Vous êtes sur la page 1sur 12

Molecular and Cellular Endocrinology 353 (2012) 45–56

Contents lists available at SciVerse ScienceDirect

Molecular and Cellular Endocrinology


journal homepage: www.elsevier.com/locate/mce

Review

Calcium signalling in astroglia


Alexei Verkhratsky a,b,c,⇑, José J. Rodríguez b,c, Vladimir Parpura b,c,d,e,⇑
a
Faculty of Life Sciences, The University of Manchester, Manchester, UK
b
IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
c
Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
d
Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy & Nanotechnology Laboratories, and
Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, USA
e
School of Medicine, University of Split, 21000 Split, Croatia

a r t i c l e i n f o a b s t r a c t

Article history: Astroglia possess excitability based on movements of Ca2+ ions between intracellular compartments and
Available online 10 September 2011 plasmalemmal Ca2+ fluxes. This ‘‘Ca2+ excitability’’ is controlled by several families of proteins located in
the plasma membrane, within the cytosol and in the intracellular organelles, most notably in the endo-
Keywords: plasmic reticulum (ER) and mitochondria. Accumulation of cytosolic Ca2+ can be caused by the entry of
Astroglia Ca2+ from the extracellular space through ionotropic receptors and store-operated channels expressed in
Calcium signalling astrocytes. Plasmalemmal Ca2+ ATP-ase and sodium–calcium exchanger extrude cytosolic Ca2+ to the
Endoplasmic reticulum
extracellular space; the exchanger can also operate in reverse, depending of the intercellular Na+ concen-
Mitochondria
Ca2+ channels
tration, to deliver Ca2+ to the cytosol. The ER internal store possesses inositol 1,4,5-trisphosphate recep-
Ionotropic receptors tors which can be activated upon stimulation of astrocytes through a multiple plasma membrane
metabotropic G-protein coupled receptors. This leads to release of Ca2+ from the ER and its elevation
in the cytosol, the level of which can be modulated by mitochondria. The mitochondrial uniporter takes
up Ca2+ into the matrix, while free Ca2+ exits the matrix through the mitochondrial Na+/Ca2+ exchanger as
well as via transient openings of the mitochondrial permeability transition pore. One of the prominent
consequences of astroglial Ca2+ excitability is gliotransmission, a release of transmitters from astroglia
which can lead to signalling to adjacent neurones.
Ó 2011 Elsevier Ireland Ltd. All rights reserved.

Contents

1. Astroglia: definition, evolution and function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45


2. Principles of calcium signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
3. Calcium signalling in astrocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.1. Endoplasmic reticulum provides for glial Ca2+ excitability. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.2. The store-operated Ca2+ entry: a role for TRPC channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
3.3. Ionotropic Ca2+ permeable receptors in astrocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
3.4. The sodium/calcium exchanger in astroglia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
3.5. Mitochondria in astroglial Ca2+ signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
3.6. Voltage-gated Ca2+ channels (VGCCs) in astroglia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
4. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53

1. Astroglia: definition, evolution and function


⇑ Corresponding authors. Addresses: The University of Manchester, Oxford Road,
Manchester M13 9PT, UK. Tel.: +44 (0)161 2757324 (A. Verkhratsky); Department The term ‘‘astroglia’’ (which was named after the word ‘‘astro-
of Neurobiology, 1719 6th Avenue South, CIRC 429, University of Alabama,
cyte’’ invented by Michael von Lenhossek in 1891 (Lenhossek,
Birmingham, AL 35294, USA. Tel.: +1 205 996 7369; fax: +1 205 975 6320 (V.
Parpura). 1891, 1893), for historic account see also (Kettenmann and
E-mail addresses: alex.verkhratsky@manchester.ac.uk (A. Verkhratsky), Verkhratsky, 2008; Verkhratsky, 2006b)) is used to define all
vlad@uab.edu (V. Parp. non-myelinating macroglial cells in the central nervous system.

0303-7207/$ - see front matter Ó 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2011.08.039
46 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

These cells are represented by many subtypes which differ sub- cells of the Caenorhabditis elegans. In this worm 302 neurones are
stantially in their morphology, location, general physiology, and assisted by 50 proto-astrocytes that cover dendrites of sensory
function (Matyash and Kettenmann, 2010; Verkhratsky and Butt, neurones and envelop the nerve ring (Oikonomou and Shaham,
2007). In fact only a minority of astrocytes have star-like morphol- 2011). The proto-astrocytes of C. elegans are still very close to
ogy, which was used a typifying criterion for their definition neurones from which they evolved; for example these glial cells
(astron kytos – star-like cells). These cells were the neuroglia that express voltage-gated Ca2+ channels, produce depolarisation-
Ramón y Cajal visualized in 1913 using a gold sublimate stain, induced Ca2+ signals (Oikonomou and Shaham, 2011; Stout and
which, as we know today targeted intermediate filaments that Parpura, 2011). At the same type proto-astrocytes of the C. elegans
consist mainly of glial fibrillary acidic protein (GFAP), a protein specialise on new homeostatic functions regulating development
contemporary used as an astroglial marker. However not all astro- and general well being of neurones. Complete ablation of glial cells
cytes are GFAP positive. Astroglia as a class of neural cells embrace from C. elegans nervous system seriously affects its development
the radial glia, classical protoplasmic and fibrous astrocytes, radial and functional abilities, and yet neurones in the worm are still able
Müller retinal glial cells, pseudo-radial cerebellar Bergmann glial to survive in the absence of astroglia as they presumably have no
cells, laminar and polarised astrocytes of the primate brain, velate much need for glial trophic support, and the glia-less animals,
astrocytes of cerebellum, tanycytes that connect ventricular walls although disabled, remain alive (Bacaj et al., 2008; Oikonomou
with parts of hypothalamus and spinal cord, pituicytes in the and Shaham, 2011; Procko and Shaham, 2010; Reichenbach and
neuro-hypophysis, and perivascular and marginal astrocytes Pannicke, 2008).
(Kimelberg, 2010; Kimelberg and Nedergaard, 2010; Verkhratsky Further evolution of the nervous system required more special-
and Butt, 2007) As if this is not sufficient, astroglia also include sev- isation form neurones and neuronal centres and coincided with a
eral types of cells that line the ventricles or the subretinal space critical increase in the importance of neuroglia (Deitmer et al.,
represented by ependymocytes, choroid plexus cells and retinal 1999; Edwards and Meinertzhagen, 2010; Laming et al., 2000). In
pigment epithelial cells. the Annelids and Arthropods astrocytes become diversified, which
There is however one basic and fundamental function which resulted in appearance of several major classes of glia (for example,
unites all these cells – their primary responsibility is the control in the insects they include surface, cortex, neuropile and tract glia;
of homeostasis of the nervous system (Heneka et al., 2010; Kimel- with further subdivision within the classes (Edwards and Mein-
berg, 2010; Verkhratsky and Butt, 2007; Verkhratsky et al., 2011). ertzhagen, 2010)). Thus evolved and more complex glia becomes
This homeostatic function is executed at many levels, and includes critical for organising the nervous system of Arthropods by isolat-
organ homeostasis (astroglia control the emergence and mainte- ing the CNS from the body through formation of blood–brain bar-
nance of brain–blood barrier), cellular homeostasis (astroglia is rier and by isolating functionally distinct nervous centres. The
intimately involved in neurogenesis as both stem element and reg- insect astroglia is fully homeostatically competent being involved
ulator) morphological homeostasis (astroglia defines the migratory in trophic support, regulation of ion and neurotransmitter concen-
pathways for neural cells during development and shapes the mi- tration, brain development, circadian rhythms, etc. Most impor-
cro-architecture of grey matter and controls synaptogenesis/syn- tantly at this evolutionary stage astroglia becomes the main
aptic pruning), molecular homeostasis (which is represented by element of brain defence as the first programmes of reactive
regulation of ion, neurotransmitter and neurohormone concentra- astrogliosis become operative. All these changes make neuroglia
tion in the CNS extracellular space), metabolic homeostasis indispensable for the nervous system survival and the deletion of
(astroglia stores energy substrates in a form of glycogen and sup- the glial cells renders the nervous system unviable. Further evolu-
plies neurones with lactate) and defensive homeostasis (astroglio- tion of the nervous system deepened the specialisation: neurones
sis is the fundamental adaptive/defensive reaction of neural became ultimately excitable elements that formed numerous func-
tissue). Moreover astrocytes are possibly the main chemosensitive tionally separated nervous centres, whereas astroglia increased
elements of the brain that perceive systemic fluctuations in CO2, their numerical presence, diversification, morphological complex-
pH and Na+ and thus regulate behavioural and systemic homeo- ity and functional competence (Kimelberg, 2010; Laming et al.,
static physiological responses (Gourine and Kasparov, 2011; 2000; Oberheim et al., 2006, 2009).
Gourine et al., 2010; Huckstepp et al., 2010; Shimizu et al., 2007). The wealth of homeostatic functions of astroglia necessitates
Astroglial cells share the common ancestry with neurones and highly sophisticated signalling system(s) that are able to monitor
oligodendrocytes all of them being scions of neuroepithelial cell, extracellular space, couple extracellular cues with cellular pro-
and as such astrocytes are capable of expressing majority of mole- cesses and coordinate astroglial networks. The neuronal and glial
cules that for a long time were believed to be exclusively present in signalling systems evolved in parallel, with neurones perfecting
neurones (the most trilling examples being neurotransmitter the electrical excitability (in close association with oligodendrog-
receptors and components of exocytotic release of neurotransmit- lia, which made possible the saltatory propagation of action poten-
ter-containing vesicles (Lalo et al., 2011b; Montana et al., 2006; tials) and refining the synaptic mechanisms (with the help of
Parpura et al., 1994, 2011; Verkhratsky and Kettenmann, 1996)). astrocytes controlling the levels of neurotransmitters in the synap-
At the same time astroglial cells (as well as all other types of neu- tic cleft). The glial cells, however, did not acquire plasmalemmal
roglia) are electrically non-excitable being thus fundamentally dif- electrical excitability, relying instead on excitable endomembranes
ferent from neurones. The electrical excitability and fast chemical and intercellular gap junctions. The intracellular calcium signalling
synaptic transmission are the defining function of neurones; the system expressed in astrocytes seemingly represents the key ele-
absolute bulk (90–95%) of energy consumed by the brain is spent ment in astroglial ‘‘excitability’’ and physiological operation.
for maintaining electrical excitability and synaptic transmission
(Magistretti, 2009). The neurones are thus perfect elements of fast
information transfer, this perfection, however, was earned at the 2. Principles of calcium signalling
expense of shifting all the house-holding, homeostatic and
defensive responsibilities to neuroglia. Calcium ions are arguably the most widespread and ubiquitous
In evolution, astrocytes appeared simultaneously with the signalling molecules operating in all types of living cells (Berridge
formation of the first centralised nervous system when neural cells et al., 2000; Carafoli, 2002; Heilbrunn, 1943; Heilbrunn and
begun to assemble into the conglomerates known as neuronal gan- Wiercinsky, 1947; Petersen et al., 2005). Calcium was appointed
glia. The most ancient proto-astrocytes known to us are the glial to this role very early in the evolution (Case et al., 2007), possibly
A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56 47

when ATP was chosen as a main energy substrate: most of these compartments and between the cell and extracellular space
reactions needed for ATP metabolism can proceed only at very produce very rapid, and quite often much localised, fluctuations
low Ca2+ concentrations (Burnstock and Verkhratsky, 2009). Fur- of free Ca2+ in different regions of the cell and within different
ther, Ca2+ ions eagerly interact with various biological molecules intracellular organelles; these fluctuations in turn exert signalling
because of special properties of Ca2+ that include flexible coordina- action (Petersen, 2002; Petersen et al., 2005; Rizzuto and Pozzan,
tion chemistry, high affinity for carboxylate oxygen, which is the 2006).
most frequent motif in amino acids, rapid binding kinetics, and ef- The living cells contain a relatively limited number of Ca2+ han-
fects on fluidity and fusion of cellular membranes (Jaiswal, 2001). dling systems. Those include membrane Ca2+ channels (both
High Ca2+ concentrations, at the same time, are harmful for biolog- plasmalemmal and intracellular), cytoplasmic Ca2+ buffers and sev-
ical material: excess of Ca2+ causes aggregation of proteins and nu- eral sets of membrane Ca2+ transporters represented by Ca2+
cleic acids, affects the integrity of lipid membranes and initiates pumps and Ca2+ exchangers (Berridge et al., 2003; Kostyuk and
the precipitation of phosphates. Therefore from the very dawn of Verkhratsky, 1994, 1995; Petersen et al., 1994). These systems
evolution the tight control over Ca2+ concentration in the cellular have ancient evolutionary roots and were conserved in phylogen-
compartments executed through containment of diffusional Ca2+ esis (Case et al., 2007; Shemarova and Nesterov, 2005a,b). By com-
fluxes paired with active transport systems that pumped Ca2+ out bining different molecular components the versatility and
of the intracellular space (Case et al., 2007; Shemarova and Neste- adaptability of Ca2+ signalling system is achieved because cells
rov, 2005a,b). This tight control over Ca2+ is present throughout all can create the context-dependent ‘‘Ca2+ signalling toolkits’’ (Ber-
the life forms, and in all the living cells cytosolic Ca2+ is maintained ridge et al., 2003), that tailor Ca2+ signalling events to match envi-
at comfortably low concentrations – somewhere around 100 nM, ronmental challenges. Indeed, Ca2+ signals resulting from cell
this being 10,000–20,000 times lower than that in the extracellu- activation appear in multiple shapes, from local microdomains,
lar milieu. which, for example regulate neurotransmitter release, postsynap-
The huge Ca2+ concentration gradient aimed at the cytoplasm tic plasticity, or cell process guidance (Bolsover, 2005; Cavazzini
forms the backbone for Ca2+ signalling by allowing diffusion of et al., 2005; Hartmann and Konnerth, 2005; Holcman et al.,
Ca2+ ions between external environment and the cell interior. 2005; Lohmann and Wong, 2005; Oertner and Matus, 2005; Red-
The second fundamental principle of Ca2+ signalling is represented mond and Ghosh, 2005; Spitzer et al., 2005), through global Ca2+
by the existence of several intracellular compartments, which con- signals, which control excitation–contraction or excitation–secre-
trol Ca2+ movements in a very different way, therefore creating tion coupling (Kiselyov et al., 2003; Petersen, 2005; Wray et al.,
steep Ca2+ concentration gradients within the cell (see e.g. Berridge 2005), regulate gene expression (Fields et al., 2005; Webb et al.,
et al., 2003; Bregestovski and Spitzer, 2005; Knot et al., 2005; Kos- 2005) and tissue development (Puceat and Jaconi, 2005), to a prop-
tyuk and Verkhratsky, 1995; Petersen, 2005; Petersen et al., 2005; agating Ca2+ waves accomplishing integration in complex multicel-
Verkhratsky et al., 1998 and Fig. 1). Movements of Ca2+ between lular structures, such as, for example, in astroglial syncytia in the

Fig. 1. Principles of calcium signalling in astroglia. [Ca2+]i accumulation could be caused by the entry of Ca2+ from the extracellular space through ionotropic receptors or
store-operated channels (SOC). Plasmalemmal Ca2+ pumps/ATP-ases (PMCA) can extrude cytosolic Ca2+, while the plasmalemmal sodium–calcium exchanger (NCX) can
operate in both directions depending of intercellular Na+ concentration. An additional source of Ca2+ is available from the ER internal store that possesses InsP3 receptors,
which can be activated by the activity of metabotropic G-protein coupled receptors (GPCRs) and PLC. The ER store is (re)filled by the activity of the store-specific Ca2+-ATPase
(SERCA). Cytosolic Ca2+ levels can be affected by a variety of cytosolic Ca2+-binding proteins (CBPs) and by the action of mitochondria. A negative membrane potential exists
across the inner mitochondrial membrane. Mitochondrial Ca2+ uptake occurs through voltage-dependent anion channels (VDAC) present in the outer membrane and by the
uniporter in the inner membrane as the electrochemical gradient drives Ca2+ into the matrix, while free Ca2+ exits the mitochondrial matrix through the mitochondrial Na+/
Ca2+ exchanger and transient opening of the mitochondrial permeability transition pore (MPTP).
48 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

brain (Verkhratsky, 2006a; Verkhratsky and Toescu, 2006). At the Kostyuk and Verkhratsky, 1995). The neurones are also in posses-
same time, the Ca2+ signalling machinery is responsible for the sion of Ca2+-induced Ca2+ release which rapidly amplifies Ca2+ sig-
control of cell survival, and numerous Ca2+-dependent pathways nals (Verkhratsky, 2005; Verkhratsky and Petersen, 2002;
trigger various modes of cell death, which are fundamental for nor- Verkhratsky and Shmigol, 1996).
mal tissue development and homeostasis (Nicotera et al., 2007; The spatial organisation of Ca2+ signals is determined by the cel-
Verkhratsky, 2007). Importantly, molecular components of Ca2+ lular distribution of Ca2+ channels and by intracellular Ca2+ biding
homeostatic/signalling system are also controlled by Ca2+ ions proteins generally known as Ca2+ buffers. In the cytosol Ca2+ buf-
themselves, so that that changes in Ca2+ concentrations in different fers are characterised by high (in a low nanomolar range) affinity
cellular compartments affect the function and/or availability of to Ca2+ ions (Ikura et al., 2002; Lewit-Bentley and Rety, 2000).
both Ca2+ channels and Ca2+ transporters (for example, plasmalem- The cytosolic high affinity Ca2+ buffers tend to localise cytosolic
mal Ca2+ channels are generally subject for Ca2+-dependent inacti- Ca2+ events and assist in forming a focal micro- or even nano-do-
vation (Imredy and Yue, 1994; Morad et al., 1988; Rycroft and mains of high (1–100 lM) cytosolic Ca2+, which in turn control
Gibb, 2004a,b)); Ca2+ concentration gradient over the ER mem- highly localised and rapid cellular reactions, such as for example
brane controls both availability of Ca2+ release channels for activa- neurotransmitter release (Neher and Sakaba, 2008; Rizzuto and
tion and efficacy of Ca2+ pumping through the endomembrane Pozzan, 2006). The Ca2+ buffers expressed in the lumen or the ER
(Burdakov et al., 2005; Guerrero-Hernandez et al., 2010; Koizumi have low affinity (KD  0.5–1 mM), which permits relatively free
et al., 1999; Oldershaw and Taylor, 1993; Shmigol et al., 1996). Ca2+ diffusion through the lumen of the endoplasmic reticulum
At the receiving end of Ca2+ signalling system ‘‘Ca2+ sensors’’ thus allowing long-range intracellular Ca2+ signalling through
control cellular reactions. These sensors are represented by a mul- ‘‘Ca2+ tunnels’’ (Petersen and Verkhratsky, 2007; Solovyova and
titude of enzymes, which all are endowed with Ca2+ binding sites, Verkhratsky, 2003).
albeit with different affinities; Ca2+ binding to these binding sites The termination of cytosolic Ca2+ signals is achieved by energy-
alters activity of the enzymes and therefore triggers or discontin- dependent Ca2+ transport against concentration gradients. In both
ues diverse biochemical processes (Burgoyne et al., 2004; Carafoli, electrically excitable and non-excitable cells Ca2+ can be expelled
2004; Carafoli et al., 2001). The differences in Ca2+ affinity of differ- to the extracellular space by plasmalemmal Ca2+ pumps/ATP-ases
ent enzymatic systems allow for amplitude coding of Ca2+ signals. (PMCA), which utilities the ATP energy directly, or by sodium–cal-
The Ca2+-sensitive enzymes are often specifically concentrated in cium exchanger (NCX), which uses the same ATP energy indirectly
cellular subcompartments, thus providing for precise spatial cod- in the form of preset transmembrane Na+ gradient (Guerini et al.,
ing of the Ca2+ signals. Finally, the duration and kinetics of intracel- 2005). Alternatively Ca2+ can be taken up back to the ER lumen,
lular Ca2+ fluctuations control the timing of Ca2+-binding/ via sarco-(endo)plasmic reticulum Ca2+ ATP-ases (SERCA) (Van-
unbinding to the ‘‘Ca2+ sensors’’ hence forming the basis for tempo- gheluwe et al., 2005).
ral coding of Ca2+ signalling events (Toescu and Verkhratsky, 1998). Another cellular organelle critical for Ca2+ signalling and Ca2+
Three main intracellular compartments in which Ca2+ signals homeostasis is represented by mitochondria (Nicholls, 2005; Toe-
emerge and fall are (Fig. 1): the cytoplasm, the endoplasmic retic- scu, 2000). The double membrane of mitochondria is endowed
ulum and the mitochondria (although some other intracellular with two main types of Ca2+-permeable channels. The outer mem-
organelles, such as e.g. Golgi complex, lysosomes and secretory brane contains the voltage-dependent anion channels that have
granules may participate in intracellular Ca2+ homeostasis and sig- considerable Ca2+ permeability, whereas the inner membrane pos-
nalling – see e.g. Gerasimenko et al., 1996; Michelangeli et al., sesses the highly selective Ca2+channel usually referred to as Ca2+
2005; Zhu et al., 2010). These compartments are formed by the uniporter (Rimessi et al., 2008). The mitochondria are hugely elec-
membranes represented by the plasmalemma that makes the out- tronegative with regard to the cytosol (the mitochondrial mem-
er cell border, by the endomembrane, which creates the endoplas- brane potential, the Dw, created by the electron transport is
mic reticulum and by the double mitochondrial membrane. Each of about 180 to 200 mV). This in turn generates the electric force
these membranes contain distinct Ca2+ channels (that allow down- for Ca2+ aimed towards the mitochondrial matrix, when cytosolic
hill Ca2+ diffusion) and transporters (which generally are responsi- Ca2+ exceeds 300–400 nM (Miyata et al., 1991; Simpson and Rus-
ble for Ca2+ transport against Ca2+ gradient); the properties of these sell, 1998). Therefore elevation of [Ca2+]i triggers substantial Ca2+
channels and transporters determine the impact on the shape of influx into the mitochondria; this Ca2+ influx depolarises the mito-
Ca2+ signals (Carafoli et al., 2001; Toescu and Verkhratsky, 1998). chondrial membrane hence stimulating ATP production (Rimessi
The second important determinant is the Ca2+ concentration and et al., 2008). In addition Ca2+ accumulation by the mitochondria
electrochemical gradient. The free Ca2+ concentration in the cytosol provides an additional Ca2+ buffer, which prevents large cytosolic
([Ca2+]i) is set in the range of 30–100 nM; the Ca2+ concentration in Ca2+ loads. The overload of mitochondria with free Ca2+ is danger-
the extracellular space normally ranges between 1 and 2 mM, ous and may initiate cell damage and various cell death programs
whereas Ca2+ concentration in the lumen of endoplasmic reticulum (Duchen et al., 2008; Nicotera and Orrenius, 1998; Toescu and Ver-
([Ca2+]L) can be as high as 0.2–1 mM (Alonso et al., 1999; Mogami khratsky, 2004).
et al., 1998; Solovyova and Verkhratsky, 2002; Solovyova et al.,
2002). The pre-existing concentration/electrochemical gradients
favour Ca2+ influx into the cytosol. Cell activation opens plasma- 3. Calcium signalling in astrocytes
lemmal or intracellular Ca2+ channels, which results in spatially
and temporally organised [Ca2+]i fluctuations. The intracellular 3.1. Endoplasmic reticulum provides for glial Ca2+ excitability
route for Ca2+ signalling generation is predominant in the electri-
cally non-excitable cells, as well as in electrically excitable cells It is generally acknowledged that all types of glia operate a spe-
such as for instance in cardiac and skeletal muscle cells; conversely cial form of Ca2+ excitability (Verkhratsky et al., 1998). Numerous
in neurones the plasmalemmal Ca2+ pathway assumes higher experiments on neuroglial cells in vitro, in situ and in vivo have
importance (Toescu and Verkhratsky, 1998). The rich complement identified the expression of multiple metabotropic receptors that,
of voltage- and ligand-operated Ca2+ channels expressed in neuro- when activated by physiological stimulation, trigger production
nal membrane allows rapid transformation of membrane events of inositol 1,4,5-trisphosphate (InsP3) and subsequent InsP3-
into cytosolic Ca2+ signals, which is critical for fast signalling idio- induced Ca2+ release from the ER (Finkbeiner, 1993; Hamilton
syncratic for neuronal networks (Bregestovski and Spitzer, 2005; et al., 2008; Kastritsis et al., 1992; Kirischuk et al., 1996; McCarthy
A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56 49

and Salm, 1991; Porter and McCarthy, 1995). Generally, Ca2+ sig- The role for Ca2+-gated Ca2+ release channels/ryanodine receptors
nals induced by activation of metabotropic receptors (most notably (RyRs) in Ca2+ signalling in astroglia remains debatable. Activation
by ATP and glutamate) share the similar set of properties. These of RyRs by caffeine was observed in astrocytes cells in thalamus
[Ca2+]i transients persist in Ca2+-free extracellular solutions and (Parri and Crunelli, 2003). In contrast, little evidence was found
are sensitive to inhibition of SERCA pumps (by thapsigargin or by for RyRs-mediated Ca2+ signalling in hippocampal astrocytes (Beck
cyclopiazonic acid) and to blockade of InsP3 receptors by intracel- et al., 2004), despite the fact that astrocytes in the hippocampus
lular administration of heparin (Fig. 2, Kirischuk et al., 1995). It was express RyRs receptors at both the mRNA and protein levels (Mat-
also found that intercellular diffusion of InsP3 and InsP3-induced yash et al., 2002; Verkhratsky et al., 2002).
Ca2+ release represent the main mechanism of glial intercellular There has been a general consensus that Ca2+ ions released from
Ca2+ waves that arguably underlie long-range signalling in glial the ER via InsP3 induced Ca2+ release are critically important for the
syncytia in vitro and in brain slices (Cornell Bell et al., 1990; Dani exocytosis of transmitters from astroglial cells (Malarkey and Par-
et al., 1992; Giaume and Venance, 1998). Recently, synchronous pura, 2009; Parpura et al., 2011). The role for the ER Ca2+ release in
Ca2+ waves that embrace hundreds of astrocytes were also re- regulated vesicular release of neurotransmitters from astroglia was
corded in vivo; these Ca2+ waves were sensitive to inhibition of initially demonstrated in cell cultures. It was found that inhibition
gap junctions and purinoceptors suggesting their InsP3/ER origin of SERCA pumps by thapsigargin, which lowers ER Ca2+ concentra-
(Kuga et al., 2011). tion due to an opposed background leakage, almost completely
The glial Ca2+ excitability is provided by the endomembrane en- eliminated Ca2+-dependent release of glutamate from astrocytes
dowed with Ca2+ pumps and Ca2+ release channels both types of (Innocenti et al., 2000; Jeremic et al., 2001). In depth analysis of
molecules being regulated by cytosolic Ca2+. Regenerative opening the contribution of ER calcium store to Ca2+-dependent glutamate
of intracellular Ca2+ channels creates the propagating wave of release from astrocytes was performed on cultured astrocytes (Hua
endomembrane excitation; subsequent activation of SERCA pumps et al., 2004). It was found that thapsigargin reduced mechanically
terminates Ca2+ signal. The InsP3 receptors are also regarded as pri- induced glutamate release. Similarly, glutamate exocytosis was
mary molecules that initiate Ca2+ release following physiological inhibited by membrane-permeable antagonist of InsP3 receptors
stimulation. It seems that the type 2 InsP3 receptor is predomi- (and of the store-operated Ca2+ entry) diphenylboric acid 2-amino-
nantly expressed in astroglial cells (Holtzclaw et al., 2002; Petra- ethyl ester (2-APB). Exposure of cultured astrocytes to 10 lM ryan-
vicz et al., 2008; Sheppard et al., 1997). The InsP3 receptors are odine for 10 min also decreased mechanically-induced glutamate
often concentrated in the distal processes of astroglial cells (Holtz- release. The pre-incubation of astrocytes with 10 mM of caffeine
claw et al., 2002), where Ca2+ signals in response to metabotropic (which acts as both inhibitor of InsP3 receptors and activator of
stimulation seems to be initiated (Fig. 2D, Kirischuk et al., 1995). RyRs (Ehrlich et al., 1994; Wakui et al., 1990)) for 10 min similarly

Fig. 2. ATP-induced Ca2+ signalling in Bergmann glial cells results exclusively from inositol 1,4,5-trisphosphate (InsP3)-mediated Ca2+ release from ER Ca2+ stores. (A) ATP-
induced [Ca2+]i transients were measured from ‘‘bulk-loaded’’ Bergmann glial cells stained by incubating cerebellar slices in fura-2 acetoxymethyl ester (AM)-containing
solutions. Addition of ATP triggered an increase in [Ca2+]i that persisted in Ca2+-free extracellular solution. (B) In a similar experiment, incubation of slice with 500 nM
thapsigargin completely and irreversibly blocked ATP-induced Ca2+ signalling. (C) Intracellular administration of heparin via intracellular dialysis with a patch pipette
inhibited [Ca2+]i increase induced by ATP. Control [Ca2+]i transient was recorded from fura 2-AM-loaded cells before commencing intracellular dialysis. (D) Illustration of
spatial distribution of [Ca2+]i at time of maximum ATP response. Note higher levels of [Ca2+]i in Bergmann glial cell processes as compared with cell body. Modified from
Kirischuk et al. (1995).
50 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

reduced glutamate release. When astrocytes were pre-treated with The store-operated Ca2+ entry (SOCE) is present in all types of
mixtures of 2-APB with ryanodine or caffeine no additive effect on neuroglial cells (Pivneva et al., 2008; Toescu et al., 1998; Tuschick
mechanically-induced glutamate release was found. Finally incu- et al., 1997). The molecular identity of SOCE-related channels in
bation of astrocytes with 100 lM of Cd2+ (Cd2+ ions are known as astrocytes are yet to be fully clarified. Expression of Orai/STIM1
a broad spectrum antagonist of Ca2+ permeable plasma membrane proteins was detected only in astroglial cell line U373 MG (Barajas
channels), reduced mechanically-induced glutamate release to 55% et al., 2008). The Ca2+-release activated Ca2+ channel currents
of control. All these pharmacological agents not only inhibited glu- (ICRAC) associated with SOCE in various immune cells also were
tamate release but also reduced the mechanically induced [Ca2+]i not detected in astrocytes; there are only some indications of sim-
transients. These data confirm that Ca2+ release from the ER repre- ilarities in the pharmacological profiles of SOCE in astroglia and in
sents the main source of Ca2+ for controlling release of glutamate, the rat basophilic leukemia cell line (Pizzo et al., 2001).
although plasmalemmal Ca2+ influx also has a role. The release of The expression of transient receptor potential (TRP) channels
glutamate and/or D-serine activated by the ER Ca2+ release was also has been identified in astrocytes (Golovina, 2005; Grimaldi et al.,
implicated in regulation of synaptic activity in Schaffer collateral- 2003; Pizzo et al., 2001), and these channels were found to partic-
CA1 neuronal synapses (Angulo et al., 2004; Fellin et al., 2004; ipate in shaping of Ca2+ signals in astroglia (Golovina, 2005;
Henneberger et al., 2010; Perea and Araque, 2005; Rusakov et al., Grimaldi et al., 2003; Pizzo et al., 2001). It appears that the brain
2011; Shigetomi et al., 2008). native and functional canonical type TRP (TRPC) channels are het-
Calcium signals originated in the ER are instrumental for astrog- eromultimeric and formed by association of an obligatory TRPC1
lial regulation of local blood flow. Glutamate-induced astroglial with ancillary TRPC4 and/or TRPC5 proteins (Hofmann et al.,
[Ca2+]i transients induce release of vasoactive substances such as 2002; Strubing et al., 2001, 2003). Thus, affecting the activity/
derivatives of arachidonic acid or carbon monoxide that in turn af- expression of TRPC1 protein alone should unveil the role of this
fect the tone of cerebral arterioles (Mulligan and MacVicar, 2004; pathway in intracellular Ca2+ dynamics in astrocytes should they
Xi et al., 2011; Zonta et al., 2003). The ER Ca2+ release can also con- express these TRPC isoforms. Indeed, freshly isolated and cultured
trol astroglial apoptosis through transactivation of proapoptotic astrocytes express all the three above TRPC isoforms (Golovina,
factor Bax (Morales et al., 2011). 2005; Malarkey et al., 2008). Looking at the function of these pro-
Recently, however, the role of metabotropic receptors as well as teins, antisense knock down of the TRPC1 gene or immunological
the InsP3-induced Ca2+ release in astroglia-dependent regulation of approach by using a blocking antibody directed at an epitope in
neuronal networks was questioned. In experiments on mice with the pore forming region of the TRPC1 protein reduced SOCE in cul-
genetically modified Ca2+ signalling pathways in astrocytes (which tured astrocytes (Golovina, 2005; Malarkey et al., 2008). Addition-
either overexpressed Mas-related gene A1, MrgA1, metabotropic ally, immunological block of TRPC1-mediated SOCE resulted in
receptor normally present only in sensory neurones, or did not ex- reduction of purinergic receptor activation induced cytoplasmic
press type 2 InsP3 receptors (Agulhon et al., 2010; Fiacco et al., Ca2+ elevations in astrocytes (Fig. 3A) (Malarkey et al., 2008).
2007; Petravicz et al., 2008)) has demonstrated that neither ampli- Hence, stimulation of astrocytes with ATP caused a biphasic
fication nor occlusion of astroglial metabotropic Ca2+ signalling af- response, where an initial transient increase in cytoplasmic Ca2+
fects synaptic transmission/plasticity in hippocampus. This level was followed by a sustained (plateau) Ca2+ elevation
however remains debatable and recent experiments described spe- (Fig. 3A), the latter known to represent Ca2+entry from the extra-
cific conditions, under which astroglial InsP3/Ca2+ signalling initi- cellular space (Tuschick et al., 1997) via stimulated SOCE channels
ates modulation of synaptic plasticity (available in preliminary (Kresse et al., 2005). Interestingly, preincubation of astrocytes with
form Haydon and Lee, 2011). TRPC1 antibody resulted in a 34% reduction in the ATP-induced
It is important to consider also that the ER is a multifunctional transient Ca2+ elevation, along with an abolishment of the Ca2+ pla-
organelle, which is involved not only in Ca2+ signalling but also teau. These data identify TRPC1 protein as a likely component of
controls various aspects of cell biochemistry and the intracellular the SOCE channel activity upon purinergic receptor stimulation
transport of molecules (Berridge, 2002; Bootman et al., 2002). in astrocytes (Malarkey et al., 2008). Similarly, mechanically-
Importantly, fluctuations on the ER Ca2+ that are affected by Ca2+ induced Ca2+ response in astrocytes was greatly reduced by an
release also act as a signalling event inside the ER by changing acute immunological inhibition of TRPC1 protein resulting in
the activity of intra-ER enzymes such as chaperones (Michalak reduced Ca2+-dependent glutamate release from astrocytes
et al., 2002; Verkhratsky and Petersen, 2002) thus providing the (Malarkey et al., 2008) (Fig. 3B and C).
link between cell activity and protein synthesis, post-translational
modification and trafficking.. 3.3. Ionotropic Ca2+ permeable receptors in astrocytes
The pre-eminence of intracellular Ca2+ as the source in glial sig-
nalling, however, does not exclude a role for plasmalemmal path- The next important pathway involved in astroglial Ca2+ signal-
ways for Ca2+ entry in shaping Ca2+ signals. ling is associated with ionotropic Ca2+ permeable receptors. The
Ca2+ permeable ligand-gated ion channels were identified in
3.2. The store-operated Ca2+ entry: a role for TRPC channels freshly isolated astroglial cells, in astrocytes in cultures and in
brain slices (Lalo et al., 2011b; Verkhratsky and Steinhauser,
In the majority of electrically non-excitable cells the depletion 2000). The main ionotropic receptors involved in astroglial Ca2+
of the ER Ca2+ store triggers secondary Ca2+ influx that involves signalling are represented by the a-amino-3-hydroxy-5-methyl-
opening of plasmalemmal channels. This mechanism is generally 4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate
referred to as store-operated or capacitative Ca2+ entry (Putney, (NMDA) glutamate receptors and P2X purinoceptors (Lalo et al.,
1986, 1990). Physiologically, the activation of the store-operated 2006, 2008, 2011b,c; Verkhratsky and Kirchhoff, 2007). In many
Ca2+ entry is important for both replenishing the ER store (capaci- types of astrocytes the AMPA receptors do not express GluR-B
tative function) and for producing the plateau phase of the Ca2+ (GluR2) subunit (Glaum et al., 1990; Muller et al., 1992), which
transient which may outlast the period of metabotropic activation. makes these receptors Ca2+ permeable (PCa/Pmonovalent  1,
At the molecular level this mechanism is operated through specific (Burnashev et al., 1992). Rapid desensitization of AMPA receptors
store-operated plasmalemmal channels (which may involve for- in physiological conditions, however severely limits Ca2+ influx.
mation of Orai/Stim1 protein complexes (Putney, 2007)) or some The second class of glutamate receptors expressed in astrocytes
types of TRP channels (Smyth et al., 2006). is the NMDA receptors. These NMDA receptors were found in
A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56 51

stimulation trigger cytosolic Ca2+ signals (Lalo et al., 2008). There


is also some evidence for functional expression of a7 Ca2+ perme-
able nicotinic cholinoreceptors in cultured astrocytes (Oikawa
et al., 2005; Sharma and Vijayaraghavan, 2001); although the pres-
ence of these receptors in astroglial cells in situ has not been hith-
erto confirmed.
The ionotropic receptors expressed in astroglia are cationic
channels, and their activation results not only in Ca2+ influx but
also in very substantial Na+ entry. Changes in cytoplasmic Na+ con-
centration modulate another important component of Ca2+ homeo-
static machinery linked to the sodium/calcium exchanger (NCX)
that is also relevant for shaping astroglial Ca2+ signals.

3.4. The sodium/calcium exchanger in astroglia

All three types of mammalian Na+/Ca2+ exchangers, the NCX1,


NCX2 and NCX3 are expressed in astroglia and are localized in peri-
synaptic processes, especially in those covering excitatory syn-
apses (Minelli et al., 2007). The NCX have complex effect on
[Ca2+]i being involved in both Ca2+ extrusion and Ca2+ entry; the
mode of the NCX operation (forward vs. reverse) is determined
by transmembrane Na+ gradient and membrane potential (increase
in [Na+]i and depolarisation favour the reverse mode). The NCX-
mediated Ca2+ fluxes in both forward and reverse modes were
found in both cultured astrocytes and astroglial cells in situ (Gold-
man et al., 1994; Kirischuk et al., 1997; Takuma et al., 1994). Acti-
vation of astroglial ionotropic receptors induce substantial [Na+]i
elevation, which turns the exchanger into the reverse mode (Kiris-
chuk et al., 1997). Similar effects could be induced by activation of
glutamate transporter that transports three Na ions with one glu-
tamate and may trigger [Na+]i increase (Kirischuk et al., 2007; Ro-
jas et al., 2007). The reverse mode of the NCX can also be triggered
Fig. 3. TRPC1 plays a role in intracellular Ca2+ elevations in astrocytes. (A)
Application of ATP (100 lM) to astrocytes results in a biphasic response: the initial by mild depolarisation of cultured astrocytes (Paluzzi et al., 2007).
transient Ca2+ elevation and sustained (plateau) Ca2+ elevation. Vertical dashed line Astroglial Ca2+ signals produced by the reverse NCX were shown to
indicates the initial point of a sustained plateau Ca2+ response. The time of ATP initiate exocytotic glutamate release (Paluzzi et al., 2007).
application is indicated by the horizontal bar. Both phases are reduced when TRPC1
containing channels are blocked with an antibody (Ab) against TRPC1. (B and C)
Mechanical stimulation of astrocytes evokes cytoplasmic Ca2+ elevations (B) and 3.5. Mitochondria in astroglial Ca2+ signalling
consequential glutamate release (C). Arrows indicate the time when the mechanical
stimulus occurred. Both measurements are reduced by incubating astrocytes with
TRPC1 antibody. Traces in (A) and (B), reporting on intracellular Ca2+ levels, Mitochondria in astrocytes were shown to sequester or release
represent mean time courses of fluo-3 and X-rhod-1 fluorescence expressed as DF/ Ca2+ to affect cytosolic Ca2+ changes and consequently modulate
F0 ± SEMs (percentage). Traces in (C) report on extracellular glutamate levels and exocytotic release of glutamate (Reyes and Parpura, 2008). When
are expressed as mean DF/F0 ± SEMs of NADH fluorescence. Modified from mitochondrial Ca2+ sequestration via the uniporter was inhibited
Malarkey et al. (2008).
by Ruthenium 360, mechanically-induced [Ca2+]i increase and glu-
tamate release were augmented in rat astrocytes. The converse
treatment of blocking the mitochondrial Ca2+ release reduced exo-
acutely isolated cortical astrocytes as well as in astrocytes in corti- cytotic release of glutamate from astrocytes. Hence, the inhibition
cal slices (Lalo et al., 2006, 2011a; Palygin et al., 2011). Astroglial of the mitochondrial Na+/Ca2+ exchanger with 7-chloro-5-(2-chlo-
NMDA receptors have specific gating and pharmacology. First they rophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one (CGP37157)
demonstrate rather weak Mg2+ block which start to develop at reduced [Ca2+]i elevation and Ca2+-regulated glutamate release.
membrane potentials negative to 80 mV (Palygin et al., 2011). Additional pathway for Ca2+ release from mitochondria is via the
Second, glial NMDA receptors were significantly more sensitive mitochondrial permeability transition pore (MPTP), a high conduc-
to memantine and GluNR2C/D subunit-selective antagonist tance channel, whose transient openings may serve a physiological
UBP141 (Palygin et al., 2011). Third, NMDA receptors in astroglia role in the rapid Ca2+ removal from mitochondria (Altschuld et al.,
had a substantially lower Ca2+ permeability (PCa/Pmonovalent 3 vs. 1992). Cyclophilin D (CypD), a matrix mitochondrial protein is in-
10 in neurones (Palygin et al., 2010)); although this was sufficient volved in modulation of the MPTP affinity for Ca2+. The agent cyclo-
to produce cytoplasmic Ca2+ transients in response to both exoge- sporin A (CsA) inhibits opening of the MPTP after binding to CypD
nous agonists and synaptic stimulation (Fig. 4, Palygin et al., 2010). (Basso et al., 2005). The application of CsA to rat astrocytes caused
The complex of biophysical and pharmacological properties indi- a reduction in [Ca2+]i elevation and Ca2+-depenent glutamate re-
cates that astroglial NMDA receptors are assembled from NR1, lease (Reyes and Parpura, 2008). Interestingly, mouse CypD
NR2C/D and NR3 subunits. knock-out astrocytes had attenuated mechanically-induced Ca2+
The ATP-gated P2X1/5 purinoceptors are also present in cortical responses, but their exocytotic release of glutamate was unexpect-
astrocytes (Lalo et al., 2008). These receptors are exceptionally sen- edly augmented (Reyes et al., 2011). This enhancement of the
sitive to ATP (EC50  50 nM), and have very weak desensitisation in glutamatergic output had been attributed to extramitochondrial
the presence of agonist. The P2X1/5 receptors are Ca2+ permeable effects via a downstream of Ca2+ modulation, likely at the level of
(PCa/Pmonovalent  2) and their activation by ATP or by synaptic secretory machinery.
52 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

Fig. 4. Synaptically induced ionotropic Ca2+ signals in protoplasmic cortical astrocytes in situ. (A and B) Cortical layer II astrocytes were loaded with Fura-2 in situ via patch
pipette. Fluorescent images were recorded simultaneously with glial currents evoked by neuronal afferent stimulation in presence of CNQX (control) and after application of
30 lM D-AP5 (A) and 10 nM NF-449, selective antagonist of P2X receptors (B). Representative images (pseudo-colour, pipette image subtracted; warmer colours correspond
to higher [Ca2+]i levels) and glial synaptic currents (right column) were recorded from two different cells before (images on the top) and at the peak of the response. [Ca2+]i-
transients (middle columns) are expressed as F340/F380 ratio. Note significant enhancement of glia currents and Ca2+-transients upon high-frequency (HFS) stimulation.
Modified from Palygin et al. (2010).

3.6. Voltage-gated Ca2+ channels (VGCCs) in astroglia ventrobasal thalamus (Parri and Crunelli, 2003; Parri et al.,
2001). In this brain region, it has been demonstrated that astro-
We have recently reviewed the expression of VGCCs in astrocytes cytes in acute slices prepared from 5- to 17-day-old rats show
(Parpura et al., 2011). Briefly, astrocytes in cell culture express five spontaneous [Ca2+]i oscillations, which dually draw Ca2+ from: (i)
main types of VGCCs: L, N, P/Q, R and T types (Barres et al., 1988, the ER intracellular store (SERCA blockers blocked them) and (ii)
1990; D’Ascenzo et al., 2004; Latour et al., 2003; MacVicar, 1984; the extracellular space by the Ca2+ entry via VGCCs (nifedipine
MacVicar et al., 1991; MacVicar and Tse, 1988). The level of expres- blocked them, while BayK8644 enhanced them). Since astrocytic
sion or functionality of VGCCs was often associated with various oscillations lead to glutamate release and consequential NMDA
treatments of astrocytes, such as addition of dibutyryl-cAMP, co- receptor-mediated neuronal excitability, it appears that, at least
culturing them with neurones, or inducing acute oxidative stress in part, astrocytic VGCCs can play a role in modulation of neuronal
(Barres et al., 1990; Bond and Greenfield, 2007; Corvalan et al., activity. NMDA receptors are essential for neuronal differentiation,
1990; MacVicar and Tse, 1988). Indeed, activity of VGCCs in cultured migration and synaptogenesis in the developing brain (Komuro
or freshly isolated astrocytes can lead to Ca2+ signalling (Duffy and and Rakic, 1993, 1998), which raises a notion that astrocytic VGCCs
MacVicar, 1994; Eriksson et al., 1993; Jalonen et al., 1997). might be important in mediating these developmental events.
Functional expression of VGCCs in astrocytes in situ has been What might be the role of astrocytic VGCCs in mature brain, it re-
studied in developing/young animals with variable outcomes. In mains elusive at the moment. However, it is tempting to speculate
acute slices from the visual cortex and the CA1 hippocampal region that VGCCs could be recruited in service at times when regenera-
of post-natal 5–18 day rats, patch-clamp whole-cell recordings tion and re-establishment of synaptic contacts are necessary. In
from astrocytes fail to detect any VGCC currents (Carmignoto support of such possibility, reactive hippocampal astrocytes of
et al., 1998). Contrary, voltage-clamp experiments on glutamine juvenile mice (12–14 weeks, based on reported weight) submit-
synthetase-positive cells, likely immature astroglia, in hippocam- ted to pilocarpine-induced status epilepticus showed the up-regu-
pus of 9- to 12-old-day mice demonstrated VGCC currents (Akopi- lation of expression of L- and P/Q-type channels at 1 week and 2
an et al., 1996). Similarly, calcium dynamics mediated by VGCCs months following such an insult (Xu et al., 2007).
were detected in astrocytes from neurogenic subventricular zone
(Young et al., 2010) and the ventrobasal thalamus (Parri and Cru- 4. Conclusions
nelli, 2003; Parri et al., 2001).
Thus far, the potential functional role of VGCCs expression in The intent of this review was to summarize the Ca2+ signalling
astrocytes in situ remains limited to the example from the in astroglia. These cells are heterogeneous in morphology and
A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56 53

function and Ca2+ signalling is no exception. We discussed general Bregestovski, P., Spitzer, N., 2005. Calcium in the function of the nervous system:
new implications. Cell Calcium 37, 371–374.
features of Ca2+ signalling mainly in astrocytes. Many of ap-
Burdakov, D., Petersen, O.H., Verkhratsky, A., 2005. Intraluminal calcium as a
proached used by astrocytes are common to those utilised by other primary regulator of endoplasmic reticulum function. Cell Calcium 38, 303–
electrically non-excitable cells. Yet, there are some specifics related 310.
to the context of the astrocytic residence in the brain parenchyma. Burgoyne, R.D., O’Callaghan, D.W., Hasdemir, B., Haynes, L.P., Tepikin, A.V., 2004.
Neuronal Ca2+-sensor proteins: multitalented regulators of neuronal function.
Hence, we briefly discussed astrocytic exocytosis which critically Trends Neurosci. 27, 203–209.
depends on intracellular Ca2+ variations. This pathway leads to Burnashev, N., Khodorova, A., Jonas, P., Helm, P.J., Wisden, W., Monyer, H., Seeburg,
gliotransmission and signalling to adjacent neurones. There are, P.H., Sakmann, B., 1992. Calcium-permeable AMPA-kainate receptors in
fusiform cerebellar glial cells. Science 256, 1566–1570.
however, many issues that remain to be resolved. For example, Burnstock, G., Verkhratsky, A., 2009. Evolutionary origins of the purinergic
the activation of ionotropic receptors expressed by astrocytes can signalling system. Acta Physiol. (Oxf.) 195, 415–447.
lead to increases in cyotosolic Ca2+, yet whether this excitation is Carafoli, E., 2002. Calcium signaling: a tale for all seasons. Proc. Natl. Acad. Sci. USA
99, 1115–1122.
coupled to the exocytotic gliotransmission from astrocytes has Carafoli, E., 2004. The ambivalent nature of the calcium signal. J. Endocrinol. Invest.
not been determined. Furthermore, it will be necessary to deter- 27, 134–136.
mine the role of VGCCs as the conduit for Ca2+ entry from the Carafoli, E., Santella, L., Branca, D., Brini, M., 2001. Generation, control, and
processing of cellular calcium signals. Crit. Rev. Biochem. Mol. Biol. 36, 107–
extracellular space. For instance, does this pathway operate under 260.
physiological conditions or it primarily operates under pathologi- Carmignoto, G., Pasti, L., Pozzan, T., 1998. On the role of voltage-dependent calcium
cal conditions, such as acute oxidative stress. It appears that channels in calcium signaling of astrocytes in situ. J. Neurosci. 18, 4637–4645.
Case, R.M., Eisner, D., Gurney, A., Jones, O., Muallem, S., Verkhratsky, A., 2007.
addressing these questions will require combining cell culture,
Evolution of calcium homeostasis: from birth of the first cell to an omnipresent
slice and in vivo approaches that will be labour intensive efforts signalling system. Cell Calcium 42, 345–350.
until we get more detailed picture. Cavazzini, M., Bliss, T., Emptage, N., 2005. Ca2+ and synaptic plasticity. Cell Calcium
38, 355–367.
Cornell Bell, A.H., Finkbeiner, S.M., Cooper, M.S., Smith, S.J., 1990. Glutamate induces
calcium waves in cultured astrocytes: long-range glial signaling. Science 247,
Acknowledgements 470–473.
Corvalan, V., Cole, R., de Vellis, J., Hagiwara, S., 1990. Neuronal modulation of
Authors research was supported by Alzheimer’s Research Trust calcium channel activity in cultured rat astrocytes. Proc. Natl. Acad. Sci. USA 87,
4345–4348.
(UK) Programme Grant (ART/PG2004A/1) to A.V. and J.J.R.; by Na- D’Ascenzo, M., Vairano, M., Andreassi, C., Navarra, P., Azzena, G.B., Grassi, C., 2004.
tional Science Foundation (CBET 0943343) grant to V.P., by the Electrophysiological and molecular evidence of L-(Cav1), N-(Cav2.2), and R-
Grant Agency of the Czech Republic (GACR 309/09/1696) to J.J.R. (Cav2.3) type Ca2+ channels in rat cortical astrocytes. Glia 45, 354–363.
Dani, J.W., Chernjavsky, A., Smith, S.J., 1992. Neuronal activity triggers calcium
and (GACR 305/08/1384) to A.V. waves in hippocampal astrocyte networks. Neuron 8, 429–440.
Deitmer, J.W., Rose, C.R., Munsch, T., Schmidt, J., Nett, W., Schneider, H.P., Lohr, C.,
1999. Leech giant glial cell: functional role in a simple nervous system. Glia 28,
References 175–182.
Duchen, M.R., Verkhratsky, A., Muallem, S., 2008. Mitochondria and calcium in
health and disease. Cell Calcium 44, 1–5.
Agulhon, C., Fiacco, T.A., McCarthy, K.D., 2010. Hippocampal short- and long-term
Duffy, S., MacVicar, B.A., 1994. Potassium-dependent calcium influx in acutely
plasticity are not modulated by astrocyte Ca2+ signaling. Science 327, 1250–
isolated hippocampal astrocytes. Neuroscience 61, 51–61.
1254.
Edwards, T.N., Meinertzhagen, I.A., 2010. The functional organisation of glia in the
Akopian, G., Kressin, K., Derouiche, A., Steinhauser, C., 1996. Identified glial cells in
adult brain of Drosophila and other insects. Prog. Neurobiol. 90, 471–497.
the early postnatal mouse hippocampus display different types of Ca2+ currents.
Ehrlich, B.E., Kaftan, E., Bezprozvannaya, S., Bezprozvanny, I., 1994. The
Glia 17, 181–194.
pharmacology of intracellular Ca2+-release channels. Trends Pharmacol. Sci.
Alonso, M.T., Barrero, M.J., Michelena, P., Carnicero, E., Cuchillo, I., Garcia, A.G.,
15, 145–149.
Garcia-Sancho, J., Montero, M., Alvarez, J., 1999. Ca2+-induced Ca2+ release in
Eriksson, P.S., Nilsson, M., Wagberg, M., Hansson, E., Ronnback, L., 1993. Kappa-
chromaffin cells seen from inside the ER with targeted aequorin. J. Cell Biol. 144,
opioid receptors on astrocytes stimulate L-type Ca2+ channels. Neuroscience 54,
241–254.
401–407.
Altschuld, R.A., Hohl, C.M., Castillo, L.C., Garleb, A.A., Starling, R.C., Brierley, G.P.,
Fellin, T., Pascual, O., Gobbo, S., Pozzan, T., Haydon, P.G., Carmignoto, G., 2004.
1992. Cyclosporin inhibits mitochondrial calcium efflux in isolated adult rat
Neuronal synchrony mediated by astrocytic glutamate through activation of
ventricular cardiomyocytes. Am. J. Physiol. 262, H1699–H1704.
extrasynaptic NMDA receptors. Neuron 43, 729–743.
Angulo, M.C., Kozlov, A.S., Charpak, S., Audinat, E., 2004. Glutamate released from
Fiacco, T.A., Agulhon, C., Taves, S.R., Petravicz, J., Casper, K.B., Dong, X., Chen, J.,
glial cells synchronizes neuronal activity in the hippocampus. J. Neurosci. 24,
McCarthy, K.D., 2007. Selective stimulation of astrocyte calcium in situ does not
6920–6927.
affect neuronal excitatory synaptic activity. Neuron 54, 611–626.
Bacaj, T., Tevlin, M., Lu, Y., Shaham, S., 2008. Glia are essential for sensory organ
Fields, R.D., Lee, P.R., Cohen, J.E., 2005. Temporal integration of intracellular Ca2+
function in C. elegans. Science 322, 744–747.
signaling networks in regulating gene expression by action potentials. Cell
Barajas, M., Andrade, A., Hernandez-Hernandez, O., Felix, R., Arias-Montano, J.A.,
Calcium 37, 433–442.
2008. Histamine-induced Ca2+ entry in human astrocytoma U373 MG cells:
Finkbeiner, S.M., 1993. Glial calcium. Glia 9, 83–104.
evidence for involvement of store-operated channels. J. Neurosci. Res. 86, 3456–
Gerasimenko, O.V., Gerasimenko, J.V., Belan, P.V., Petersen, O.H., 1996. Inositol
3468.
trisphosphate and cyclic ADP-ribose-mediated release of Ca2+ from single
Barres, B.A., Chun, L.L., Corey, D.P., 1988. Ion channel expression by white matter
isolated pancreatic zymogen granules. Cell 84, 473–480.
glia: I. Type 2 astrocytes and oligodendrocytes. Glia 1, 10–30.
Giaume, C., Venance, L., 1998. Intercellular calcium signaling and gap junctional
Barres, B.A., Koroshetz, W.J., Chun, L.L., Corey, D.P., 1990. Ion channel expression by
communication in astrocytes. Glia 24, 50–64.
white matter glia: the type-1 astrocyte. Neuron 5, 527–544.
Glaum, S.R., Holzwarth, J.A., Miller, R.J., 1990. Glutamate receptors activate Ca2+
Basso, E., Fante, L., Fowlkes, J., Petronilli, V., Forte, M.A., Bernardi, P., 2005. Properties
mobilization and Ca2+ influx into astrocytes. Proc. Natl. Acad. Sci. USA 87, 3454–
of the permeability transition pore in mitochondria devoid of Cyclophilin D. J.
3458.
Biol. Chem. 280, 18558–18561.
Goldman, W.F., Yarowsky, P.J., Juhaszova, M., Krueger, B.K., Blaustein, M.P., 1994.
Beck, A., Nieden, R.Z., Schneider, H.P., Deitmer, J.W., 2004. Calcium release from
Sodium/calcium exchange in rat cortical astrocytes. J. Neurosci. 14, 5834–5843.
intracellular stores in rodent astrocytes and neurons in situ. Cell Calcium 35,
Golovina, V.A., 2005. Visualization of localized store-operated calcium entry in
47–58.
mouse astrocytes. Close proximity to the endoplasmic reticulum. J. Physiol. 564,
Berridge, M.J., 2002. The endoplasmic reticulum: a multifunctional signaling
737–749.
organelle. Cell Calcium 32, 235–249.
Gourine, A.V., Kasparov, S., 2011. Astrocytes as brain interoceptors. Exp. Physiol. 96,
Berridge, M.J., Bootman, M.D., Roderick, H.L., 2003. Calcium signalling: dynamics,
411–416.
homeostasis and remodelling. Nat. Rev. Mol. Cell Biol. 4, 517–529.
Gourine, A.V., Kasymov, V., Marina, N., Tang, F., Figueiredo, M.F., Lane, S.,
Berridge, M.J., Lipp, P., Bootman, M.D., 2000. The versatility and universality of
Teschemacher, A.G., Spyer, K.M., Deisseroth, K., Kasparov, S., 2010. Astrocytes
calcium signalling. Nat. Rev. Mol. Cell Biol. 1, 11–21.
control breathing through pH-dependent release of ATP. Science 329, 571–575.
Bolsover, S.R., 2005. Calcium signalling in growth cone migration. Cell Calcium 37,
Grimaldi, M., Maratos, M., Verma, A., 2003. Transient receptor potential channel
395–402.
activation causes a novel form of [Ca2+]i oscillations and is not involved in
Bond, C.E., Greenfield, S.A., 2007. Multiple cascade effects of oxidative stress on
capacitative Ca2+ entry in glial cells. J. Neurosci. 23, 4737–4745.
astroglia. Glia 55, 1348–1361.
Guerini, D., Coletto, L., Carafoli, E., 2005. Exporting calcium from cells. Cell Calcium
Bootman, M.D., Petersen, O.H., Verkhratsky, A., 2002. The endoplasmic reticulum is
38, 281–289.
a focal point for co-ordination of cellular activity. Cell Calcium 32, 231–234.
54 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

Guerrero-Hernandez, A., Dagnino-Acosta, A., Verkhratsky, A., 2010. An intelligent Kresse, W., Sekler, I., Hoffmann, A., Peters, O., Nolte, C., Moran, A., Kettenmann, H.,
sarco-endoplasmic reticulum Ca2+ store: release and leak channels have 2005. Zinc ions are endogenous modulators of neurotransmitter-stimulated
differential access to a concealed Ca2+ pool. Cell Calcium 48, 143–149. capacitative Ca2+ entry in both cultured and in situ mouse astrocytes. Eur. J.
Hamilton, N., Vayro, S., Kirchhoff, F., Verkhratsky, A., Robbins, J., Gorecki, D.C., Butt, Neurosci. 21, 1626–1634.
A.M., 2008. Mechanisms of ATP- and glutamate-mediated calcium signaling in Kuga, N., Sasaki, T., Takahara, Y., Matsuki, N., Ikegaya, Y., 2011. Large-scale calcium
white matter astrocytes. Glia 56, 734–749. waves traveling through astrocytic networks in vivo. J. Neurosci. 31, 2607–2614.
Hartmann, J., Konnerth, A., 2005. Determinants of postsynaptic Ca2+ signaling in Lalo, U., Palygin, O., North, R.A., Verkhratsky, A., Pankratov, Y., 2011a. Age-
Purkinje neurons. Cell Calcium 37, 459–466. dependent remodelling of ionotropic signalling in cortical astroglia. Aging Cell
Haydon, P.G., Lee, S., 2011. The alleged glial controversy is no more: conditions 10, 392–402.
under which astrocytic IP3/Ca2+ signals do modulate synaptic plasticity. Trans. Lalo, U., Pankratov, Y., Kirchhoff, F., North, R.A., Verkhratsky, A., 2006. NMDA
Am. Soc. Neurochem. 15. receptors mediate neuron-to-glia signaling in mouse cortical astrocytes. J.
Heilbrunn, L.V., 1943. An Outline of General Physiology. Saunders, Philadelphia. Neurosci. 26, 2673–2683.
Heilbrunn, L.V., Wiercinsky, F.J., 1947. Action of various cations on muscle Lalo, U., Pankratov, Y., Parpura, V., Verkhratsky, A., 2011b. Ionotropic receptors in
protoplasm. J. Cell. Comp. Physiol. 19, 15–32. neuronal–astroglial signalling: what is the role of ‘‘excitable’’ molecules in non-
Heneka, M.T., Rodriguez, J.J., Verkhratsky, A., 2010. Neuroglia in neurodegeneration. excitable cells. Biochim. Biophys. Acta 1813, 992–1002.
Brain Res. Rev. 63, 189–211. Lalo, U., Pankratov, Y., Wichert, S.P., Rossner, M.J., North, R.A., Kirchhoff, F.,
Henneberger, C., Papouin, T., Oliet, S.H., Rusakov, D.A., 2010. Long-term potentiation Verkhratsky, A., 2008. P2X1 and P2X5 subunits form the functional P2X
depends on release of D-serine from astrocytes. Nature 463, 232–236. receptor in mouse cortical astrocytes. J. Neurosci. 28, 5473–5480.
Hofmann, T., Schaefer, M., Schultz, G., Gudermann, T., 2002. Subunit composition of Lalo, U., Verkhratsky, A., Pankratov, Y., 2011c. Ionotropic ATP receptors in neuronal–
mammalian transient receptor potential channels in living cells. Proc. Natl. glial communication. Semin. Cell Dev. Biol. 22, 220–228.
Acad. Sci. USA 99, 7461–7466. Laming, P.R., Kimelberg, H., Robinson, S., Salm, A., Hawrylak, N., Muller, C., Roots, B.,
Holcman, D., Korkotian, E., Segal, M., 2005. Calcium dynamics in dendritic spines, Ng, K., 2000. Neuronal–glial interactions and behaviour. Neurosci. Biobehav.
modeling and experiments. Cell Calcium 37, 467–475. Rev. 24, 295–340.
Holtzclaw, L.A., Pandhit, S., Bare, D.J., Mignery, G.A., Russell, J.T., 2002. Astrocytes in Latour, I., Hamid, J., Beedle, A.M., Zamponi, G.W., Macvicar, B.A., 2003. Expression of
adult rat brain express type 2 inositol 1,4,5-trisphosphate receptors. Glia 39, voltage-gated Ca2+ channel subtypes in cultured astrocytes. Glia 41, 347–353.
69–84. Lenhossek, M.v., 1891. Zur Kenntnis der Neuroglia des menschlichen
Hua, X., Malarkey, E.B., Sunjara, V., Rosenwald, S.E., Li, W.H., Parpura, V., 2004. Ca2+- Ruckenmarkes. Verh. Anat. Ges. 5, 193–221.
dependent glutamate release involves two classes of endoplasmic reticulum Lenhossek, M.v., 1893. Der feinere Bau des Nervensystems im Lichte neuester
Ca2+ stores in astrocytes. J. Neurosci. Res. 76, 86–97. Forschung. Fischer’s Medicinische Buchhandlung H. Kornfield, Berlin.
Huckstepp, R.T., id Bihi, R., Eason, R., Spyer, K.M., Dicke, N., Willecke, K., Marina, N., Lewit-Bentley, A., Rety, S., 2000. EF-hand calcium-binding proteins. Curr. Opin.
Gourine, A.V., Dale, N., 2010. Connexin hemichannel-mediated CO2-dependent Struct. Biol. 10, 637–643.
release of ATP in the medulla oblongata contributes to central respiratory Lohmann, C., Wong, R.O., 2005. Regulation of dendritic growth and plasticity by
chemosensitivity. J. Physiol. 588, 3901–3920. local and global calcium dynamics. Cell Calcium 37, 403–409.
Ikura, M., Osawa, M., Ames, J.B., 2002. The role of calcium-binding proteins in the MacVicar, B.A., 1984. Voltage-dependent calcium channels in glial cells. Science
control of transcription: structure to function. Bioessays 24, 625–636. 226, 1345–1347.
Imredy, J.P., Yue, D.T., 1994. Mechanism of Ca2+-sensitive inactivation of L-type Ca2+ MacVicar, B.A., Hochman, D., Delay, M.J., Weiss, S., 1991. Modulation of intracellular
channels. Neuron 12, 1301–1318. Ca++ in cultured astrocytes by influx through voltage-activated Ca++ channels.
Innocenti, B., Parpura, V., Haydon, P.G., 2000. Imaging extracellular waves of Glia 4, 448–455.
glutamate during calcium signaling in cultured astrocytes. J. Neurosci. 20, MacVicar, B.A., Tse, F.W., 1988. Norepinephrine and cyclic adenosine 30 :50 -cyclic
1800–1808. monophosphate enhance a nifedipine-sensitive calcium current in cultured rat
Jaiswal, J.K., 2001. Calcium – how and why? J. Biosci. 26, 357–363. astrocytes. Glia 1, 359–365.
Jalonen, T.O., Margraf, R.R., Wielt, D.B., Charniga, C.J., Linne, M.L., Kimelberg, H.K., Magistretti, P.J., 2009. Neuroscience. Low-cost travel in neurons. Science 325, 1349–
1997. Serotonin induces inward potassium and calcium currents in rat cortical 1351.
astrocytes. Brain Res. 758, 69–82. Malarkey, E.B., Ni, Y., Parpura, V., 2008. Ca2+ entry through TRPC1 channels
Jeremic, A., Jeftinija, K., Stevanovic, J., Glavaski, A., Jeftinija, S., 2001. ATP stimulates contributes to intracellular Ca2+ dynamics and consequent glutamate release
calcium-dependent glutamate release from cultured astrocytes. J. Neurochem. from rat astrocytes. Glia 56, 821–835.
77, 664–675. Malarkey, E.B., Parpura, V., 2009. Mechanisms of transmitter release from
Kastritsis, C.H., Salm, A.K., McCarthy, K., 1992. Stimulation of the P2Y purinergic astrocytes. In: Parpura, V., Haydon, P.G. (Eds.), Astrocytes in
receptor on type 1 astroglia results in inositol phosphate formation and calcium (Patho)physiology of the Nervous System. Springer, New York, pp. 301–350.
mobilization. J. Neurochem. 58, 1277–1284. Matyash, M., Matyash, V., Nolte, C., Sorrentino, V., Kettenmann, H., 2002.
Kettenmann, H., Verkhratsky, A., 2008. Neuroglia: the 150 years after. Trends Requirement of functional ryanodine receptor type 3 for astrocyte migration.
Neurosci. 31, 653–659. FASEB J. 16, 84–86.
Kimelberg, H.K., 2010. Functions of mature mammalian astrocytes: a current view. Matyash, V., Kettenmann, H., 2010. Heterogeneity in astrocyte morphology and
Neuroscientist 16, 79–106. physiology. Brain Res. Rev. 63, 2–10.
Kimelberg, H.K., Nedergaard, M., 2010. Functions of astrocytes and their potential as McCarthy, K.D., Salm, A.K., 1991. Pharmacologically-distinct subsets of astroglia can
therapeutic targets. Neurotherapeutics 7, 338–353. be identified by their calcium response to neuroligands. Neuroscience 41, 325–
Kirischuk, S., Kettenmann, H., Verkhratsky, A., 1997. Na+/Ca2+ exchanger modulates 333.
kainate-triggered Ca2+ signaling in Bergmann glial cells in situ. FASEB J. 11, 566– Michalak, M., Robert Parker, J.M., Opas, M., 2002. Ca2+ signaling and calcium binding
572. chaperones of the endoplasmic reticulum. Cell Calcium 32, 269–278.
Kirischuk, S., Kettenmann, H., Verkhratsky, A., 2007. Membrane currents and Michelangeli, F., Ogunbayo, O.A., Wootton, L.L., 2005. A plethora of interacting
cytoplasmic sodium transients generated by glutamate transport in Bergmann organellar Ca2+ stores. Curr. Opin. Cell Biol. 17, 135–140.
glial cells. Pflugers Arch. 454, 245–252. Minelli, A., Castaldo, P., Gobbi, P., Salucci, S., Magi, S., Amoroso, S., 2007. Cellular and
Kirischuk, S., Moller, T., Voitenko, N., Kettenmann, H., Verkhratsky, A., 1995. ATP- subcellular localization of Na+–Ca2+ exchanger protein isoforms, NCX1, NCX2,
induced cytoplasmic calcium mobilization in Bergmann glial cells. J. Neurosci. and NCX3 in cerebral cortex and hippocampus of adult rat. Cell Calcium 41,
15, 7861–7871. 221–234.
Kirischuk, S., Tuschick, S., Verkhratsky, A., Kettenmann, H., 1996. Calcium signalling Miyata, H., Silverman, H.S., Sollott, S.J., Lakatta, E.G., Stern, M.D., Hansford, R.G.,
in mouse Bergmann glial cells mediated by a1-adrenoreceptors and H1 1991. Measurement of mitochondrial free Ca2+ concentration in living single rat
histamine receptors. Eur. J. Neurosci. 8, 1198–1208. cardiac myocytes. Am. J. Physiol. 261, H1123–H1134.
Kiselyov, K., Shin, D.M., Muallem, S., 2003. Signalling specificity in GPCR-dependent Mogami, H., Tepikin, A.V., Petersen, O.H., 1998. Termination of cytosolic Ca2+
Ca2+ signalling. Cell. Signal. 15, 243–253. signals: Ca2+ reuptake into intracellular stores is regulated by the free Ca2+
Knot, H.J., Laher, I., Sobie, E.A., Guatimosim, S., Gomez-Viquez, L., Hartmann, H., concentration in the store lumen. EMBO J. 17, 435–442.
Song, L.S., Lederer, W.J., Graier, W.F., Malli, R., Frieden, M., Petersen, O.H., 2005. Montana, V., Malarkey, E.B., Verderio, C., Matteoli, M., Parpura, V., 2006. Vesicular
Twenty years of calcium imaging: cell physiology to dye for. Mol. Interv. 5, 112– transmitter release from astrocytes. Glia 54, 700–715.
127. Morad, M., Davies, N.W., Kaplan, J.H., Lux, H.D., 1988. Inactivation and block of
Koizumi, S., Lipp, P., Berridge, M.J., Bootman, M.D., 1999. Regulation of ryanodine calcium channels by photo-released Ca2+ in dorsal root ganglion neurons.
receptor opening by lumenal Ca2+ underlies quantal Ca2+ release in PC12 cells. J. Science 241, 842–844.
Biol. Chem. 274, 33327–33333. Morales, A.P., Carvalho, A.C., Monteforte, P.T., Hirata, H., Han, S.W., Hsu, Y.T., Smaili,
Komuro, H., Rakic, P., 1993. Modulation of neuronal migration by NMDA receptors. S.S., 2011. Endoplasmic reticulum calcium release engages Bax translocation in
Science 260, 95–97. cortical astrocytes. Neurochem. Res. 36, 829–838.
Komuro, H., Rakic, P., 1998. Orchestration of neuronal migration by activity of ion Muller, T., Moller, T., Berger, T., Schnitzer, J., Kettenmann, H., 1992. Calcium entry
channels, neurotransmitter receptors, and intracellular Ca2+ fluctuations. J. through kainate receptors and resulting potassium-channel blockade in
Neurobiol. 37, 110–130. Bergmann glial cells. Science 256, 1563–1566.
Kostyuk, P., Verkhratsky, A., 1994. Calcium stores in neurons and glia. Neuroscience Mulligan, S.J., MacVicar, B.A., 2004. Calcium transients in astrocyte endfeet cause
63, 381–404. cerebrovascular constrictions. Nature 431, 195–199.
Kostyuk, P., Verkhratsky, A., 1995. Calcium Signalling in the Nervous System. Wiley Neher, E., Sakaba, T., 2008. Multiple roles of calcium ions in the regulation of
& Sons, Chichester. neurotransmitter release. Neuron 59, 861–872.
A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56 55

Nicholls, D.G., 2005. Mitochondria and calcium signaling. Cell Calcium 38, 311–317. Rizzuto, R., Pozzan, T., 2006. Microdomains of intracellular Ca2+: molecular
Nicotera, P., Orrenius, S., 1998. The role of calcium in apoptosis. Cell Calcium 23, determinants and functional consequences. Physiol. Rev. 86, 369–408.
173–180. Rojas, H., Colina, C., Ramos, M., Benaim, G., Jaffe, E.H., Caputo, C., DiPolo, R., 2007.
Nicotera, P., Petersen, O.H., Melino, G., Verkhratsky, A., 2007. Janus a god with two Na+ entry via glutamate transporter activates the reverse Na+/Ca2+ exchange
faces: death and survival utilise same mechanisms conserved by evolution. Cell and triggers Cai2+-induced Ca2+ release in rat cerebellar type-1 astrocytes. J.
Death Differ. 14, 1235–1236. Neurochem. 100, 1188–1202.
Oberheim, N.A., Takano, T., Han, X., He, W., Lin, J.H., Wang, F., Xu, Q., Wyatt, J.D., Rusakov, D.A., Zheng, K., Henneberger, C., 2011. Astrocytes as regulators of synaptic
Pilcher, W., Ojemann, J.G., Ransom, B.R., Goldman, S.A., Nedergaard, M., 2009. function: a quest for the Ca2+ master key. Neuroscientist, in press.
Uniquely hominid features of adult human astrocytes. J. Neurosci. 29, 3276– Rycroft, B.K., Gibb, A.J., 2004a. Inhibitory interactions of calcineurin (phosphatase
3287. 2B) and calmodulin on rat hippocampal NMDA receptors. Neuropharmacology
Oberheim, N.A., Wang, X., Goldman, S., Nedergaard, M., 2006. Astrocytic complexity 47, 505–514.
distinguishes the human brain. Trends Neurosci. 29, 547–553. Rycroft, B.K., Gibb, A.J., 2004b. Regulation of single NMDA receptor channel activity
Oertner, T.G., Matus, A., 2005. Calcium regulation of actin dynamics in dendritic by alpha-actinin and calmodulin in rat hippocampal granule cells. J. Physiol.
spines. Cell Calcium 37, 477–482. (Lond.) 557, 795–808.
Oikawa, H., Nakamichi, N., Kambe, Y., Ogura, M., Yoneda, Y., 2005. An increase in Sharma, G., Vijayaraghavan, S., 2001. Nicotinic cholinergic signaling in hippocampal
intracellular free calcium ions by nicotinic acetylcholine receptors in a single astrocytes involves calcium-induced calcium release from intracellular stores.
cultured rat cortical astrocyte. J. Neurosci. Res. 79, 535–544. Proc. Natl. Acad. Sci. USA 98, 4148–4153.
Oikonomou, G., Shaham, S., 2011. The glia of Caenorhabditis elegans. Glia 59, 1253– Shemarova, I.V., Nesterov, V.P., 2005a. Evolution of Ca2+ signaling mechanisms. Role
1263. of calcium ions in signal transduction in lower eukaryotes. Zh. Evol. Biokhim.
Oldershaw, K.A., Taylor, C.W., 1993. Luminal Ca2+ increases the affinity of inositol Fiziol. 41, 303–313.
1,4,5-trisphosphate for its receptor. Biochem. J. 292 (Pt. 3), 631–633. Shemarova, I.V., Nesterov, V.P., 2005b. Evolution of mechanisms of calcium
Paluzzi, S., Alloisio, S., Zappettini, S., Milanese, M., Raiteri, L., Nobile, M., Bonanno, G., signaling: the role of calcium ions in signal transduction in prokaryotes. Zh.
2007. Adult astroglia is competent for Na+/Ca2+ exchanger-operated exocytotic Evol. Biokhim. Fiziol. 41, 12–17.
glutamate release triggered by mild depolarization. J. Neurochem. 103, 1196– Sheppard, C.A., Simpson, P.B., Sharp, A.H., Nucifora, F.C., Ross, C.A., Lange, G.D.,
1207. Russell, J.T., 1997. Comparison of type 2 inositol 1,4,5-trisphosphate receptor
Palygin, O., Lalo, U., Pankratov, Y., 2011. Distinct pharmacological and functional distribution and subcellular Ca2+ release sites that support Ca2+ waves in
properties of NMDA receptors in mouse cortical astrocytes. Br. J. Pharmacol. cultured astrocytes. J. Neurochem. 68, 2317–2327.
163, 1755–1766. Shigetomi, E., Bowser, D.N., Sofroniew, M.V., Khakh, B.S., 2008. Two forms of
Palygin, O., Lalo, U., Verkhratsky, A., Pankratov, Y., 2010. Ionotropic NMDA and P2X1/ astrocyte calcium excitability have distinct effects on NMDA receptor-mediated
2+
5 receptors mediate synaptically induced Ca signalling in cortical astrocytes. slow inward currents in pyramidal neurons. J. Neurosci. 28, 6659–6663.
Cell Calcium 48, 225–231. Shimizu, H., Watanabe, E., Hiyama, T.Y., Nagakura, A., Fujikawa, A., Okado, H.,
Parpura, V., Basarsky, T.A., Liu, F., Jeftinija, K., Jeftinija, S., Haydon, P.G., 1994. Yanagawa, Y., Obata, K., Noda, M., 2007. Glial Nax channels control lactate
Glutamate-mediated astrocyte–neuron signalling. Nature 369, 744–747. signaling to neurons for brain [Na+] sensing. Neuron 54, 59–72.
Parpura, V., Grubisic, V., Verkhratsky, A., 2011. Ca2+ sources for the exocytotic Shmigol, A., Svichar, N., Kostyuk, P., Verkhratsky, A., 1996. Gradual caffeine-induced
release of glutamate from astrocytes. Biochim. Biophys. Acta 1813, 984–991. Ca2+ release in mouse dorsal root ganglion neurons is controlled by cytoplasmic
Parri, H.R., Crunelli, V., 2003. The role of Ca2+ in the generation of spontaneous and luminal Ca2+. Neuroscience 73, 1061–1067.
astrocytic Ca2+ oscillations. Neuroscience 120, 979–992. Simpson, P.B., Russell, J.T., 1998. Role of mitochondrial Ca2+ regulation in neuronal
Parri, H.R., Gould, T.M., Crunelli, V., 2001. Spontaneous astrocytic Ca2+ oscillations and glial cell signalling. Brain Res. Brain Res. Rev. 26, 72–81.
in situ drive NMDAR-mediated neuronal excitation. Nat. Neurosci. 4, 803–812. Smyth, J.T., Dehaven, W.I., Jones, B.F., Mercer, J.C., Trebak, M., Vazquez, G., Putney Jr.,
Perea, G., Araque, A., 2005. Properties of synaptically evoked astrocyte calcium J.W., 2006. Emerging perspectives in store-operated Ca2+ entry: roles of Orai,
signal reveal synaptic information processing by astrocytes. J. Neurosci. 25, Stim and TRP. Biochim. Biophys. Acta 1763, 1147–1160.
2192–2203. Solovyova, N., Verkhratsky, A., 2002. Monitoring of free calcium in the neuronal
Petersen, O.H., 2002. Calcium signal compartmentalization. Biol. Res. 35, 177–182. endoplasmic reticulum: an overview of modern approaches. J. Neurosci.
Petersen, O.H., 2005. Ca2+ signalling and Ca2+-activated ion channels in exocrine Methods 122, 1–12.
acinar cells. Cell Calcium 38, 171–200. Solovyova, N., Verkhratsky, A., 2003. Neuronal endoplasmic reticulum acts as a
Petersen, O.H., Michalak, M., Verkhratsky, A., 2005. Calcium signalling: past, present single functional Ca2+ store shared by ryanodine and inositol-1,4,5-
and future. Cell Calcium 38, 161–169. trisphosphate receptors as revealed by intra-ER [Ca2+] recordings in single rat
Petersen, O.H., Petersen, C.C., Kasai, H., 1994. Calcium and hormone action. Annu. sensory neurones. Pflugers Arch. 446, 447–454.
Rev. Physiol. 56, 297–319. Solovyova, N., Veselovsky, N., Toescu, E.C., Verkhratsky, A., 2002. Ca2+ dynamics in
Petersen, O.H., Verkhratsky, A., 2007. Endoplasmic reticulum calcium tunnels the lumen of the endoplasmic reticulum in sensory neurons: direct
integrate signalling in polarised cells. Cell Calcium 42, 373–378. visualization of Ca2+-induced Ca2+ release triggered by physiological Ca2+
Petravicz, J., Fiacco, T.A., McCarthy, K.D., 2008. Loss of IP3 receptor-dependent Ca2+ entry. EMBO J. 21, 622–630.
increases in hippocampal astrocytes does not affect baseline CA1 pyramidal Spitzer, N.C., Borodinsky, L.N., Root, C.M., 2005. Homeostatic activity-dependent
neuron synaptic activity. J. Neurosci. 28, 4967–4973. paradigm for neurotransmitter specification. Cell Calcium 37, 417–423.
Pivneva, T., Haas, B., Reyes-Haro, D., Laube, G., Veh, R.W., Nolte, C., Skibo, G., Stout Jr., R.F., Parpura, V., 2011. Voltage-gated calcium channel types in cultured C.
Kettenmann, H., 2008. Store-operated Ca2+ entry in astrocytes: different spatial elegans CEPsh glial cells. Cell Calcium 50, 98–108.
arrangement of endoplasmic reticulum explains functional diversity in vitro Strubing, C., Krapivinsky, G., Krapivinsky, L., Clapham, D.E., 2001. TRPC1 and TRPC5
and in situ. Cell Calcium 43, 591–601. form a novel cation channel in mammalian brain. Neuron 29, 645–655.
Pizzo, P., Burgo, A., Pozzan, T., Fasolato, C., 2001. Role of capacitative calcium entry Strubing, C., Krapivinsky, G., Krapivinsky, L., Clapham, D.E., 2003. Formation of novel
on glutamate-induced calcium influx in type-I rat cortical astrocytes. J. TRPC channels by complex subunit interactions in embryonic brain. J. Biol.
Neurochem. 79, 98–109. Chem. 278, 39014–39019.
Porter, J.T., McCarthy, K.D., 1995. Adenosine receptors modulate [Ca2+]i in Takuma, K., Matsuda, T., Hashimoto, H., Asano, S., Baba, A., 1994. Cultured rat
hippocampal astrocytes in situ. J. Neurochem. 65, 1515–1523. astrocytes possess Na+–Ca2+ exchanger. Glia 12, 336–342.
Procko, C., Shaham, S., 2010. Assisted morphogenesis: glial control of dendrite Toescu, E.C., 2000. Mitochondria and Ca2+ signaling. J. Cell. Mol. Med. 4, 164–175.
shapes. Curr. Opin. Cell Biol. 22, 560–565. Toescu, E.C., Moller, T., Kettenmann, H., Verkhratsky, A., 1998. Long-term activation
Puceat, M., Jaconi, M., 2005. Ca2+ signalling in cardiogenesis. Cell Calcium 38, 383– of capacitative Ca2+ entry in mouse microglial cells. Neuroscience 86, 925–935.
389. Toescu, E.C., Verkhratsky, A., 1998. Principles of calcium signalling. In: Verkhratsky,
Putney Jr., J.W., 1986. A model for receptor-regulated calcium entry. Cell Calcium 7, A., Toescu, E.C. (Eds.), Integrative Aspects of Calcium Signalling. Plenum Press,
1–12. New York, pp. 2–22.
Putney Jr., J.W., 1990. Capacitative calcium entry revisited. Cell Calcium 11, 611– Toescu, E.C., Verkhratsky, A., 2004. Ca2+ and mitochondria as substrates for deficits
624. in synaptic plasticity in normal brain ageing. J. Cell. Mol. Med. 8, 181–190.
Putney Jr., J.W., 2007. Recent breakthroughs in the molecular mechanism of Tuschick, S., Kirischuk, S., Kirchhoff, F., Liefeldt, L., Paul, M., Verkhratsky, A.,
capacitative calcium entry (with thoughts on how we got here). Cell Calcium 42, Kettenmann, H., 1997. Bergmann glial cells in situ express endothelin B
103–110. receptors linked to cytoplasmic calcium signals. Cell Calcium 21, 409–419.
Redmond, L., Ghosh, A., 2005. Regulation of dendritic development by calcium Vangheluwe, P., Raeymaekers, L., Dode, L., Wuytack, F., 2005. Modulating
signaling. Cell Calcium 37, 411–416. sarco(endo)plasmic reticulum Ca2+ ATPase 2 (SERCA2) activity: cell biological
Reichenbach, A., Pannicke, T., 2008. Neuroscience. A new glance at glia. Science 322, implications. Cell Calcium 38, 291–302.
693–694. Verkhratsky, A., 2005. Physiology and pathophysiology of the calcium store in the
Reyes, R.C., Parpura, V., 2008. Mitochondria modulate Ca2+-dependent glutamate endoplasmic reticulum of neurons. Physiol. Rev. 85, 201–279.
release from rat cortical astrocytes. J. Neurosci. 28, 9682–9691. Verkhratsky, A., 2006a. Calcium ions and integration in neural circuits. Acta Physiol.
Reyes, R.C., Perry, G., Lesort, M., Parpura, V., 2011. Immunophilin deficiency (Oxf.) 187, 357–369.
augments Ca2+-dependent glutamate release from mouse cortical astrocytes. Verkhratsky, A., 2006b. Patching the glia reveals the functional organisation of the
Cell Calcium 49, 23–34. brain. Pflugers Arch. 453, 411–420.
Rimessi, A., Giorgi, C., Pinton, P., Rizzuto, R., 2008. The versatility of mitochondrial Verkhratsky, A., 2007. Calcium and cell death. Subcell. Biochem. 45, 465–480.
calcium signals: from stimulation of cell metabolism to induction of cell death. Verkhratsky, A., Butt, A., 2007. Glial Neurobiology. A Textbook. John Wiley & Sons,
Biochim. Biophys. Acta 1777, 808–816. Chichester.
56 A. Verkhratsky et al. / Molecular and Cellular Endocrinology 353 (2012) 45–56

Verkhratsky, A., Kettenmann, H., 1996. Calcium signalling in glial cells. Trends Webb, S.E., Moreau, M., Leclerc, C., Miller, A.L., 2005. Calcium transients and neural
Neurosci. 19, 346–352. induction in vertebrates. Cell Calcium 37, 375–385.
Verkhratsky, A., Kirchhoff, F., 2007. NMDA receptors in glia. Neuroscientist 13, 28– Wray, S., Burdyga, T., Noble, K., 2005. Calcium signalling in smooth muscle. Cell
37. Calcium 38, 397–407.
Verkhratsky, A., Orkand, R.K., Kettenmann, H., 1998. Glial calcium: homeostasis and Xi, Q., Tcheranova, D., Basuroy, S., Parfenova, H., Jaggar, J.H., Leffler, C.W., 2011.
signaling function. Physiol. Rev. 78, 99–141. Glutamate-induced calcium signals stimulate CO production in piglet
Verkhratsky, A., Parpura, V., Rodriguez, J.J., 2011. Where the thoughts dwell: the astrocytes. Am. J. Physiol. Heart Circ. Physiol. 301, H428–433.
physiology of neuronal–glial ‘‘diffuse neural net’’. Brain Res. Rev. 66, 133–151. Xu, J.H., Long, L., Tang, Y.C., Hu, H.T., Tang, F.R., 2007. Cav1.2, Cav1.3, and Cav2.1 in
Verkhratsky, A., Petersen, O.H., 2002. The endoplasmic reticulum as an integrating the mouse hippocampus during and after pilocarpine-induced status
signalling organelle: from neuronal signalling to neuronal death. Eur. J. epilepticus. Hippocampus 17, 235–251.
Pharmacol. 447, 141–154. Young, S.Z., Platel, J.C., Nielsen, J.V., Jensen, N.A., Bordey, A., 2010. GABA(A) increases
Verkhratsky, A., Shmigol, A., 1996. Calcium-induced calcium release in neurones. calcium in subventricular zone astrocyte-like cells through L- and T-type
Cell Calcium 19, 1–14. voltage-gated calcium channels. Front. Cell. Neurosci. 4, 8.
Verkhratsky, A., Solovyova, N., Toescu, E.C., 2002. Calcium excitability of glial cells. Zhu, M.X., Ma, J., Parrington, J., Galione, A., Evans, A.M., 2010. TPCs: endolysosomal
In: Volterra, A., Haydon, P., Magistretti, P. (Eds.), Glia in Synaptic Transmission. channels for Ca2+ mobilization from acidic organelles triggered by NAADP. FEBS
OUP, Oxford, pp. 99–109. Lett. 584, 1966–1974.
Verkhratsky, A., Steinhauser, C., 2000. Ion channels in glial cells. Brain Res. Brain Zonta, M., Angulo, M.C., Gobbo, S., Rosengarten, B., Hossmann, K.A., Pozzan, T.,
Res. Rev. 32, 380–412. Carmignoto, G., 2003. Neuron-to-astrocyte signaling is central to the dynamic
Verkhratsky, A., Toescu, E.C., 2006. Neuronal–glial networks as substrate for CNS control of brain microcirculation. Nat. Neurosci. 6, 43–50.
integration. J. Cell. Mol. Med. 10, 826–836.
Wakui, M., Osipchuk, Y.V., Petersen, O.H., 1990. Receptor-activated cytoplasmic Ca2+
spiking mediated by inositol trisphosphate is due to Ca2+-induced Ca2+ release.
Cell 63, 1025–1032.

Vous aimerez peut-être aussi