ArticlePDF AvailableLiterature Review

Integrated Brain Circuits: Astrocytic Networks Modulate Neuronal Activity and Behavior

Authors:

Abstract

The past decade has seen an explosion of research on roles of neuron-astrocyte interactions in the control of brain function. We highlight recent studies performed on the tripartite synapse, the structure consisting of pre- and postsynaptic elements of the synapse and an associated astrocytic process. Astrocytes respond to neuronal activity and neurotransmitters, through the activation of metabotropic receptors, and can release the gliotransmitters ATP, d-serine, and glutamate, which act on neurons. Astrocyte-derived ATP modulates synaptic transmission, either directly or through its metabolic product adenosine. d-serine modulates NMDA receptor function, whereas glia-derived glutamate can play important roles in relapse following withdrawal from drugs of abuse. Cell type-specific molecular genetics has allowed a new level of examination of the function of astrocytes in brain function and has revealed an important role of these glial cells that is mediated by adenosine accumulation in the control of sleep and in cognitive impairments that follow sleep deprivation.
Integrated Brain Circuits: Astrocytic Networks Modulate
Neuronal Activity and Behavior
Michael M. Halassa1,2 and Philip G. Haydon3
Philip G. Haydon: philip.haydon@tufts.edu
1 Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts 02114
2 Department of Psychiatry, McLean Hospital, Belmont, Massachusetts 02478
3 Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
02111
Abstract
The past decade has seen an explosion of research on roles of neuron-astrocyte interactions in the
control of brain function. We highlight recent studies performed on the tripartite synapse, the
structure consisting of pre- and postsynaptic elements of the synapse and an associated astrocytic
process. Astrocytes respond to neuronal activity and neuro-transmitters, through the activation of
metabotropic receptors, and can release the gliotransmitters ATP, D-serine, and glutamate, which
act on neurons. Astrocyte-derived ATP modulates synaptic transmission, either directly or through
its metabolic product adenosine. D-serine modulates NMDA receptor function, whereas glia-
derived glutamate can play important roles in relapse following withdrawal from drugs of abuse.
Cell type–specific molecular genetics has allowed a new level of examination of the function of
astrocytes in brain function and has revealed an important role of these glial cells that is mediated
by adenosine accumulation in the control of sleep and in cognitive impairments that follow sleep
deprivation.
Keywords
sleep; ATP; adenosine; NMDA; astrocyte; synapse
INTRODUCTION
In 1895 Santiago Ramóny Cajal proposed that astrocytes, the major subtype of glial cell in
the brain, control sleep and waking states. He specifically proposed that astrocytic processes
are electrical insulators that, when extended between neurons, act as circuit breakers to
facilitate sleep but, when retracted, allow neuronal circuits to communicate, facilitating
wakefulness (1). Now, more than a century after Cajal’s proposal, we know that his intuition
was, in part, correct; astrocytes have an intimate structural and functional association with
neurons and, by virtue of their complex physiology, are able to modulate behaviors such as
sleep (2).
Copyright © 2010 by Annual Reviews. All rights reserved
DISCLOSURE STATEMENT
The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the
objectivity of this review.
NIH Public Access
Author Manuscript
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
Published in final edited form as:
Annu Rev Physiol
. 2010 March 17; 72: 335–355. doi:10.1146/annurev-physiol-021909-135843.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Cajal was the first to systematically study astrocytes from a structural standpoint (1), and
until very recently, our view of astrocytic morphology has been based both on Cajal’s metal
impregnation methods and on glial fibrillary acidic protein (GFAP) staining. Advances in
cellular labeling and imaging technologies showed that astrocytic morphology is far more
complicated than previously thought. By filling single astrocytes with fluorescent dyes,
researchers in one study showed that GFAP staining reveals only 15% of the astrocytic
volume and that astrocytes extend fine processes that occupy the surrounding neuropil (3).
Astrocytes occupy nonoverlapping spatial territories in which a single astrocyte contacts
hundreds of neuronal processes and multiple neuronal cell bodies (3, 4). The processes of
one astrocyte contact tens of thousands of synapses, with more than 50% of hippocampal
excitatory synapses, for example, being closely opposed to an astrocytic process (5) at a
structure termed the tripartite synapse to recognize the structural and functional relationship
between the astrocyte and the pre- and postsynaptic terminals (6). Astrocytes are also
intimately associated with the cerebral microvasculature, onto which they extend several
endfeet (7). By being strategically positioned between synapses and blood vessels,
astrocytes are thought to be mediators of neurovascular coupling, the process by which
neuronal activity is coupled to cerebral blood flow and the cellular substrate of functional
brain imaging (8–12).
Astrocytes interact with neurons at multiple spatial and temporal scales. By controlling the
ionic and metabolic environment of the neuropil, astrocytes can dramatically impact
neuronal activity. In addition, astrocytes listen and talk to synapses via regulated pathways
of transmitter release (13, 14). The use of molecular genetics has revolutionized the study of
astrocytic physiology and, as we discuss below, has provided an unprecedented
understanding of how these cells impact brain function at the levels of synapses, circuits,
and behavior. The view that the brain is a collection of integrated circuits of astrocytes and
neurons that control thought and behavior may not only enhance our understanding of this
fascinating organ but also have many implications for the treatment of neurological and
psychiatric disorders.
ASTROCYTIC MEMBRANE PROPERTIES SUPPORT NEURONAL
PHYSIOLOGY
Although not equipped with the cellular machinery necessary for generating action
potentials, an astrocyte exhibits changes in its electrical properties that are essential for
supporting normal neuronal activity. Astrocytes express inward rectifier K+ (Kir) channels
(15), which maintain the astrocytic membrane potential close to the equilibrium potential of
K+. Thus, when extracellular K+ rises, K+ ions flow into astrocytes through these inward
rectifiers. Astrocyte-specific knockout of Kir4.1, the major Kir channel in these glia, results
in seizure activity and premature death (16). Astrocytes also express Ca2+-activated
potassium channels (BK channels), which allow for coupling of astrocytic Ca2+ signaling
(discussed below) to the release of K+ from astrocytic endfeet onto blood vessels (17).
Astrocytes express a number of electrogenic neurotransmitter transporters such as glutamate
(18), GABA (19), and glycine (20). Not only is the uptake of transmitters necessary for
maintaining the fidelity of synaptic transmission, but this process is part of a cycle that
provides presynaptic terminals with a renewable source of these transmitters. For example,
when glutamate is taken up by astrocytes, the enzyme glutamine synthetase (GS) converts it
to glutamine (21). Astrocytes then release glutamine onto both glutamatergic and
GABAergic terminals. Because GABAergic terminals have a limited glutamine store,
pharmacological inhibition of GS results in failure of inhibitory synaptic transmission under
conditions of elevated neuronal activity (22). One consequence of such failure can be the
development of seizures (23, 24). Another can be the dysregulation of neuronal coding and
Halassa and Haydon Page 2
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
information transfer among networks; because GABAergic inhibition acts as an oscillatory
timing signal for neuronal network activity (25–27), failure of inhibition profoundly affects
information processing in the brain and is thought to contribute to a number of psychiatric
states (28, 29). Thus, by being the sole supplier of glutamine to synapses, astrocytes have the
potential to shape neuronal activity and information processing by controlling neuronal
inhibition.
ASTROCYTES FORM METABOLIC NETWORKS THAT RESPOND TO
EXTERNAL STIMULI
Astrocytes play essential roles in brain energy homeostasis and metabolism (30). These glial
cells express transporters that mediate the uptake of glucose from cerebral microvessels. An
important structural/signaling feature of astrocytes is their extensive intercellular coupling,
mediated mainly by connexin 43– and connexin 30–containing gap junctions (31). Such gap
junctions allow for the diffusion of glucose among many astrocytes (32), and the genetic
ablation of these connexins profoundly impacts neuronal activity and metabolism.
Glucose, which is taken up by the endfeet of the astrocytes, can diffuse through the
multicellular astrocytic network through gap junctions. This process is regulated because the
diffusion of fluorescent glucose analogs among hippocampal astrocytes is enhanced by
neuronal activity (32). This process is sensitive to AMPA receptor blockade, suggesting the
involvement of postsynaptic mechanisms in recruiting astrocytic metabolic coupling. Under
conditions of high neuronal activity and low metabolic supply, astrocytic metabolic coupling
is essential for maintaining glutamatergic synaptic transmission. Delivering glucose
specifically to a single astrocyte rescues synaptic activity when extracellular glucose is
lowered, but this process is inhibited by astrocyte-specific genetic ablation of connexin 43
and connexin 30.
Following its uptake by astrocytes, glucose is either stored as glycogen or metabolized to
lactate (33). Astrocytic glycogen metabolism is regulated by neuronal mechanisms. For
example, under conditions of increased metabolic demand, noradrenaline activates astrocytic
cAMP signaling, resulting in the breakdown of intra-cellular glycogen. The resulting
glucose is further metabolized by astrocytes to lactate and is released to the extracellular
space, where it is taken up by neurons (34). This metabolic relationship between neurons
and astrocytes, known as the lactate shuttle, is a highly dynamic process that can be
regulated by a number of physiological processes, including the sleep/wake cycle (30).
Thus, as we discuss below, the function of astrocytes as modulators of sleep in mammals
may link their metabolic functions to their neuromodulatory roles.
ASTROCYTES EXHIBIT Ca2+ EXCITABILITY
Astrocytes respond to transmitter spillover from nearby synaptic activity with an elevation
of their Ca2+ level. Neuronally released transmitters can engage astrocytic metabotropic
receptors, a subset of which couples through Gq to phospholipase C (PLC), resulting in the
accumulation of diacylglycerol (DAG) and IP3. IP3 binds to its receptor and causes the
release of Ca2+ from internal stores. The activation of this signaling pathway has now been
demonstrated to occur in vivo in response to sensory stimulation. For example, stimulation
of an individual mouse whisker causes Ca2+ signals in astrocytes located in the
corresponding cortical barrel. This response, which has a latency of ~3 s, is dependent on
activation of astrocytic metabotropic glutamate receptors (35). In the visual cortex of the
ferret, astrocytic activity is driven by visual gratings that trigger the activity of nearby
neurons, suggesting a spatial alignment between these two cell types (36). Interestingly,
astrocytes show sharper tuning to orientation and spatial frequency than do neurons, and a
Halassa and Haydon Page 3
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
group of 2 to <10 astrocytes respond to a particular one-stimulus orientation, suggesting a
network organization of visual cortical astrocytes (36). Because sensory tuning of cortical
neurons is shaped by the interplay between excitation and inhibition (37), this experiment
raises the possibility that astrocytes are tracking the output of these cortical pyramidal
neurons rather than their input. Pharmacological agents that inhibit astrocytic Ca2+ signaling
while leaving the neuronal response intact, such as increasing the level of isofluorane
anesthesia, also inhibit the activity-dependent vascular response in the visual cortex (36).
Interestingly, the astrocytic response is inhibited not by metabotropic glutamate receptor
antagonists in the visual cortex of the ferret, but instead by glutamate transporter inhibitors,
suggesting that the sensory-evoked response of astrocytes in the visual cortex is distinct
from that in the barrel cortex. However, caution must be exerted in interpreting such an
observation because the resulting increase in ambient glutamate may lead to desensitization
of metabotropic receptors.
Similar to the findings from the visual cortex of the ferret, olfactory bulb astrocytes in the
mouse respond to odor-evoked synaptic input, and their response coincides with the vascular
response (38). These converging findings from different brain regions and different species
strongly link astrocytic Ca2+ signaling to the activity-dependent neurovascular coupling.
Interestingly, recent findings have shown decoupling of cerebral blood flow from local
neuronal activity (39) under certain conditions, suggesting a nonneuronal mediator of the
cerebrovascular signal and that under such conditions the vascular response plays an
anticipatory role in brain state changes, rather than act as a follower for neuronal activity.
ASTROCYTES RELEASE CHEMICAL TRANSMITTERS TO MODULATE
NEURONAL ACTIVITY
In addition to regulating neuronal signaling by controlling the ionic environment of the
neuropil and controlling the supply of neuro-transmitters to synapses, astrocytes can directly
activate neuronal receptors by releasing gliotransmitters. Gliotransmission is an umbrella
term that includes the release of many chemical transmitters from astrocytes, including
classical transmitters, peptides, chemokines, and cytokines, through a number of different
mechanisms.
Exocytosis mediates one of the better-characterized pathways of gliotransmission. Regulated
exocytosis is dependent on the docking and fusion of vesicles to the plasma membrane,
which is mediated via the formation of the soluble N-ethylmaleimide-sensitive fusion
protein attachment protein receptor (SNARE) complex. Astrocytes express the core
machinery proteins involved in forming the SNARE complex, such as synaptobrevin II (40,
41) [and its homolog cellubrevin (42)] and SNAP-23 (40, 41), and ancillary proteins to this
complex, such as Munc 18 (40, 41), complexin 2 (40, 41), and synaptotagmin IV (43).
Molecular genetic manipulations directed at the SNARE complex and to perturb
synaptotagmin IV have shown their requirement for Ca2+-regulated vesicular
gliotransmission.
In cultured astrocytes, SNARE proteins colocalize with a number of vesicular organelles,
including small vesicles positive for vesicular glutamate transporters (VGlut 1–3) (40–42),
ATP-storing vesicles (44, 45), and D-serine-containing vesicles (46), suggesting the
involvement of vesicular mechanisms in the release of these gliotransmitters. The size of
these vesicular organelles ranges between 30 and 100 nm in diameter in cultured astrocytes,
and immunoelectronmicroscopy (IEM) in situ has shown the existence of 30-nm clear
astrocytic vesicles opposed to presynaptic terminals (47). This latter demonstration strongly
supports the existence of a vesicular pathway of gliotransmission in the intact brain.
Halassa and Haydon Page 4
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
ASTROCYTES RELEASE GLUTAMATE, WHICH MODULATES NEURONAL
NMDA RECEPTOR FUNCTION
Glutamate was the first gliotransmitter shown to be released by astrocytes. The first
evidence for its release came from high-performance liquid chromatography (HPLC) in
cultured astrocytes, where bradykinin-evoked Ca2+ signaling was followed by the release of
glutamate from these cells (48). Astrocytic glutamate was also detected by cocultured
neurons, where bradykinin evoked N-methyl-D-aspartate (NMDA)-dependent Ca2+
transients in neurons only in the presence of astrocytes. In addition to vesicular
gliotransmission of glutamate, other pathways of glutamate gliotransmission have been
proposed, including reversal of uptake by glutamate transporters (49), anion channel–
mediated efflux (50), release by the glutamate/cystine antiporter (51), diffusion through
ionotropic purinergic receptors (52), and hemichannel (connexin/pannexin)-mediated efflux
(53). Although all pathways can mediate release of glutamate, it is extremely important to
identify the conditions that activate these release pathways so that it will become possible to
understand when they are utilized in the nervous system.
Although much of our discussion here focuses around Ca2+-regulated exocytosis, we discuss
one transporter, the cystine/glutamate transporter, as it has great relevance for addiction and
brain tumors. Astrocytes express a cystine/glutamate transporter that is responsible for the
uptake of cystine, which is subsequently used in the synthesis of glutathione. Following a
subject’s withdrawal from cocaine administration, the extracellular levels of glutamate in the
nucleus accumbens fall due to reduced activity of this transporter, which exchanges
glutamate for cystine. To determine whether the reduced activity of this transporter is
important for relapse and reinstatement of drug-seeking behavior following subsequent
exposure to cocaine, Kalivas’s group experimentally activated this transporter while
monitoring behavior. Either direct infusion of cystine or systemic administration of N-
acetylcysteine, which activates the transporter and increases extracellular glutamate,
prevented relapse (54).
The same cystine/glutamate transporter is receiving considerable scrutiny in the study of
glioblastoma multiforme. Glioma cells release excess glutamate as a consequence of the
activation of the cystine/glutamate transporter. Because glioma do not express Na+-
dependent glutamate transporters, which normally take up glutamate, the presence of
gliomal cells leads to enhanced extracellular glutamate, which causes exocytotic death of
neurons, which presumably helps clear space for tumor growth. Cystine uptake is rate
limiting the production of glutathione. Sontheimer’s group has investigated the importance
of cystine/glutamate transport in tumor growth and found that inhibition of the transporter
leads to a rapid depletion of glutathione and a reduction in tumor growth (55). On the basis
of these results, clinical trials are under way, with the objectives of stimulating the
transporter to prevent relapse and inhibiting the transporter as a treatment for brain tumors.
Ca2+-regulated glutamatergic gliotransmission targets neuronal extrasynaptic NR2B-
containing NMDA receptors, resulting in changes in neuronal excitability and modulating
synaptic transmission. Two forms of modulation have been observed, discrete transient
slow-inward currents (SICs) and a tonic modulation of synaptic transmission mediated
through NMDA receptors. SICs have been observed in physiological conditions of normal
extracellular Mg2+ in brain slices isolated from the thalamus, hippocampus, cortex, nucleus
accumbens, and olfactory bulb (56–62). These events have distinct kinetics that
distinguishes them from excitatory postsynaptic currents (EPSCs), and a unique
pharmacological signature that is consistent with their nonneuronal origin (57). SICs are not
blocked by tetrodotoxin (TTX) or perfusion with tetanus toxin (TeNT) at a concentration
that results in inhibition of EPSCs (57). Whole-cell patch clamp of hippocampal pyramidal
Halassa and Haydon Page 5
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
neurons has shown that these events can occur synchronously in two neurons within 100 μm
from one another, suggesting that a single astrocyte can synchronize a group of neurons it
contacts via glutamatergic gliotransmission (4). Several groups have shown that, in addition
to an increase in SICs, glutamatergic gliotransmission results in an increase in the frequency
of EPSCs in nearby neurons (47, 63) or can tonically activate postsynaptic NMDA receptors
(64). Astrocytic modulation of synaptic transmission is dependent on a vesicular mechanism
because it is attenuated by the dialysis of the light chain of TeNT into individual astrocytes
and because small-electron lucent vesicles are apposed to astrocytic membranes adjacent to
presynaptic NR2B receptors that mediate the synaptic modulation (47).
THE DEVIL IS IN THE DETAILS: DISCREPANCIES IN THE FIELD
Although it is clear that glutamatergic gliotransmission occurs under physiological
conditions in situ (57, 59), this research is now entering a phase in which it is necessary to
understand the details of the process so that we may gain a thorough understanding of its
function in vivo. There are several questions that need to be confronted in order to gain a
more thorough appreciation of the significance of glutamatergic gliotransmission. Here we
discuss three prominent concerns in the field.
First is the neurochemical concern: Because astrocytes metabolize glutamate to glutamine,
there may be insufficient residual glutamate for gliotransmission. However, two arguments
indicate that this is not the case: (a) Because astrocytes are living, there is sufficient residual
glutamate for protein synthesis despite the conversion of glutamate to glutamine by the
enzyme glutamine synthetase. (b) The Km of vesicular glutamate transporters is lower than
that of glutamine synthetase, providing the opportunity for glutamate transport into vesicles.
Glutamine synthetase has a Km of ~7.0 mM for glutamate (65). Vesicular glutamate
transporters have Kms of ~0.6–4.7 mM for glutamate (66–68), indicating that these
molecules can package glutamate in astrocytic vesicles under physiological conditions.
Second, it is important to determine which molecules are expressed in astrocytes in vivo and
whether they are sufficient to support gliotransmission. Numerous approaches have been
used to study glutamate, and the majority support the idea that astrocytes contain the
necessary machinery for glutamatergic gliotransmission. Studies performed in cell culture,
in brain slices, with acutely isolated astrocytes, and with tissue sections provide compelling
support for the presence of vesicular machinery for glutamatergic gliotransmission. Studies
in cell culture have provided the highest resolution for the study of glutamatergic
gliotransmission. To name a few, astrocytes both in vitro and in vivo express vesicular
glutamate transporters (69, 70), SNARE proteins that are necessary for membrane fusion, as
well as synaptotagmin IV (43, 69). Because of the concern that cultured cells may express
aberrant proteins, considerable work has been performed in vivo and with freshly isolated
astrocytes. Such research is in agreement with the detection of the message (based on RT-
PCR) and the protein. However, in contrast, two studies, each using microarrays, have not
detected the message for vesicular glutamate transporters (71, 72). The reason for this
apparent discrepancy is not clear. However, it is important to appreciate some of the
concerns about the interpretation of microarray data. One pressing issue is the lack of
consensus in data acquired by commercially available microarray platforms (73) and the
high variability in relative gene expression profiles obtained from different laboratories
performing seemingly identical experiments (74). This issue is particularly important in light
of the microarray data, which report a lack of enrichment of Entpd2, a 5 ectonucleotidase
gene, in astrocytes (71), disagreeing with decades of biochemical and immunohistochemical
findings that show that this protein is expressed primarily by astrocytes (75). Because
microarray studies set cut-off criteria for the number of copies, a gene must be considered
“enriched” in a specific cell type, and such studies run the risk of overlooking genes with a
Halassa and Haydon Page 6
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
relatively low abundance. Certain neuronal genes, such as β-neurexin, have low message
abundance but long-lived protein expression, which makes them particularly resistant to
RNAi-mediated gene knockdown (76). In contrast, the more sensitive method of RT-PCR
has detected these messages. However, RT-PCR can also be criticized, although in contrast
to microarrays, on the basis of its high sensitivity and potential to amplify contaminating
mRNA. Perhaps the most critical observation is based on the use of IEM, which
demonstrates the presence of VG-LUT on astrocytic vesicles adjacent to synapses (69).
Further work is required to determine the reasons for such discrepancies.
A third concern is that astrocytic Ca2+ signals regulate the dilatation/constriction of
microvessels as well as gliotransmission. A priori one would envision that it is important to
have layers of control mechanisms that would allow differential signaling to these two
targets—the vasculature and synapses. Considerable evidence supports the presence of
localized Ca2+ signals within a subregion of an individual astrocyte. Such microdomain
Ca2+ signals would permit local synaptic modulation without regulating the vasculature,
which requires a Ca2+ signal to propagate along the process of the astrocyte that links to the
endfoot. When a cell-wide Ca2+ signal that will regulate the vasculature is evoked, will
gliotransmission necessarily be activated? Perhaps not. Although a wealth of experimental
data in tissue culture and in brain slice preparations supports a model in which astrocytic
Ca2+ signaling is necessary and sufficient for glutamate release, recent studies show that
there are conditions in which an astrocytic Ca2+ signal does not initiate gliotransmission.
Despite the ability of astrocytic flash photolysis of caged Ca2+ and caged IP3 to evoke SICs
(57) and increases in the frequency of EPSCs (63) in nearby neurons, a recent study has
shown that the regulation of this process is more complex than initially appreciated because
not all agonist-induced Ca2+ signals trigger glutamatergic gliotransmission (61) (see below
for a discussion).
Molecular genetic techniques have been used to express novel G protein–coupled proteins
(GPCRs) in astrocytes to ask whether their selective activation triggers glutamatergic
gliotransmission (77). One of these receptors, MrgA1, is normally expressed by dorsal root
ganglion neurons but not expressed in the central nervous system (CNS). Expression of this
receptor in astrocytes results in Ca2+ transients in these glia in response to the peptide ligand
FLRFa. However, despite a robust volume-averaged Ca2+ signal, astrocyte-dependent
modulation of neighboring neurons was not detected. The use of molecular genetics to
introduce a foreign receptor into astrocytes for the study of receptor-activated
gliotransmission is highly innovative but is not without potential concerns. For example, the
expression of a foreign receptor, RASSL, in astrocytes by the same group caused
hydrocephalus, even in the absence of the foreign ligand (78). Whether the expression of the
foreign receptor MrgA1, which was used to activate astrocytic Ca2+ signals, perturbed the
physiological state of the astrocyte is not known. However, it is of concern that this
publication does not report the use of doxycycline to prevent transgene expression during
development, raising the possibility that the inability of this laboratory to reproduce their
earlier results and those of others may be a product of the experimental approach. Further
support for this possibility is that, in the same study, photolysis of caged IP3 was shown
capable of evoking gliotransmission.
An alternative interpretation is that there is more to a Ca2+ signal than just its amplitude.
Support for this possibility is provided by the work of Khakh’s laboratory, which has shown
a differential coupling between distinct astrocytic receptors and gliotransmission. Although
the activation of two astrocyte-enriched receptors, PAR-1 and P2YR, resulted in Ca2+
signals of similar amplitudes but slightly different kinetics, only PAR-1 receptor activation
triggered SICs in nearby neurons (61). Evidence for an astrocytic origin of this process is
provided by dialysis of the Ca2+ chelator BAPTA into the astrocytic syncytium, which
Halassa and Haydon Page 7
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
inhibits PAR-1-induced SICs. The optical activation of surface-expressed ion channels in
astrocytes can evoke the release of ATP from astrocytes in vivo, raising the possibility that
astrocytes use different sources of Ca2+ to support different modes of gliotransmission. The
discovery that astrocytes express a number of TRP channels (79) supports the possibility
that influx of Ca2+ from the extracellular space may support certain forms of
gliotransmission. These studies raise the exciting possibility of distinct astrocytic Ca2+
signals, ones that couple with gliotransmission and others that may mediate other processes
such as the control of the vasculature.
ASTROCYTES RELEASE D-SERINE, WHICH MODULATES NEURONAL
NMDA RECEPTOR FUNCTION
NMDA receptor gating is regulated by several signals. Although glutamate binding is
essential for NMDA receptor activation, ion permeation requires coincident depolarization.
This coincident-detector property of the NMDA receptor is dependent on Mg2+ ions
blocking the receptor pore at negative membrane potentials. In addition, receptor activity is
regulated by a coagonist binding site, the so-called glycine binding site (80). In many brain
areas D-serine is an endogenous ligand for this site. D-serine is synthesized by the enzyme
serine racemase, which is expressed predominantly by astrocytes (81). Astrocytes release D-
serine-containing vesicles in a Ca2+-dependent manner (46), and this form of
gliotransmission is thought to regulate NMDA receptor function and synaptic plasticity (82).
Long-term depression (LTD) and long-term potentiation (LTP) are two opposing forms of
synaptic plasticity requiring different degrees of NMDA receptor activation. The innovative
use of the hypothalamus as a model for astrocyte-neuron interaction has provided
considerable insight into the role of D-serine in synaptic plasticity. For example, in the
supraoptic nucleus, astrocytic ensheathment of synapses is reduced during lactation (82). As
a consequence, less D-serine is provided to NMDA receptors, resulting in a switch in the
ability of a neuronal stimulus to induce plasticity. Virgin rodents with a higher degree of
astrocytic coverage of synapses and, thus, relatively high synaptic D-serine levels exhibit
LTP, whereas lactating rodents with reduced synaptic coverage by astrocytes, D-serine
levels, and NMDA receptor activation exhibit LTD. Thus, the degree of astrocyte-induced
D-serine-dependent coactivation of the NMDA receptor confers metaplasticity to the
synapse. Given that astrocytic processes are highly dynamic and capable of extending and
retracting on the timescale of minutes (83, 84), an exciting possibility is that this form of
astrocyte-induced metaplasticity is a widespread process in the CNS.
THE RELEASE OF ATP BY ASTROCYTES
Shortly after the observation that astrocytes can release glutamate in culture, several groups
demonstrated that ATP can also be released from astrocytes and that it mediates coupling
between astrocytes at least in culture. In culture and in vivo, elevation of the Ca2+ signal
within one astrocyte can lead to a Ca2+ wave that propagates through the coupled glial
network. High-resolution cell culture studies showed that ATP is the signal mediating this
Ca2+ wave (85). Indeed, considerable evidence shows that ATP is a significant extracellular
signaling molecule that is utilized by astrocytes to signal with one another as well as to
neurons. Below, we discuss astrocytic purinergic release mechanisms, roles of purines in
synaptic modulation, as well as the recently discovered role of an astrocytic ATP metabolic
product, adenosine, in the modulation of sleep phenotypes.
Incubation of astrocyte cultures with styryl dye results in labeling of a recycling pool of
vesicles that include lysosomes (44, 86). Although traditionally thought of as organelles of
degradation, lysosomes of a number of cells, including melanocytes and hematopoietic cells,
are secretion competent (87). In one study, lysosomes were imaged in living astrocytes by
Halassa and Haydon Page 8
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
transfecting an EGFP-tagged version of the lysosomal marker CD-63. This fusion protein,
colocalized with a number of styryl dyes, including FM1-43 and FM2-10, but did not
colocalize with vesicular glutamate transporters, suggesting that these organelles do not
support glutamatergic gliotransmission (44). Astrocytic stimulation by ATP or glutamate
resulted in partial destaining of these CD-63+, FM2-10-labeled puncta, suggesting a kiss-
and-run mode of fusion for lysosomes. Furthermore, incubating astrocytes with the cell-
permeable Ca2+ chelator BAPTA-AM inhibited lysosomal fusion, suggesting a Ca2+-
dependent mechanism for lysosomal exocytosis by astrocytes. In another study, astrocytes
were labeled with FM4-64 in an activity-independent manner, and the resulting puncta
colocalized with the lysosomal marker lysotracker and with a number of vesicular SNARE
proteins, including synaptobrevin II and cellubrevin. This study also showed a Ca2+-
dependent mode of exocytosis of astrocytic lysosomes (86).
What, if any, sort of gliotransmission do lysosomes support? Biochemical and imaging
experiments show that these organelles are highly enriched in ATP. Subcellular
fractionation experiments revealed that the highest levels of ATP are found in the fractions
containing the lysosomal markers LAMP-1 and β-hexosaminidase. In contrast, confocal
imaging showed that incubation of astrocytes with Mant-ATP, a fluorescent ATP analog,
results in colocalization with EGFP-CD63 and with FM2-10 (44). Interestingly, astrocytes
appear to be highly enriched in Bloc1s1, a gene involved in lysosome biogenesis (71),
raising the possibility that astrocytes require a constant generation of this organelle for
continuous release of purines.
A previous study has shown that ATP is enriched in astrocytic dense-core vesicles that are
positive for secreteogranin II (45). This study also showed that the release of ATP from
astrocytes is Ca2+ dependent and is inhibited by TeNT. Thus, the evidence is strong for a
Ca2+-dependent, vesicular pathway of ATP release from astrocytes. However, whether such
release is predominantly mediated by lysosomes or dense-core vesicles is still not clear. The
recent discovery of the vesicular nucleotide transporter (VNUT) (88) and its enrichment in
brain astrocytes may be helpful in answering this important question and others related to
the mechanism of vesicular ATP release by astrocytes.
PHYSIOLOGICAL CONSEQUENCES OF ASTROCYTIC PURINES ON
SYNAPSES AND EXCITABILITY
In hypothalamic slices, astrocytic release of ATP is necessary and sufficient for
noradrenaline-dependent synaptic potentiation (89). Hypothalamic astrocytes express α1-
adrenergic receptors, and in response to adrenergic input they release ATP onto nearby
magnocellular neurosecretory neurons. Activation of P2X7 receptors on these neurons, as a
result, causes the enhancement of AMPA receptor surface expression and an increase in
miniature excitatory postsynaptic current (mEPSC) amplitude. In contrast, studies in the
mammalian retina demonstrated the role of astrocytic purinergic signaling in suppressing
neuronal activity (90). Using retinal whole-mounts, Newman showed that stimulation of
photoreceptors using light causes glial Ca2+ signaling (91) and that such Ca2+ signals lead to
ATP release from Muller cells (92). Patch clamp recordings showed that the resulting
actions on ganglion cells are mediated by adenosine acting through adenosine 1 (A1)
receptors to suppress neuronal activity. Both studies demonstrated ATP release, but one
study showed that ATP has direct actions, whereas the latter showed that this gliotransmitter
acts through a metabolite, adenosine. It is unclear whether there is an absence of
ectonucleotidases in the hypothalamus or whether there is a tight spatial association of glial
ATP release sites and neuronal P2X7 receptors to allow direct actions of ATP in this system.
Halassa and Haydon Page 9
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Similar suppressive actions of glia-derived adenosine have been observed in the
hippocampus (93–95). Hippocampal astrocytes release ATP, which, following its
degradation by extra-cellular nucleotidases to adenosine, causes an A1-dependent
presynaptic inhibition of synaptic transmission. Because astrocytes express receptors and
use signaling pathways that are shared with neurons, it is difficult to use pharmacological
manipulations to discern the role of these glia in the modulation of neuronal physiology.
However, the use of glia-specific toxins (95) and the selective loading of the Ca2+ chelator
BAPTA into astrocytes (94) have shown that the aforementioned suppressive actions of
adenosine on hippocampal synaptic transmission are of glial origin.
PURINERGIC GLIOTRANSMISSION MODULATES SYNAPTIC NETWORKS
In search of an understanding of the importance of gliotransmission, we developed
conditional astrocyte-specific molecular genetic manipulations based on our initial cell
culture studies so that we could study the importance of gliotransmission in situ and in vivo.
Several studies have shown that astrocytes express SNARE proteins that are required for
membrane-membrane fusion. In cultures the overexpression of the cytoplasmic SNARE
domain of synaptobrevin (in the absence of the vesicular tail) led to reduced glutamate
release from astrocytes. We applied this perturbation in situ and in vivo through the
development of a tet-off astrocyte-specific mouse that expresses dnSNARE only in
astrocytes. Molecular genetic inhibition of SNARE-dependent gliotransmission resulted in
the discovery that ATP is a major gliotransmitter in vivo (2). In our initial studies we were
surprised to find that the magnitude of Schaffer collateral CA1 synaptic transmission is
enhanced when astrocytes express dnSNARE. Fortuitously, we discovered that the well-
known tonic A1 receptor–mediated presynaptic inhibition of excitatory synaptic
transmission is mediated by adenosine derived from an astrocytic source (93).
Astrocytic specificity of this manipulation was validated in vitro (93) and in vivo (2). The
observation that SNAP-23 appears in the cytoplasmic fraction when dnSNARE is expressed
in astrocytes confirmed the hypothesis that this molecular manipulation targets vesicular
gliotransmission. One concern about this approach is that it should selectively perturb one
specific pathway of membrane trafficking—the regulated release of gliotransmitter—and not
impact trafficking of receptors, channels, transporters, or enzymes to the membrane.
Previous studies examining the specificity of SNARE domain interactions indicate that
specificity should be achieved with this approach because SNARE domains compete in a
highly SNARE protein–specific manner. This possibility was borne out by control
experiments showing that dnSNARE expression does not affect astrocytic membrane
physiology, including K+ buffering, glutamate transport activity, and agonist-dependent
Ca2+ signaling. Moreover, ectonucleotidases that are responsible for the hydrolysis of ATP
to adenosine are intact, as the exogenous addition of ATP fully reconstitutes A1-dependent
signaling in transgenic mice expressing dnSNARE.
Hippocampal slices derived from animals expressing dnSNARE in astrocytes (dnSNARE
animals) during postnatal life following weaning show enhanced basal synaptic
transmission. Luciferin/luciferase bioluminescence studies showed that astrocytic dnSNARE
expression leads to reduced extracellular ATP as well as adenosine and that in wild-type
mice the inhibition of ectonucleotidases leads to enhanced ATP-dependent signaling.
Because exogenous ATP can reconstitute the A1-dependent signaling in transgenic mice, we
concluded that the tonic source of adenosine that is responsible for A1 receptor activation is
astrocytic ATP. This is an important observation because it was previously thought that
adenosine is derived from a metabolic source that is cell type independent: During activity
adenosine accumulates intracellularly and exits into the extracellular space through
equilibrative nucleotide transporters. In actuality, under physiological conditions adenosine
Halassa and Haydon Page 10
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
is derived from astrocytic ATP that is released into the extracellular space, and under
hypoxic conditions the metabolic source predominates (96).
In addition to this tonic control of neuronal A1 receptor functions, astrocytic Ca2+ signaling
enhances the vesicular pathway of ATP gliotransmission. Tetanic stimulation of a subset of
CA3-CA1 synapses leads to A1 receptor–dependent heterosynaptic depression of
neighboring (untetanized) synapses (97) that requires the recruitment of astrocytes, which
provide adenosine (93). Thus, as the activity of neurons waxes and wanes, astrocytes have
the potential to dynamically modulate neuronal network function.
PURINERGIC GLIOTRANSMISSION MODULATES SLEEP
A1 receptor function regulates a number of mammalian behaviors including sleep (98).
Sleep is a fundamental behavior that is universal among multicellular animals (99).
Evidence suggests that if an organism has a nervous system, then that nervous system must
sleep (100). Although modulated by the circadian clock, which entrains many physiological
processes to salient environmental cues, such as the light/dark cycle (101), sleep is
additionally controlled by a homeostatic process (also known as Process S), which is
completely dissociable from circadian control. The fact that, when prevented, sleep is
subsequently compensated suggests that important processes take place during this state, and
may be the strongest argument against the null hypothesis of sleep function (that sleep has
evolved as an inactive state to hide organisms from their predators).
The molecular and cellular process underlying sleep homeostasis has been under
investigation for more than 90 years. This investigation started with transfer experiments in
which cerebrospinal fluid (CSF) extracts and cerebral venous blood from sleep-deprived
animals were injected into control animals to determine whether they could induce sleep.
Although these experiments were unsuccessful at identifying endogenous sleep factors, they
were essential in establishing that such factors are generated locally within the brain (102).
More recently, studies have implicated adenosine as an endogenous sleep factor. Porkka-
Heiskanen and colleagues have determined that adenosine levels vary with sleep propensity;
during wakefulness, adenosine levels progressively increase, whereas during sleep they
subside. Antagonizing adenosine (both A1 and A2A receptors) promotes wakefulness (103),
and injecting adenosine or its agonists into the brain promotes sleep (104). Thus, adenosine
may be not only a sleep factor but also a mediator of the homeostatic sleep response.
Criteria for identifying sleep were initially developed for the study of mammals and thus are
both behavioral and electrophysiological (105). During sleep, organisms exhibit an
increased threshold to sensory stimulation (99), and in mammals, specific changes occur in
the electroencephalogram (EEG) (106). On the basis of a combination of these criteria,
mammalian sleep can be subdivided into rapid-eye-movement (REM) sleep and nonrapid-
eye-movement (NREM) sleep.
During NREM sleep, the EEG is dominated by slow rhythms (<5.0 Hz) (100, 107, 108)
whose power positively correlates with the accumulation of sleep pressure. The longer the
animal is kept awake, the larger and more frequent these slower rhythms become in
subsequent sleep (105, 109, 110); this phenomenon is referred to as the slow-wave activity
(SWA) of the EEG and is thought to be an electrophysiological marker of sleep pressure.
The first clue to the involvement of purinergic gliotransmission in sleep regulation was that
animals exhibit a blunting of their SWA when dnSNARE is expressed in astrocytes (2). One
concern with the inactivation of a tonic A1 receptor–dependent pathway is that such
inactivation may lead to neurodegeneration given the powerful neuroprotective role of this
Halassa and Haydon Page 11
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
signaling pathway (111). However, this is not the case in this system because all sleep
phenotypes are reversed when transgene expression is subsequently turned off in the same
animals.
When animals were subjected to 6 h of sleep deprivation, their homeostatic response (both
the electrophysiological and behavioral components) was intact only when normal
gliotransmission was allowed to occur. The reversible inhibition of gliotransmission
attenuated the increase in SWA and the increase in total sleep time that follows a period of
sleep deprivation. Interestingly, when animals were sleep deprived after novel object
recognition (NOR) training, a task whose memory is sensitive to the effects of sleep
pressure, NOR memory was intact when purinergic gliotransmission was blocked. This
suggests that cognitive impairment following short-term sleep loss is an active process and
is dependent on astrocytic purinergic signaling.
These behavioral effects that were perturbed by astrocytic dnSNARE expression were fully
reconstituted in wild-type mice in which we introduced the A1 receptor antagonist CPT.
Using osmotic minipumps to administer CPT intracerebroventricularly, we similarly found
reduced SWA, reduced responses to sleep deprivation, as well as the maintenance of NOR
memory following sleep deprivation. Further support for astrocytic adenosine signaling in
vivo is provided by recent studies using optogenetics in vivo. The expression and activation
of the blue light–sensitive cation channel channelrhodopsin-2 (ChR-2) in astrocytes of the
subthalamic nucleus (STN) result in a robust suppression of nearby neuronal spiking in vivo
(112). Astrocyte-induced neuronal suppression has an onset of ~400 ms and an offset of
~800 ms following optical activation, consistent with the possibility that astrocytic ChR-2
activation results in the release of ATP and its degradation to adenosine to suppress nearby
spiking (112).
The role of astrocytic gliotransmission in sleep modulation is the first experimental
demonstration of active glial involvement in mammalian behavior. Because adenosinergic
gliotransmission operates over significantly longer timescales than does synaptic
transmission, it is an ideal cellular candidate for mechanisms underlying the control of
slowly evolving behaviors such as sleep. The accumulation of adenosine and/or its
downstream A1-mediated effects in thalamocortical circuits during wake-fulness over the
course of wakefulness may promote the generation of slow oscillations in these structures.
Interestingly, adenosine accumulation in the cortex exerts negative feedback on brainstem
circuits involved in promoting wake-fulness (113). These findings support the possibility
that cortical activity is an important determinant of global vigilance state and provide a
possible link between local sleep and global sleep. Because slow oscillations can occur in
quiet wakefulness (114) and because many cortical regions appear to generate this rhythm,
the involvement of a certain number of thalamocortical circuits in this rhythm may promote
the switch from wakefulness to sleep.
Theoretical and empirical studies suggest that the increase in the genesis and spread of
cortical slow waves is dependent on the increase in synaptic strength of the thalamocortical
system (107). Biochemical and electrophysiological recordings of rats in vivo show that
wakefulness is associated with net synaptic potentiation, whereas sleep is associated with net
synaptic depression (115). One hypothesis of sleep function suggests that one of the primary
functions of sleep is homeostatic synaptic downscaling, in which the overall strength of
synapses is decreased while preserving relative synaptic weights (100). Synaptic
downscaling may be essential for a system such as the brain, which, when engaged in
sensory processing (during wake), exhibits a progressive strengthening of its connections.
Wake-dependent increase in synaptic strength would increase energy and space demand,
requiring a process to offset synaptic potentiation. Thus, sleep may have evolved to scale
Halassa and Haydon Page 12
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
synapses back and allow the brain to function as a learning/ adaptive machine in the face of
space and energy constraints. Additionally, it may have evolved to offset the effects of
increased excitability of neural circuits, which if left unchecked may result in epileptiform
activity. Given that sleep deprivation is a known precipitator of seizures in patients with
epileptic disorders (116), this hypothesis may have therapeutic implications.
ASTROCYTES IN NEUROLOGICAL DISEASES
Astrocytes become “reactive” in response to a number of brain insults, including trauma,
infection, epilepsy, and neurodegeneration (117). Reactive astrocytes are characterized by
well-described morphological changes, but less understood physiological alterations.
Whether the reactive astrocyte phenotype is a single biological state or a family of related
changes is not clear.
A major feature of reactive astrocytes is the upregulation of the intermediate filament
proteins GFAP and vimentin. This upregulation is associated with thickening of the main
processes of the astrocyte (astrocytic hypertrophy) (118). Genetic ablation of these
astrocytic intermediate filaments results in exacerbation of neuronal injury following middle
cerebral artery occlusion, suggesting that the upregulation of these molecules is beneficial in
the context of certain brain insults (119). Status epilepticus (SE) causes the loss of astrocytic
domains and the overlapping of astrocytic processes (120). This last morphological feature
is not seen in the context of other brain injuries such as neurodegenerative disorders.
Certain studies suggest that reactive astrocytosis is accompanied by astrocytic proliferation,
and recent cell fate mapping studies suggest that adult astrocytes are able to divide following
brain injury (121). Inhibition of astrocytic proliferation using molecular genetics results in a
poorer outcome of acute injury models, including forebrain stab wound (122), traumatic
brain injury (123), and stab or crush spinal cord injury (124). Introducing an NMDA
receptor antagonist into the forebrain in the stab wound model ameliorated its severity,
suggesting that astrocytic proliferation is important for maintaining glutamate homeostasis
(122). In another set of studies, the molecular ablation of astrocytic signal transducer and
activator of transcription 3 (STAT3) resulted in the impairment of astrocytic reactivity,
astrocytic migration toward injury, and the worsening of SCI functional outcome (125).
These studies demonstrate that astrocytic reactivity and proliferation are beneficial in the
context of certain models of acute brain injury.
Depending on the nature of the insult, astrocytic reactivity can be associated with
physiological alterations of glutamate uptake, K+ buffering, and/or H2O homeostasis (126).
Both epilepsy and neurodegenerative disorders such as Alzheimer’s disease appear to be
associated with an alteration in astrocytic glutamate uptake and K+ buffering. The vascular
β-amyloid load in mouse models of Alzheimer’s disease correlates with the loss of Kir, BK,
and aquaporin 4 channels in astrocytes (127). Furthermore, these channels are reduced in
postmortem brains of patients with moderate to severe vascular amyloid deposition (127).
Postmortem tissues derived from patients with Alzheimer’s disease show evidence for
reduced astrocytic glutamate transporter expression and reduced glutamate uptake by
biochemical assays. It will be important to investigate whether reversing these molecular
alterations in astrocytes slows the progression of Alzheimer’s disease in animals models and
ultimately in humans.
Until recently, the neurodevelopmental disorder Rett syndrome (RTT) was thought to be a
result of cell-autonomous, lack-of-function expression of the transcriptional factor methyl-
CpG binding protein 2 (MeCP2) in neurons. However, a careful study by Ballas et al. (128)
revealed that this transcription factor is expressed in brain astrocytes, albeit at lower levels
than in neurons. Furthermore, wild-type neurons exhibit aberrant dendritic morphology and
Halassa and Haydon Page 13
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
synapse numbers when cocultured with MeCP2-deficient astrocytes, reminiscent of
morphological abnormalities of RTT. These changes are recaptured when wild-type neurons
are incubated in MeCP2-deficient astrocyte-conditioned medium, suggesting that astrocytes
release a soluble factor to support neuronal development, which is inhibited by the RTT
mutation. The growing list of astrocytic factors important for synaptogenesis, including
thrombospondins and cholesterol (129), suggests that disruption of this important astrocytic
function contributes to a number of neurodevelopmental disorders.
Do changes in gliotransmission contribute to the development of neurological disorders? A
growing body of evidence suggests that astrocytic Ca2+ signaling is enhanced in the context
of certain neurological diseases (130, 131). For example, in mouse models of Alzheimer’s
disease, astrocytes appear to exhibit higher resting levels of Ca2+ and to generate
synchronous Ca2+ oscillations (130). In addition, following pilocarpine-induced SE,
astrocytic Ca2+ signaling is also enhanced; this enhancement lasts for days after the
termination of SE (131). The pharmacological antagonism of both mGluR5 and NR2B
provides protection from the delayed neuronal death that occurs following SE, suggesting
that enhanced Ca2+ signaling in astrocytes results in enhanced glutamatergic
gliotransmission, which targets NR2B-containing NMDA receptors (discussed above). The
activation of extrasynaptic NR2B-containing NMDA receptors stimulates CREB
dephosphorylation and neuronal death (132). Thus, enhanced astrocytic Ca2+ promotes
neuronal death via glutamatergic gliotransmission. This conclusion is supported by the
observation that the loading of cortical astrocytes with a Ca2+ chelator is neuroprotective in
the context of SE (131). Whether molecular manipulations that target astrocytic Ca2+
signaling promote neuronal survival following SE is an open question and an active area of
investigation.
ASTROCYTES IN PSYCHIATRIC DISEASES
In contrast to neurological disorders, which show glial upregulation of intermediate filament
proteins, two psychiatric disorders, namely major depressive disorder (MDD) and
schizophrenia, are characterized by a decrease in GFAP expression (and perhaps a decrease
in glial cell number) in the prefrontal cortex, as revealed by postmortem studies (133).
Whether these changes are causal in the context of these disorders or a consequence of the
illness is not clear.
Studies have shown altered gene expression of GFAP, glutamate transporters, and GS in
MDD (134). Chronic unpredictable stress (CUS) of rodents, an animal model of depression,
mimics the human MDD changes in astrocytic gene expression (134, 135). Certain
functional consequences of these gene expression changes result from the disruption of
astrocytic support of GABAergic transmission. For example, ex vivo nuclear magnetic
resonance (NMR) reveals reduced metabolic cycling of glutamate in the prefrontal cortex of
rats undergoing CUS. Riluzole, a chemical that facilitates glutamate uptake by astrocytes,
reverses CUS-induced anhedonia and helplessness, suggesting that boosting the glutamate/
glutamine cycle can be therapeutic in the context of MDD. Support for this hypothesis
comes from studies showing that patients with MDD exhibit reduced cortical GABA levels,
similar to rats undergoing CUS (136), and that normalizing GABA levels in these
individuals correlates with their clinical improvement. Thus, it appears that a dysregulation
of astrocytic support of GABAergic transmission contributes to the pathophysiology of
MDD and that this process may provide novel therapeutic targets for the treatment of this
debilitating state. Because one of the leading hypotheses regarding the pathophysiology of
schizophrenia suggests a disruption in the function of a subclass of inhibitory interneurons
(namely the parvalbumin-positive phenotype of interneurons), which leads to a
dysregulation of γ oscillations and related temporal coding (137), it would be intriguing to
Halassa and Haydon Page 14
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
investigate whether the astrocytic support of GABAergic transmission (22) is important for
the generation of fast-cortical rhythms and whether its disruption can contribute to
schizophrenia endophenotypes.
Do changes in gliotransmission contribute to psychiatric disorders? Although the answer to
this important question is not known, it is intriguing that cycloserine, an analog of the
gliotransmitter D-serine, is used as a therapeutic agent in schizophrenia (138). In a previous
perspective, we speculated that a hypoactive astrocyte phenotype may exist in certain
psychiatric disorders, which would result in the attenuation of astrocytic activation of
neuronal NMDA receptors (139). Because a hypo-NMDA phenotype is thought to be a final
common pathway of the pathophysiology of schizophrenia, disrupted gliotransmission may
also contribute to this debilitating disorder. Future experiments will test this hypothesis and,
more importantly, provide a framework for the role of astrocytes in psychiatric disorders.
Glossary
Astrocyte the dominant subclass of nonneuronal glial cell of the nervous
system
Gliotransmission the release of chemical transmitters from glial cells, with
particular reference to astrocytes
SNARE soluble N-ethylmaleimide-sensitive fusion protein attachment
protein receptor
NMDA N-methyl-D-aspartate
Ectonucleotidases cell surface–expressed enzymes that stimulate the hydrolysis of
ATP to adenosine
Photolysis an organic molecule is attached to a bioactive molecule to yield
a biologically inactive product. Absorption of a UV photon
leads to the photolysis of this molecule, yielding the
biologically active species
A1 adenosine 1
Heterosynaptic
depression the ability of the activity of one synapse to lead to the
depression of a neighboring independent synapse
Sleep homeostat sleep is regulated by a circadian oscillator (which controls
timing) and a sleep homeostat, which integrates the period of
prior wakefulness and provides the drive for sleep
REM rapid eye movement
NREM nonrapid eye movement
Slow-wave activity
(SWA) low-frequency activity in the range of 0.5 to 4 Hz detected by
EEG recordings during NREM sleep
LITERATURE CITED
1. Garcia-Marin V, Garcia-Lopez P, Freire M. Cajal’s contributions to glia research. Trends Neurosci.
2007; 30:479–87. [PubMed: 17765327]
2. Halassa MM, Florian C, Fellin T, Munoz JR, Lee SY, et al. Astrocytic modulation of sleep
homeostasis and cognitive consequences of sleep loss. Neuron. 2009; 61:213–19. [PubMed:
19186164]
Halassa and Haydon Page 15
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
3. Bushong EA, Martone ME, Jones YZ, Ellisman MH. Protoplasmic astrocytes in CA1 stratum
radiatum occupy separate anatomical domains. J Neurosci. 2002; 22:183–92. [PubMed: 11756501]
4. Halassa MM, Fellin T, Takano H, Dong JH, Haydon PG. Synaptic islands defined by the territory of
a single astrocyte. J Neurosci. 2007; 27:6473–77. [PubMed: 17567808]
5. Ventura R, Harris KM. Three-dimensional relationships between hippocampal synapses and
astrocytes. J Neurosci. 1999; 19:6897–906. [PubMed: 10436047]
6. Araque A, Parpura V, Sanzgiri RP, Haydon PG. Tripartite synapses: glia, the unacknowledged
partner. Trends Neurosci. 1999; 22:208–15. [PubMed: 10322493]
7. Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. Signaling at the gliovascular interface. J
Neurosci. 2003; 23:9254–62. [PubMed: 14534260]
8. Takano T, Tian GF, Peng W, Lou N, Libionka W, et al. Astrocyte-mediated control of cerebral
blood flow. Nat Neurosci. 2006; 9:260–67. [PubMed: 16388306]
9. Mulligan SJ, MacVicar BA. Calcium transients in astrocyte endfeet cause cerebrovascular
constrictions. Nature. 2004; 431:195–99. [PubMed: 15356633]
10. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, et al. Neuron-to-astrocyte
signaling is central to the dynamic control of brain microcirculation. Nat Neurosci. 2003; 6:43–50.
[PubMed: 12469126]
11. Filosa JA, Bonev AD, Nelson MT. Calcium dynamics in cortical astrocytes and arterioles during
neurovascular coupling. Circ Res. 2004; 95:e73–81. [PubMed: 15499024]
12. Metea MR, Newman EA. Glial cells dilate and constrict blood vessels: a mechanism of
neurovascular coupling. J Neurosci. 2006; 26:2862–70. [PubMed: 16540563]
13. Haydon PG. GLIA: listening and talking to the synapse. Nat Rev Neurosci. 2001; 2:185–93.
[PubMed: 11256079]
14. Volterra A, Meldolesi J. Astrocytes, from brain glue to communication elements: the revolution
continues. Nat Rev Neurosci. 2005; 6:626–40. [PubMed: 16025096]
15. Butt AM, Kalsi A. Inwardly rectifying potassium channels (Kir) in central nervous system glia: a
special role for Kir4.1 in glial functions. J Cell Mol Med. 2006; 10:33–44. [PubMed: 16563220]
16. Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir4.1 leads
to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced
short-term synaptic potentiation. J Neurosci. 2007; 27:11354–65. [PubMed: 17942730]
17. Gordon GR, Mulligan SJ, MacVicar BA. Astrocyte control of the cerebrovasculature. Glia. 2007;
55:1214–21. [PubMed: 17659528]
18. Anderson CM, Swanson RA. Astrocyte glutamate transport: review of properties, regulation, and
physiological functions. Glia. 2000; 32:1–14. [PubMed: 10975906]
19. Schousboe A. Pharmacological and functional characterization of astrocytic GABA transport: a
short review. Neurochem Res. 2000; 25:1241–44. [PubMed: 11059798]
20. Jursky F, Nelson N. Developmental expression of the glycine transporters GLYT1 and GLYT2 in
mouse brain. J Neurochem. 1996; 67:336–44. [PubMed: 8667011]
21. Hertz L, Zielke HR. Astrocytic control of glutamatergic activity: astrocytes as stars of the show.
Trends Neurosci. 2004; 27:735–43. [PubMed: 15541514]
22. Liang SL, Carlson GC, Coulter DA. Dynamic regulation of synaptic GABA release by the
glutamate-glutamine cycle in hippocampal area CA1. J Neurosci. 2006; 26:8537–48. [PubMed:
16914680]
23. Bernard-Helary K, Lapouble E, Ardourel M, Hevor T, Cloix JF. Correlation between brain
glycogen and convulsive state in mice submitted to methionine sulfoximine. Life Sci. 2000;
67:1773–81. [PubMed: 11021361]
24. Bernard-Helary K, Ardourel MY, Hevor T, Cloix JF. In vivo and in vitro glycogenic effects of
methionine sulfoximine are different in two inbred strains of mice. Brain Res. 2002; 929:147–55.
[PubMed: 11864619]
25. Gonzalez-Burgos G, Hashimoto T, Lewis DA. Inhibition and timing in cortical neural circuits. Am
J Psychiatry. 2007; 164:12. [PubMed: 17202537]
26. Sohal VS, Zhang F, Yizhar O, Deisseroth K. Parvalbumin neurons and gamma rhythms enhance
cortical circuit performance. Nature. 2009; 459:698–702. [PubMed: 19396159]
Halassa and Haydon Page 16
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
27. Cardin JA, Carlen M, Meletis K, Knoblich U, Zhang F, et al. Driving fast-spiking cells induces
gamma rhythm and controls sensory responses. Nature. 2009; 459:663–67. [PubMed: 19396156]
28. Uhlhaas PJ, Haenschel C, Nikolic D, Singer W. The role of oscillations and synchrony in cortical
networks and their putative relevance for the pathophysiology of schizophrenia. Schizophr Bull.
2008; 34:927–43. [PubMed: 18562344]
29. Lewis DA, Hashimoto T. Deciphering the disease process of schizophrenia: the contribution of
cortical gaba neurons. Int Rev Neurobiol. 2007; 78:109–31. [PubMed: 17349859]
30. Magistretti PJ. Neuron-glia metabolic coupling and plasticity. J Exp Biol. 2006; 209:2304–11.
[PubMed: 16731806]
31. Theis M, Sohl G, Eiberger J, Willecke K. Emerging complexities in identity and function of glial
connexins. Trends Neurosci. 2005; 28:188–95. [PubMed: 15808353]
32. Rouach N, Koulakoff A, Abudara V, Willecke K, Giaume C. Astroglial metabolic networks sustain
hippocampal synaptic transmission. Science. 2008; 322:1551–55. [PubMed: 19056987]
33. Tsacopoulos M, Magistretti PJ. Metabolic coupling between glia and neurons. J Neurosci. 1996;
16:877–85. [PubMed: 8558256]
34. Sorg O, Magistretti PJ. Characterization of the glycogenolysis elicited by vasoactive intestinal
peptide, noradrenaline and adenosine in primary cultures of mouse cerebral cortical astrocytes.
Brain Res. 1991; 563:227–33. [PubMed: 1664773]
35. Wang X, Lou N, Xu Q, Tian GF, Peng WG, et al. Astrocytic Ca2+ signaling evoked by sensory
stimulation in vivo. Nat Neurosci. 2006; 9:816–23. [PubMed: 16699507]
36. Schummers J, Yu H, Sur M. Tuned responses of astrocytes and their influence on hemodynamic
signals in the visual cortex. Science. 2008; 320:1638–43. [PubMed: 18566287]
37. Higley MJ, Contreras D. Balanced excitation and inhibition determine spike timing during
frequency adaptation. J Neurosci. 2006; 26:448–57. [PubMed: 16407542]
38. Petzold GC, Albeanu DF, Sato TF, Murthy VN. Coupling of neural activity to blood flow in
olfactory glomeruli is mediated by astrocytic pathways. Neuron. 2008; 58:897–910. [PubMed:
18579080]
39. Sirotin YB, Das A. Anticipatory haemodynamic signals in sensory cortex not predicted by local
neuronal activity. Nature. 2009; 457:475–79. [PubMed: 19158795]
40. Zhang Q, Pangric T, Kreft M, Kran M, Li N, et al. Fusion-related release of glutamate from
astrocytes. J Biol Chem. 2004; 279:12724–33. [PubMed: 14722063]
41. Montana V, Ni Y, Sunjara V, Hua X, Parpura V. Vesicular glutamate transporter-dependent
glutamate release from astrocytes. J Neurosci. 2004; 24:2633–42. [PubMed: 15028755]
42. Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhauser C, et al. Astrocytes contain a vesicular
compartment that is competent for regulated exocytosis of glutamate. Nat Neurosci. 2004; 7:613–
20. [PubMed: 15156145]
43. Zhang Q, Fukuda M, Van Bockstaele E, Pascual O, Haydon PG. Synaptotagmin IV regulates glial
glutamate release. Proc Natl Acad Sci USA. 2004; 101:9441–46. [PubMed: 15197251]
44. Zhang Z, Chen G, Zhou W, Song A, Xu T, et al. Regulated ATP release from astrocytes through
lysosome exocytosis. Nat Cell Biol. 2007; 9:945–53. [PubMed: 17618272]
45. Coco S, Calegari F, Pravettoni E, Pozzi D, Taverna E, et al. Storage and release of ATP from
astrocytes in culture. J Biol Chem. 2003; 278:1354–62. [PubMed: 12414798]
46. Mothet JP, Pollegioni L, Ouanounou G, Martineau M, Fossier P, Baux G. Glutamate receptor
activation triggers a calcium-dependent and SNARE protein-dependent release of the
gliotransmitter D-serine. Proc Natl Acad Sci USA. 2005; 102:5606–11. [PubMed: 15800046]
47. Jourdain P, Bergersen LH, Bhaukaurally K, Bezzi P, Santello M, et al. Glutamate exocytosis from
astrocytes controls synaptic strength. Nat Neurosci. 2007; 10:331–39. [PubMed: 17310248]
48. Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG. Glutamate-mediated astrocyte-
neuron signalling. Nature. 1994; 369:744–47. [PubMed: 7911978]
49. Szatkowski M, Barbour B, Attwell D. Non-vesicular release of glutamate from glial cells by
reversed electrogenic glutamate uptake. Nature. 1990; 348:443–46. [PubMed: 2247147]
Halassa and Haydon Page 17
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
50. Kimelberg HK, Goderie SK, Higman S, Pang S, Waniewski RA. Swelling-induced release of
glutamate, aspartate, and taurine from astrocyte cultures. J Neurosci. 1990; 10:1583–91. [PubMed:
1970603]
51. Warr O, Takahashi M, Attwell D. Modulation of extracellular glutamate concentration in rat brain
slices by cystine-glutamate exchange. J Physiol. 1999; 514(Pt. 3):783–93. [PubMed: 9882750]
52. Duan S, Anderson CM, Keung EC, Chen Y, Chen Y, Swanson RA. P2X7 receptor-mediated
release of excitatory amino acids from astrocytes. J Neurosci. 2003; 23:1320–28. [PubMed:
12598620]
53. Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR. Functional hemichannels in astrocytes: a novel
mechanism of glutamate release. J Neurosci. 2003; 23:3588–96. [PubMed: 12736329]
54. Baker DA, McFarland K, Lake RW, Shen H, Tang XC, et al. Neuroadaptations in cystine-
glutamate exchange underlie cocaine relapse. Nat Neurosci. 2003; 6:743–49. [PubMed: 12778052]
55. Sontheimer H. A role for glutamate in growth and invasion of primary brain tumors. J Neurochem.
2008; 105:287–95. [PubMed: 18284616]
56. Fellin T, Gomez-Gonzalo M, Gobbo S, Carmignoto G, Haydon PG. Astrocytic glutamate is not
necessary for the generation of epileptiform neuronal activity in hippocampal slices. J Neurosci.
2006; 26:9312–22. [PubMed: 16957087]
57. Fellin T, Pascual O, Gobbo S, Pozzan T, Haydon PG, Carmignoto G. Neuronal synchrony
mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron.
2004; 43:729–43. [PubMed: 15339653]
58. Parri HR, Gould TM, Crunelli V. Spontaneous astrocytic Ca2+ oscillations in situ drive NMDAR-
mediated neuronal excitation. Nat Neurosci. 2001; 4:803–12. [PubMed: 11477426]
59. D’Ascenzo M, Fellin T, Terunuma M, Revilla-Sanchez R, Meaney DF, et al. mGluR5 stimulates
gliotransmission in the nucleus accumbens. Proc Natl Acad Sci USA. 2007; 104:1995–2000.
[PubMed: 17259307]
60. Angulo MC, Kozlov AS, Charpak S, Audinat E. Glutamate released from glial cells synchronizes
neuronal activity in the hippocampus. J Neurosci. 2004; 24:6920–27. [PubMed: 15295027]
61. Shigetomi E, Bowser DN, Sofroniew MV, Khakh BS. Two forms of astrocyte calcium excitability
have distinct effects on NMDA receptor-mediated slow inward currents in pyramidal neurons. J
Neurosci. 2008; 28:6659–63. [PubMed: 18579739]
62. Navarrete M, Araque A. Endocannabinoids mediate neuron-astrocyte communication. Neuron.
2008; 57:883–93. [PubMed: 18367089]
63. Fiacco TA, McCarthy KD. Intracellular astrocyte calcium waves in situ increase the frequency of
spontaneous AMPA receptor currents in CA1 pyramidal neurons. J Neurosci. 2004; 24:722–32.
[PubMed: 14736858]
64. Lee CJ, Mannaioni G, Yuan H, Woo DH, Gingrich MB, Traynelis SF. Astrocytic control of
synaptic NMDA receptors. J Physiol. 2007; 581:1057–81. [PubMed: 17412766]
65. Yudkoff M, Daikhin Y, Nissim I, Pleasure D, Stern J, Nissim I. Inhibition of astrocyte glutamine
production by alpha-ketoisocaproic acid. J Neurochem. 1994; 63:1508–15. [PubMed: 7931304]
66. Bellocchio EE, Reimer RJ, Fremeau RT Jr, Edwards RH. Uptake of glutamate into synaptic
vesicles by an inorganic phosphate transporter. Science. 2000; 289:957–60. [PubMed: 10938000]
67. Fremeau RT, Troyer MD, Pahner I, Nygaard GO, Tran CH, et al. The expression of vesicular
glutamate transporters defines two classes of excitatory synapse. Neuron. 2001; 31:247–60.
[PubMed: 11502256]
68. Fremeau RT Jr, Burman J, Qureshi T, Tran CH, Proctor J, et al. The identification of vesicular
glutamate transporter 3 suggests novel modes of signaling by glutamate. Proc Natl Acad Sci USA.
2002; 99:14488–93. [PubMed: 12388773]
69. Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhauser C, et al. Astrocytes contain a vesicular
compartment that is competent for regulated exocytosis of glutamate. Nat Neurosci. 2004; 7:613–
20. [PubMed: 15156145]
70. Zhang Q, Pangrsic T, Kreft M, Krzan M, Li N, et al. Fusion-related release of glutamate from
astrocytes. J Biol Chem. 2004; 279:12724–33. [PubMed: 14722063]
Halassa and Haydon Page 18
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
71. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, et al. A transcriptome database for
astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development
and function. J Neurosci. 2008; 28:264–78. [PubMed: 18171944]
72. Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, et al. The transcriptome and
metabolic gene signature of protoplasmic astrocytes in the adult murine cortex. J Neurosci. 2007;
27:12255–66. [PubMed: 17989291]
73. Tan PK, Downey TJ, Spitznagel EL Jr, Xu P, Fu D, et al. Evaluation of gene expression
measurements from commercial microarray platforms. Nucleic Acids Res. 2003; 31:5676–84.
[PubMed: 14500831]
74. Shi L, Perkins RG, Fang H, Tong W. Reproducible and reliable microarray results through quality
control: Good laboratory proficiency and appropriate data analysis practices are essential. Curr
Opin Biotechnol. 2008; 19:10–18. [PubMed: 18155896]
75. Langer D, Hammer K, Koszalka P, Schrader J, Robson S, Zimmermann H. Distribution of
ectonucleotidases in the rodent brain revisited. Cell Tissue Res. 2008; 334:199–217. [PubMed:
18843508]
76. Futai K, Kim MJ, Hashikawa T, Scheiffele P, Sheng M, Hayashi Y. Retrograde modulation of
presynaptic release probability through signaling mediated by PSD-95-neuroligin. Nat Neurosci.
2007; 10:186–95. [PubMed: 17237775]
77. Fiacco TA, Agulhon C, Taves SR, Petravicz J, Casper KB, et al. Selective stimulation of astrocyte
calcium in situ does not affect neuronal excitatory synaptic activity. Neuron. 2007; 54:611–26.
[PubMed: 17521573]
78. Sweger EJ, Casper KB, Scearce-Levie K, Conklin BR, McCarthy KD. Development of
hydrocephalus in mice expressing the Gi-coupled GPCR Ro1 RASSL receptor in astrocytes. J
Neurosci. 2007; 27:2309–17. [PubMed: 17329428]
79. Malarkey EB, Ni Y, Parpura V. Ca2+ entry through TRPC1 channels contributes to intracellular
Ca2+ dynamics and consequent glutamate release from rat astrocytes. Glia. 2008; 56:821–35.
[PubMed: 18338793]
80. Mothet JP, Parent AT, Wolosker H, Brady RO Jr, Linden DJ, et al. D-serine is an endogenous
ligand for the glycine site of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci USA. 2000;
97:4926–31. [PubMed: 10781100]
81. Schell MJ, Molliver ME, Snyder SH. D-serine, an endogenous synaptic modulator: localization to
astrocytes and glutamate-stimulated release. Proc Natl Acad Sci USA. 1995; 92:3948–52.
[PubMed: 7732010]
82. Panatier A, Theodosis DT, Mothet JP, Touquet B, Pollegioni L, et al. Glia-derived D-serine
controls NMDA receptor activity and synaptic memory. Cell. 2006; 125:775–84. [PubMed:
16713567]
83. Haber M, Murai KK. Reshaping neuron-glial communication at hippocampal synapses. Neuron
Glia Biol. 2006; 2:59–66. [PubMed: 18634591]
84. Haber M, Zhou L, Murai KK. Cooperative astrocyte and dendritic spine dynamics at hippocampal
excitatory synapses. J Neurosci. 2006; 26:8881–91. [PubMed: 16943543]
85. Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater SB. ATP released from
astrocytes mediates glial calcium waves. J Neurosci. 1999; 19:520–28. [PubMed: 9880572]
86. Li D, Ropert N, Koulakoff A, Giaume C, Oheim M. Lysosomes are the major vesicular
compartment undergoing Ca2+-regulated exocytosis from cortical astrocytes. J Neurosci. 2008;
28:7648–58. [PubMed: 18650341]
87. Blott EJ, Griffiths GM. Secretory lysosomes. Nat Rev Mol Cell Biol. 2002; 3:122–31. [PubMed:
11836514]
88. Sawada K, Echigo N, Juge N, Miyaji T, Otsuka M, et al. Identification of a vesicular nucleotide
transporter. Proc Natl Acad Sci USA. 2008; 105:5683–86. [PubMed: 18375752]
89. Gordon GR, Baimoukhametova DV, Hewitt SA, Rajapaksha WR, Fisher TE, Bains JS. Nore-
pinephrine triggers release of glial ATP to increase postsynaptic efficacy. Nat Neurosci. 2005;
8:1078–86. [PubMed: 15995701]
90. Newman EA. Calcium signaling in retinal glial cells and its effect on neuronal activity. Prog Brain
Res. 2001; 132:241–54. [PubMed: 11544993]
Halassa and Haydon Page 19
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
91. Newman EA. Calcium increases in retinal glial cells evoked by light-induced neuronal activity. J
Neurosci. 2005; 25:5502–10. [PubMed: 15944378]
92. Newman EA. Glial cell inhibition of neurons by release of ATP. J Neurosci. 2003; 23:1659–66.
[PubMed: 12629170]
93. Pascual O, Casper KB, Kubera C, Zhang J, Revilla-Sanchez R, et al. Astrocytic purinergic
signaling coordinates synaptic networks. Science. 2005; 310:113–16. [PubMed: 16210541]
94. Serrano A, Haddjeri N, Lacaille JC, Robitaille R. GABAergic network activation of glial cells
underlies hippocampal heterosynaptic depression. J Neurosci. 2006; 26:5370–82. [PubMed:
16707789]
95. Zhang JM, Wang HK, Ye CQ, Ge W, Chen Y, et al. ATP released by astrocytes mediates
glutamatergic activity-dependent heterosynaptic suppression. Neuron. 2003; 40:971–82. [PubMed:
14659095]
96. Martin ED, Fernandez M, Perea G, Pascual O, Haydon PG, et al. Adenosine released by astrocytes
contributes to hypoxia-induced modulation of synaptic transmission. Glia. 2007; 55:36–45.
[PubMed: 17004232]
97. Manzoni O, Bockaert J. Metabotropic glutamate receptors inhibiting excitatory synapses in the
CA1 area of rat hippocampus. Eur J Neurosci. 1995; 7:2518–23. [PubMed: 8845958]
98. Basheer R, Strecker RE, Thakkar MM, McCarley RW. Adenosine and sleep-wake regulation. Prog
Neurobiol. 2004; 73:379–96. [PubMed: 15313333]
99. Cirelli C, Tononi G. Is sleep essential? PLoS Biol. 2008; 6:e216. [PubMed: 18752355]
100. Tononi G, Cirelli C. Sleep function and synaptic homeostasis. Sleep Med Rev. 2006; 10:49–62.
[PubMed: 16376591]
101. Borbely AA. A two process model of sleep regulation. Hum Neurobiol. 1982; 1:195–204.
[PubMed: 7185792]
102. Krueger JM, Obal F Jr. Sleep function. Front Biosci. 2003; 8:d511–19. [PubMed: 12700033]
103. Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW.
Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science. 1997;
276:1265–68. [PubMed: 9157887]
104. Porkka-Heiskanen T, Strecker RE, McCarley RW. Brain site-specificity of extracellular
adenosine concentration changes during sleep deprivation and spontaneous sleep: an in vivo
microdialysis study. Neuroscience. 2000; 99:507–17. [PubMed: 11029542]
105. Franken P, Dijk DJ, Tobler I, Borbely AA. Sleep deprivation in rats: effects on EEG power
spectra, vigilance states, and cortical temperature. Am J Physiol. 1991; 261:R198–208. [PubMed:
1858947]
106. Franken P, Malafosse A, Tafti M. Genetic determinants of sleep regulation in inbred mice. Sleep.
1999; 22:155–69. [PubMed: 10201060]
107. Esser SK, Hill SL, Tononi G. Sleep homeostasis and cortical synchronization: I. Modeling the
effects of synaptic strength on sleep slow waves. Sleep. 2007; 30:1617–30. [PubMed: 18246972]
108. Tononi G, Cirelli C. The frontiers of sleep. Trends Neurosci. 1999; 22:417–18. [PubMed:
10577249]
109. Franken P, Tobler I, Borbely AA. Sleep homeostasis in the rat: simulation of the time course of
EEG slow-wave activity. Neurosci Lett. 1991; 130:141–44. [PubMed: 1795873]
110. Franken P, Chollet D, Tafti M. The homeostatic regulation of sleep need is under genetic control.
J Neurosci. 2001; 21:2610–21. [PubMed: 11306614]
111. Stone TW, Ceruti S, Abbracchio MP. Adenosine receptors and neurological disease:
neuroprotection and neurodegeneration. Handb Exp Pharmacol. 2009; 193:535–87. [PubMed:
19639293]
112. Gradinaru V, Mogri M, Thompson KR, Henderson JM, Deisseroth K. Optical deconstruction of
parkinsonian neural circuitry. Science. 2009; 324:354–59. [PubMed: 19299587]
113. Van Dort CJ, Baghdoyan HA, Lydic R. Adenosine A1 and A2A receptors in mouse prefrontal
cortex modulate acetylcholine release and behavioral arousal. J Neurosci. 2009; 29:871–81.
[PubMed: 19158311]
Halassa and Haydon Page 20
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
114. Poulet JF, Petersen CC. Internal brain state regulates membrane potential synchrony in barrel
cortex of behaving mice. Nature. 2008; 454:881–85. [PubMed: 18633351]
115. Vyazovskiy VV, Cirelli C, Pfister-Genskow M, Faraguna U, Tononi G. Molecular and
electrophysiological evidence for net synaptic potentiation in wake and depression in sleep. Nat
Neurosci. 2008; 11:200–8. [PubMed: 18204445]
116. Kotagal P, Yardi N. The relationship between sleep and epilepsy. Semin Pediatr Neurol. 2008;
15:42–49. [PubMed: 18555190]
117. Ridet JL, Malhotra SK, Privat A, Gage FH. Reactive astrocytes: cellular and molecular cues to
biological function. Trends Neurosci. 1997; 20:570–57. [PubMed: 9416670]
118. Escartin C, Brouillet E, Gubellini P, Trioulier Y, Jacquard C, et al. Ciliary neurotrophic factor
activates astrocytes, redistributes their glutamate transporters GLAST and GLT-1 to raft
microdomains, and improves glutamate handling in vivo. J Neurosci. 2006; 26:5978–89.
[PubMed: 16738240]
119. Li L, Lundkvist A, Andersson D, Wilhelmsson U, Nagai N, et al. Protective role of reactive
astrocytes in brain ischemia. J Cereb Blood Flow Metab. 2008; 28:468–81. [PubMed: 17726492]
120. Oberheim NA, Tian GF, Han X, Peng W, Takano T, et al. Loss of astrocytic domain organization
in the epileptic brain. J Neurosci. 2008; 28:3264–76. [PubMed: 18367594]
121. Buffo A, Rite I, Tripathi P, Lepier A, Colak D, et al. Origin and progeny of reactive gliosis: a
source of multipotent cells in the injured brain. Proc Natl Acad Sci USA. 2008; 105:3581–86.
[PubMed: 18299565]
122. Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, et al. Leukocyte infiltration,
neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes
in adult transgenic mice. Neuron. 1999; 23:297–308. [PubMed: 10399936]
123. Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive
astrocytes in traumatic brain injury. Brain. 2006; 129:2761–72. [PubMed: 16825202]
124. Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive astrocytes
protect tissue and preserve function after spinal cord injury. J Neurosci. 2004; 24:2143–55.
[PubMed: 14999065]
125. Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, et al. Conditional ablation of Stat3 or
Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med. 2006;
12:829–34. [PubMed: 16783372]
126. Sofroniew MV. Reactive astrocytes in neural repair and protection. Neuroscientist. 2005; 11:400–
7. [PubMed: 16151042]
127. Wilcock DM, Vitek MP, Colton CA. Vascular amyloid alters astrocytic water and potassium
channels in mouse models and humans with Alzheimer’s disease. Neuroscience. 2009;
159:1055–69. [PubMed: 19356689]
128. Ballas N, Lioy DT, Grunseich C, Mandel G. Non-cell autonomous influence of MeCP2-deficient
glia on neuronal dendritic morphology. Nat Neurosci. 2009; 12:311–17. [PubMed: 19234456]
129. Barres BA. The mystery and magic of glia: a perspective on their roles in health and disease.
Neuron. 2008; 60:430–40. [PubMed: 18995817]
130. Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous hyperactivity and
intercellular calcium waves in astrocytes in Alzheimer mice. Science. 2009; 323:1211–15.
[PubMed: 19251629]
131. Ding S, Fellin T, Zhu Y, Lee SY, Auberson YP, et al. Enhanced astrocytic Ca2+ signals
contribute to neuronal excitotoxicity after status epilepticus. J Neurosci. 2007; 27:10674–84.
[PubMed: 17913901]
132. Hardingham GE, Fukunaga Y, Bading H. Extrasynaptic NMDARs oppose synaptic NMDARs by
triggering CREB shut-off and cell death pathways. Nat Neurosci. 2002; 5:405–14. [PubMed:
11953750]
133. Cotter DR, Pariante CM, Everall IP. Glial cell abnormalities in major psychiatric disorders: the
evidence and implications. Brain Res Bull. 2001; 55:585–95. [PubMed: 11576755]
134. Choudary PV, Molnar M, Evans SJ, Tomita H, Li JZ, et al. Altered cortical glutamatergic and
GABAergic signal transmission with glial involvement in depression. Proc Natl Acad Sci USA.
2005; 102:15653–58. [PubMed: 16230605]
Halassa and Haydon Page 21
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
135. Banasr M, Chowdhury GM, Terwilliger R, Newton SS, Duman RS, et al. Glial pathology in an
animal model of depression: reversal of stress-induced cellular, metabolic and behavioral deficits
by the glutamate-modulating drug riluzole. Mol Psychiatry. 2008 In press.
136. Sanacora G, Saricicek A. GABAergic contributions to the pathophysiology of depression and the
mechanism of antidepressant action. CNS Neurol Disord Drug Targets. 2007; 6:127–40.
[PubMed: 17430150]
137. Lisman J, Buzsaki G. A neural coding scheme formed by the combined function of gamma and
theta oscillations. Schizophr Bull. 2008; 34:974–80. [PubMed: 18559405]
138. Tuominen HJ, Tiihonen J, Wahlbeck K. Glutamatergic drugs for schizophrenia: a systematic
review and meta-analysis. Schizophr Res. 2005; 72:225–34. [PubMed: 15560967]
139. Halassa MM, Fellin T, Haydon PG. The tripartite synapse: roles for gliotransmission in health
and disease. Trends Mol Med. 2007; 13:54–63. [PubMed: 17207662]
Halassa and Haydon Page 22
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
SUMMARY POINTS
1. Astrocytes are physically associated with synapses and provide the structural
substrate for reciprocal chemical signaling between neurons and glia.
2. Astrocytes provide important metabolic support to the neuron that is required to
maintain physiological properties of neuronal networks.
3. The astrocyte is chemically excitable and expresses a plethora of receptors that
allow the detection of neuronal activity and induce second messenger signaling
within these glial cells.
4. Chemical gliotransmitters can be released from astrocytes through a variety of
mechanisms. These pathways lead to the release of glutamate, D-serine, and
ATP.
5. Astrocyte-derived glutamate plays important roles in behavioral responses to
cocaine and the growth of glioblastoma. D-serine modulates NMDA receptor
function, whereas ATP and its metabolic product adenosine modulate synaptic
transmission and promote sleep drive.
Halassa and Haydon Page 23
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FUTURE ISSUES
1. How do astrocytes and gliotransmission modulate inhibitory synaptic
transmission?
2. Could astrocytes act as therapeutic targets in which one hijacks the glial
signaling pathways to modulate and repair defective synaptic transmission,
circuit function, and behavior?
3. What is the range of behaviors for which astrocytes modulate neuronal network
activity?
4. How do astrocytes integrate the period of wakefulness to promote sleep drive?
Halassa and Haydon Page 24
Annu Rev Physiol. Author manuscript; available in PMC 2011 June 17.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... Mounting experimental evidence suggests a significant role for glial cells, particularly astrocytes, in shaping neuronal activity in both cell cultures and the brain, driving interest in exploring multi-component network dynamics [1]. These systems are modeled at varying levels of physiological detail, ranging from the Hodgkin-Huxley and Ullah models for neurons and astrocytes, respectively, to simple integrate-and-fire models or phase oscillators. ...
... АННОТАЦИЯ Экспериментальные результаты, указывающие на роль глиальных клеток, в особенности астроцитов, в формировании нейрональной активности как в культуральных средах, так и в мозге подкрепляют интерес к изучению динамических свойств многокомпонетных сетей [1]. Для исследования таких систем используются математические модели различной степени физиологической детализации: от моделей Ходжкина-Хаксли и Уллаха для нейронов и астроцитов соответственно до сильно упрощённых систем типа накопление-сброс и фазовых осцилляторов. ...
Article
Full-text available
Mounting experimental evidence suggests a significant role for glial cells, particularly astrocytes, in shaping neuronal activity in both cell cultures and the brain, driving interest in exploring multi-component network dynamics [1]. These systems are modeled at varying levels of physiological detail, ranging from the Hodgkin-Huxley and Ullah models for neurons and astrocytes, respectively, to simple integrate-and-fire models or phase oscillators. Multiplex networks are commonly used to model generic dynamical phenomena, with an emphasis on network connectivity effects rather than the complexity of an individual cell’s behavior. This model is particularly advantageous due to its use of a two-layer network structure [2]. This structure accurately describes neural connectivity through the long-range random coupling, while glial cells interact locally through the diffusion of mediatory molecules. Furthermore, the oscillatory timescales between neural and glial cells differ by at least one order of magnitude. In this context, the analysis of synchronization is a focal point as it is a crucial process that underpins information processing, decision making, and movement control in living neural systems [3]. Network topology is known to be essential, where all-to-all coupling and random Erdos–Renyi connectivity support the second-order phase transition to global phase coherence. In contrast, regular local coupling such as a 2D lattice only permits frequency and phase locking. Additionally, symplectic networks are capable of demonstrating a first-order phase transition, which is commonly referred to as “explosive” synchronization [4]. Recently, studies have shown that combining model neural (random) and glial (regular lattice) oscillatory layers can lead to a variety of outcomes [5]. Specifically, this combination was found to induce a second-order phase transition in the regular glial layer. In addition, synchronization in both layers may be preceded by desynchronization as the coupling between the layers becomes stronger. Here we investigate synchronization in the multiplex neural-glial network, where the neural layer is symplectic and contains triadic interactions. We demonstrate that symplectic coupling in the neural layer can induce the first order transition in the regular (glial) layer. Additionally, we show that such a transition can be induced by strengthening the glial and interlayer coupling, even if the symplectic neural layer alone is below the synchronization threshold. Desynchronization in the neuronal layer, resulting from the moderated coupling to the glial layer, never occurs abruptly.
... 54 In addition, astrocytes secrete cytokines that regulate the survival and differentiation of neurons and the formation and elimination of synapses. 55,56 During postnatal development, astrocytes eliminate synapses by phagocytosis through the EGF-like domain 10 (MEGF10) and MERTK receptors and actively contribute to activity-dependent synaptic pruning and developmental refinement of circuits. 57 Moreover, contrary to the previous notion that microglia are the sole mediators of synapse elimination, astrocytes have been shown to play an important role in synaptic pruning during brain development. ...
Article
Full-text available
Background Stroke is an acute cerebrovascular disease in which brain tissue is damaged due to sudden obstruction of blood flow to the brain or the rupture of blood vessels in the brain, which can prompt ischemic or hemorrhagic stroke. After stroke onset, ischemia, hypoxia, infiltration of blood components into the brain parenchyma, and lysed cell fragments, among other factors, invariably increase blood–brain barrier (BBB) permeability, the inflammatory response, and brain edema. These changes lead to neuronal cell death and synaptic dysfunction, the latter of which poses a significant challenge to stroke treatment. Results Synaptic dysfunction occurs in various ways after stroke and includes the following: damage to neuronal structures, accumulation of pathologic proteins in the cell body, decreased fluidity and release of synaptic vesicles, disruption of mitochondrial transport in synapses, activation of synaptic phagocytosis by microglia/macrophages and astrocytes, and a reduction in synapse formation. Conclusions This review summarizes the cellular and molecular mechanisms related to synapses and the protective effects of drugs or compounds and rehabilitation therapy on synapses in stroke according to recent research. Such an exploration will help to elucidate the relationship between stroke and synaptic damage and provide new insights into protecting synapses and restoring neurologic function.
... Therefore, astrocytes encode and decode neuronal information by exhibiting Ca 2+ transients of various frequency and amplitude depending on the neighboring synaptic activities (Volterra et al., 2014). Moreover, as a consequence of the intracellular Ca 2+ elevations, astrocytes release various chemical substances termed as gliotransmitters to modulate the function of closely located neurons and glial cells (Araque et al., 2001;Halassa and Haydon, 2010;Parpura and Zorec, 2010;Parpura and Verkhratsky, 2012;Harada et al., 2015). Microglia also express receptors for various neurotransmitters which can maintain their quiescent state or promote a phenotypic switch (Pocock and Kettenmann, 2007;Liu et al., 2016). ...
Article
Full-text available
The endogenous cannabinoid 2-arachidonoylglycerol (2-AG) influences neurotransmission in the central nervous system mainly by activating type 1 cannabinoid receptor (CB1). Following its release, 2-AG is broken down by hydrolases to yield arachidonic acid, which may subsequently be metabolized by cyclooxygenase-2 (COX-2). COX-2 converts arachidonic acid and also 2-AG into prostanoids, well-known inflammatory and pro-nociceptive mediators. Here, using immunohistochemical and biochemical methods and pharmacological manipulations, we found that reactive spinal astrocytes and microglia increase the expression of COX-2 and the production of prostaglandin E2 when exposed to 2-AG. Both 2-AG and PGE2 evoke calcium transients in spinal astrocytes, but PGE2 showed 30% more efficacy and 55 times more potency than 2-AG. Unstimulated spinal dorsal horn astrocytes responded to 2-AG with calcium transients mainly through the activation of CB1. 2-AG induced exaggerated calcium transients in reactive astrocytes, but this increase in the frequency and area under the curve of calcium signals was only partially dependent on CB1. Instead, aberrant calcium transients were almost completely abolished by COX-2 inhibition. Our results suggest that both reactive spinal astrocytes and microglia perform an endocannabinoid-prostanoid switch to produce PGE2 at the expense of 2-AG. PGE2 in turn is responsible for the induction of aberrant astroglial calcium signals which, together with PGE2 production may play role in the development and maintenance of spinal neuroinflammation-associated disturbances such as central sensitization.
... Despite the large number of identified neuronal mRNA targets of FMRP, the mechanisms underlying the critical dysfunctions in FXS are still unclear and no effective therapy exists so far, making the search for novel targets of prime importance. Interestingly, non-neuronal cells such as astrocytes, the most abundant brain glial cells, intimately interact with neurons at the synaptic and network levels and actively regulate their activities via various mechanisms 18,19 . Astrocytes thus participate to brain physiology and pathologies 20,21 , including several neurodevelopmental disorders such as FXS 22,23 . ...
Article
Full-text available
Fragile X syndrome (FXS) is an inherited form of intellectual disability caused by the loss of the mRNA-binding fragile X mental retardation protein (FMRP). FXS is characterized by neuronal hyperexcitability and behavioral defects, however the mechanisms underlying these critical dysfunctions remain unclear. Here, using male Fmr1 knockout mouse model of FXS, we identify abnormal extracellular potassium homeostasis, along with impaired potassium channel Kir4.1 expression and function in astrocytes. Further, we reveal that Kir4.1 mRNA is a binding target of FMRP. Finally, we show that the deficit in astroglial Kir4.1 underlies neuronal hyperexcitability and several behavioral defects in Fmr1 knockout mice. Viral delivery of Kir4.1 channels specifically to hippocampal astrocytes from Fmr1 knockout mice indeed rescues normal astrocyte potassium uptake, neuronal excitability, and cognitive and social performance. Our findings uncover an important role for astrocyte dysfunction in the pathophysiology of FXS, and identify Kir4.1 channel as a potential therapeutic target for FXS.
... They influence the activity of individual synapses, and recent evidence shows that they act as key elements in experience-dependent wiring of brain circuits (Araque and others 2014;Pascual and others 2005;Zhang and others 2015) and in the closing of critical periods of plasticity (Andrade-Talavera, Pérez-Rodríguez and others 2023; Falcón-Moya and others 2020; Martínez-Gallego, Pérez-Rodríguez, Coatl-Cuaya, and others 2022; Martínez-Gallego and Rodríguez-Moreno 2023; Pérez-Rodríguez and others 2019). These glial cells have receptors of neurotransmitters, being able to detect and respond to neural activity releasing gliotransmitters, which may act on neurons (Haydon 2001;Halassa and Haydon 2010;Volterra and Meldolesi 2005). Indeed, one of the central themes of the tripartite synapse is that astrocytes sense the synaptic inputs and respond with intracellular Ca 2 + elevations, which can elicit the release of gliotransmitters (Perea and Araque 2005 (Haydon and Carmignoto 2006;Volterra and Steinhauser 2004), and in synaptic plasticity (for review, see Durkee and others 2021). ...
Article
Brain plasticity is the ability of the nervous system to change its structure and functioning in response to experiences. These changes occur mainly at synaptic connections, and this plasticity is named synaptic plasticity. During postnatal development, environmental influences trigger changes in synaptic plasticity that will play a crucial role in the formation and refinement of brain circuits and their functions in adulthood. One of the greatest challenges of present neuroscience is to try to explain how synaptic connections change and cortical maps are formed and modified to generate the most suitable adaptive behavior after different external stimuli. Adenosine is emerging as a key player in these plastic changes at different brain areas. Here, we review the current knowledge of the mechanisms responsible for the induction and duration of synaptic plasticity at different postnatal brain development stages in which adenosine, probably released by astrocytes, directly participates in the induction of long-term synaptic plasticity and in the control of the duration of plasticity windows at different cortical synapses. In addition, we comment on the role of the different adenosine receptors in brain diseases and on the potential therapeutic effects of acting via adenosine receptors.
... The role of astrocytes in post-traumatic epileptogenesis (PTE) development has seen a significant paradigm shift, with a growing understanding that extends beyond their traditional role as mere support cells [17]. Recent research underscores their multifaceted functions, particularly in the pathogenic inflammatory response and direct involvement in various neuronal functions, underscoring their integral role in brain activities such as learning, memory, and sleep [18][19][20][21]. Astrocytes, the most prevalent cell type in the brain, are pivotal in maintaining ionic homeostasis, preserving the integrity of the blood-brain barrier, regulating neurotransmitter metabolism, and ensuring neuronal energy supply. ...
Article
Full-text available
This paper presents an in-depth exploration of Post-Traumatic Epilepsy (PTE), a complex neurological disorder following traumatic brain injury (TBI), characterized by recurrent, unprovoked seizures. With TBI being a global health concern, understanding PTE is crucial for effective diagnosis, management, and prognosis. This study aims to provide a comprehensive overview of the epidemiology, risk factors, and emerging biomarkers of PTE, thereby informing clinical practice and guiding future research. The epidemiological aspect of the study reveals PTE as a significant contributor to acquired epilepsies, with varying incidence influenced by injury severity, age, and intracranial pathologies. The paper delves into the multifactorial nature of PTE risk factors, encompassing clinical, demographic, and genetic elements. Key insights include the association of injury severity, intracranial hemorrhages, and early seizures with increased PTE risk, and the roles of age, gender, and genetic predispositions. Advancements in neuroimaging, electroencephalography, and molecular biology are presented, highlighting their roles in identifying potential PTE biomarkers. These biomarkers, ranging from radiological signs to electroencephalography EEG patterns and molecular indicators, hold promise for enhancing PTE pathogenesis understanding, early diagnosis, and therapeutic guidance. The paper also discusses the critical roles of astrocytes and microglia in PTE, emphasizing the significance of neuroinflammation in PTE development. The insights from this review suggest potential therapeutic targets in neuroinflammation pathways. In conclusion, this paper synthesizes current knowledge in the field, emphasizing the need for continued research and a multidisciplinary approach to effectively manage PTE. Future research directions include longitudinal studies for a better understanding of TBI and PTE outcomes, and the development of targeted interventions based on individualized risk profiles. This research contributes significantly to the broader understanding of epilepsy and TBI.
Article
It was shown that abnormalities in astrocyte functions can cause alterations in neuronal excitability and promote the development of epilepsy, however, this question claims further investigation. The aim of the present work was to analyze the functional state of astrocytes in the hippocampus of Krushinsky-Molodkina (KM) rats genetically predisposed to the reflex audiogenic seizures. Adult naïve animals with fully-established audiogenic sensitivity were recruited in our experiments. Rats of parental Wistar strain were used as a control. Obtained results revealed unchanged protein expression of such astocytic markers as GFAP, ALDH1L1, and NFIA suggesting the absence of the reactive astrogliosis in the hippocampus of KM rats. SPARC and aquaporin 4 expression also did not differ from the control indicating no violations in astroglia regulation of synaptogenesis and water balance. Analysis of glial proteins responsible for reuptake and metabolism of neurotransmitters revealed normal expression of GABA transporter GAT-3 and glutamine synthetase, while glutamate transporters EAAT1 and 2 were significantly elevated pointing on enhanced activity of glutamate removal from synapses. In addition, decreased expression of the key glycolytic enzyme aldolase C, probably, indicated insufficient activity of glucose metabolism in astrocytes. Thus, obtained data pointed on genetically determined alterations of astroglia functions in the hippocampus of rats with inherited reflex epilepsy.
Article
Full-text available
Animals often behave repetitively and predictably. These repetitive behaviors can have a component that is learned and ingrained as habits, which can be evolutionarily advantageous as they reduce cognitive load and the expenditure of attentional resources. Repetitive behaviors can also be conscious and deliberate, and may occur in the absence of habit formation, typically when they are a feature of normal development in children, or neuropsychiatric disorders. They can be considered pathological when they interfere with social relationships and daily activities. For instance, people affected by obsessive-compulsive disorder, autism spectrum disorder, Huntington’s disease and Gilles de la Tourette syndrome can display a wide range of symptoms like compulsive, stereotyped and ritualistic behaviors. The striatum nucleus of the basal ganglia is proposed to act as a master regulator of these repetitive behaviors through its circuit connections with sensorimotor, associative, and limbic areas of the cortex. However, the precise mechanisms within the striatum, detailing its compartmental organization, cellular specificity, and the intricacies of its downstream connections, remain an area of active research. In this review, we summarize evidence across multiple scales, including circuit-level, cellular, and molecular dimensions, to elucidate the striatal mechanisms underpinning repetitive behaviors and offer perspectives on the implicated disorders. We consider the close relationship between behavioral output and transcriptional changes, and thereby structural and circuit alterations, including those occurring through epigenetic processes.
Article
Full-text available
Astrocyte glutamate release can modulate synaptic activity and participate in brain intercellular signaling. P2X7 receptors form large ion channels when activated by ATP or other ligands. Here we show that P2X7 receptors provide a route for excitatory amino acid release from astrocytes. Studies were performed using murine cortical astrocyte cultures. ATP produced an inward current in patch-clamped astrocytes with properties characteristic of P2X7 receptor activation: the current was amplified in low divalent cation medium, blocked by pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), and more potently activated by 3'-O-(4-benzoyl)benzoyl ATP (BzATP) than by ATP itself. Measurement of current reversal potentials showed the relative BzATP-induced permeabilities to different substrates to be Na+, 1 > Cl-, 0.34 > N-methyl-D-glucamine, 0.27 > L-glutamate, 0.15 approximately D-aspartate, 0.16. Astrocytes exposed to BzATP also became permeable to Lucifer yellow, indicating a large channel opening. Release of L-glutamate and D-aspartate through P2X7 channels was confirmed using radiolabeled tracers. As with the inward current, release of glutamate and D-aspartate was induced by BzATP more potently than ATP, amplified in Ca2+/Mg2+-free medium, and blocked by PPADS or oxidized ATP. Efflux through P2X7 channels is a previously unrecognized route of ligand-stimulated, nonvesicular astrocyte glutamate release.
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
Genetic variation in the expression and regulation of sleep was assessed in six inbred mice strains (AK, C, B6, BR, D2, 129). The amount, distribution, and fragmentation of the behavioral states wakefulness (W), slow-wave sleep (SWS), and paradoxical sleep (PS), as well as EEG delta power in SWS, were determined and compared among strains and between baseline and recovery from a 6-hour sleep deprivation (SD) starting at lights-on. In baseline, the most striking strain differences concerned sleep amount, the onset and duration of the main rest period, and SWS fragmentation. The time course of delta power in SWS during the main rest period was similar between strains. Immediately following the SD, high delta power values were reached (higher for AK than for 129). However, the relative increase in delta power, compared to the first 6 hours of the baseline rest period, was not strain-specific. Over the first 6 hours of recovery, W was decreased and PS increased in AK, B6, BR, and 129. In C and D2, time spent in any of the states was not affected by the SD. In contrast, in the recovery dark period, SWS and PS were invariably increased. In recovery, SWS fragmentation was strongly reduced for D2, resulting in the disappearance of the strain differences observed in baseline. Since these inbred strains are fully homozygous and thus can be considered genetic clones, the sleep-related strain differences reported here can be attributed to differences in genotype. Therefore, this study provides a basis for the identification of genetic factors underlying sleep and its regulation.