You are on page 1of 10

Review

Astrocytes and the regulation of


cerebral blood flow
Raymond C. Koehler1, Richard J. Roman2 and David R. Harder2,3
1
Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
2
Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
3
Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA

Moment-to-moment changes in local neuronal activity hippocampus [2–4]. In the human cortex, a single astrocyte
lead to dynamic changes in cerebral blood flow. Emer- might sense the activity and regulate the function of more
ging evidence implicates astrocytes as one of the key than one million synapses within its domain [4]. Moreover,
players in coordinating this neurovascular coupling. each astrocyte has at least one process with endfeet sur-
Astrocytes are poised to sense glutamatergic synaptic rounding a blood vessel [5]. By contrast, processes from
activity over a large spatial domain via activation of neurons are rarely in direct contact with intraparenchymal
metabotropic glutamate receptors and subsequent blood vessels. This anatomical relationship has led to the
calcium signaling and via energy-dependent glutamate hypothesis that astrocytes have a pivotal role in the
transport. Astrocyte foot processes can signal vascular dynamic regulation of the cerebral circulation [6,7].
smooth muscle by arachidonic acid pathways involving Blood delivered to the major cerebral arteries is dis-
astrocytic cytochrome P450 epoxygenase, astrocytic tributed over the cortical surface through pial arteries that
cyclooxygenase-1 and smooth muscle cytochrome give rise to penetrating arterioles. Pial arteries are in close
P450 v-hydroxylase activities, and by astrocytic and proximity to the underlying glia limitans, and intrapar-
smooth muscle potassium channels. Non-glutamatergic enchymal vessels emerging from penetrating arterioles
transmitters released from neurons, such as nitric oxide, beyond the space of Virchow are in direct contact with foot
cyclooxygenase-2 metabolites and vasoactive intestinal processes of astrocytes. In capillaries and venules without
peptide, might modulate neurovascular signaling at the smooth muscle, integrins maintain adhesion of foot pro-
level of the astrocyte or smooth muscle. Thus, astrocytes cesses with the basal lamina surrounding the endothelium
have a pivotal role in dynamic signaling within the and pericytes. Signaling between astrocytes and endo-
neurovascular unit. Important questions remain on thelium is important for angiogenesis, maintenance of
how this signaling is integrated with other pathways endothelial tight junctions and transport across the
in health and disease. blood–brain barrier. More recently, signaling between
astrocytes and vascular smooth muscle (VSM) in intrapar-
Introduction enchymal arterioles and between glia limitans and pial
Steady-state regional cerebral blood flow (CBF) is matched arteries has assumed increasing importance in the regu-
to regional energy metabolism. However, increases in local lation of CBF. Changes in CBF are actively controlled by
neuronal activity lead to dynamic increases in CBF that changes in VSM tone, leading to dilation or constriction of
exceed the increases in oxidative metabolism. This relative pial arteries and intraparenchymal arterioles. With wide-
hyperemic response provides an increased oxygen gradient spread neuronal activation, dilation of pial arteries is
between blood vessels and tissue for assuring adequate coordinated with intraparenchymal dilation to help main-
oxygen diffusion to the most distant mitochondria when tain a constant perfusion pressure in penetrating arter-
oxygen demand fluctuates. Here, we focus on the role of ioles and prevent a passive decrease in CBF in other brain
astrocytes in coordinating the vascular response to regions. This coordination is thought to depend on sig-
neuronal activation. naling through astrocytes [8] and possibly occur by ascend-
Our understanding of the multifunctional role of astro- ing dilation propagated through the endothelium [9].
cytes and their relationships with neurons, blood vessels Measurements with laser-Doppler flowmetry (LDF),
and other astrocytes has grown substantially in recent which measures red-blood-cell flux integrated in cortical
years. Astrocytes are now known to have more of a spongi- tissue typically over 1 mm depth, show increases in CBF
form appearance than stellate appearance because of the in the primary somatosensory cortex within 1–2 s of sen-
many fine processes that extend beyond the large processes sory stimulation [10]. Similar response times are evident
that typically stain for intermediate filaments. These fine with intrinsic monitoring of tissue hemoglobin concen-
processes have minimal spatial overlap with the processes tration and with functional magnetic resonance imaging
of other astrocytes and result in an individual astrocytic (fMRI), which uses the blood-oxygen-level-dependent sig-
domain that is thought to contain 300–600 neuronal nal as a surrogate for CBF coupled to neuronal activity [11–
dendrites [1] and 105 synapses in the rodent cortex and 13]. More recent work has employed Ca2+ imaging in brain
slices and two-photon imaging in the intact brain to reveal
Corresponding author: Koehler, R.C. (rkoehler@jhmi.edu). involvement of astrocytes in the coupling of CBF with
160 0166-2236/$ – see front matter ß 2008 Elsevier Ltd. All rights reserved. doi:10.1016/j.tins.2008.11.005 Available online 21 January 2009
Review Trends in Neurosciences Vol.32 No.3

neuronal activation. One way astrocytes might monitor peripheral circulation began to reveal an important role of
excitatory presynaptic activity is by glutamate activation arachidonic acid metabolites derived from specific cyto-
of metabotropic glutamate (mGlu) receptors, which are chrome P450 (CYP) proteins [18]. One class of CYP-derived
known to stimulate phospholipase C and increase astro- eicosanoids formed by epoxygenation of arachidonic acid
cytic Ca2+. Here, we summarize some of these recent comprises the four regioisomers of epoxyeicosatrienoic
findings that illustrate the complexity of dynamic com- acids (EETs). EETs were shown to hyperpolarize VSM
munication between cell types within the neurovascular by opening calcium-sensitive potassium (KCa) channels
unit with an emphasis on signaling by arachidonic acid [19]. The mechanism is thought to involve EETs activation
metabolites and the integration with other signaling path- of vanilloid transient receptor potential 4 (TRPV4) chan-
ways. nels, which produce ryanodine-receptor-dependent
increases in local Ca2+ sufficient to activate nearby KCa
Cytochrome P450 epoxygenase metabolites channels [20], although ADP ribosylation and phosphoryl-
By the mid-1990s, several groups suggested a role for nitric ation mechanisms might also be involved [21,22]. In the
oxide (NO) derived from neurons and for adenosine in peripheral circulation, EETs might serve as one of several
contributing to vasodilation during neural activation endothelial-derived hyperpolarizing factors that relax
[14–16]. However, these molecules did not account for VSM. In the brain, astrocytes might serve a similar role
the entire vascular response [17]. In 1998, Harder et al. in providing glial-derived hyperpolarizing factors. EETs
[7] proposed that astrocyte signaling involving epoxygen- are synthesized in astrocytes and can be further metab-
ase metabolites of arachidonic acid could have an import- olized to less bioactive dihydroxyeicosatrienoic acids by
ant role in neurovascular coupling. Cyclooxygenase (COX) soluble epoxide hydrolase [23] or stored in membrane
products of arachidonic acid metabolism had traditionally phospholipid pools [24]. The epoxygenase enzyme CYP
been thought to be the predominant eicosanoids involved 2C11 is expressed in rat astrocytes and produces EETs
in vascular signaling. However, in the 1980s, work in the [25], whereas soluble epoxide hydrolase is enriched in

Figure 1. Simplified schematic diagram of selected aspects of glutamatergic signaling in an astrocytic synaptic process that results in arachidonic acid (AA) metabolite
signaling in an astrocyte foot process with vascular smooth muscle (VSM). Presynaptic release of glutamate (Glu) acts on postsynaptic AMPA receptors (GluRs), NMDA
receptors (NMDARs) and metabotropic glutamate receptors (mGluRs). Glu is taken up in the synaptic astrocyte process by the glutamate transporter GLT-1 with cotransport
of Na+, which stimulates energy-dependent Na+,K+-ATPase and consequent glucose uptake. Glu also activates astrocytic mGluR coupled to phospholipase C (PLC) and
Ins(1,4,5)P3 (IP3)-induced Ca2+ increases, which can be propagated throughout the astrocyte by release of ATP through hemichannels. Extracellular ATP acts on astrocytic
purinergic-2 receptors (P2Rs) to promote further intracellular Ca2+ release and is also metabolized to adenosine, which can stimulate astrocytic A2B receptors (A2BRs) and
which might also promote Ca2+ release. Increased Ca2+ in the astrocyte foot process is thought to stimulate phospholipase A2 (PLA2) and mobilize AA. Mobilized AA can
serve as a substrate for (i) astrocytic cyclooxygenase-1 (COX-1) and generate prostaglandin E2 (PGE2), which can stimulate vasodilatory cAMP formation in VSM, (ii)
cytochrome P450 2C11 (CYP 2C11) and generate vasodilatory epoxyeicosatrienoic acids (EETs), which are known to activate hyperpolarizing Ca2+-sensitive K+ (KCa)
channels, and (iii) cytochrome P450 4A (CYP 4A) in VSM and generate 20-HETE, which closes KCa channels and results in depolarization and constriction. In a small fraction
of postsynaptic neurons, NMDARs are coupled to neuronal nitric oxide (NO) synthase (nNOS), which generates highly diffusible NO that stimulates VSM guanylyl cyclase
(GC) and inhibits CYP 4A, thereby enabling astrocytic EETs to activate KCa channels. Baseline VSM constriction is also countered by release of NO from endothelial NOS
(eNOS) and by prostaglandin I2 (PGI2) from endothelial COX-1, both of which are sensitive to the mechanical shear stress of moving blood on the luminal wall. The broken
arrow indicates an inhibitory effect.

161
Review Trends in Neurosciences Vol.32 No.3

neurons [26]. Addition of glutamate to the media of cul- [45] reported that neuronal stimulation resulted in an
tured astrocytes releases both arachidonic acid and EETs increase in Ca2+ in a fraction of the astrocytes surrounding
into the media [27]. It was observed that, in isolated blood vessels and that the increase in Ca2+ was associated
cerebral VSM, EETs open KCa channels and result in with an increase in blood vessel diameter. Subtype 1 and 5
hyperpolarizion and vasorelaxation [7,19,25] and that, in of group I mGlu receptor antagonists blocked the increase
vivo, topical application of EETs on pial arterioles in astrocyte Ca2+ and the vasodilation without attenuating
increases their diameter [28–30]. These observations led the increase in neuronal Ca2+. Because aspirin attenuated
to the hypothesis that stimulation of glutamate receptors the dilation [45] and because cultured astrocytes release
on astrocytes leads to the formation and release of EETs, prostaglandin (PG)E2 in response to glutamate [46], COX-
which then act on cerebral VSM to cause vasodilation derived PGE2 is thought to trigger the vasodilation. One
(Figure 1). limitation of this study was that the time course of the
In vivo support for this hypothesis is derived from astrocyte Ca2+ response and vasodilation was much slower
several studies on rats. N-methyl-D-aspartic acid (NMDA) than the in vivo response. Nevertheless, work in vivo
perfusion through microdialysis probes implanted in the demonstrated that the mGlu receptor antagonists attenu-
striatum elicited an increase in local CBF that was blocked ated the increase in CBF during whisker stimulation
not only by an inhibitor of NO synthase (NOS) but also by [34,45]. The role for astrocytic ionotropic glutamate recep-
inhibitors of epoxygenase [31]. Physiologic activation of the tors in neurovascular coupling has yet to be determined.
cerebral cortex by movement of the whiskers or by elec- Mechanisms of Ca2+ signaling might differ in astrocyte
trical forepaw stimulation produced increases in cortical processes directed at blood vessels and in processes
CBF that were attenuated by inhibitors of EETs synthesis directed at synapses or in the astrocyte soma. Straub
[32,33]. Moreover, an EET antagonist also suppressed the et al. [47] focused on Ca2+ responses within endfeet and
evoked increase in CBF [34]. Therefore, EETs participate observed them to be spatially heterogeneous and to
in the signaling that couples neuronal activation with precede vasodilation in mouse cortical slices. The
vasodilation. responses depended on inositol (1,4,5)-trisphosphate
In addition to acting as a paracrine signaling molecule, [Ins(1,4,5)P3] rather than on ryanodine receptors [47].
EETs might also serve as an intracellular messenger. One Retrograde Ca2+ waves were not observed. The increase
of the regioisomers, 5,6-EET, acts as a calcium influx factor in Ca2+ spread through the foot process longitudinally
to restore Ca2+ stores after depletion by thapsigargin in along the vessel wall but seemed to recover last at the
astrocytes [30]. Application of 11,12-EET to cortical slices point of origin, which is presumed to be a locus of high
increased astrocytic Ca2+ [35], although the mechanism for Ins(1,4,5)P3 receptor density. Whether these loci colocalize
the increase remains to be elucidated. Other evidence with the expression of phospholipase A2 and KCa channels
indicates that EETs facilitate the opening of KCa channels remains to be determined.
within astrocytes exposed to glutamate [36] or hypoxia
[37]. Intermittent hypoxia increases the expression of CYP Potassium signaling
2C11 and the formation of EETs in astrocytes [37,38]. This Astrocytes have long been known to have high per-
increase might contribute to the increased tolerance to meability to K+. Indeed, maintenance of a constant level
oxygen-glucose deprivation and ischemia [38,39]. More- of extracellular K+ during neuronal activity is an import-
over, increased formation of EETs in hypoxic astrocytes ant function of astrocytes for maintaining neuronal excit-
is associated with the formation of superoxide anion [37], ability. Early observations that astrocyte foot processes on
which might then serve as an intracellular signaling mol- blood vessels had a high density of K+ channels led to the
ecule or, under severe conditions, produce oxidative stress. hypothesis that neuronal release of K+ during action poten-
Therefore, the role of EETs as an intracellular messenger tials resulted in the uptake of K+ by astrocyte processes at
within astrocytes and as an intercellular messenger be- synapses that was counterbalanced by release of K+ at
tween astrocytes and other cells is complex and requires astrocyte foot processes surrounding blood vessels [6,48].
further investigation. Because of the tight interstitial space between astrocyte
foot processes and blood vessels [49], small amounts of K+
Signaling initiated by mGlu receptor activation release could result in transient elevation of extracellular
More direct evidence of a role for astrocytes in neurovas- K+ to levels sufficient to produce hyperpolarization of VSM
cular coupling has relied on observations in brain slices. In by activation of inward rectifier potassium (Kir) channels.
vitro, astrocytes are known to propagate Ca2+ waves This process of K+ siphoning was based on work in Mueller
throughout their processes by the release of ATP through cells in the retina. However, the more recent work demon-
connexin hemichannels and consequent activation of pur- strating intact neurovascular coupling in retina after gene
inergic receptors on their own processes and on adjacent deletion of the Kir 4.1 channel subtype expressed in Muel-
astrocytes [40–42]. Adjacent foot processes can also com- ler cells casts some doubt on the K+ siphoning mechanism
municate through gap junctions and lead to the spread of as an important player in neurovascular coupling in retina
signals along the vascular wall [5]. By acting in a coordi- [50].
nated manner, astrocytes might integrate neuronal Astrocyte foot processes are enriched with multiple
activity over large spatial domains and thereby coordinate types of K+ channels, including Kir, voltage-dependent
vasodilation within microcirculatory units. Neuronal K+ (Kv) and KCa channels [51,52]. A set of key studies
stimulation of brain slices was known to produce dilation elucidating direct communication between astrocytes and
of non-perfused blood vessels [43,44]. In 2003, Zonta et al. the vasculature via K+ was performed by Filosa et al.

162
Review Trends in Neurosciences Vol.32 No.3

[53,54]. By increasing vascular tone with the use of the no effect. Therefore, the working hypothesis derived from
thromboxane agonist U46619 in cortical slices, spon- this work is that activation of the astrocyte mGlu receptor
taneous oscillations in Ca2+ were induced in VSM. With produces an increased Ca2+ that is transmitted to the
neuronal stimulation or application of a mGlu receptor astrocyte foot process, where KCa channels are activated
agonist, increases in astrocyte Ca2+ were observed on a and sufficient K+ is released to open Kir 2.1 channels
timescale of a few seconds [53], which was more rapid than expressed on adjacent VSM, thereby leading to vasorelaxa-
that reported by Zonta et al. [45] who studied slices from tion (Figure 2).
younger animals at lower temperature. Interestingly, the
increase in astrocyte Ca2+ was associated with an immedi- 20-Hydroxyeicosatetraenoic acid signaling
ate suppression of Ca2+ oscillations in VSM and with Another eicosanoid, known as 20-hydroxyeicosatetraenoic
vasodilation. To further understand the mechanism of acid (20-HETE), produces depolarization of VSM at sub-
astrocyte–VSM coupling, the authors performed a patch nanomolar concentrations by closing KCa channels and
clamp study of the astrocyte foot process [54]. As expected, opening voltage-dependent Ca2+ channels [56,57]. CYP
large K+ currents were obtained in the foot process. How- enzymes that possess v-hydroxylase activity generate
ever, a significant portion of K+ current was attributed to 20-HETE from arachidonic acid. Isoforms of the CYP 4A
KCa channels. Others have also shown that inhibitors of family possess v-hydroxylase activity and are expressed in
KCa channels reduce the CBF response in vivo to whisker VSM, including cerebral VSM, where they produce 20-
stimulation [55]. Release of K+ through KCa channels on HETE [58,59]. Interestingly, work in hippocampal slices
foot processes is postulated to increase extracellular K+ in by Mulligan and MacVicar [60] demonstrated that pho-
the small space surrounding VSM. tolysis of astrocytic caged Ca2+ often produces constriction
To evaluate the role of Kir channels in the vascular of neighboring arterioles that were blocked by either a
response, Filosa et al. [54] showed that barium, an inhibi- phospholipase A2 or a 20-HETE-synthesis inhibitor. The
tor of Kir channels, reduces the evoked dilation in cortical predominance of a constrictor rather than a dilator
brain slices, whereas the COX inhibitor indomethacin has response in this preparation was probably related to the

Figure 2. Expanded view of selected aspects of potassium signaling in the astrocyte foot process and potential neuronal modulation. Increased Ca2+ in the astrocyte foot
process mediated by Ins(1,4,5)P3 (IP3) can activate KCa channels on the endfoot and release K+ into the small extracellular space, where it is postulated to transiently
accumulate to levels sufficient to stimulate VSM inward-rectifier K+ (Kir) channels and lead to VSM hyperpolarization and relaxation. EETs are also thought to promote
opening of astrocytic KCa channels in addition to VSM KCa channels. Because stimulation of perivascular interneurons containing vasoactive intestinal peptide (VIP) are
known to cause vasodilation and because VIP can increase Ca2+ in astrocyte endfeet, activation of these neurons might produce vasodilation through astrocytic Ca2+-
dependent mechanisms, such as activation of KCa channels and the PLA2 cascade. Subpopulations of neurons also express COX-2, which is required for the overall vascular
response to sensory activation, but the particular prostaglandin metabolite, receptor subtype and location of action on the astrocyte process, endfoot, vascular smooth
muscle or other neurons remains to be determined (as indicated by ‘?’). The broken arrow indicates an inhibitory effect.

163
Review Trends in Neurosciences Vol.32 No.3

low vascular tone of non-pressurized vessels because antagonist on the CBF response to whisker stimulation is
increasing vascular tone with an NOS inhibitor converted lost when NOS is inhibited and 20-HETE synthesis is
the constrictor response to a dilator response. Although the postulated to be disinhibited, whereas efficacy of the EETs
low vascular tone in brain slices is not physiologic, these antagonist is restored when both nNOS and 20-HETE
results raise the possibility that an increase in astrocytic synthesis are simultaneously inhibited [10]. Thus, unop-
Ca2+ can stimulate Ca2+-sensitive phospholipase A2 to posed 20-HETE synthesis might keep EETs from opening
mobilize sufficient arachidonic acid for diffusion to VSM, vasodilatory KCa channels. By suppressing 20-HETE syn-
where it can act as a substrate for 20-HETE synthesis thesis, NO might permit EETs-dependent vasodilation,
(Figure 1). Further support for the involvement of CYP thereby indicating a complex interplay of signaling among
metabolites of arachidonic acid has been obtained in neurons, astrocytes and VSM (Figure 1). Spatial hetero-
retinal slices, in which light or glial activation resulted geneity in local vasodilator and constrictor responses
in 20-HETE-dependent constriction in a portion of arter- might be related to spatial heterogeneity in the local NO
ioles and EETs-dependent dilation in a different set of concentrations above and below that necessary to inhibit
arterioles [61]. More recently, others have demonstrated CYP 4A.
that application of a mGlu receptor agonist in cortical slices
produces constriction in vessels with low baseline tone and Adenosine signaling
dilation in vessels preconstricted with a thromboxane Adenosine is a potent vasodilator and has long been impli-
analog [35]. Here, the constrictor response was reduced cated as a mediator of neurovascular coupling [15,17].
by inhibitors of phospholipase A2, 20-HETE synthesis and Indeed, adenosine A2A receptors have a role in dilation
COX-1, whereas the dilator responses were suppressed by of upstream pial arterioles during neuronal activation or
inhibitors of phospholipase A2, EETs synthesis and COX-1. direct activation of glutamatergic receptors [68,69]. In
Thus, the type of response elicited by astrocyte activation addition to acting on vascular adenosine receptors, adeno-
depends on the baseline activation state of the arteriole. sine could act on astrocytes, which also express various
These findings in brain slices raise the question of the types of adenosine receptors [70]. Activation of A2B recep-
relative importance of these mediators in vivo. Application tors can increase intracellular Ca2+ in astrocytes and
of a 20-HETE synthesis inhibitor in vivo had no effect on might participate in the propogation of increased Ca2+
the increase in CBF measured by LDF during whisker throughout the astrocyte processes [71]. Ordinarily, an
stimulation in mice and had only a small effect on enhan- increase in Ca2+ is associated with release of ATP through
cing the response in rats [10]. Thus, 20-HETE does not connexin hemichannels, and A2B receptors potentiate the
substantially constrain the evoked dilatory response in ATP-evoked Ca2+ response [72,73]. In addition to acting on
intact cortex. However, activation of discrete cortical col- astrocyte purinergic receptors, ATP can be hydrolyzed
umns can result in decreased perfusion in inhibitory-sur- sequentially to adenosine by ecto-enzymes that are
round cortex [62]. Even within the activated region, the enriched at hemichannel sites [70,74]. Localized increases
changes in red-blood-cell flux among capillaries are hetero- in extracellular adenosine might be sufficient to activate
geneous and a small proportion of capillaries exhibit a the A2B receptors (Figure 1), which have lower affinity for
decreased flux [63]. Whether the decreased perfusion is adenosine than do A1 and A2A receptors. Support for a role
the result of decreased tonic release of vasodilators or of A2B receptors in neurovascular coupling is derived from
increased release of vasoconstrictors or both is unknown, the use of different A2B antagonists that inhibit the CBF
but a role for 20-HETE in the inhibitory-surround response response to whisker stimulation [34]. Moreover, the lack of
or in the heterogenous response in the activated region is an additive effect of an EETs antagonist with an A2B
plausible. antagonist in reducing the CBF response is consistent with
The lack of a large effect of a 20-HETE inhibitor on the the A2B signaling acting sequentially with EETs signaling
overall CBF response in vivo could be the result of other within astrocytes. Another possibility is that ATP is
mechanisms suppressing 20-HETE synthesis or the effi- released at astrocyte endfeet and hydrolyzed to adenosine
cacy of 20-HETE on downstream signaling. After inhi- [5,75], which could then activate VSM A2A or A2B receptors.
bition of neuronal NOS (nNOS), the CBF response to However, a VSM A2A-dependent mechanism might be
whisker stimulation is decreased, and 20-HETE-synthesis more important in pial arterioles than in parenchymal
inhibition has a larger effect in enhancing the response VSM because A2A antagonists limit pial arteriolar dilation
than without nNOS inhibition [10]. Moreover, the 20- [76] but not CBF measured by LDF [34] during sensory
HETE-synthesis inhibitor was more effective in restoring activation. Furthermore, they do not limit ATP-induced
the CBF response after nNOS inhibition than after gua- dilation of penetrating arterioles [77].
nylyl cyclase inhibition. Because NO can inhibit CYP v-
hydroxylase activity [64,65] and can dilate cerebral Carbon monoxide signaling
arteries by both increasing cGMP and by inhibiting 20- Carbon monoxide (CO) derived from heme oxygenase
HETE synthesis [66,67], one interpretation is that neu- metabolism of heme has a prominent role in various
ronally derived NO during activation increases VSM NO aspects of cerebrovascular regulation in the newborn pig
concentration above the basal level provided by endothelial [78]. Application of glutamate to the cortical surface of
NOS and suppresses 20-HETE synthesis locally in neigh- piglet brain in vivo generates CO and dilates pial arteries
boring arterioles. Suppressed 20-HETE production per- in a glial-dependent manner [79]. The dilation depends on
mits vasodilation evoked by other mediators. Consistent activation of VSM KCa channels [80], where CO binds to a
with this interpretation, the inhibitory effect of an EETs heme ligand [81]. In cortical slices from piglet brain,

164
Review Trends in Neurosciences Vol.32 No.3

production of CO, activation of VSM KCa channels and In addition to pressure-induced changes in capillary
intraparenchymal arteriolar dilation in response to gluta- volume due to upstream dilation of arterioles, pericytes
mate are dependent on heme oxygenase in astrocytes [82]. have also been implicated in controlling vascular dimen-
Thus, CO might have a role as an astrocyte-derived sig- sions and flow in capillaries [88,89]. Pericytes are in great
naling molecule in the cerebral circulation of the newborn. abundance around capillaries and small venules, where
The role of CO in neurovascular coupling in mature brain is they often have a longitudinal orientation and might
less certain, although some evidence indicates a role migrate axially along a blood vessel. However, a subset
during seizures [83] and activation of a-amino-3- has a circumferential orientation, and these pericytes
hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) recep- selectively contain the smooth muscle isoform of actin
tors [69]. [90]. Often observed at branch points, this subset is specu-
lated to adjust the distribution of red-blood-cell flux among
Neuronal modulation of astrocyte responses capillaries. Because pericytes and astrocyte foot processes
During neuronal stimulation, NMDA receptors coupled to are separated by only the basal lamina, communication
nNOS are activated and lead to NO production by a between these cell types is likely to be crucially important
mechanism dependent on tissue plasminogen activator but remains a relatively unexplored area of research.
[84]. Neuronally derived NO is usually considered to Flow-induced changes in shear stress along the endo-
directly signal the VSM. However, NO might also modu- thelial wall are sensed by mechanoreceptors. In arterioles,
late signaling within astrocytes, although this possibility the endothelium can signal surrounding smooth muscle
has not been well investigated. Other neurotransmitters and change vascular tone to help restore shear stress.
might also interact with astrocyte signaling. In cortical Whether the endothelium in capillaries sends similar sig-
slices, stimulation of vasoactive intestinal peptide (VIP)- nals to surrounding astrocytes, which conceivably could
positive interneurons in the vicinity of microvessels often then signal neurons, is an interesting hypothesis that has
produces vasodilation, whereas stimulation of somato- recently been proposed [91]. Furthermore, dilation of the
statin- or neuropeptides-Y-positive interneurons some- various segments of the microcirculation leads to distortion
times produces vasoconstriction [85]. Neuronal processes of surrounding cells, including astrocytes. Whether
from these interneurons can make close contact with astro- mechanoreceptors are present in endfeet and sense the
cyte endfeet [86]. Both VIP and somatostatin application distortion is another open question. Endfeet are enriched
increase Ca2+ in astrocyte endfeet [47], although VIP in aquaporin-4 water channels, which seem to colocalize
causes dilation and somatostatin causes constriction. In with K+ channels [49,52]. Transient release of K+ during
areas enriched with dopaminergic neurons, dopamine is activation might be accompanied by transient shifts in
thought to modulate CBF, and the mechanism could water, which would be consistent with MRI data that
involve interactions with perivascular astrocytes [87]. Nor- indicate that water is relocated out of tissue and into
epinephrine has been demonstrated to increase Ca2+ in the vascular compartment during functional activation
astrocyte endfeet [60], which raises the possibility that [92]. Thus, many unanswered questions remain about
vasoconstriction evoked by locus coeruleus stimulation is the role of astrocytes in sensing or responding to segmental
modulated by astrocyte coupling. Likewise, diffuse cholin- changes in vascular volume.
ergic vasodilation evoked by basal forebrain stimulation
might also be modulated by astrocyte coupling. Thus, an Astrocyte responses in vivo
interesting possibility is that different populations of Two-photon imaging has enabled a closer examination of
neurons modulate the vasodilator and vasoconstrictor the relationship of neuronal activation, astrocytic Ca2+ and
responses to local and diffuse activation differentially by vasodilation in the cerebral cortex of intact anesthetized
modulating signaling within astrocyte endfeet (Figure 2). animals. Photolysis of caged Ca2+ in mouse cortical astro-
cytes produced dilation of some of the neighboring arter-
Segmental changes in vascular blood volume ioles with a latency of 1–2 s and a peak response within 4 s
The vascular volume within arterioles comprises a small [93]. In contrast to some studies using brain slices, con-
fraction of the total cerebral blood volume, yet substantial striction was not observed in vivo. Perivascular astrocytes
changes in blood volume occur during neuronal activation. expressed COX-1, and the dilation to Ca2+ uncaging was
Increases in blood volume within capillaries and small inhibited by a COX-1 inhibitor. Thus, a COX-1 metabolite,
venules have been reported to accompany the increase such as PGE2, is presumed to participate in the dilator
in red-blood-cell flux [11]. Because the increase in capillary response to increased astrocytic Ca2+.
and venular volume lag behind the brisk increase in arter- Whisker stimulation in mouse produced an increase in
iolar volume, the increased volume in the these down- astrocytic Ca2+ with a latency of 3 s to the initial increase,
stream vessels without smooth muscle is likely to be the 6–12 s to the peak response in the cell body and a slightly
result of an increase in intraluminal distending pressure longer delay in the foot process [94]. This response to
secondary to upstream arteriolar dilation. The change in physiologic activation in vivo was blocked by mGlu re-
diameter of capillaries during neuronal activation has been ceptor antagonists. Others using mechanical limb stimu-
estimated to be 10% [63]. Although this dilation amounts lation observed faster Ca2+ responses within 1 s from
to only 0.5 mm in a typical 5-mm-diameter capillary, this stimulation in the cell body and a variable delay in the
change could have a physiological effect on decreasing the endfeet [12]. In the visual cortex of ferrets, astrocytic Ca2+
viscous drag required for passage of a 7-mm-diameter red increased with an onset latency of 3–4 s after the start of
blood cell or a larger and less deformable leukocyte. the visual stimulus and reached a peak at 6–7 s [13].

165
Review Trends in Neurosciences Vol.32 No.3

Interestingly, astrocytic Ca2+ responses displayed sharper Third, administration of mGlu receptor antagonists,
tuning to orientation and spatial frequency than did EETs synthesis inhibitors and antagonists, and A2B
neuronal Ca2+ responses, possibly because an astrocyte antagonists, individually or in combination, inhibit the
might sample presynaptic glutamate release from a large steady-state CBF response to whisker stimulation by
number of synapses or have a lower threshold for eliciting a 50% without affecting the CBF response in the first
Ca2+ response throughout the cell. Another unexpected few seconds [34]. Thus, drugs that are likely to disrupt
finding was that the astrocytes responded to visual acti- astrocyte signaling do not interfere with the initial vaso-
vation individually and not as a spatial syncytium. It dilation, which might account for approximately half of the
remains to be determined whether this lack of spread of response. Because neuronal activation is often episodic,
astrocytic Ca2+ waves is specific for the primary visual understanding the mechanisms of initial vasodilation
cortex or a secondary effect of the isoflurane anesthetic. A remains an important subject of research. With sustained
key finding of this study was that the hemodynamic activation, the percentage increase in CBF and glucose
response tracked the astrocytic Ca2+ response better than consumption exceeds the percentage increase in oxygen
the neuronal Ca2+ response during visual stimulation consumption. Some of the excess glucose consumption and
when the astrocyte response was selectively suppressed lactate production is purported to occur in astrocytes. The
by a glutamate transport inhibitor or increased depth of stimulus for increased astrocytic glucose consumption is
anesthesia. Elegant work in glomeruli in the olfactory bulb unclear but might be related to electrolyte and water
indicated that the hemodynamic response to odor stimu- shifts, glutamate uptake, glutamate metabolism and glu-
lation is specifically related to presynaptic glutamate tamate release through hemichannels and the associated
release rather than to electrical activity per se. [95]. The increase in Na+ [97], which is likely to stimulate glycolytic-
hemodynamic response partly depended sequentially on dependent Na+,K+-ATPase activity. The observations that
activation of astrocytic mGlu5 receptors, increased Ca2+, a astrocytic glutamate release is associated with increased
COX-1 metabolite and partly on activation of glutamate NADH fluorescence [98] and that blocking glutamate
transporters. However, signaling among specialized cell uptake reduces the vasodilatory response [13,95] are con-
types in sensory nuclei, such as those in the olfactory bulb, sistent with a metabolic signal initiating part of the vas-
might not necessarily be generalized to other brain regions cular response. In this regard, the NADH:NAD+ ratio has
where postsynaptic effects on interneurons are more com- been proposed as a metabolic sensor [99].
plex [96]. Nevertheless, the results from the various in vivo Fourth, whereas COX-1 localized in astrocytes has been
studies confirm the observations in brain slices and demon- implicated in astrocyte–vascular coupling [93,95], inhibi-
strate that cortical astrocytes can be activated with physio- tors of COX-2 (which is localized in a subset of neurons
logic sensory stimulation and that dilation of arterioles is [100]) are actually more effective in reducing the CBF
linked to the astrocytic Ca2+ response. response to sensory activation than are inhibitors of
COX-1 [101–103]. Furthermore, COX-1-null mice have
Issues to be resolved an intact response, whereas COX-2-null mice have a
Although the preponderance of evidence summarized decreased response. High concentrations of a COX-1
before indicates a crucial role for astrocytes in the control inhibitor increase baseline vascular tone, which could
of CBF, many questions remain to be answered. reduce the dynamic response to activation. For example,
First, vasodilation, as assessed by LDF, blood-oxygen- the tonic release of a COX-1 metabolite from the endo-
level-dependent signals or the intrinsic optical signal, thelium might be required to maintain the minimal level of
begins to occur in <2 s from the onset of sensory stimu- cAMP in VSM that might be necessary for full expression of
lation, whereas the astrocytic Ca2+ response often has a the dynamic response to neuronal activation. Moreover,
delay of >2 s and a peak response that occurs a few seconds SC-560, which is selective for inhibiting COX-1 in isolated
later [13,94]. Therefore, the initial vasodilation seems to systems, seems to lose its selectivity in cell-culture systems
precede the astrocytic Ca2+ response and, hence, does not and inhibits COX-2 at nanomolar concentrations [104].
seem to require an increase in astrocytic Ca2+. The astro- Thus, the 500 mM concentration of SC-560 used in some
cyte Ca2+ response might be more important for sustaining studies [93,95] might result in nonspecific effects. More-
the vasodilation during prolonged activation. over, non-astrocytic expression of COX-1 and non-neuronal
Second, the observation that a glutamate-transport expression of COX-2 complicate interpretation of the role of
inhibitor blocked the astrocytic Ca2+ response to visual each isoform [105]. Hence, the precise role of each COX
stimulation [13] is somewhat unexpected because excessive isoform in each cell type and their associated metabolites
synaptic glutamate resulting from transport inhibition need to be better characterized, and the role of neuronally
would be expected to enhance astrocytic mGlu receptor derived COX-2 metabolites on astrocyte and VSM PG re-
stimulation. This result indicates that the astrocytic Ca2+ ceptor subtypes needs to be better discerned (Figure 2).
response to mGlu receptor stimulation is coordinated with Fifth, the precise role of EETs as an autocrine-signaling
glutamate uptake and possibly metabolism by glutamine molecule within astrocytes versus a paracrine-signaling
synthetase or glutamate export through connexin hemi- molecule with VSM needs to be better defined. In contrast
channels and reuptake by glutamate transporters [97]. In to COX metabolites, de novo synthesis of EETs might not
the olfactory bulb, the astrocyte Ca2+ response does not be required during each episode of activation because
depend completely on glutamate transporter activity, which preformed EETs are stored in the phospholipid membrane
can regulate the hemodynamic response independently of [24]. Hence, further elucidation is necessary concerning the
the astrocyte Ca2+ response [95]. relative role of newly synthesized EETs versus EETs

166
Review Trends in Neurosciences Vol.32 No.3

released from the phospholipid membrane and their mech- coupling is a highly integrated process involving multiple
anism of release. signaling pathways.
Sixth, most work on neurovascular coupling has focused Understanding how these mechanisms might be dis-
on the cerebral cortex. However, the signaling mechanisms turbed in various neurologic and vascular disease states is
differ in different brain regions. For example, NO is an important area of current research. Because inflam-
required as a mediator of neurovascular coupling in the mation induces an increase in COX-2, inducible NOS and
cerebellum [106], whereas NO acts as a modulator in the reactive oxygen species, diseases that are associated with
cerebral cortex [10,107]. Non-glutamate neurotransmit- inflammation are likely to disrupt normal neurovascular
ters in different brain regions might have selective roles coupling responses. Increases in angiotensin-associated
in modulating astrocyte responses. Moreover, the state of arterial hypertension and in b-amyloid associated with
attention, anticipation or stress might change the release Alzheimer’s disease can stimulate NADPH oxidase gener-
of neuronal and glial modulators and thereby modulate the ation of superoxide in animal models and interfere with
gain of neurovascular coupling. vascular dilation to neuronal activation [110–112]. More-
Seventh, anesthesia affects the magnitude of the CBF over, aged rodents without overt disease have an impaired
response [108] and could affect the processing of signals in vascular response that is related to increased NADPH
selective pathways. For example, isoflurane has been oxidase activity [113]. Impaired vascular responses to
reported to suppress the astrocytic Ca2+ response more neuronal activation in various disease states could influ-
than the neuronal Ca2+ response [13]. In the cerebral ence the progression of the disease process. Furthermore,
cortex, hemodynamic responses seem to depend on local fMRI is being used extensively for cognitive mapping in
field potentials with high-frequency activity in the gamma various disease states. Knowing whether the disease state
range [109]. Background neuronal activity is affected by alters neuronal activation or neurovascular signaling will
anesthetics, and dynamic changes in neuronal activity on be crucial for proper interpretation of fMRI results.
different background activities are likely to alter the gain
of the responses in a nonlinear manner. Acknowledgements
We are supported in this area of research by a grant from the National
Conclusions Institutes of Health (HL59996; www.nih.gov).
Emerging evidence from work in brain slices and in intact
anesthetized animals indicates that astrocytes play a References
crucial part in gathering information on presynaptic 1 Halassa, M.M. et al. (2007) Synaptic islands defined by the territory of
a single astrocyte. J. Neurosci. 27, 6473–6477
activity integrated over a large and discrete spatial
2 Bushong, E.A. et al. (2002) Protoplasmic astrocytes in CA1 stratum
domain and in using that information to dynamically radiatum occupy separate anatomical domains. J. Neurosci. 22, 183–
regulate local perfusion. The astrocyte signaling mechan- 192
isms are complex but seem to involve increases in astro- 3 Wilhelmsson, U. et al. (2006) Redefining the concept of reactive
cytic Ca2+ that are initiated by activation of the mGlu astrocytes as cells that remain within their unique domains
upon reaction to injury. Proc. Natl. Acad. Sci. U. S. A. 103, 17513–
receptor, phospholipase C and Ins(1,4,5)P3 and that are 17518
transmitted by release of ATP, which acts on astrocyte 4 Oberheim, N.A. et al. (2006) Astrocytic complexity distinguishes the
purinergic receptors, and possibly by catabolism of ATP to human brain. Trends Neurosci. 29, 547–553
adenosine, which then acts on A2B receptors. An increase 5 Simard, M. et al. (2003) Signaling at the gliovascular interface. J.
in Ins(1,4,5)P3 within astrocyte endfeet causes the spread Neurosci. 23, 9254–9262
6 Paulson, O.B. and Newman, E.A. (1987) Does the release of potassium
of Ca2+ along the endfoot process, activation of phospho- from astrocyte endfeet regulate cerebral blood flow? Science 237, 896–
lipase A2 and mobilization of arachidonic acid. The ara- 897
chidonic acid can be used to support synthesis of (i) EETs, 7 Harder, D.R. et al. (1998) Functional hyperemia in the brain.
which can help to sustain increases in astrocyte Ca2+ and Hypothesis for astrocyte-derived vasodilator metabolites. Stroke 29,
229–234
open KCa channels within astrocytes or on VSM to produce
8 Xu, H.L. et al. (2008) Astrocytes are a key conduit for upstream
vasorelaxation, (ii) PGE2, which can act on VSM to signaling of vasodilation during cerebral cortical neuronal
increase cAMP, and (iii) 20-HETE, which closes KCa chan- activation in vivo. Am. J. Physiol. Heart Circ. Physiol. 294, H622–
nels on VSM and produces vasoconstriction, especially H632
when the concentration of NO is too low to inhibit 20- 9 Dietrich, H.H. et al. (1996) Local and conducted vasomotor responses
in isolated rat cerebral arterioles. Am. J. Physiol. 271, H1109–H1116
HETE synthesis. The increase in Ca2+ in astrocyte endfeet
10 Liu, X. et al. (2008) Interaction of nitric oxide, 20-HETE, and EETs
also promotes opening of astrocytic KCa channels and during functional hyperemia in whisker barrel cortex. Am. J. Physiol.
release of K+, which then could act on vascular Kir chan- Heart Circ. Physiol. 295, H619–H631
nels to produce vasorelaxation. 11 Vanzetta, I. et al. (2005) Compartment-resolved imaging of activity-
However, these mechanisms that are dependent on dependent dynamics of cortical blood volume and oximetry. J.
Neurosci. 25, 2233–2244
astrocytic Ca2+ signaling seem to account for only approxi- 12 Winship, I.R. et al. (2007) Rapid astrocyte calcium signals correlate
mately half of the steady-state increase in CBF, and other with neuronal activity and onset of the hemodynamic response in vivo.
mechanisms might initiate vasorelaxation within the first J. Neurosci. 27, 6268–6272
few seconds. Other astrocytic mechanisms might include 13 Schummers, J. et al. (2008) Tuned responses of astrocytes and their
energy-metabolism-dependent signals, such as those aris- influence on hemodynamic signals in the visual cortex. Science 320,
1638–1643
ing from glutamate transport. Other mechanisms include 14 Dirnagl, U. et al. (1993) Role of nitric oxide in the coupling of cerebral
neuronal release of COX-2 metabolites, NO and polypep- blood flow to neuronal activation in rats. Neurosci. Lett. 149,
tide neurotransmitters such as VIP. Thus, neurovascular 43–46

167
Review Trends in Neurosciences Vol.32 No.3

15 Ko, K.R. et al. (1990) Role of adenosine in regulation of regional 39 Alkayed, N.J. et al. (2002) Neuroprotection and P450 2C11
cerebral blood flow in sensory cortex. Am. J. Physiol. 259, H1703– upregulation after experimental transient ischemic attack. Stroke
H1708 33, 1677–1684
16 Irikura, K. et al. (1994) Importance of nitric oxide synthase inhibition 40 Wang, Z. et al. (2000) Direct observation of calcium-independent
to the attenuated vascular responses induced by topical L- intercellular ATP signaling in astrocytes. Anal. Chem. 72, 2001–2007
nitroarginine during vibrissal stimulation. J. Cereb. Blood Flow 41 Haydon, P.G. and Carmignoto, G. (2006) Astrocyte control of synaptic
Metab. 14, 45–48 transmission and neurovascular coupling. Physiol. Rev. 86, 1009–
17 Dirnagl, U. et al. (1994) Coupling of cerebral blood flow to neuronal 1031
activation: role of adenosine and nitric oxide. Am. J. Physiol. 267, 42 Arcuino, G. et al. (2002) Intercellular calcium signaling mediated by
H296–H301 point-source burst release of ATP. Proc. Natl. Acad. Sci. U. S. A. 99,
18 Roman, R.J. (2002) P-450 metabolites of arachidonic acid in the 9840–9845
control of cardiovascular function. Physiol. Rev. 82, 131–185 43 Lovick, T.A. et al. (1999) Neurovascular relationships in hippocampal
19 Gebremedhin, D. et al. (1992) Mechanism of action of cerebral slices: physiological and anatomical studies of mechanisms
epoxyeicosatrienoic acids on cerebral arterial smooth muscle. Am. underlying flow-metabolism coupling in intraparenchymal
J. Physiol. 263, H519–H525 microvessels. Neuroscience 92, 47–60
20 Earley, S. et al. (2005) TRPV4 forms a novel Ca2+ signaling complex 44 Brown, L.A. et al. (2002) Inhibition of vasomotion in hippocampal
with ryanodine receptors and BKCa channels. Circ. Res. 97, 1270– cerebral arterioles during increases in neuronal activity. Auton.
1279 Neurosci. 95, 137–140
21 Li, P.L. et al. (2002) Role of ADP-ribose in 11,12-EET-induced 45 Zonta, M. et al. (2003) Neuron-to-astrocyte signaling is central to
activation of KCa channels in coronary arterial smooth muscle cells. the dynamic control of brain microcirculation. Nat. Neurosci. 6,
Am. J. Physiol. Heart Circ. Physiol. 282, H1229–H1236 43–50
22 Imig, J.D. et al. (2008) Afferent arteriolar dilation to 11,12-EET 46 Zonta, M. et al. (2003) Glutamate-mediated cytosolic calcium
analogs involves PP2A activity and Ca2+-activated K+ Channels. oscillations regulate a pulsatile prostaglandin release from
Microcirculation 15, 137–150 cultured rat astrocytes. J. Physiol. 553, 407–414
23 Amruthesh, S.C. et al. (1993) Metabolism of arachidonic acid to 47 Straub, S.V. et al. (2006) Dynamic inositol trisphosphate-mediated
epoxyeicosatrienoic acids, hydroxyeicosatetraenoic acids, and calcium signals within astrocytic endfeet underlie vasodilation of
prostaglandins in cultured rat hippocampal astrocytes. J. cerebral arterioles. J. Gen. Physiol. 128, 659–669
Neurochem. 61, 150–159 48 Newman, E.A. (1986) High potassium conductance in astrocyte
24 Shivachar, A.C. et al. (1995) Effect of protein kinase C modulators on endfeet. Science 233, 453–454
14,15-epoxyeicosatrienoic acid incorporation into astroglial 49 Nagelhus, E.A. et al. (2004) Aquaporin-4 in the central nervous
phospholipids. J. Neurochem. 65, 338–346 system: cellular and subcellular distribution and coexpression with
25 Alkayed, N.J. et al. (1996) Molecular characterization of an KIR4.1. Neuroscience 129, 905–913
arachidonic acid epoxygenase in rat brain astrocytes. Stroke 27, 50 Metea, M.R. et al. (2007) Neurovascular coupling is not mediated
971–979 by potassium siphoning from glial cells. J. Neurosci. 27, 2468–
26 Zhang, W. et al. (2007) Soluble epoxide hydrolase: a novel therapeutic 2471
target in stroke. J. Cereb. Blood Flow Metab. 27, 1931–1940 51 Roy, M.L. et al. (1996) Manipulation of the delayed rectifier Kv1.5
27 Alkayed, N.J. et al. (1997) Role of P-450 arachidonic acid expoygenase potassium channel in glial cells by antisense oligodeoxynucleotides.
in the response of cerebral blood flow to glutamate in rats. Stroke 28, Glia 18, 177–184
1066–1072 52 Price, D.L. et al. (2002) Distribution of rSlo Ca2+-activated K+
28 Ellis, E.F. et al. (1990) Dilation of cerebral arterioles by cytochrome P- channels in rat astrocyte perivascular endfeet. Brain Res. 956,
450 metabolites of arachidonic acid. Am. J. Physiol. 259, H1171– 183–193
H1177 53 Filosa, J.A. et al. (2004) Calcium dynamics in cortical astrocytes and
29 Leffler, C.W. and Fedinec, A.L. (1997) Newborn piglet cerebral arterioles during neurovascular coupling. Circ. Res. 95, e73–e81
microvascular responses to epoxyeicosatrienoic acids. Am. J. 54 Filosa, J.A. et al. (2006) Local potassium signaling couples neuronal
Physiol. 273, H333–H338 activity to vasodilation in the brain. Nat. Neurosci. 9, 1397–1403
30 Rzigalinski, B.A. et al. (1999) Calcium influx factor, further evidence it 55 Gerrits, R.J. et al. (2002) Ca2+-activated potassium (KCa) channel
is 5,6-epoxyeicosatrienoic acid. J. Biol. Chem. 274, 175–182 inhibition decreases neuronal activity-blood flow coupling. Brain
31 Bhardwaj, A. et al. (2000) P-450 epoxygenase and NO synthase Res. 948, 108–116
inhibitors reduce cerebral blood flow response to N-methyl-D- 56 Lange, A. et al. (1997) 20-Hydroxyeicosatetraenoic acid-induced
aspartate. Am. J. Physiol. Heart Circ. Physiol. 279, H1616–H1624 vasoconstriction and inhibition of potassium current in cerebral
32 Peng, X. et al. (2002) Suppression of cortical functional hyperemia to vascular smooth muscle is dependent on activation of protein
vibrissal stimulation in the rat by epoxygenase inhibitors. Am. J. kinase C. J. Biol. Chem. 272, 27345–27352
Physiol. Heart Circ. Physiol. 283, H2029–H2037 57 Gebremedhin, D. et al. (1998) Cat cerebral arterial smooth muscle
33 Peng, X. et al. (2004) Dependency of cortical functional hyperemia to cells express cytochrome P450 4A2 enzyme and produce the
forepaw stimulation on epoxygenase and nitric oxide synthase vasoconstrictor 20-HETE which enhances L-type Ca2+ current. J.
activities in rats. J. Cereb. Blood Flow Metab. 24, 509–517 Physiol. 507, 771–781
34 Shi, Y. et al. (2008) Interaction of mechanisms involving 58 Gebremedhin, D. et al. (2000) Production of 20-HETE and its role in
epoxyeicosatrienoic acids, adenosine receptors, and metabotropic autoregulation of cerebral blood flow. Circ. Res. 87, 60–65
glutamate receptors in neurovascular coupling in rat whisker 59 Dunn, K.M. et al. (2008) Elevated production of 20-HETE in the
barrel cortex. J. Cereb. Blood Flow Metab. 28, 111–125 cerebral vasculature contributes to severity of ischemic stroke and
35 Blanco, V.M. et al. (2008) Tone-dependent vascular responses to oxidative stress in spontaneously hypertensive rats. Am. J. Physiol.
astrocyte-derived signals. Am. J. Physiol. Heart Circ. Physiol. 294, Heart Circ. Physiol. 295, H2455–H2465
H2855–H2863 60 Mulligan, S.J. and MacVicar, B.A. (2004) Calcium transients in
36 Gebremedhin, D. et al. (2003) Metabotropic glutamate receptor astrocyte endfeet cause cerebrovascular constrictions. Nature 431,
activation enhances the activities of two types of Ca2+-activated K+ 195–199
channels in rat hippocampal astrocytes. J. Neurosci. 23, 1678–1687 61 Metea, M.R. and Newman, E.A. (2006) Glial cells dilate and constrict
37 Yamaura, K. et al. (2006) Contribution of epoxyeicosatrienoic acids to blood vessels: a mechanism of neurovascular coupling. J. Neurosci. 26,
the hypoxia-induced activation of Ca2+-activated K+ channel current 2862–2870
in cultured rat hippocampal astrocytes. Neuroscience 143, 703–716 62 Devor, A. et al. (2007) Suppressed neuronal activity and concurrent
38 Liu, M. and Alkayed, N.J. (2005) Hypoxic preconditioning and arteriolar vasoconstriction may explain negative blood oxygenation
tolerance via hypoxia inducible factor (HIF) 1a-linked induction of level-dependent signal. J. Neurosci. 27, 4452–4459
P450 2C11 epoxygenase in astrocytes. J. Cereb. Blood Flow Metab. 25, 63 Stefanovic, B. et al. (2008) Functional reactivity of cerebral capillaries.
939–948 J. Cereb. Blood Flow Metab. 28, 961–972

168
Review Trends in Neurosciences Vol.32 No.3

64 Sun, C-W. et al. (1998) Nitric oxide-20-hydroxyeicosatetraenoic acid 88 Peppiatt, C.M. et al. (2006) Bidirectional control of CNS capillary
interaction in the regulation of K+ channel activity and vascular tone diameter by pericytes. Nature 443, 700–704
in renal arterioles. Circ. Res. 83, 1069–1079 89 Yamanishi, S. et al. (2006) Extracellular lactate as a dynamic
65 Oyekan, A.O. et al. (1999) Renal cytochrome P450 v-hydroxylase and vasoactive signal in the rat retinal microvasculature. Am. J.
epoxygenase activity are differentially modified by nitric oxide and Physiol. Heart Circ. Physiol. 290, H925–H934
sodium chloride. J. Clin. Invest. 104, 1131–1137 90 Dore-Duffy, P. (2008) Pericytes: pluripotent cells of the blood brain
66 Alonso-Galicia, M. et al. (1999) Contribution of 20-HETE to barrier. Curr. Pharm. Des. 14, 1581–1593
vasodilator actions of nitric oxide in the cerebral microcirculation. 91 Moore, C.I. and Cao, R. (2008) The hemo-neural hypothesis: on the
Stroke 30, 2727–2734 role of blood flow in information processing. J. Neurophysiol. 99, 2035–
67 Sun, C.W. et al. (2000) Role of cGMP versus 20-HETE in the 2047
vasodilator response to nitric oxide in rat cerebral arteries. Am. J. 92 Lu, H. et al. (2003) Functional magnetic resonance imaging based on
Physiol. Heart Circ. Physiol. 279, H339–H350 changes in vascular space occupancy. Magn. Reson. Med. 50, 263–274
68 Iliff, J.J. et al. (2003) Adenosine receptors mediate glutamate-evoked 93 Takano, T. et al. (2006) Astrocyte-mediated control of cerebral blood
arteriolar dilation in the rat cerebral cortex. Am. J. Physiol. Heart flow. Nat. Neurosci. 9, 260–267
Circ. Physiol. 284, H1631–H1637 94 Wang, X. et al. (2006) Astrocytic Ca2+ signaling evoked by sensory
69 Ohata, H. et al. (2006) Contribution of adenosine A2A and A2B stimulation in vivo. Nat. Neurosci. 9, 816–823
receptors and heme oxygenase to AMPA-induced dilation of pial 95 Petzold, G.C. et al. (2008) Coupling of neural activity to blood flow in
arterioles in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. olfactory glomeruli is mediated by astrocytic pathways. Neuron 58,
291, R728–R735 897–910
70 Fields, R.D. and Burnstock, G. (2006) Purinergic signalling in neuron- 96 Lauritzen, M. (2005) Reading vascular changes in brain imaging: is
glia interactions. Nat. Rev. Neurosci. 7, 423–436 dendritic calcium the key? Nat. Rev. Neurosci. 6, 77–85
71 Pilitsis, J.G. and Kimelberg, H.K. (1998) Adenosine receptor mediated 97 Bernardinelli, Y. et al. (2004) Astrocytes generate Na+-mediated
stimulation of intracellular calcium in acutely isolated astrocytes. metabolic waves. Proc. Natl. Acad. Sci. U. S. A. 101, 14937–14942
Brain Res. 798, 294–303 98 Innocenti, B. et al. (2000) Imaging extracellular waves of glutamate
72 Jimenez, A.I. et al. (1999) Potentiation of ATP calcium responses by during calcium signaling in cultured astrocytes. J. Neurosci. 20, 1800–
A2B receptor stimulation and other signals coupled to Gs proteins in 1808
type-1 cerebellar astrocytes. Glia 26, 119–128 99 Mintun, M.A. et al. (2004) Increased lactate/pyruvate ratio augments
73 Alloisio, S. et al. (2004) Differential modulation of ATP-induced blood flow in physiologically activated human brain. Proc. Natl. Acad.
calcium signalling by A1 and A2 adenosine receptors in cultured Sci. U. S. A. 101, 659–664
cortical astrocytes. Br. J. Pharmacol. 141, 935–942 100 Wang, H. et al. (2005) Synaptic and vascular associations of neurons
74 Joseph, S.M. et al. (2003) Colocalization of ATP release sites and ecto- containing cyclooxygenase-2 and nitric oxide synthase in rat
ATPase activity at the extracellular surface of human astrocytes. J. somatosensory cortex. Cereb. Cortex 15, 1250–1260
Biol. Chem. 278, 23331–23342 101 Niwa, K. et al. (2000) Cyclooxygenase-2 contributes to functional
75 Xu, H.L. and Pelligrino, D.A. (2007) ATP release and hydrolysis hyperemia in whisker-barrel cortex. J. Neurosci. 20, 763–770
contribute to rat pial arteriolar dilatation elicited by neuronal 102 Niwa, K. et al. (2001) Cyclooxygenase-1 participates in selected
activation. Exp. Physiol. 92, 647–651 vasodilator responses of the cerebral circulation. Circ. Res. 88,
76 Meno, J.R. et al. (2001) Effect of adenosine receptor blockade on pial 600–608
arteriolar dilation during sciatic nerve stimulation. Am. J. Physiol. 103 Stefanovic, B. et al. (2006) Modulatory role of cyclooxygenase-2 in
Heart Circ. Physiol. 281, H2018–H2027 cerebrovascular coupling. Neuroimage 32, 23–32
77 Ngai, A.C. et al. (2001) Receptor subtypes mediating adenosine- 104 Brenneis, C. et al. (2006) Inhibition of prostaglandin E2 synthesis by
induced dilation of cerebral arterioles. Am. J. Physiol. Heart Circ. SC-560 is independent of cyclooxygenase 1 inhibition. FASEB J. 20,
Physiol. 280, H2329–H2335 1352–1360
78 Leffler, C.W. et al. (2006) Carbon monoxide and hydrogen sulfide: 105 Parfenova, H. et al. (1997) Upregulation of COX-2 in cerebral
gaseous messengers in cerebrovascular circulation. J. Appl. Physiol. microvascular endothelial cells by smooth muscle cell signals. Am.
100, 1065–1076 J. Physiol. 273, C277–C288
79 Leffler, C.W. et al. (2006) Contributions of astrocytes and CO to pial 106 Yang, G. and Iadecola, C. (1997) Obligatory role of NO in glutamate-
arteriolar dilation to glutamate in newborn pigs. Am. J. Physiol. Heart dependent hyperemia evoked from cerebellar parallel fibers. Am. J.
Circ. Physiol. 291, H2897–H2904 Physiol. 272, R1155–R1161
80 Kanu, A. and Leffler, C.W. (2007) Carbon monoxide and Ca2+- 107 Lindauer, U. et al. (1999) Nitric oxide: a modulator, but not a
activated K+ channels in cerebral arteriolar responses to glutamate mediator, of neurovascular coupling in rat somatosensory cortex.
and hypoxia in newborn pigs. Am. J. Physiol. Heart Circ. Physiol. 293, Am. J. Physiol. Heart Circ. Physiol. 277, H799–H811
H3193–H3200 108 Nakao, Y. et al. (2001) Effects of anesthesia on functional activation of
81 Jaggar, J.H. et al. (2005) Heme is a carbon monoxide receptor for cerebral blood flow and metabolism. Proc. Natl. Acad. Sci. U. S. A. 98,
large-conductance Ca2+-activated K+ channels. Circ. Res. 97, 805–812 7593–7598
82 Li, A. et al. (2008) Astrocyte-derived CO is a diffusible messenger that 109 Niessing, J. et al. (2005) Hemodynamic signals correlate tightly with
mediates glutamate-induced cerebral arteriolar dilation by activating synchronized gamma oscillations. Science 309, 948–951
smooth muscle cell KCa channels. Circ. Res 102, 234–241 110 Kazama, K. et al. (2004) Angiotensin II impairs neurovascular
83 Montecot, C. et al. (1998) Carbon monoxide regulates cerebral blood flow coupling in neocortex through NADPH oxidase-derived radicals.
in epileptic seizures but not in hypercapnia. Neuroreport 9, 2341–2346 Circ. Res 95, 1019–1026
84 Park, L. et al. (2008) Key role of tissue plasminogen activator in 111 Park, L. et al. (2005) NADPH oxidase-derived reactive oxygen species
neurovascular coupling. Proc. Natl. Acad. Sci. U. S. A. 105, 1073–1078 mediate the cerebrovascular dysfunction induced by the amyloid b
85 Cauli, B. et al. (2004) Cortical GABA interneurons in neurovascular peptide. J. Neurosci. 25, 1769–1777
coupling: relays for subcortical vasoactive pathways. J. Neurosci 24, 112 Park, L. et al. (2008) Nox2-derived radicals contribute to
8940–8949 neurovascular and behavioral dysfunction in mice overexpressing
86 Hamel, E. (2006) Perivascular nerves and the regulation of the amyloid precursor protein. Proc. Natl. Acad. Sci. U. S. A. 105,
cerebrovascular tone. J. Appl. Physiol. 100, 1059–1064 1347–1352
87 Choi, J.K. et al. (2006) Brain hemodynamic changes mediated by 113 Park, L. et al. (2007) Nox2-derived reactive oxygen species mediate
dopamine receptors: role of the cerebral microvasculature in neurovascular dysregulation in the aging mouse brain. J. Cereb.
dopamine-mediated neurovascular coupling. Neuroimage 30, 700–712 Blood Flow Metab. 27, 1908–1918

169

You might also like