and release may be mediated by HSPGs [143,144]. P2X7R also regulates HSPG expression and localization, at least in the cornea [145], and we suggest that exploration of the potential for P2X7R signalling to similarly affect HSPGs in astrocytes and alter tau clearance is warranted.

In summary, we provide an overview of synapse-related and protein clearance functions of astrocytes that can be modulated by P2X7R signalling in AD. P2X7R has gained much attention in recent years as a possible therapeutic target. Genetic deletion of P2X7R in APP/PS1 mice improved long-term synaptic plasticity, spatial learning and memory dysfunction relative to wild-type littermates [71], and P2X7R antagonism in mouse models of tauopathy harbouring the frontotemporal dementia (FTD)-causing MAPT mutations G272V and/or P301S ameliorates cognitive and behavioural deficits as well as synaptic dysfunction [72,101,136]. We suggest that further exploration of P2X7R-driven effects on biological processes linked with astrocyte contributions to AD may uncover novel targets for therapeutic intervention.

  • Astrocytes play critical roles in maintaining a healthy brain environment. This is mediated through multiple homeostatic transporters, interactions with neurons and microglia, and functions at the blood brain barrier and synapses.

  • Some astrocytes become ‘reactive’ in AD, while others show asthenia and loss of homeostatic functions. Astrocytes in AD reduce their support for synapses and show deficits in endolysosomal pathway components.

  • There are increases in P2X7R mRNA and protein in AD, particularly in the vicinity of plaques. P2X7R activation in astrocytes influences synaptic activity and protein clearance pathways, and this may be one route by which P2X7R affects AD progression.

  • Further exploration of the functional consequences of astrocytic P2X7R in AD may reveal novel cell-type specific targets for intervention.

The authors declare that there are no competing interests associated with the manuscript.

Our work is funded by Alzheimer’s Research UK [grant numbers ARUK-PhD2018-002 and ARUK-PG2019A-004 (to W.N. and B.G.P.-N.)] and a Medical Research Council Transition Award [MR/V036947/1 (to M.J.-S.)].

All authors wrote and edited the manuscript. For the purpose of open access, the author has applied a Creative Commons Attribution (CC BY) licence to any Author Accepted Manuscript version arising.

A1R

adenosine A1 receptor

AD

Alzheimer’s disease

ADP

adenosine diphosphate

AMP

adenosine monophosphate

ApoE

apolipoprotein E

APP

amyloid precursor protein

AQP4

aquaporin-4

ATP

adenosine triphosphate

β-amyloid

CNS

central nervous system

CSF

cerebrospinal fluid

EAAT (1/2)

excitatory amino acid transporter (1/2)

FAK

focal adhesion kinase

GABA

γ amino-butyric acid

GABA-R

GABA receptor

GAT1/3

GABA transporter type 1/3

GFAP

glial fibrillary acidic protein

HSPB1

heat shock protein B1

HSPB25

heat shock protein B25

HSPG

heparin sulphate proteoglycan

LAMP1

lysosome associated membrane protein 1

LRP1

low density lipoprotein receptor-related protein 1

NFTs

neurofibrillary tangles

NMDAR

N-methyl-D-aspartate receptor

PET

positron electron tomography

PS1

presenilin 1

TFEB

transcription factor EB

TRPA1

transient receptor potential cation channel, subfamily A, member 1

UDP

uridine diphosphate

1.
Henstridge
C.M.
,
Hyman
B.T.
and
Spires-Jones
T.L.
(
2019
)
Beyond the neuron-cellular interactions early in Alzheimer disease pathogenesis
.
Nat. Rev. Neurosci.
20
,
94
108
[PubMed]
2.
Molofsky
A.V.
and
Deneen
B.
(
2015
)
Astrocyte development: a guide for the perplexed
.
Glia
63
,
1320
1329
[PubMed]
3.
von Bartheld
C.S.
,
Bahney
J.
and
Herculano-Houzel
S.
(
2016
)
The search for true numbers of neurons and glial cells in the human brain: A review of 150 years of cell counting
.
J. Comp. Neurol.
524
,
3865
3895
[PubMed]
4.
Batiuk
M.Y.
,
Martirosyan
A.
,
Wahis
J.
,
de Vin
F.
,
Marneffe
C.
,
Kusserow
C.
et al.
(
2020
)
Identification of region-specific astrocyte subtypes at single cell resolution
.
Nat. Commun.
11
,
1220
[PubMed]
5.
Bayraktar
O.A.
,
Bartels
T.
,
Holmqvist
S.
,
Kleshchevnikov
V.
,
Martirosyan
A.
,
Polioudakis
D.
et al.
(
2020
)
Astrocyte layers in the mammalian cerebral cortex revealed by a single-cell in situ transcriptomic map
.
Nat. Neurosci.
23
,
500
509
[PubMed]
6.
Oberheim
N.A.
,
Takano
T.
,
Han
X.
,
He
W.
,
Lin
J.H.
,
Wang
F.
et al.
(
2009
)
Uniquely hominid features of adult human astrocytes
.
J. Neurosci.
29
,
3276
3287
[PubMed]
7.
Perez-Nievas
B.G.
and
Serrano-Pozo
A.
(
2018
)
Deciphering the astrocyte reaction in Alzheimer's disease
.
Front. Aging Neurosci.
10
,
114
[PubMed]
8.
Allen
N.J.
,
Bennett
M.L.
,
Foo
L.C.
,
Wang
G.X.
,
Chakraborty
C.
,
Smith
S.J.
et al.
(
2012
)
Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors
.
Nature
486
,
410
414
[PubMed]
9.
Chung
W.S.
,
Clarke
L.E.
,
Wang
G.X.
,
Stafford
B.K.
,
Sher
A.
,
Chakraborty
C.
et al.
(
2013
)
Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways
.
Nature
504
,
394
400
[PubMed]
10.
Aten
S.
,
Kiyoshi
C.M.
,
Arzola
E.P.
,
Patterson
J.A.
,
Taylor
A.T.
,
Du
Y.
et al.
(
2022
)
Ultrastructural view of astrocyte arborization, astrocyte-astrocyte and astrocyte-synapse contacts, intracellular vesicle-like structures, and mitochondrial network
.
Prog. Neurobiol.
213
,
102264
[PubMed]
11.
Semyanov
A.
and
Verkhratsky
A.
(
2021
)
Astrocytic processes: from tripartite synapses to the active milieu
.
Trends Neurosci.
44
,
781
792
[PubMed]
12.
Verkhratsky
A.
and
Nedergaard
M.
(
2014
)
Astroglial cradle in the life of the synapse
.
Philos. Trans. R. Soc. Lond. B Biol. Sci.
369
,
20130595
[PubMed]
13.
Augusto-Oliveira
M.
,
Arrifano
G.P.
,
Takeda
P.Y.
,
Lopes-Araujo
A.
,
Santos-Sacramento
L.
,
Anthony
D.C.
et al.
(
2020
)
Astroglia-specific contributions to the regulation of synapses, cognition and behaviour
.
Neurosci. Biobehav. Rev.
118
,
331
357
[PubMed]
14.
Andersen
J.V.
,
Schousboe
A.
and
Verkhratsky
A.
(
2022
)
Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle
.
Prog. Neurobiol.
102331
[PubMed]
15.
Henneberger
C.
,
Papouin
T.
,
Oliet
S.H.
and
Rusakov
D.A.
(
2010
)
Long-term potentiation depends on release of D-serine from astrocytes
.
Nature
463
,
232
236
[PubMed]
16.
Pascual
O.
,
Casper
K.B.
,
Kubera
C.
,
Zhang
J.
,
Revilla-Sanchez
R.
,
Sul
J.Y.
et al.
(
2005
)
Astrocytic purinergic signaling coordinates synaptic networks
.
Science
310
,
113
116
[PubMed]
17.
Seifert
G.
,
Henneberger
C.
and
Steinhauser
C.
(
2018
)
Diversity of astrocyte potassium channels: An update
.
Brain Res. Bull.
136
,
26
36
[PubMed]
18.
Mishra
A.
(
2017
)
Binaural blood flow control by astrocytes: listening to synapses and the vasculature
.
J. Physiol.
595
,
1885
1902
[PubMed]
19.
Machler
P.
,
Wyss
M.T.
,
Elsayed
M.
,
Stobart
J.
,
Gutierrez
R.
,
von Faber-Castell
A.
et al.
(
2016
)
In vivo evidence for a lactate gradient from astrocytes to neurons
.
Cell Metab.
23
,
94
102
[PubMed]
20.
Morgello
S.
,
Uson
R.R.
,
Schwartz
E.J.
and
Haber
R.S.
(
1995
)
The human blood-brain barrier glucose transporter (GLUT1) is a glucose transporter of gray matter astrocytes
.
Glia
14
,
43
54
[PubMed]
21.
Iliff
J.
and
Simon
M.
(
2019
)
CrossTalk proposal: The glymphatic system supports convective exchange of cerebrospinal fluid and brain interstitial fluid that is mediated by perivascular aquaporin-4
.
J. Physiol.
597
,
4417
4419
[PubMed]
22.
Escartin
C.
,
Galea
E.
,
Lakatos
A.
,
O'Callaghan
J.P.
,
Petzold
G.C.
,
Serrano-Pozo
A.
et al.
(
2021
)
Reactive astrocyte nomenclature, definitions, and future directions
.
Nat. Neurosci.
24
,
312
325
[PubMed]
23.
Santos
G.W.Q.
,
Araujo
J.F.
,
Cunha
M.J.B.
,
Costa
S.O.
,
Barbosa
A.L.C.
,
Mesquita
J.B.
et al.
(
2005
)
Circadian variation in GFAP immunoreactivity in the mouse suprachiasmatic nucleus
.
Biol. Rhythm. Res.
36
,
141
150
24.
Rodriguez
J.J.
,
Terzieva
S.
,
Olabarria
M.
,
Lanza
R.G.
and
Verkhratsky
A.
(
2013
)
Enriched environment and physical activity reverse astrogliodegeneration in the hippocampus of AD transgenic mice
.
Cell Death Dis.
4
,
e678
[PubMed]
25.
Carvalho-Paulo
D.
,
de Morais Magalhaes
N.G.
,
de Almeida Miranda
D.
,
Diniz
D.G.
,
Henrique
E.P.
,
Moraes
I.A.M.
et al.
(
2017
)
Hippocampal astrocytes in migrating and wintering semipalmated sandpiper calidris pusilla
.
Front. Neuroanat.
11
,
126
[PubMed]
26.
Mouser
P.E.
,
Head
E.
,
Ha
K.H.
and
Rohn
T.T.
(
2006
)
Caspase-mediated cleavage of glial fibrillary acidic protein within degenerating astrocytes of the Alzheimer's disease brain
.
Am. J. Pathol.
168
,
936
946
[PubMed]
27.
Kamphuis
W.
,
Middeldorp
J.
,
Kooijman
L.
,
Sluijs
J.A.
,
Kooi
E.J.
,
Moeton
M.
et al.
(
2014
)
Glial fibrillary acidic protein isoform expression in plaque related astrogliosis in Alzheimer’s disease
.
Neurobiol. Aging
35
,
492
510
[PubMed]
28.
Galea
E.
,
Morrison
W.
,
Hudry
E.
,
Arbel-Ornath
M.
,
Bacskai
B.J.
,
Gomez-Isla
T.
et al.
(
2015
)
Topological analyses in APP/PS1 mice reveal that astrocytes do not migrate to amyloid-beta plaques
.
Proc. Natl. Acad. Sci. U.S.A.
112
,
15556
15561
[PubMed]
29.
Irwin
D.J.
,
Cohen
T.J.
,
Grossman
M.
,
Arnold
S.E.
,
Xie
S.X.
,
Lee
V.M.
et al.
(
2012
)
Acetylated tau, a novel pathological signature in Alzheimer’s disease and other tauopathies
.
Brain
135
,
807
818
[PubMed]
30.
Ikeda
K.
,
Haga
C.
,
Akiyama
H.
,
Kase
K.
and
Iritani
S.
(
1992
)
Coexistence of paired helical filaments and glial filaments in astrocytic processes within ghost tangles
.
Neurosci. Lett.
148
,
126
128
[PubMed]
31.
Ikeda
K.
,
Haga
C.
,
Oyanagi
S.
,
Iritani
S.
and
Kosaka
K.
(
1992
)
Ultrastructural and immunohistochemical study of degenerate neurite-bearing ghost tangles
.
J. Neurol.
239
,
191
194
[PubMed]
32.
Probst
A.
,
Ulrich
J.
and
Heitz
P.U.
(
1982
)
Senile dementia of Alzheimer type: astroglial reaction to extracellular neurofibrillary tangles in the hippocampus. An immunocytochemical and electron-microscopic study
.
Acta Neuropathol.
57
,
75
79
[PubMed]
33.
Perez-Nievas
B.G.
,
Stein
T.D.
,
Tai
H.C.
,
Dols-Icardo
O.
,
Scotton
T.C.
,
Barroeta-Espar
I.
et al.
(
2013
)
Dissecting phenotypic traits linked to human resilience to Alzheimer's pathology
.
Brain
136
,
2510
2526
[PubMed]
34.
Carter
S.F.
,
Scholl
M.
,
Almkvist
O.
,
Wall
A.
,
Engler
H.
,
Langstrom
B.
et al.
(
2012
)
Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG
.
J. Nucl. Med.
53
,
37
46
[PubMed]
35.
Benedet
A.L.
,
Mila-Aloma
M.
,
Vrillon
A.
,
Ashton
N.J.
,
Pascoal
T.A.
,
Lussier
F.
et al.
(
2021
)
Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer Disease continuum
.
JAMA Neurol.
78
,
1471
1483
[PubMed]
36.
Ferrari-Souza
J.P.
,
Ferreira
P.C.L.
,
Bellaver
B.
,
Tissot
C.
,
Wang
Y.T.
,
Leffa
D.T.
et al.
(
2022
)
Astrocyte biomarker signatures of amyloid-beta and tau pathologies in Alzheimer's disease
.
Mol. Psychiatry
[PubMed]
37.
Carter
S.F.
,
Herholz
K.
,
Rosa-Neto
P.
,
Pellerin
L.
,
Nordberg
A.
and
Zimmer
E.R.
(
2019
)
Astrocyte biomarkers in Alzheimer’s disease
.
Trends Mol. Med.
25
,
77
95
[PubMed]
38.
Querol-Vilaseca
M.
,
Colom-Cadena
M.
,
Pegueroles
J.
,
San Martin-Paniello
C.
,
Clarimon
J.
,
Belbin
O.
et al.
(
2017
)
YKL-40 (Chitinase 3-like I) is expressed in a subset of astrocytes in Alzheimer’s disease and other tauopathies
.
J. Neuroinflammation
14
,
118
[PubMed]
39.
Muszynski
P.
,
Groblewska
M.
,
Kulczynska-Przybik
A.
,
Kulakowska
A.
and
Mroczko
B.
(
2017
)
YKL-40 as a potential biomarker and a possible target in therapeutic strategies of Alzheimer’s disease
.
Curr. Neuropharmacol.
15
,
906
917
[PubMed]
40.
Gispert
J.D.
,
Monte
G.C.
,
Falcon
C.
,
Tucholka
A.
,
Rojas
S.
,
Sanchez-Valle
R.
et al.
(
2016
)
CSF YKL-40 and pTau181 are related to different cerebral morphometric patterns in early AD
.
Neurobiol. Aging
38
,
47
55
[PubMed]
41.
Liddelow
S.A.
,
Guttenplan
K.A.
,
Clarke
L.E.
,
Bennett
F.C.
,
Bohlen
C.J.
,
Schirmer
L.
et al.
(
2017
)
Neurotoxic reactive astrocytes are induced by activated microglia
.
Nature
541
,
481
487
[PubMed]
42.
Galea
E.
,
Weinstock
L.D.
,
Larramona-Arcas
R.
,
Pybus
A.F.
,
Gimenez-Llort
L.
,
Escartin
C.
et al.
(
2022
)
Multi-transcriptomic analysis points to early organelle dysfunction in human astrocytes in Alzheimer’s disease
.
Neurobiol. Dis.
166
,
105655
[PubMed]
43.
Heneka
M.T.
,
Carson
M.J.
,
El Khoury
J.
,
Landreth
G.E.
,
Brosseron
F.
,
Feinstein
D.L.
et al.
(
2015
)
Neuroinflammation in Alzheimer’s disease
.
Lancet Neurol.
14
,
388
405
[PubMed]
44.
Bours
M.J.
,
Swennen
E.L.
,
Di Virgilio
F.
,
Cronstein
B.N.
and
Dagnelie
P.C.
(
2006
)
Adenosine 5'-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation
.
Pharmacol. Ther.
112
,
358
404
[PubMed]
45.
Chen
G.Y.
and
Nunez
G.
(
2010
)
Sterile inflammation: sensing and reacting to damage
.
Nat. Rev. Immunol.
10
,
826
837
[PubMed]
46.
Wang
P.
and
Ye
Y.
(
2021
)
Filamentous recombinant human Tau activates primary astrocytes via an integrin receptor complex
.
Nat. Commun.
12
,
95
[PubMed]
47.
Liu
C.C.
,
Hu
J.
,
Zhao
N.
,
Wang
J.
,
Wang
N.
,
Cirrito
J.R.
et al.
(
2017
)
Astrocytic LRP1 mediates brain abeta clearance and impacts amyloid deposition
.
J. Neurosci.
37
,
4023
4031
[PubMed]
48.
Li
D.
and
Wu
M.
(
2021
)
Pattern recognition receptors in health and diseases
.
Signal Transduct Target Ther.
6
,
291
[PubMed]
49.
Price
B.R.
,
Johnson
L.A.
and
Norris
C.M.
(
2021
)
Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease
.
Ageing Res. Rev.
68
,
101335
[PubMed]
50.
Burnstock
G.
(
1976
)
Purinergic receptors
.
J. Theor. Biol.
62
,
491
503
[PubMed]
51.
Verkhratsky
A.
and
Burnstock
G.
(
2014
)
Purinergic and glutamatergic receptors on astroglia
.
Adv. Neurobiol.
11
,
55
79
[PubMed]
52.
Knospe
M.
,
Muller
C.E.
,
Rosa
P.
,
Abdelrahman
A.
,
von Kugelgen
I.
,
Thimm
D.
et al.
(
2013
)
The rat adenine receptor: pharmacological characterization and mutagenesis studies to investigate its putative ligand binding site
.
Purinergic Signal.
9
,
367
381
[PubMed]
53.
Gorzalka
S.
,
Vittori
S.
,
Volpini
R.
,
Cristalli
G.
,
von Kugelgen
I.
and
Muller
C.E.
(
2005
)
Evidence for the functional expression and pharmacological characterization of adenine receptors in native cells and tissues
.
Mol. Pharmacol.
67
,
955
964
[PubMed]
54.
Abbracchio
M.P.
and
Burnstock
G.
(
1994
)
Purinoceptors: are there families of P2X and P2Y purinoceptors?
Pharmacol. Ther.
64
,
445
475
[PubMed]
55.
Fields
R.D.
and
Burnstock
G.
(
2006
)
Purinergic signalling in neuron-glia interactions
.
Nat. Rev. Neurosci.
7
,
423
436
[PubMed]
56.
Surprenant
A.
,
Rassendren
F.
,
Kawashima
E.
,
North
R.A.
and
Buell
G.
(
1996
)
The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7)
.
Science
272
,
735
738
[PubMed]
57.
Di Virgilio
F.
,
Dal Ben
D.
,
Sarti
A.C.
,
Giuliani
A.L.
and
Falzoni
S.
(
2017
)
The P2X7 receptor in infection and inflammation
.
Immunity
47
,
15
31
[PubMed]
58.
Harkat
M.
,
Peverini
L.
,
Cerdan
A.H.
,
Dunning
K.
,
Beudez
J.
,
Martz
A.
et al.
(
2017
)
On the permeation of large organic cations through the pore of ATP-gated P2X receptors
.
Proc. Natl. Acad. Sci. U.S.A.
114
,
E3786
E3795
[PubMed]
59.
McCarthy
A.E.
,
Yoshioka
C.
and
Mansoor
S.E.
(
2019
)
Full-length P2X7 structures reveal how palmitoylation prevents channel desensitization
.
Cell
179
,
659e13
670e13
60.
Zhou
M.
,
Du
Y.
,
Aten
S.
and
Terman
D.
(
2021
)
On the electrical passivity of astrocyte potassium conductance
.
J. Neurophysiol.
126
,
1403
1419
[PubMed]
61.
Bartlett
R.
,
Stokes
L.
and
Sluyter
R.
(
2014
)
The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease
.
Pharmacol. Rev.
66
,
638
675
[PubMed]
62.
Gu
B.J.
,
Sluyter
R.
,
Skarratt
K.K.
,
Shemon
A.N.
,
Dao-Ung
L.P.
,
Fuller
S.J.
et al.
(
2004
)
An Arg307 to Gln polymorphism within the ATP-binding site causes loss of function of the human P2X7 receptor
.
J. Biol. Chem.
279
,
31287
31295
[PubMed]
63.
Wiley
J.S.
,
Dao-Ung
L.P.
,
Li
C.
,
Shemon
A.N.
,
Gu
B.J.
,
Smart
M.L.
et al.
(
2003
)
An Ile-568 to Asn polymorphism prevents normal trafficking and function of the human P2X7 receptor
.
J. Biol. Chem.
278
,
17108
17113
[PubMed]
64.
Gu
B.J.
,
Zhang
W.
,
Worthington
R.A.
,
Sluyter
R.
,
Dao-Ung
P.
,
Petrou
S.
et al.
(
2001
)
A Glu-496 to Ala polymorphism leads to loss of function of the human P2X7 receptor
.
J. Biol. Chem.
276
,
11135
11142
[PubMed]
65.
Stokes
L.
,
Fuller
S.J.
,
Sluyter
R.
,
Skarratt
K.K.
,
Gu
B.J.
and
Wiley
J.S.
(
2010
)
Two haplotypes of the P2X(7) receptor containing the Ala-348 to Thr polymorphism exhibit a gain-of-function effect and enhanced interleukin-1beta secretion
.
FASEB J.
24
,
2916
2927
[PubMed]
66.
Adinolfi
E.
,
Cirillo
M.
,
Woltersdorf
R.
,
Falzoni
S.
,
Chiozzi
P.
,
Pellegatti
P.
et al.
(
2010
)
Trophic activity of a naturally occurring truncated isoform of the P2X7 receptor
.
FASEB J.
24
,
3393
3404
[PubMed]
67.
Cheewatrakoolpong
B.
,
Gilchrest
H.
,
Anthes
J.C.
and
Greenfeder
S.
(
2005
)
Identification and characterization of splice variants of the human P2X7 ATP channel
.
Biochem. Biophys. Res. Commun.
332
,
17
27
[PubMed]
68.
Khakh
B.S.
and
North
R.A.
(
2012
)
Neuromodulation by extracellular ATP and P2X receptors in the CNS
.
Neuron
76
,
51
69
[PubMed]
69.
Darmanis
S.
,
Sloan
S.A.
,
Zhang
Y.
,
Enge
M.
,
Caneda
C.
,
Shuer
L.M.
et al.
(
2015
)
A survey of human brain transcriptome diversity at the single cell level
.
Proc. Natl. Acad. Sci. U.S.A.
112
,
7285
7290
[PubMed]
70.
Zhang
Y.
,
Chen
K.
,
Sloan
S.A.
,
Bennett
M.L.
,
Scholze
A.R.
,
O'Keeffe
S.
et al.
(
2014
)
An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex
.
J. Neurosci.
34
,
11929
11947
[PubMed]
71.
Martin
E.
,
Amar
M.
,
Dalle
C.
,
Youssef
I.
,
Boucher
C.
,
Le Duigou
C.
et al.
(
2019
)
New role of P2X7 receptor in an Alzheimer’s disease mouse model
.
Mol. Psychiatry
24
,
108
125
[PubMed]
72.
Carvalho
K.
,
Martin
E.
,
Ces
A.
,
Sarrazin
N.
,
Lagouge-Roussey
P.
,
Nous
C.
et al.
(
2021
)
P2X7-deficiency improves plasticity and cognitive abilities in a mouse model of Tauopathy
.
Prog. Neurobiol.
206
,
102139
[PubMed]
73.
Matute
C.
,
Torre
I.
,
Perez-Cerda
F.
,
Perez-Samartin
A.
,
Alberdi
E.
,
Etxebarria
E.
et al.
(
2007
)
P2X(7) receptor blockade prevents ATP excitotoxicity in oligodendrocytes and ameliorates experimental autoimmune encephalomyelitis
.
J. Neurosci.
27
,
9525
9533
[PubMed]
74.
Pietrowski
M.J.
,
Gabr
A.A.
,
Kozlov
S.
,
Blum
D.
,
Halle
A.
and
Carvalho
K.
(
2021
)
Glial purinergic signaling in neurodegeneration
.
Front Neurol.
12
,
654850
[PubMed]
75.
Illes
P.
,
Khan
T.M.
and
Rubini
P.
(
2017
)
Neuronal P2X7 receptors revisited: do they really exist?
J. Neurosci.
37
,
7049
7062
[PubMed]
76.
Miras-Portugal
M.T.
,
Sebastian-Serrano
A.
,
de Diego Garcia
L.
and
Diaz-Hernandez
M.
(
2017
)
Neuronal P2X7 receptor: involvement in neuronal physiology and pathology
.
J. Neurosci.
37
,
7063
7072
[PubMed]
77.
Bianco
F.
,
Pravettoni
E.
,
Colombo
A.
,
Schenk
U.
,
Moller
T.
,
Matteoli
M.
et al.
(
2005
)
Astrocyte-derived ATP induces vesicle shedding and IL-1 beta release from microglia
.
J. Immunol.
174
,
7268
7277
[PubMed]
78.
Fischer
W.
,
Appelt
K.
,
Grohmann
M.
,
Franke
H.
,
Norenberg
W.
and
Illes
P.
(
2009
)
Increase of intracellular Ca2+ by P2X and P2Y receptor-subtypes in cultured cortical astroglia of the rat
.
Neuroscience
160
,
767
783
[PubMed]
79.
Fumagalli
M.
,
Brambilla
R.
,
D'Ambrosi
N.
,
Volonte
C.
,
Matteoli
M.
,
Verderio
C.
et al.
(
2003
)
Nucleotide-mediated calcium signaling in rat cortical astrocytes: Role of P2X and P2Y receptors
.
Glia
43
,
218
303
[PubMed]
80.
Parvathenani
L.K.
,
Tertyshnikova
S.
,
Greco
C.R.
,
Roberts
S.B.
,
Robertson
B.
and
Posmantur
R.
(
2003
)
P2X7 mediates superoxide production in primary microglia and is up-regulated in a transgenic mouse model of Alzheimer’s disease
.
J. Biol. Chem.
278
,
13309
13317
[PubMed]
81.
Duan
S.
,
Anderson
C.M.
,
Keung
E.C.
,
Chen
Y.
,
Chen
Y.
and
Swanson
R.A.
(
2003
)
P2X7 receptor-mediated release of excitatory amino acids from astrocytes
.
J. Neurosci.
23
,
1320
1328
[PubMed]
82.
Pannicke
T.
,
Fischer
W.
,
Biedermann
B.
,
Schadlich
H.
,
Grosche
J.
,
Faude
F.
et al.
(
2000
)
P2X7 receptors in Muller glial cells from the human retina
.
J. Neurosci.
20
,
5965
5972
[PubMed]
83.
Norenberg
W.
,
Schunk
J.
,
Fischer
W.
,
Sobottka
H.
,
Riedel
T.
,
Oliveira
J.F.
et al.
(
2010
)
Electrophysiological classification of P2X7 receptors in rat cultured neocortical astroglia
.
Br. J. Pharmacol.
160
,
1941
1952
[PubMed]
84.
Oliveira
J.F.
,
Riedel
T.
,
Leichsenring
A.
,
Heine
C.
,
Franke
H.
,
Krugel
U.
et al.
(
2011
)
Rodent cortical astroglia express in situ functional P2X7 receptors sensing pathologically high ATP concentrations
.
Cereb. Cortex
21
,
806
820
[PubMed]
85.
Illes
P.
,
Verkhratsky
A.
,
Burnstock
G.
and
Franke
H.
(
2012
)
P2X receptors and their roles in astroglia in the central and peripheral nervous system
.
Neuroscientist
18
,
422
438
[PubMed]
86.
Kesavan
J.
,
Watters
O.
,
Dinkel
K.
,
Hamacher
M.
,
Prehn
J.H.M.
,
Henshall
D.C.
et al.
(
2021
)
Functional expression of the ATP-gated P2X7 receptor in human iPSC-derived neurons and astrocytes
.
BioRxiv
87.
Murakami
K.
,
Nakamura
Y.
and
Yoneda
Y.
(
2003
)
Potentiation by ATP of lipopolysaccharide-stimulated nitric oxide production in cultured astrocytes
.
Neuroscience
117
,
37
42
[PubMed]
88.
Munoz
F.M.
,
Patel
P.A.
,
Gao
X.
,
Mei
Y.
,
Xia
J.
,
Gilels
S.
et al.
(
2020
)
Reactive oxygen species play a role in P2X7 receptor-mediated IL-6 production in spinal astrocytes
.
Purinergic Signal.
16
,
97
107
[PubMed]
89.
Panenka
W.
,
Jijon
H.
,
Herx
L.M.
,
Armstrong
J.N.
,
Feighan
D.
,
Wei
T.
et al.
(
2001
)
P2X7-like receptor activation in astrocytes increases chemokine monocyte chemoattractant protein-1 expression via mitogen-activated protein kinase
.
J. Neurosci.
21
,
7135
7142
[PubMed]
90.
Bianco
F.
,
Colombo
A.
,
Saglietti
L.
,
Lecca
D.
,
Abbracchio
M.P.
,
Matteoli
M.
et al.
(
2009
)
Different properties of P2X(7) receptor in hippocampal and cortical astrocytes
.
Purinergic Signal.
5
,
233
240
[PubMed]
91.
Jacques-Silva
M.C.
,
Rodnight
R.
,
Lenz
G.
,
Liao
Z.
,
Kong
Q.
,
Tran
M.
et al.
(
2004
)
P2X7 receptors stimulate AKT phosphorylation in astrocytes
.
Br. J. Pharmacol.
141
,
1106
1117
[PubMed]
92.
Walter
L.
,
Dinh
T.
and
Stella
N.
(
2004
)
ATP induces a rapid and pronounced increase in 2-arachidonoylglycerol production by astrocytes, a response limited by monoacylglycerol lipase
.
J. Neurosci.
24
,
8068
8074
[PubMed]
93.
Ballerini
P.
,
Ciccarelli
R.
,
Caciagli
F.
,
Rathbone
M.P.
,
Werstiuk
E.S.
,
Traversa
U.
et al.
(
2005
)
P2X7 receptor activation in rat brain cultured astrocytes increases the biosynthetic release of cysteinyl leukotrienes
.
Int. J. Immunopathol. Pharmacol.
18
,
417
430
[PubMed]
94.
Wei
W.
,
Ryu
J.K.
,
Choi
H.B.
and
McLarnon
J.G.
(
2008
)
Expression and function of the P2X(7) receptor in rat C6 glioma cells
.
Cancer Lett.
260
,
79
87
[PubMed]
95.
Pan
H.C.
,
Chou
Y.C.
and
Sun
S.H.
(
2015
)
P2X7 R-mediated Ca(2+) -independent d-serine release via pannexin-1 of the P2X7 R-pannexin-1 complex in astrocytes
.
Glia
63
,
877
893
[PubMed]
96.
Suadicani
S.O.
,
Brosnan
C.F.
and
Scemes
E.
(
2006
)
P2X7 receptors mediate ATP release and amplification of astrocytic intercellular Ca2+ signaling
.
J. Neurosci.
26
,
1378
1385
[PubMed]
97.
Sanz
J.M.
,
Falzoni
S.
,
Rizzo
R.
,
Cipollone
F.
,
Zuliani
G.
and
Di Virgilio
F.
(
2014
)
Possible protective role of the 489C>T P2X7R polymorphism in Alzheimer's disease
.
Exp. Gerontol.
60
,
117
119
[PubMed]
98.
McLarnon
J.G.
,
Ryu
J.K.
,
Walker
D.G.
and
Choi
H.B.
(
2006
)
Upregulated expression of purinergic P2X(7) receptor in Alzheimer disease and amyloid-beta peptide-treated microglia and in peptide-injected rat hippocampus
.
J. Neuropathol. Exp. Neurol.
65
,
1090
1097
[PubMed]
99.
Martinez-Frailes
C.
,
Di Lauro
C.
,
Bianchi
C.
,
de Diego-Garcia
L.
,
Sebastian-Serrano
A.
,
Bosca
L.
et al.
(
2019
)
Amyloid peptide induced neuroinflammation increases the P2X7 receptor expression in microglial cells, impacting on its functionality
.
Front. Cell Neurosci.
13
,
143
[PubMed]
100.
Lee
H.G.
,
Won
S.M.
,
Gwag
B.J.
and
Lee
Y.B.
(
2011
)
Microglial P2X(7) receptor expression is accompanied by neuronal damage in the cerebral cortex of the APPswe/PS1dE9 mouse model of Alzheimer's disease
.
Exp. Mol. Med.
43
,
7
14
[PubMed]
101.
Di Lauro
C.
,
Bianchi
C.
,
Sebastian-Serrano
A.
,
Soria-Tobar
L.
,
Alvarez-Castelao
B.
,
Nicke
A.
et al.
(
2022
)
P2X7 receptor blockade reduces tau induced toxicity, therapeutic implications in tauopathies
.
Prog. Neurobiol.
208
,
102173
[PubMed]
102.
Chow
S.K.
,
Yu
D.
,
Macdonald
C.L.
,
Buibas
M.
and
Silva
G.A.
(
2010
)
Amyloid beta-peptide directly induces spontaneous calcium transients, delayed intercellular calcium waves and gliosis in rat cortical astrocytes
.
ASN Neuro
2
,
e00026
[PubMed]
103.
Haughey
N.J.
and
Mattson
M.P.
(
2003
)
Alzheimer's amyloid beta-peptide enhances ATP/gap junction-mediated calcium-wave propagation in astrocytes
.
Neuromolecular Med.
3
,
173
180
[PubMed]
104.
Paumier
A.
,
Boisseau
S.
,
Jacquier-Sarlin
M.
,
Pernet-Gallay
K.
,
Buisson
A.
and
Albrieux
M.
(
2022
)
Astrocyte-neuron interplay is critical for Alzheimer's disease pathogenesis and is rescued by TRPA1 channel blockade
.
Brain
145
,
388
405
[PubMed]
105.
Wolosker
H.
,
Balu
D.T.
and
Coyle
J.T.
(
2016
)
The Rise and fall of the d-serine-mediated gliotransmission hypothesis
.
Trends Neurosci.
39
,
712
721
[PubMed]
106.
Mosconi
L.
(
2005
)
Brain glucose metabolism in the early and specific diagnosis of Alzheimer's disease. FDG-PET studies in MCI and AD
.
Eur. J. Nucl. Med. Mol. Imaging
32
,
486
510
[PubMed]
107.
An
Y.
,
Varma
V.R.
,
Varma
S.
,
Casanova
R.
,
Dammer
E.
,
Pletnikova
O.
et al.
(
2018
)
Evidence for brain glucose dysregulation in Alzheimer's disease
.
Alzheimers Dement
14
,
318
329
[PubMed]
108.
Bonvento
G.
,
Oliet
S.H.R.
and
Panatier
A.
(
2022
)
Glycolysis-derived L-serine levels versus PHGDH expression in Alzheimer's disease
.
Cell Metab.
34
,
654
655
[PubMed]
109.
Le Douce
J.
,
Maugard
M.
,
Veran
J.
,
Matos
M.
,
Jego
P.
,
Vigneron
P.A.
et al.
(
2020
)
Impairment of Glycolysis-Derived l-Serine Production in Astrocytes Contributes to Cognitive Deficits in Alzheimer's Disease
.
Cell Metab.
31
,
503e8
517e8
110.
Hefendehl
J.K.
,
LeDue
J.
,
Ko
R.W.
,
Mahler
J.
,
Murphy
T.H.
and
MacVicar
B.A.
(
2016
)
Mapping synaptic glutamate transporter dysfunction in vivo to regions surrounding Abeta plaques by iGluSnFR two-photon imaging
.
Nat Commun.
7
,
13441
[PubMed]
111.
Li
S.
,
Jin
M.
,
Koeglsperger
T.
,
Shepardson
N.E.
,
Shankar
G.M.
and
Selkoe
D.J.
(
2011
)
Soluble Abeta oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors
.
J. Neurosci.
31
,
6627
6638
[PubMed]
112.
Kobayashi
E.
,
Nakano
M.
,
Kubota
K.
,
Himuro
N.
,
Mizoguchi
S.
,
Chikenji
T.
et al.
(
2018
)
Activated forms of astrocytes with higher GLT-1 expression are associated with cognitive normal subjects with Alzheimer pathology in human brain
.
Sci. Rep.
8
,
1712
[PubMed]
113.
Sanz
J.M.
,
Chiozzi
P.
,
Ferrari
D.
,
Colaianna
M.
,
Idzko
M.
,
Falzoni
S.
et al.
(
2009
)
Activation of microglia by amyloid {beta} requires P2X7 receptor expression
.
J. Immunol.
182
,
4378
4385
[PubMed]
114.
Fellin
T.
,
Pozzan
T.
and
Carmignoto
G.
(
2006
)
Purinergic receptors mediate two distinct glutamate release pathways in hippocampal astrocytes
.
J. Biol. Chem.
281
,
4274
4284
[PubMed]
115.
Ficker
C.
,
Rozmer
K.
,
Kato
E.
,
Ando
R.D.
,
Schumann
L.
,
Krugel
U.
et al.
(
2014
)
Astrocyte-neuron interaction in the substantia gelatinosa of the spinal cord dorsal horn via P2X7 receptor-mediated release of glutamate and reactive oxygen species
.
Glia
62
,
1671
1686
[PubMed]
116.
Wu
Z.
,
Guo
Z.
,
Gearing
M.
and
Chen
G.
(
2014
)
Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in an Alzheimer's [corrected] disease model
.
Nat. Commun.
5
,
4159
[PubMed]
117.
Khan
M.T.
,
Deussing
J.
,
Tang
Y.
and
Illes
P.
(
2019
)
Astrocytic rather than neuronal P2X7 receptors modulate the function of the tri-synaptic network in the rodent hippocampus
.
Brain Res. Bull.
151
,
164
173
[PubMed]
118.
Sung
K.
and
Jimenez-Sanchez
M.
(
2020
)
Autophagy in Astrocytes and its Implications in Neurodegeneration
.
J. Mol. Biol.
432
,
2605
2621
[PubMed]
119.
Mathur
R.
,
Ince
P.G.
,
Minett
T.
,
Garwood
C.J.
,
Shaw
P.J.
,
Matthews
F.E.
et al.
(
2015
)
A reduced astrocyte response to beta-amyloid plaques in the ageing brain associates with cognitive impairment
.
PloS ONE
10
,
e0118463
[PubMed]
120.
Lasagna-Reeves
C.A.
and
Kayed
R.
(
2011
)
Astrocytes contain amyloid-beta annular protofibrils in Alzheimer's disease brains
.
FEBS Lett.
585
,
3052
3057
[PubMed]
121.
Davis
N.
,
Mota
B.C.
,
Stead
L.
,
Palmer
E.O.C.
,
Lombardero
L.
,
Rodriguez-Puertas
R.
et al.
(
2021
)
Pharmacological ablation of astrocytes reduces Abeta degradation and synaptic connectivity in an ex vivo model of Alzheimer's disease
.
J. Neuroinflammation
18
,
73
[PubMed]
122.
Pihlaja
R.
,
Koistinaho
J.
,
Malm
T.
,
Sikkila
H.
,
Vainio
S.
and
Koistinaho
M.
(
2008
)
Transplanted astrocytes internalize deposited beta-amyloid peptides in a transgenic mouse model of Alzheimer's disease
.
Glia
56
,
154
163
[PubMed]
123.
Kraft
A.W.
,
Hu
X.
,
Yoon
H.
,
Yan
P.
,
Xiao
Q.
,
Wang
Y.
et al.
(
2013
)
Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice
.
FASEB J.
27
,
187
198
[PubMed]
124.
Furman
J.L.
,
Sama
D.M.
,
Gant
J.C.
,
Beckett
T.L.
,
Murphy
M.P.
,
Bachstetter
A.D.
et al.
(
2012
)
Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease
.
J. Neurosci.
32
,
16129
16140
[PubMed]
125.
Koistinaho
M.
,
Lin
S.
,
Wu
X.
,
Esterman
M.
,
Koger
D.
,
Hanson
J.
et al.
(
2004
)
Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides
.
Nat. Med.
10
,
719
726
[PubMed]
126.
Xiao
Q.
,
Yan
P.
,
Ma
X.
,
Liu
H.
,
Perez
R.
,
Zhu
A.
et al.
(
2014
)
Enhancing astrocytic lysosome biogenesis facilitates Abeta clearance and attenuates amyloid plaque pathogenesis
.
J. Neurosci.
34
,
9607
9620
[PubMed]
127.
Leal
M.C.
,
Dorfman
V.B.
,
Gamba
A.F.
,
Frangione
B.
,
Wisniewski
T.
,
Castano
E.M.
et al.
(
2006
)
Plaque-associated overexpression of insulin-degrading enzyme in the cerebral cortex of aged transgenic tg2576 mice with Alzheimer pathology
.
J. Neuropathol. Exp. Neurol.
65
,
976
987
[PubMed]
128.
Palmer
J.C.
,
Baig
S.
,
Kehoe
P.G.
and
Love
S.
(
2009
)
Endothelin-converting enzyme-2 is increased in Alzheimer's disease and up-regulated by Abeta
.
Am. J. Pathol.
175
,
262
270
[PubMed]
129.
de Calignon
A.
,
Polydoro
M.
,
Suarez-Calvet
M.
,
William
C.
,
Adamowicz
D.H.
,
Kopeikina
K.J.
et al.
(
2012
)
Propagation of tau pathology in a model of early Alzheimer's disease
.
Neuron
73
,
685
697
[PubMed]
130.
Martini-Stoica
H.
,
Cole
A.L.
,
Swartzlander
D.B.
,
Chen
F.
,
Wan
Y.W.
,
Bajaj
L.
et al.
(
2018
)
TFEB enhances astroglial uptake of extracellular tau species and reduces tau spreading
.
J. Exp. Med.
215
,
2355
2377
[PubMed]
131.
Perea
J.R.
,
Lopez
E.
,
Diez-Ballesteros
J.C.
,
Avila
J.
,
Hernandez
F.
and
Bolos
M.
(
2019
)
Extracellular monomeric tau is internalized by astrocytes
.
Front. Neurosci.
13
,
442
[PubMed]
132.
Puangmalai
N.
,
Bhatt
N.
,
Montalbano
M.
,
Sengupta
U.
,
Gaikwad
S.
,
Ventura
F.
et al.
(
2020
)
Internalization mechanisms of brain-derived tau oligomers from patients with Alzheimer's disease, progressive supranuclear palsy and dementia with Lewy bodies
.
Cell Death Dis.
11
,
314
[PubMed]
133.
Rauch
J.N.
,
Luna
G.
,
Guzman
E.
,
Audouard
M.
,
Challis
C.
,
Sibih
Y.E.
et al.
(
2020
)
LRP1 is a master regulator of tau uptake and spread
.
Nature
580
,
381
385
[PubMed]
134.
Reid
M.J.
,
Beltran-Lobo
P.
,
Johnson
L.
,
Perez-Nievas
B.G.
and
Noble
W.
(
2020
)
Astrocytes in tauopathies
.
Front Neurol.
11
,
572850
[PubMed]
135.
Diaz-Hernandez
J.I.
,
Gomez-Villafuertes
R.
,
Leon-Otegui
M.
,
Hontecillas-Prieto
L.
,
Del Puerto
A.
,
Trejo
J.L.
et al.
(
2012
)
In vivo P2X7 inhibition reduces amyloid plaques in Alzheimer's disease through GSK3beta and secretases
.
Neurobiol. Aging
33
,
1816
1828
[PubMed]
136.
Ruan
Z.
,
Delpech
J.C.
,
Venkatesan Kalavai
S.
,
Van Enoo
A.A.
,
Hu
J.
,
Ikezu
S.
et al.
(
2020
)
P2RX7 inhibitor suppresses exosome secretion and disease phenotype in P301S tau transgenic mice
.
Mol. Neurodegener
15
,
47
[PubMed]
137.
Takenouchi
T.
,
Fujita
M.
,
Sugama
S.
,
Kitani
H.
and
Hashimoto
M.
(
2009
)
The role of the P2X7 receptor signaling pathway for the release of autolysosomes in microglial cells
.
Autophagy
5
,
723
724
[PubMed]
138.
Takenouchi
T.
,
Nakai
M.
,
Iwamaru
Y.
,
Sugama
S.
,
Tsukimoto
M.
,
Fujita
M.
et al.
(
2009
)
The activation of P2X7 receptor impairs lysosomal functions and stimulates the release of autophagolysosomes in microglial cells
.
J. Immunol.
182
,
2051
2062
[PubMed]
139.
Kim
J.E.
,
Ko
A.R.
,
Hyun
H.W.
,
Min
S.J.
and
Kang
T.C.
(
2018
)
P2RX7-MAPK1/2-SP1 axis inhibits MTOR independent HSPB1-mediated astroglial autophagy
.
Cell Death Dis.
9
,
546
[PubMed]
140.
Lee
D.S.
and
Kim
J.E.
(
2020
)
P2 x 7 receptor inhibits astroglial autophagy via regulating FAK- and PHLPP1/2-Mediated AKT-S473 phosphorylation following kainic acid-induced seizures
.
Int. J. Mol. Sci.
21
,
6476
,
[PubMed]
141.
Ryu
H.J.
,
Kim
J.E.
,
Yeo
S.I.
and
Kang
T.C.
(
2011
)
p65/RelA-Ser529 NF-kappaB subunit phosphorylation induces autophagic astroglial death (Clasmatodendrosis) following status epilepticus
.
Cell. Mol. Neurobiol.
31
,
1071
1078
[PubMed]
142.
Kim
J.E.
,
Ryu
H.J.
,
Yeo
S.I.
and
Kang
T.C.
(
2011
)
P2X7 receptor differentially modulates astroglial apoptosis and clasmatodendrosis in the rat brain following status epilepticus
.
Hippocampus
21
,
1318
1333
[PubMed]
143.
Holmes
B.B.
,
DeVos
S.L.
,
Kfoury
N.
,
Li
M.
,
Jacks
R.
,
Yanamandra
K.
et al.
(
2013
)
Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds
.
Proc. Natl. Acad. Sci. U.S.A.
110
,
E3138
E3147
[PubMed]
144.
Merezhko
M.
,
Brunello
C.A.
,
Yan
X.
,
Vihinen
H.
,
Jokitalo
E.
,
Uronen
R.L.
et al.
(
2018
)
Secretion of tau via an unconventional non-vesicular mechanism
.
Cell Rep.
25
,
2027e4
2035e4
145.
Mankus
C.
,
Chi
C.
,
Rich
C.
,
Ren
R.
and
Trinkaus-Randall
V.
(
2012
)
The P2X(7) receptor regulates proteoglycan expression in the corneal stroma
.
Mol. Vis.
18
,
128
138
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).