ArticlePDF Available

Vesicular Glutamate Transporter-Dependent Glutamate Release from Astrocytes

Authors:

Abstract and Figures

Astrocytes exhibit excitability based on variations of their intracellular Ca2+ concentrations, which leads to glutamate release, that in turn can signal to adjacent neurons. This glutamate-mediated astrocyte-neuron signaling occurs at physiological intracellular Ca2+ levels in astrocytes and includes modulation of synaptic transmission. The mechanism underlying Ca2+-dependent glutamate release from astrocytes is most likely exocytosis, because astrocytes express the protein components of the soluble N-ethyl maleimide-sensitive fusion protein attachment protein receptors complex, including synaptobrevin 2, syntaxin, and synaptosome-associated protein of 23 kDa. Although these proteins mediate Ca2+-dependent glutamate release from astrocytes, it is not well understood whether astrocytes express functional vesicular glutamate transporters (VGLUTs) that are critical for vesicle refilling. Here, we find in cultured and freshly isolated astrocytes the presence of brain-specific Na+-dependent inorganic phosphate cotransporter and differentiation-associated Na+-dependent inorganic phosphate cotransporter that have recently been identified as VGLUTs 1 and 2. Indirect immunocytochemistry showed a punctate pattern of VGLUT immunoreactivity throughout the entire cell body and processes, whereas pharmacological inhibition of VGLUTs abolished mechanically and agonist-evoked Ca2+-dependent glutamate release from astrocytes. Taken together, these data indicate that VGLUTs play a functional role in exocytotic glutamate release from astrocytes.
VGLUTs mediate exocytotic glutamate release from cultured astrocytes. Mechanical stimulation evokes glutamate release ( A–D) caused by the internal Ca 2 elevations in astrocytes ( E–H). Glutamate release from astrocyte is shown (raw data) in A (rest) and B (after stimulation). The pseudocolor scale is a linear representation of the fluorescence intensities ranging from 1240 to 1350. C, Time lapse of NADH fluorescence, reporting on glutamate release. Mechanical stimulation caused glutamate release (black squares) that was greatly reduced when cells were preincubated with Rose Bengal (open circles; 0.5 M; 30 min), an allosteric site modulator of VGLUTs. D, Peak values of mechanically induced glutamate release. Incubation of astrocytes with BAPTA-AM (50 M; 30 min), a membrane-permeable Ca 2 chelator, reduced the mechanically induced glutamate release. Pretreatment of astrocytes with a holoprotein of tetanus toxin (TeTx; 15 g/ml; 24 – 48 hr), which cleaves synaptobrevin 2, or bafilomycin A 1 (5 M, 1 hr), a specific inhibitor of V-ATPase, reduces mechanically induced glutamate release consistent with exocytotic mechanism underlying Ca 2 -dependent glutamate release from astrocytes. Changes in NADH fluorescence are shown as dF/Fo (percentage) after background subtraction and correction for bleaching. E–H, In experiments parallel to those in A–D, astrocytes were mechanically stimulated while measuring intracellular Ca 2 levels using fluo-3. This stimulation reliably increases the intracellular Ca 2 levels ( F) from its resting level ( E) (raw data). The pseudocolor scale is a linear representation of the fluorescence intensities ranging from 0 to 255. G, Time lapse of fluo-3 fluorescence, reporting on Ca 2 levels in astrocytes. Mechanical stimulation caused an increase in astrocytic intracellular Ca 2 levels (black squares) that were unaffected when cells were preincubated with Rose Bengal (open circles; 0.5 M; 30 min). H, Peak values of mechanically induced Ca 2 elevations are greatly reduced when cells are preincubated with BAPTA but not when treated with TeTx, bafilomycin A 1 , or Rose Bengal. Changes in fluo-3 fluorescence are expressed as dF/Fo (percentage) after background subtraction. Arrows (C, G) indicate the time when the pipette–astrocyte contact occurred. Points and bars represent the means and SEMs. In C and G, SEMs are shown in single directions for clarity. Asterisks indicate a significant change of measurements compared with the control group (one-way ANOVA, followed by post hoc Fisher's LSD test; *p 0.05; **p 0.01). We found no significant difference between various treatments in D, whereas in H, preincubation with BAPTA, in addition to a significant change in measurement compared with the control group, showed a statistically significant difference when compared with all other treatments ( p 0.05).
… 
Content may be subject to copyright.
Cellular/Molecular
Vesicular Glutamate Transporter-Dependent Glutamate
Release from Astrocytes
Vedrana Montana,
1
Yingchun Ni,
1
Vice Sunjara,
2
Xue Hua,
1
and Vladimir Parpura
1
1
Department of Cell Biology and Neuroscience and Center for Nanoscale Science and Engineering,
2
International Scholars Program, University of
California, Riverside, California 92521
Astrocytes exhibit excitability based on variations of their intracellular Ca
2
concentrations, which leads to glutamate release, that in
turn can signal to adjacent neurons. This glutamate-mediated astrocyte–neuron signaling occurs at physiological intracellular Ca
2
levels in astrocytes and includes modulation of synaptic transmission. The mechanism underlying Ca
2
-dependent glutamate release
from astrocytes is most likely exocytosis, because astrocytes express the protein components of the soluble N-ethyl maleimide-sensitive
fusion protein attachment protein receptors complex, including synaptobrevin 2, syntaxin, and synaptosome-associated protein of 23
kDa. Although these proteins mediate Ca
2
-dependent glutamate release from astrocytes, it is not well understood whether astrocytes
express functional vesicular glutamate transporters (VGLUTs) that are critical for vesicle refilling. Here, we find in cultured and freshly
isolated astrocytes the presence of brain-specific Na
-dependent inorganic phosphate cotransporter and differentiation-associated
Na
-dependent inorganic phosphate cotransporter that have recently been identified as VGLUTs 1 and 2. Indirect immunocytochem-
istry showed a punctate pattern of VGLUT immunoreactivity throughout the entire cell body and processes, whereas pharmacological
inhibition of VGLUTs abolished mechanically and agonist-evoked Ca
2
-dependent glutamate release from astrocytes. Taken together,
these data indicate that VGLUTs play a functional role in exocytotic glutamate release from astrocytes.
Key words: astrocytes; vesicular glutamate transporters; SNAREs; V-ATPase; calcium-dependent glutamate release; exocytosis; signaling;
synapse
Introduction
The ultrastructure of the CNS suggests that astrocytes might reg-
ulate synaptic neurotransmission. Astrocytes enwrap nerve ter-
minals (Peters et al., 1991), which makes them perfectly posi-
tioned to exchange information with synapses. Indeed, it has
been demonstrated that astrocytes can respond to synaptic acti-
vation (Dani et al., 1992) and that they can modulate synaptic
neurotransmission by releasing glutamate in a Ca
2
-dependent
manner (Araque et al., 1998a; Kang et al., 1998). Because astro-
cytic internal Ca
2
levels necessary for glutamate release are
within the physiological range (Parpura and Haydon, 2000), this
release can be used as a signaling pathway that may influence
synaptic neurotransmission and plasticity within the CNS.
Although the mechanism of Ca
2
-dependent release of glu-
tamate from astrocytes is not fully defined, the protein compo-
nents of the neuronal exocytotic machinery are expressed in as-
trocytes (Parpura et al., 1995b; Jeftinija et al., 1997; Bezzi et al.,
1998; Hepp et al., 1999; Maienschein et al., 1999; Araque et al.,
2000; Pasti et al., 2001). Additionally, Clostridial toxins, which
cleave some of these exocytosis-related proteins, inhibit Ca
2
-
dependent release of glutamate from astrocytes (Jeftinija et al.,
1997; Bezzi et al., 1998; Araque et al., 2000), indicating that reg-
ulated exocytosis most likely mediates glutamate release from
astrocytes.
Immunoelectron microscopic study indicated that exocytotic
proteins in astrocytes can be associated with electronlucent or
dense-core vesicular structures, the diameters of which are less
uniform than those reported in neurons (Maienschein et al.,
1999). The storage of glutamate in synaptic vesicles requires the
presence of V-type H
-ATPase (V-ATPase) and vesicular gluta-
mate transporters (VGLUTs). Hence, astrocytic Ca
2
-
dependent glutamate release can be blocked with bafilomycin A
1
(Araque et al., 2000; Bezzi et al., 2001; Pasti et al., 2001), which
specifically interferes with V-ATPase, leading to alkalinization of
vesicular lumen and collapsing the proton gradient necessary for
VGLUTs to transport glutamate into vesicles. Brain tissue ex-
presses VGLUT 1 and 2 isoforms (Ni et al., 1994, 1995; Hisano et
al., 2000; Bai et al., 2001) in glutamatergic neurons (Bellocchio et
Received Aug. 12, 2003; revised Jan. 30, 2004; accepted Jan. 30, 2004.
This work was supported by a grant from the Department of Defense/Defense Advanced Research Planning
Agency/Defense Microelectronics Activity under Award DMEA90-02-2-0216 and the Whitehall Foundation (Award
2000-05-17). V.P. is an Institute for Complex Adaptive Matter Senior Fellow. We thank Dr. Karl Bauer (Max-Planck-
Institute for Experimental Endocrinology, Hannover, Germany) for kindly providing
-Ala-Lys-N
-AMCA peptide,
Dr. James E. Rothman (Memorial Sloan-Kettering Cancer Center, New York, NY) for generously providing a plasmid
encoding for super-ecliptic synapto-pHluorin, and Dr. Robert H. Edwards (University of California San Francisco, San
Francisco, CA) for graciously providing polyclonal antibodies against VGLUTs 1 and 3 and plasmids encoding for
GST-VGLUTs 1 and 3 fusion proteins. We are grateful for Dr. Glenn I. Hatton’s arrangements regarding supply of
“Bauer’s” peptide and for providing an Alexa Fluor 488 goat anti-mouse antibody. We also thank Dr. Hatton and
Todd A. Ponzio for comments on previous versions of this manuscript, for engaging discussions related to this work,
and help with brain dissection of adult rats.
Correspondenceshould beaddressed toDr. VladimirParpura, Departmentof CellBiology andNeuroscience,1208
Spieth Hall, University of California, Riverside, CA 92521. E-mail: vlad@citrus.ucr.edu.
V. Sunjara’s present address: School of Medicine, University of Zagreb, 10000 Zagreb, Croatia.
DOI:10.1523/JNEUROSCI.3770-03.2004
Copyright © 2004 Society for Neuroscience 0270-6474/04/232633-10$15.00/0
The Journal of Neuroscience, March 17, 2004 24(11):2633–2642 2633
al., 1998; Fremeau et al., 2001; Fujiyama et al., 2001; Herzog et al.,
2001; Sakata-Haga et al., 2001; Kaneko et al., 2002; Varoqui et al.,
2002). Recently, there have been reports indicating the presence
of a VGLUT 3 isoform in subpopulations of GABAergic
(Fremeau et al., 2002), cholinergic, and monoaminergic neurons
(Fremeau et al., 2002; Gras et al., 2002; Schafer et al., 2002) and
some astrocytes (Fremeau et al., 2002). However, whether astro-
cytes can express the VGLUT 1 and 2 isoforms, which account for
the release of glutamate by all known excitatory neurons, is not
yet known.
In this study, we found the presence of VGLUTs 1 and 2 in
cultured and freshly isolated astrocytes from rat visual cortices,
exhibiting a subcellular localization pattern consistent with their
vesicular association. Because pharmacological inhibition of
VGLUTs greatly reduced Ca
2
-dependent exocytotic release of
glutamate from astrocytes, these proteins can play a functional
role in astrocytic glutamate release in the CNS.
Some of these data appeared in preliminary form (Ni et al.,
2003).
Materials and Methods
Cell cultures. We prepared enriched astrocytic cultures using a modifica-
tion (Parpura et al., 1995a) of the originally described shaking procedure
(McCarthy and deVellis, 1980). Visual cortices isolated from 0- to 2-d-
old Sprague Dawley rats were treated enzymatically (papain, 20 IU/ml; 1
hr at 36.8°C). After subsequent treatment with trypsin inhibitor (10 mg/
ml; type II-O; 5 min at room temperature) to terminate the enzymatic
reaction, tissue was dispersed mechanically by triturating through a glass
pipette. Cells were initially plated into tissue culture flasks (25 cm
2
) and
maintained at 36.8°C in a humidified 5% CO
2
/95% air atmosphere in a
complete culture medium that consisted of
-MEM (without phenol
red; Invitrogen, Carlsbad, CA) supplemented with 10% heat-inactivated
FBS (HyClone, Logan, UT), L-glutamine (2 mM; Invitrogen), D-glucose
(20 mM; Sigma-Aldrich, St. Louis, MO), sodium pyruvate (1 mM; Invitro-
gen), penicillin (100 IU/ml), streptomycin (100
g/ml), and sodium
bicarbonate (14 mM; Invitrogen), pH 7.4.
After 6 –24 d in culture, the cells were shaken twice (260 rpm at
36.8°C), first for 1.5–2 hr and then, after exchange of complete medium,
again for 18 –20 hr. At that time, the remaining attached cells were de-
tached from flasks using trypsin [10,000 N
-benzoyl-L-arginine ethyl
ester hydrochloride (BAEE) units/ml; Sigma-Aldrich] and replated onto
12 mm round glass coverslips precoated with polyethyleneimine (PEI; 1
mg/ml; Sigma-Aldrich). Resulting purified astrocytes were kept in cul-
ture for 1–10 d (8 –29 d after initial plating) until used in experiments.
The purity (99%) of astrocytic culture was confirmed by anti-GFAP
antibody and indirect immunocytochemistry. In some experiments be-
fore GFAP immunocytochemisty, cells were incubated with a dipeptide
-Ala-Lys conjugated to 7-amino-4methylcoumarin-3-acetic acid
(AMCA), kindly provided by Dr. Karl Bauer (Max-Planck-Institute for
Experimental Endocrinology, Hannover, Germany). This peptide selec-
tively accumulates in astrocytes after its uptake is mediated by the PepT2
peptide transporter (Dieck et al., 1999). Incubation with
-Ala-Lys-N
-
AMCA (20
Mat 36.8°C for 2 hr) labels astrocytes, because this peptide
accumulates only within GFAP-positive cells (n68 of 68 tested).
Freshly isolated cells. Cells were freshly isolated from visual cortices of
1-, 2-, 8-, and 55-d-old Sprague Dawley rats using a modification of a
previously described procedure (Zhou and Kimelberg, 2000). Briefly,
visual cortices isolated from rats were subjected to papain treatment,
followed by a trituration as described for cell cultures. After trituration,
cells were applied onto PEI-coated coverslips and allowed to adhere while
they were incubated with
-Ala-Lys-N
-AMCA (20
Mat 36.8°C for 1
hr). After washing, cells were fixed for immunocytochemisty.
Immunocytochemistry and nuclear staining. For confirmation of astro-
cytic culture purity, cells were exposed to Dent’s fixative at room tem-
perature for 30 min (Parpura and Haydon, 2000). A monoclonal anti-
body (catalog #69110; 1:500 dilution; 1 hr at room temperature or
overnight at 4°C; ICN Biomedicals, Aurora, OH), followed by a
rhodamine-conjugated secondary antibody (1 hr at room temperature),
was used to probe for GFAP. In some experiments, we subsequently
colabeled astrocytic nuclei using 4,6-diamidino-2-phenylindole dilac-
tate (DAPI dilactate; 3
M; Molecular Probes, Eugene, OR) for 5 min at
room temperature.
Subcellular localization of VGLUTs 1, 2, and 3, synaptobrevin 2, and
synaptosome-associated protein of 23 kDa (SNAP-23) was determined
by indirect immunocytochemistry, in which cells were incubated with
primary antibodies overnight at 4°C. A monoclonal antibody against
synaptobrevin 2 (catalog #104201; 1:250 dilution; Synaptic Systems,
Goettingen, Germany), a polyclonal antibody against SNAP-23 (catalog
#111202; 1:50; Synaptic Systems), and seven different antibodies against
VGLUTs (Table 1) were used. After washout of the primary antibody,
TRITC (rhodamine)-conjugated secondary antibodies were applied, and
preparation was incubated for 1 hr at room temperature. In double-
labeling experiments, an Alexa Fluor 488-conjugated secondary antibody
(Molecular Probes) was used for visualization of synaptobrevin 2 immu-
noreactivity. In all experiments, we performed controls in which primary
antibodies were omitted to test for nonspecific binding of secondary
antibodies. We classified cells as immunoreactive if the average fluores-
cence intensity for a probed protein, within a cellular region of interest
(at least 40 40 pixels), was at least 2 SDs above the average signals
acquired from control cells. In a subset of experiments, we performed
adsorption controls, in which primary antibodies were preincubated
overnight at 4°C with respective antigens, proteins, or peptides (Table 1),
to test for specificity of primary antibodies (Bellocchio et al., 1998;
Fremeau et al., 2001, 2002; Takamori et al., 2001). All imaging data were
background subtracted using fluorescence emission originating from a
region on the coverslip containing no cells.
Preparation of subcellular fractions and Western blotting. Synaptosomal
Table 1. Antibodies raised against VGLUTs and their respective antigens used in this study
Final dilutions
Antibody Source Type
Catalog number
or reference ICC WB Adsorption antigen
Catalog number
or reference
Final
concentration
(
g/ml)
Anti-VGLUT 1 Synaptic Systems Mouse monoclonal 135001 1:1000 1:1000 GST-rVGLUT1 (aa 523–560) 135– 0P 5
Synaptic Systems Rabbit polyclonal 135002 1:500 –1:1000 1:1000–1:2000 GST-rVGLUT1 (aa 523–560) 135– 0P 2.5–5
Dr. R. H. Edwards Rabbit polyclonal
a
Bellocchio et al.
(1998)
1:500 1:1000 GST-rVGLUT1 (aa 493–560) Bellocchio et al.
(1998)
10.5
Anti-VGLUT 2 Synaptic Systems Rabbit polyclonal
a
135102 1:500 1:1000 GST-rVGLUT2 (aa 510 –582) 135–1P 2–5
Chemicon Guinea pig polyclonal AB5907 1:2000 1:2000 rVGLUT2 (aa 565–582), Sp AG209 2.5
Anti-VGLUT 3 Chemicon Guinea pig polyclonal AB5421 1:5000 1:5000 rVGLUT3 (aa 569 –588), Sp
b
AG320 1–5
Dr. R. H. Edwards Rabbit polyclonal Fremeau et al.
(2002)
1:500 1:1000 GST-rVGLUT3 (aa 530 –588) Fremeau et al.
(2002)
11.5
ICC, Immunocytochemistry; WB, Western blots; GST, glutathione S-tranferase; r, rat; Sp, synthetic peptide. Parentheses indicate the portion of VGLUTs, based on rat amino acid sequences.
a
No cross-reactivity to VGLUT 3.
b
Used in cross-reactivity experiments.
2634 J. Neurosci., March 17, 2004 24(11):2633–2642 Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes
membrane-enriched [lysate pellet (LP-1)] preparations were done ac-
cording to the standard procedures (Huttner et al., 1983). We obtained
non-nuclear membranes and vesicular extracts from purified astrocytes
[astrocytic preparation (AsP)] as described previously (Parpura et al.,
1995b). LP-1 and AsP were subjected to 15% SDS-PAGE, followed by
transfer to nitrocellulose membranes that were probed with VGLUT
antibodies used in immunocytochemistry, but at different dilutions (Ta-
ble 1), and antibodies against synaptobrevin 2 and SNAP-23, which we
used at 1:1000 and 1:400, respectively. Additionally, we used monoclonal
antibodies against SNAP-25 (catalog #111001; 1:500; Synaptic Systems)
and syntaxin 1 (catalog #S0664; 1:1000; Sigma-Aldrich). Immunoreac-
tivity of bands was detected using ECL (Amersham Biosciences, Piscat-
away, NJ). In a subset of experiments, we performed adsorption controls
for primary antibodies against VGLUTs (Table 1) as described for im-
munocytochemistry (Bellocchio et al., 1998; Fremeau et al., 2001, 2002;
Takamori et al., 2001). In experiments testing for cross-reactivity of an-
tibodies raised against VGLUTs 1 and 2 to their heterologous VGLUT 3
antigen, we preincubated different VGLUT antibodies with VGLUT 3
antigen.
Reverse transcription-PCR. Total RNA was extracted from purified as-
trocytic culture and cortical tissue of postnatal Sprague Dawley rats (0
2-d-old) using TRIzol Reagent (Invitrogen) and protocols provided by
the manufacturer. Five micrograms of total RNA were used for reverse tran-
scription using Oligo(dT)
1218
and superscript II reverse transcriptase (In-
vitrogen). Several pairs of primers were used to amplify cDNA using PCR
(each pair, 35 cycles). Primers for SNAP-25A (GenBank accession number
L19760) amplification were 5-CTGGAAAGCACCCGTCGTATG-3and
5-GCACGTTGGTTGGCTTCATCA-3, whereas for
-actin the primers
were 5-TCATGAAGTGTGACGTTGACATCCGT-3and 5-CCTAGAA-
GCATTTGCGGTGCACGATG-3(catalog #G5740; Promega, Madison,
WI). In some experiments, we used two rounds of amplification for SNAP-
25A (GenBank accession number NM_030991). In the first round, we used
5-ATGGCCGAGGACGCAGACA-3and 5-ACCACTTCCCAGCA-
TCTTTGT-3(618 bp product). Amplified DNA was used as the template
for the second round of PCR using nested primers 5-CTGGAAAGCACCC-
GTCGCATG-3and 5-GCACGTTGGTTGGCTTCATCA-3(521 bp
product). RT-PCR reaction in two subsequent rounds was also performed
for VGLUT 2 (GenBank accession number NM_053427) using 5-
AGCAAGGTTGGCATGTTGTCTG-3and 5-CGGTCCTTATAGGAGT-
ACGCGT-3(698 bp product), followed by amplification of the product
using nested primers 5-TGGTGCAATGACGAAGAACAAG-3and 5-
TCC TTTTTCTCCCAGCCGTT-3(294 bp product). Primers for VGLUT
1 (GenBank accession number NM_053859) amplification were 5-
GAGAAACAGCCGTGGGCAGAG-3and 5-TCAGTAGTCCCGGACA-
GGGGGTGG-3(207 bp product), whereas for VGLUT 3 (GenBank acces-
sion number NM_153725) the primers were 5-ACCCGGGAAGAATGG-
C A GAATGTG-3and 5-ATGGGAAAAGCAATGGGTGTGGAG-3(399
bp product).
Calcium measurements. We monitored astrocytic intracellular Ca
2
levels using a Ca
2
indicator, fluo-3 (Parpura et al., 1994). Cells were
loaded in a complete culturing medium containing an acetoxymethyl
(AM) ester derivative of fluo-3 (10
g/ml; Molecular Probes) and plu-
ronic acid (0.025% w/v; Molecular Probes), at 36.8°C for 30 min. After
washing in normal external solution, de-esterification of the dye was
permitted for 30 min at room temperature. The normal external solution
contained (in mM) 140 NaCl, 5 KCl, 2 CaCl
2
, 2 MgCl
2
, 5 glucose, and 10
HEPES, pH 7.4. Coverslips containing fluo-3-loaded cells were mounted
into a recording chamber and imaged. All data were background sub-
tracted and expressed as dF/Fo (percentage), where Fo represents the
fluorescent level before cell stimulation, and dF represents the change in
fluorescence.
Glutamate measurements. We optically monitored extracellular gluta-
mate levels using an L-glutamate dehydrogenase (GDH; Sigma-Aldrich)-
linked assay (Bezzi et al., 1998; Innocenti et al., 2000). GDH generates
NADH from NAD
(
-nicotinamide adenine dinucleotide; Sigma-
Aldrich) in the presence of glutamate. Provided that GDH and NAD
are added to the solution in which astrocytes are bathed, glutamate re-
leased in the extracellular space can be detected as an increase in NADH
fluorescence. Astrocytes were bathed in an enzymatic assay solution con-
taining normal external solution supplemented with NAD
(1 mM) and
GDH (53 IU/ml; pH 7.4). Every experiment was preceded by a sham
run on cells bathed in solution lacking GDH and NAD
, which was used
to correct for photobleaching and background subtraction. The reduc-
tion of fluorescence because of photobleaching in the sham run exhibited
the same time course as in the matching experimental run. All imaging
data, corrected for photobleaching and background subtracted, were ex-
pressed as dF/Fo (percentage).
Transfection. After the purification of astrocytes, cells in flasks were
transfected with a plasmid encoding for super-ecliptic synapto-pHluorin
(Sankaranarayanan et al., 2000), kindly provided by Dr. James E. Roth-
man (Memorial Sloan-Kettering Cancer Center, New York, NY), each
flask receiving 6
g of plasmid premixed with 12
l of TransIT-293
transfection reagent (catalog #2700; Mirus, Madison, WI), which aids
plasmid entry to cells for 34 hr. At that time, we replaced culturing
medium and returned cells to the culture incubator. Cells were main-
tained at 36.8°C in a humidified 5% CO
2
/95% air atmosphere for 2 d,
when they were replated onto PEI-coated coverslips and kept in culture
until used in experiments.
Imaging acquisition and processing. All experiments were done at room
temperature (20 24°C). We used an inverted microscope (TE 300; Ni-
kon, Melville, NY) equipped with wide-field epifluorescence. Visualiza-
tion of indirect immunocytochemistry of GFAP, VGLUTs, and SNARE
proteins was accomplished using a standard rhodamine/TRITC filter set
(Chroma Technology, Rockingham, VT), except in double-labeling ex-
periments in which immunoreactivity of synaptobrevin 2 was visualized
using a standard fluorescein/FITC filter set (Chroma Technology),
whereas for nuclear staining (DAPI) and
-Ala-Lys-N
-AMCA we used a
standard DAPI filter set (Chroma Technology). For calcium imaging and
synapto-pHluorin, we used a standard fluorescein/FITC filter set. Images
were captured through a 60plan-achromatic oil-immersion objective
[numerical aperture (NA), 1.4; Nikon) using either a CoolSNAP-HQ
cooled CCD camera (Roper Scientific, Tucson, AZ) or an intensified
CCD camera (IC-300; Photon Technology International, Lawrenceville,
NJ) driven by V⫹⫹ imaging software (Digital Optics, Auckland, New
Zealand) or LabView/IMAQ (National Instruments, Austin, TX), re-
spectively. For glutamate imaging experiments, we used a 40SFluor
objective (NA, 1.3; Nikon) and a DAPI filter set (Chroma Technology).
For time-lapse image acquisition, a camera and an electronic shutter
(Vincent Associates, Rochester, NY) inserted in the excitation pathway
were controlled by software. A Xenon arc lamp (100 W) was used as a
light source. In a subset of double-labeling experiments, we used a C1
modular confocal microscope system (Nikon) configured with an in-
Figure 1. Purified astrocytic cultures are immunopositive for GFAP but do not express SNAP-
25. A, GFAP was used to identify astrocytes (red), with nuclei that are counterstained with DAPI
(blue).B,Top, AfterSDS-PAGE (1
gperlane forLP-1 and15
gperlane forAsP), immunoblots
indicate the absence of SNAP-25 in the astrocytic non-nuclear cell membrane extract AsP, while
this protein was detected in synaptosomal membrane-enriched LP-1 preparations. Bottom,
RT-PCR using mRNA isolated from purified astrocytes (As) reveals the presence of
-actin and
lack of SNAP-25 PCR products.
-Actin and SNAP-25 PCR products (285 and 521 bp, respec-
tively) were present when mRNA isolated from rat brain (Br) was used in RT-PCR. L represents a
molecular size marker (100 bp DNA ladder), whereas NC represents a negative control for reac-
tion. Scale bar, 20
m.
Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes J. Neurosci., March 17, 2004 24(11):2633–2642 2635
verted microscope (TE 2000; Nikon) and equipped with a VioFlame Plus
solid state laser (405 nm; for DAPI and AMCA excitation), an argon laser
(488 nm; for Alexa Fluor 488 excitation; Melles Griot, Carlsbad, CA) and
a Spectra Physics green helium neon laser (543 nm; for TRITC excita-
tion). All images shown in the figures represent raw data.
Stimulation of astrocytes. To evoke an increase in the internal Ca
2
concentrations in astrocytes and consequential glutamate release, we ei-
ther bath applied agonists (duration, 55 sec) bradykinin (90 nM) and ATP
(9
M) or mechanically stimulated astrocytes using patch pipettes (Par-
pura et al., 1994; Araque et al., 2000). To control the establishment of the
contact between the pipette and an astrocyte, we monitored pipette re-
sistance during delivery of 20 mV, 10 msec square pulses by a patch-
clamp amplifier (PC-ONE; Dagan, Minneapolis, MN) equipped with a
whole-cell headstage (PC-ONE-30; 1 G). Pipette resistances measured
2.33.3 Mwhen pipette tips were immersed in an external solution,
which increased to 2.74.0 M(721% increase) during transient con-
tacts with astrocytes, lasting 1 sec.
Pharmacological agents. In experiments using a Ca
2
chelator,
BAPTA, cells were loaded in a complete culturing medium containing
BAPTA-AM (50
M; Molecular Probes) and pluronic acid (0.025% w/v)
at 36.8°C for 30 min. After washing in normal external solution, de-
esterification of the chelator was permitted for 30 min at room temper-
ature before use of cells in imaging experiments. Holoprotein of tetanus
toxin (15
g/ml; List Biological Laboratories, Campbell, CA) was applied
to the cells in a complete culturing medium for 24 48 hr at 36.8°C, at
which time cells were rinsed three times with an external solution and
used in imaging experiments. Astrocytes were preincubated with bafilo-
Figure 2. Astrocytes in culture express VGLUTs 1 and 2. A, After SDS-PAGE, immunoblots
indicate the presence of VGLUTs 1, 2, and 3 in astrocytes (AsP) as well as SNARE proteins,
synaptobrevin 2, syntaxin, and SNAP-23. In LP-1 preparations, SNAP-23 was not detected.
When probing SNARE proteins, LP-1 was loaded 1–2
g per lane, whereas AsP was loaded
10 –15
g per lane. However, when probing VGLUTs, LP-1 and AsP were equally loaded at 15
g per lane for VGLUTs 1 and 2 or 30
g per lane for VGLUT 3. B, The immunoreactive bands in
AsPrecognizedby antibodiesraisedagainst VGLUT1,2, and3isoforms (Ag)werecompletely
abolished when antibodies were preincubated with their homologous antigens (Ag). C, An-
tibodies raised against VGLUTs 1 and 2 do not cross-react with VGLUT 3 because their pread-
sorption with this heterologous antigen does not abolish the immunoreactive bands, although
the same VGLUT 3 antigen in preadsorption with anti-VGLUT 3, as seen in B, abolishes its ability
for detection. The arrows in Band Cindicate molecular weight markers. D, RT-PCR using mRNA
isolated from purified astrocytes (As) or from brain (Br) reveals the presence of VGLUT 1, 2, and
3 PCR products (207, 294, and 399 bp, respectively). L and arrows indicate molecular size
markers (100 bp DNA ladder).
Figure 3. Subcellular localization of synaptobrevin 2 ( A), SNAP-23 ( B), VGLUT 1 (C), VGLUT
2(D), and VGLUT 3 ( E). VGLUT immunoreactivity was completely abolished when primary
antibodies (1
o
Ab) were preadsorbed with their respective antigens (Ag) in cultured astrocytes
(C–E,F). Scale bar: A,B,20
m; C–E,C–E,10
m. Fluorescent immunoreactivity is ex-
pressed in intensity units (i.u.). Bars represent means SEMs of measurements from the
number of individual astrocytes (n). Asterisks indicate a significant change of measurements
compared with the control group (1
o
Ab,Ag;one-way ANOVA, followed by post hoc Fish-
er’s LSD test; **p0.01). We found no difference between measurements in preadsorption
controls(1
o
Ab,Ag)when comparedwith controls inwhich primaryantibodieswere omit-
ted (1
o
Ab,Ag).
2636 J. Neurosci., March 17, 2004 24(11):2633–2642 Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes
mycin A
1
(5
M; 30 or 60 min; Sigma-Aldrich) and Rose Bengal (0.1 or
0.5
M; 30 min; Sigma-Aldrich) at room temperature.
Statistical analysis. The effect of agonists and mechanical stimulation
on intracellular Ca
2
levels and consequential glutamate release, as well
as the effects of Rose Bengal on synapto-pHluorin fluorescence, were
assessed using a paired ttest. When testing the time-dependent effects of
bafilomycin A
1
on synapto-pHluorin fluorescence, we used a paired ttest
with Bonferroni adjustment for multiple comparisons. The effects of
pharmacological agents on glutamate and Ca
2
measurements, as well as
the effects of preadsorption of primary antibodies with respective anti-
gens on VGLUT immunoreactivity, were determined using one-way
ANOVA, followed by Fishers least significant difference (LSD) test. Stu-
dentsttest was used for assessing the effects of Rose Bengal on ATP- or
bradykinin-induced Ca
2
increases and glutamate release.
Results
Differential interference contrast microscopy of astrocyte-
enriched cultures indicated that these cultures were neuron free.
This was confirmed using indirect immunocytochemistry, im-
munoblotting, and RT-PCR. Astrocytic cell cultures were immu-
nopositive for GFAP (245 of 245 cells tested), a characteristic
astrocytic marker, revealing the purity of this culture to be 99%
(Fig. 1A). To assess the extent of putative neuronal contamina-
tion, we used immunoblots and RT-PCR. We prepared a non-
nuclear membrane extract from cultured astrocytes (AsP) that
was subjected to SDS-PAGE (Fig. 1B, top). We used synaptoso-
mal plasma membrane-enriched prepara-
tion (LP-1) as a positive control. Immuno-
blot analysis revealed the absence of a
neuron-specific protein, SNAP-25 (Par-
pura et al., 1995b; Latour et al., 2003), in
astrocytic culture, although SNAP-25 was
reliably detected in LP-1 preparations.
Next, we performed RT-PCR using mRNA
isolated from rat brain and from astrocytic
cultures (Fig. 1 B, bottom). The PCR prod-
uct for SNAP-25 was not detected in astro-
cytic mRNA, whereas the positive control
was obtained from brain mRNA. Taken
together, immunocytochemisty, Western
blots, and RT-PCR demonstrate that the
cultures used in this study were devoid of
neurons and contain GFAP-positive po-
lygonal astrocytes.
To determine for the presence of
VGLUTs 1 and 2, we submitted AsP and
LP-1 to SDS-PAGE, followed by Western
blots (Fig. 2A). We found that astrocytic
preparations showed expression of
VGLUTs 1 and 2 and also VGLUT 3, the
presence of which had been recently re-
ported in astrocytes in vivo (Fremeau et al.,
2002). VGLUTs 1 and 2 were much less
abundant in AsP than in LP-1, whereas
VGLUT3 was more abundant in AsP. Ad-
ditionally, astrocytes contained SNARE
proteins, synaptobrevin 2, syntaxin, and
SNAP-23 (Parpura et al., 1995b; Hepp et
al., 1999). To assess the specificity of anti-
bodies raised against VGLUTs (Table 1),
we first performed preadsorption controls
(Fig. 2B). We subjected AsP to the SDS-
PAGE, followed by Western blot analysis
using antibodies alone or antibodies that
were preincubated with their respective
antigens (Table 1). We detected VGLUTs only when probing
with antibodies alone but not in preadsorption controls. Next, we
performed controls for cross-reactivity of antibodies raised
against VGLUTs 1 and 2 to a VGLUT 3 antigen (Fig. 2C). Lack of
cross-reactivity of anti-VGLUTs 1 and 2 to VGLUTs 2 and 1,
respectively, had been demonstrated previously (Bellocchio et al.,
1998; Takamori et al., 2001), but not their possible cross-
reactivity to VGLUT 3. Because astrocytes in vivo express VGLUT
3, it is possible that the antibodies we used against VGLUTs 1 and
2 cross-react with VGLUT 3. To address this issue, we preincu-
bated antibodies against VGLUTs 1, 2, and 3 with a VGLUT 3
antigen. We used antibodies alone or preincubated antibodies to
probe membranes containing AsP. We found that preadsorption
with VGLUT 3 antigens abolished VGLUT detection on Western
blots only when using anti-VGLUT 3 (99% of control; n3)
but not for anti-VGLUT 1 (32% of control; n3) and anti-
VGLUT 2 (11% of control; n3), indicating that these anti-
bodies to not exhibit cross-reactivity to VGLUT 3 (Fig. 2C). After
assessment of the antibody specificity, we performed RT-PCR
using mRNA isolated from rat brain and from astrocytic cultures
(Fig. 2 D). PCR products for VGLUTs 1, 2, and 3 were detected in
astrocytic mRNA. Thus, Western blots and RT-PCR demonstrate
that the purified astrocytic cultures express all three known iso-
forms of VGLUTs.
Figure 4. VGLUT immunoreactivity colocalizes with vesicular marker synaptobrevin 2. A, Astrocytes were identified based on
their ability to intracellularly accumulate
-Ala-Lys-N
-AMCA (blue). Double labeling using indirect immunocytochemistry
against VGLUT 2 (B; red) reveals punctate immunoreactivity that colocalizes with
-Ala-Lys-N
-AMCA (C; overlay of Aand B; pink
indicates colocalization), as shown in X-Z (C; bottom) and Y-Z (C; right) projections from the regions identified by dotted lines.
Double labeling of astrocytes with synaptobrevin 2 (D; green) and a mixture of antibodies labeling all three isoforms of VGLUTs (E;
red) indicates that VGLUT immunoreactivity substantially overlaps with vesicular marker synaptobrevin 2 (F; overlay of Dand E;
yellow indicates colocalization). Scale bars: C, X-Z and Y-Z projections, 5
m; A–C,10
m; D–F,20
m.
Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes J. Neurosci., March 17, 2004 24(11):2633–2642 2637
The intracellular localization of proteins was studied using
indirect immunocytochemistry (Fig. 3). Staining with anti-
VGLUT 1 (Fig. 3C), anti-VGLUT 2 (Fig. 3D), or anti-VGLUT 3
(Fig. 3E) showed a punctate pattern of immunoreactivity that was
present throughout the entire cell body and processes, and it was
completely abolished when antibodies were preadsorbed with
their respective antigens (Fig. 3CE,F). However, consistent
with Western blot data, staining of astrocytes with anti-VGLUT 1
(n68) or anti-VGLUT 2 (n46) showed no reduction in
fluorescence intensities, when these antibodies were preincu-
bated with a VGLUT3 antigen. A similar pattern of punctate
immunoreactivity was observed using anti-SNAP-23 (Fig. 3B).
Synaptobrevin 2, however, was primarily located at the leading
edge of the cells, although there were puncta present throughout
the entire cell body (Figs. 3A,4D).
To further study intracellular distribution of VGLUTs, we
used double labeling. First, we used
-Ala-Lys-N
-AMCA (Dieck
et al., 1999) (also see Materials and Methods) to label astrocytes,
followed by colabeling using indirect immunocytochemistry
against VGLUTs 1, 2, or 3. In Figure 4, we show a typical distri-
bution of VGLUT 2 immunoreactivity that colocalizes with in-
tracellular astrocytic tag
-Ala-Lys-N
-AMCA, as revealed by
volume rendering after image acquisition using a laser confocal
scanning microscope (Fig. 4 A–C). Similar distributions were ob-
served for colabeling VGLUT 1 or 3 with
-Ala-Lys-N
-AMCA,
indicating predominant intracellular, rather than plasma mem-
brane, association of VGLUTs. Because VGLUTs show synaptic
vesicular localization in neurons, next we used indirect immuno-
cytochemisty to label astrocytic vesicular pool using a synaptic
vesicle marker synaptobrevin 2, while counterstaining VGLUTs
using a mixture of antibodies encompassing all three isoforms
(Fig. 4D–F). We found a substantial colocalization of VGLUTs
with synaptobrevin 2, although some of the synaptobrevin 2 im-
munoreactivity was devoid of VGLUT immunoreactivity. Be-
cause astrocytes, in addition to glutamate, can exocytotically re-
lease ATP (Coco et al., 2003) and atrial natriuretic peptide (Krzan
et al., 2003), this is possibly attributable to vesicular packaging of
these segretagogues in the absence of intravesicular glutamate,
hence some synaptobrevin 2-positive vesicles lacking VGLUTs.
Because astrocytes could de-differentiate while cultured, pos-
sibly leading to gene expression of VGLUTs, we next used freshly
(acutely) isolated astrocytes that provide information of proper-
ties without the changes in gene expression (Kimelberg et al.,
2000a,b, 2001). Astrocytes were freshly isolated from rat visual
cortices of 1-, 2-, 8-, and 55-d-old rats. To determine the identity
of astrocytes in cell mixture, we used
-Ala-Lys-N
-AMCA. We
colabeled cells using indirect immunocytochemistry against ei-
ther VGLUT 1, VGLUT 2, or VGLUT 3. Once we localized astro-
cytes of interest based on their ability to accumulate
-Ala-Lys-
N
-AMCA (Fig. 5A–C), we checked for expression of VGLUTs in
these cells. We found that freshly isolated astrocytes contained
VGLUTs in a punctate pattern similar to that seen in cultured
astrocytes (Fig. 5 A–C). This immunoreactivity was completely
abolished when antibodies were preadsorbed with their respec-
tive antigens (Fig. 5D) but not in astrocytes probed with anti-
VGLUT 1 (n54) or anti-VGLUT 2 (n56) antibodies when
these antibodies were preincubated with a VGLUT 3 antigen. The
positive immunoreactivity exhibited fluorescence intensity that
was five (for VGLUTs 1 and 2) to six (for VGLUT 3) times higher
than the intensity recorded from control astrocytes in which pri-
mary antibodies were omitted. This signal/noise ratio may indi-
cate that the expression of VGLUTs in freshly isolated astrocytes
is low. The proportion of astrocytes expressing VGLUTs declined
with the increased age of the animals (Table 2). Together, these
data demonstrate that astrocytes express VGLUTs.
Having determined that astrocytes in culture as well as freshly
isolated astrocytes possess VGLUTs, we asked whether VGLUTs
play a role in Ca
2
-dependent exocytotic release of glutamate
from astrocytes. We optically monitored glutamate release into
extracellular space surrounding cultured astrocytes using GDH-
linked assay, based on accumulation of the fluorescent product
NADH. To evoke Ca
2
-dependent glutamate release from astro-
Figure 5. Freshly isolated astrocytes contain VGLUTs. A–C, Astrocytes were identified based
on their ability to accumulate
-Ala-Lys-N
-AMCA. Double labeling using indirect immunocy-
tochemistry against VGLUT1 (A), VGLUT2 (B), or VGLUT 3 (C) revealed punctate immunore-
activity that was completely abolished ( D) when primary antibodies (1
o
Ab) were preadsorbed
with their respective antigens (Ag). Fluorescent immunoreactivity is expressed in intensity
units(i.u.).Bars representmeans SEMs ofmeasurements fromanumber ofindividual freshly
isolated astrocytes (n). Asterisks indicate a significant change of measurements compared with
the control group (1
o
Ab,Ag; one-way ANOVA, followed by post hoc Fisher’s LSD test;
**p0.01). We found no difference between measurements in preadsorption controls
(1
o
Ab,Ag) when compared with controls in which primary antibodies were omitted
(1
o
Ab,Ag).Scale bar, 10
m.
Table 2. Expression of VGLUTs 1, 2, and 3 in cultured and freshly isolated (FIA)
astrocytes
Astrocytes Cultured FIA
Animal age (days) 0 –1 1–2 8 55
VGLUT 1
Tested 335 156 22 22
Positive 316 134 16 7
Percentage positive 94 86 73 32
VGLUT 2
Tested 404 113 25 26
Positive 381 112 17 9
Percentage positive 94 99 68 35
VGLUT 3
Tested 144 74 19 22
Positive 136 55 10 9
Percentage positive 94 74 53 41
2638 J. Neurosci., March 17, 2004 24(11):2633–2642 Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes
cytes, we used mechanical stimulation, known to cause an in-
crease in astrocytic intracellular Ca
2
levels (Nedergaard, 1994;
Araque et al., 1998a), leading to glutamate release (Parpura et al.,
1994; Araque et al., 1998a). To apply mechanical stimulus, we
established a contact between an astrocyte and a patch pipette
under control of a patch-clamp amplifier. This direct stimulation
reliably caused glutamate release from as-
trocytes as indicated by a transient increase
in NADH fluorescence surrounding astro-
cytes (Fig. 6A-D) (control group, n41;
peak response dF/Fo 90 12%; paired t
test; p0.01).
We confirmed the Ca
2
and SNARE
dependency of this glutamate release (Par-
pura et al., 1994, 1995a,b; Jeftinija et al.,
1997; Bezzi et al., 1998; Araque et al., 2000;
Parpura and Haydon, 2000; Pasti et al.,
2001). We monitored the intracellular
Ca
2
levels using the Ca
2
indicator
fluo-3. Mechanical stimulation caused
transient Ca
2
elevations (Fig. 6 EH)
(control group, n7; peak response dF/
Fo 208 58%; paired ttest; p0.015),
which was greatly reduced when we prein-
cubated astrocytes with BAPTA-AM (50
M; 30 min; 23% of control; n8; peak
response dF/Fo 47 11%; Fishers LSD;
p0.01) (Fig. 6H). Consistent with Ca
2
dependency of mechanically induced glu-
tamate release, this Ca
2
chelator greatly
reduced the extracellular NADH fluores-
cence accumulation reporting on gluta-
mate release (29% of control; n4; peak
response dF/Fo 26 8%; Fishers LSD
test; p0.05) (Fig. 6D).
Growing evidence indicates that the
mechanism underlying the Ca
2
-
dependent glutamate release from astro-
cytes is exocytosis (Jeftinija et al., 1997;
Bezzi et al., 1998; Araque et al., 2000; Pasti
et al., 2001). Consequently, we tested the
sensitivity of mechanically induced gluta-
mate release from astrocytes to tetanus
toxin and bafilomycin A
1
.Ca
2
-
dependent glutamate release from astro-
cytes can be reduced when astrocytes are
exposed to tetanus toxin that specifically
cleaves synaptobrevin 2 (Jeftinija et al.,
1997; Bezzi et al., 1998; Pasti et al., 2001).
We confirmed the presence of synaptobre-
vin 2 (Parpura et al., 1995b) in astrocytes
using Western blots (Fig. 2) and indirect
immunocytochemistry (Fig. 3). Addition-
ally, we tested functional involvement of
synaptobrevin 2 in Ca
2
-dependent glu-
tamate release from astrocytes by pretreat-
ing these cells with holoprotein of tetanus
toxin (15
g/ml for 24 48 hr). After incu-
bation with this toxin, astrocytes had re-
duced ability to release glutamate in re-
sponse to mechanical stimulation (41% of
control; n13; peak response dF/Fo
37 5%; Fishers LSD test; p0.01) (Fig.
6D), although toxin treatment did not affect mechanically in-
duced Ca
2
elevations (Fig. 6H)(n6).
Bafilomycin A
1
, a specific inhibitor of V-ATPase (Bowman et
al., 1988), has been shown to reduce the extent of Ca
2
-
dependent glutamate release from astrocytes, by collapsing the
proton gradient necessary for vesicular storage of glutamate
Figure 6. VGLUTs mediate exocytotic glutamate release from cultured astrocytes. Mechanical stimulation evokes glutamate
release ( A–D) caused by the internal Ca
2
elevations in astrocytes ( E–H). Glutamate release from astrocyte is shown (raw data)
in A(rest) and B(after stimulation). The pseudocolor scale is a linear representation of the fluorescence intensities ranging from
1240 to 1350. C, Time lapse of NADH fluorescence, reporting on glutamate release. Mechanical stimulation caused glutamate
release (black squares) that was greatly reduced when cells were preincubated with Rose Bengal (open circles; 0.5
M; 30 min),
an allosteric site modulator of VGLUTs. D, Peak values of mechanically induced glutamate release. Incubation of astrocytes with
BAPTA-AM (50
M; 30 min), a membrane-permeable Ca
2
chelator, reduced the mechanically induced glutamate release.
Pretreatment of astrocytes with a holoprotein of tetanus toxin (TeTx; 15
g/ml; 24 –48 hr), which cleaves synaptobrevin 2, or
bafilomycin A
1
(5
M, 1 hr), a specific inhibitor of V-ATPase, reduces mechanically induced glutamate release consistent with
exocytoticmechanismunderlying Ca
2
-dependentglutamaterelease fromastrocytes.Changes inNADHfluorescence areshown
as dF/Fo (percentage) after background subtraction and correction for bleaching. E–H, In experiments parallel to those in A–D,
astrocytes were mechanically stimulated while measuring intracellular Ca
2
levels using fluo-3. This stimulation reliably in-
creasestheintracellular Ca
2
levels ( F) from its resting level ( E)(rawdata). Thepseudocolor scale isa linearrepresentationof the
fluorescence intensities ranging from 0 to 255. G, Time lapse of fluo-3 fluorescence, reporting on Ca
2
levels in astrocytes.
Mechanical stimulation caused an increase in astrocytic intracellular Ca
2
levels (black squares) that were unaffected when cells
were preincubated with Rose Bengal (open circles; 0.5
M; 30 min). H, Peak values of mechanically induced Ca
2
elevations are
greatlyreducedwhen cellsare preincubatedwithBAPTA butnot whentreatedwith TeTx,bafilomycin A
1
,orRose Bengal.Changes
in fluo-3 fluorescence are expressed as dF/Fo (percentage) after background subtraction. Arrows (C,G) indicate the time when the
pipette–astrocytecontactoccurred. Pointsand barsrepresentthe meansand SEMs. InCand G,SEMsare shownin singledirections
for clarity. Asterisks indicate a significant change of measurements compared with the control group (one-way ANOVA, followed
by post hoc Fisher’s LSD test; *p0.05; **p0.01). We found no significant difference between various treatments in D,
whereas in H, preincubation with BAPTA, in addition to a significant change in measurement compared with the control group,
showed a statistically significant difference when compared with all other treatments ( p0.05).
Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes J. Neurosci., March 17, 2004 24(11):2633–2642 2639
(Araque et al., 2000; Pasti et al., 2001). Consistent with previous
observations, incubation of astrocytes with bafilomycin A
1
(5
M; 1 hr) greatly reduced mechanically induced glutamate release
(22% of control; n9; peak response dF/Fo 20 5%; Fishers
LSD test; p0.01) (Fig. 6D), without significantly affecting
Ca
2
elevations (Fig. 6H;n6).
After demonstration that mechanically induced glutamate re-
lease is mediated by exocytosis, we incubated astrocytes with Rose
Bengal (0.5
M; 30 min), an inhibitor of vesicular glutamate up-
take in synaptic vesicles via allosteric modulation of VGLUTs
(Ogita et al., 2001; Schafer et al., 2002). We found that this treat-
ment abolished mechanically induced glutamate release from as-
trocytes (Fig. 6C,D) (3% of control; n6; dF/Fo 22%;
Fishers LSD; p0.01) without affecting Ca
2
elevations (Fig.
6G;n7). Together, these data indicate that VGLUTs mediate
mechanically induced exocytotic release from astrocytes.
Although the presented data are consistent with the notion
that glutamate released from astrocyte is stored in acidic com-
partments that are sensitive to bafilomycin A
1
and Rose Bengal, it
is important to confirm specificity of these pharmacological
agents. To address this issue, we transfected the cultured cortical
astrocytes using a plasmid encoding for synapto-pHluorin (San-
karanarayanan et al., 2000), a fusion protein that contains a pH-
sensitive and fluorescent protein, pHluorin, attached to the C
terminus of synaptobrevin 2. Because pHluorin is attached to the
lumenal (intravesicular) portion of synaptobrevin, it is sensing
intravesicular pH with its fluorescence mainly quenched at rest-
ing intravesicular pH (5.5). However, blockade of V-ATPase
using bafilomycin A
1
can collapse proton gradient, leading to
alklinization of intravesicular lumen and increase in pHluorins
fluorescence (Sankaranarayanan and Ryan, 2001). Incubation of
astrocytes with bafilomycin A
1
(5
M; 30 and 60 min) caused a
time-dependent increase in synapto-pHluorin fluorescence
(each group, n8; paired ttest with Bonferroni adjustment for
multiple comparisons; p0.01) (Fig. 7 A,B,E). Furthermore,
synapto-pHluorin fluorescence showed a punctate pattern con-
sistent with the vesicular nature of exocytosis (Fig. 7B). Although
it has been reported that Rose Bengal, at submicromolar concen-
trations used in the present study, has a much higher affinity for
VGLUTs than for V-ATPase (Ogita et al., 2001), it is necessary to
confirm that Rose Bengal does not reduce glutamate release as a
result of inhibition of V-ATPase, thereby reducing the proton
gradient necessary for glutamate uptake into vesicular lumen, as
opposed to its action on allosteric site of VGLUTs. Vesicular
proton shunt through VGLUTs reduces the intravascular proton
concentration created by V-ATPase. By inhibiting VGLUTs and
their proton-shunting activity, Rose Bengal would lead to vesic-
ular acidification and the reduction of synapto-pHluorin fluores-
cence. Incubation of astrocytes with Rose Bengal (0.1 or 0.5
M;
30 min) caused a reduction of pHluorin fluorescence (0.1
M,
n6; paired ttest; p0.05; 0.5
M,n10; paired ttest; p
0.01), consistent with its action on VGLUTs (Fig. 7CE).
Whereas mechanical stimulation offers direct stimulation of
astrocytes without receptor activation and mimics the action of
endogenous ligands (Sanzgiri et al., 1999), it is not a physiological
stimulus. Consequently, to further study VGLUT-dependent
exocytotic glutamate release from astrocytes, we determined the
effects of Rose Bengal on astrocytes stimulated by bradykinin (90
nM; 55 sec) or ATP (9
M; 55 sec), endogenous ligands known to
stimulate Ca
2
-dependent glutamate release (Parpura et al.,
1994, 1995a; Jeftinija et al., 1997; Jeremic et al., 2001). Both bra-
Figure 7. Bafilomycin A
1
alkalinizes vesicular lumen, whereas Rose Bengal acidifies it, con-
sistent with the actions of these agents on V-ATPase and VGLUTs, respectively. A–D, Astrocytes
expressing synapto-pHluorin (raw data). The addition of bafilomycin A
1
to the astrocytes at rest
expressing synapto-pHluorin ( A) causes an increase in pHluorin fluorescence (B,E). Quite the
contrary, the addition of Rose Bengal to synapto-pHluorin-expressing astrocytes at rest (C)
causes decrease of pHluorin fluorescence (D,E). The image in Bis acquired after incubation with
bafilomycin A
1
(5
M; 60 min), whereas the image in Dis acquired after incubation with Rose
Bengal (0.5
M; 30 min). The pseudocolor scale is a linear representation of the fluorescence
intensities ranging from 0 to 255. Changes in synapto-pHluorin fluorescence are expressed as
dF/Fo (percentage) after background subtraction. Bars represent the means and SEMs. Concen-
trations (c) and exposure times (t) of pharmacological agents are given in micromolars and
minutes, respectively.
Figure 8. Rose Bengal reduces bradykinin- and ATP-evoked Ca
2
-dependent glutamate
release from astrocytes. A, Stimulation of astrocytes with bath-applied (55 sec) bradykinin (90
nM) or ATP (9
M) causes glutamate release (A, black bars) and the increase in internal Ca
2
levels (B, black bars). A,B, This agonist-induced glutamate release from astrocytes was greatly
reduced when astrocytes were preincubated with Rose Bengal (0.1
Mfor 30 min; A, open
bars),althoughastrocytic internalCa
2
elevationswerenot affected(B, open bars).Changes in
NADH fluorescence, reporting on glutamate release, are shown as dF/Fo (percentage) after
background subtraction and correction for bleaching. Changes in fluo-3 fluorescence ( B), re-
porting on internal Ca
2
levels, are expressed as dF/Fo (percentage) after background subtrac-
tion. Bars represent the means and SEMs. Asterisks indicate a significant change of measure-
ments compared with the matching control group (Student’s ttest; **p0.01).
2640 J. Neurosci., March 17, 2004 24(11):2633–2642 Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes
dykinin (n19) and ATP (n33) caused glutamate release
from astrocytes (paired ttests; p0.01); release magnitudes were
reduced to 44% (n15) and 47% (n21) of control, respec-
tively (Studentsttests; p0.01), when we preincubated astro-
cytes with Rose Bengal (0.1
M; 30 min) (Fig. 8A). Consistent
with preservation of Ca
2
mobilization by bradykinin or ATP,
Rose Bengal did not affect ligand-induced Ca
2
elevations in
astrocytes (Fig. 8 B)(n10 for bradykinin alone and n9 for
bradykinin with Rose Bengal; n26 for ATP alone and n18
for ATP with Rose Bengal; Studentsttests; p0.4). Thus, three
distinct stimuli, mechanical contact, bradykinin, and ATP, can
raise intracellular Ca
2
levels and cause glutamate release from
astrocytes; release magnitudes can be significantly reduced when
we pharmacologically interfered with VGLUTs.
Discussion
Our findings indicate that the expression of VGLUTs 1 and 2 is
not restricted to neurons but that these proteins necessary for
glutamate accumulation in synaptic vesicles are also found in
astrocytes, as determined by using purified astrocytic culture and
Western blots. Immunocytochemisty performed on both cul-
tured and freshly isolated astrocytes reveals punctate pattern of
immunoreactivity consistent with the possible role of VGLUTs in
exocytotic glutamate release from astrocytes. Furthermore, me-
chanically, bradykinin-, and ATP-induced glutamate release
from astrocytes was greatly reduced when astrocytes were prein-
cubated with Rose Bengal, a broad spectrum modulator of
VGLUTs allosteric site, indicating a functional role of VGLUTs in
this release.
Interestingly, in freshly isolated astrocytes originating from 1-
to 2-d-old rat pups, there is a high likelihood that VGLUTs 1, 2,
and 3 are co expressed in a single cell (Table 2). Although we did
not directly demonstrate the presence of these proteins in single
cells, the proportion of cells expressing individual proteins (86%
for VGLUT 1, 99% for VGLUT 2, and 74% for VGLUT 3) sup-
ports this inference. However, the probability of dual or triple
expression of VGLUTs in individual astrocytes was greatly re-
duced when we studied astrocytes isolated from 55-d-old ani-
mals, as was the proportion of astrocytes expressing individual
VGLUTs. Why would individual astrocytes in the developing
brains of postnatal animals simultaneously express two or three
different VGLUTs? One possible explanation is that the Ca
2
-
dependent release of glutamate is important for astrocytic roles in
modulation of synaptic transmission and that it is necessary to
have protein redundancy to ensure astrocytic contribution to the
physiology in the CNS. An alternative explanation could be that
different VGLUTs have specialized functions in astrocytic gluta-
mate metabolism and/or release. For example, it is entirely pos-
sible that VGLUT isoforms are exclusively expressed in a different
subpopulation of vesicles within individual astrocytes. These
subpopulations of vesicles could represent distinct releasable
pools that could control spatiotemporal characteristics of gluta-
mate release. Such a possibility could explain different forms of
neuronal responses to glutamate released from astrocytes, in-
cluding internal Ca
2
increases (Parpura et al., 1994), slow-
inward currents (Araque et al., 1998a; Parpura and Haydon,
2000), and modulation of spontaneous and action potential-
evoked synaptic transmissions (Araque et al., 1998b; Kang et al.,
1998). In mature animals, however, VGLUT isoforms 1 and 2 are
each expressed in approximately one-third of the astrocytic pop-
ulation (32 and 35%, respectively). Because VGLUT 3 is ex-
pressed in a similar proportion (41%), it is tempting to suggest
that astrocytes could show highly complementary distributions
of these three proteins, such as is found in neurons (Herzog et al.,
2001; Fremeau et al., 2002; Schafer et al., 2002). An alternative
extreme possibility is that only one-third of astrocytes express
VGLUTs but all isoforms. Subsequent work will be necessary to
resolve these issues. Consequently, it should become possible to
selectively manipulate individual VGLUTs in astrocytes to test
for their roles in glutamate-mediated astrocyte-neuron signaling
in health and disease.
References
Araque A, Parpura V, Sanzgiri RP, Haydon PG (1998a) Glutamate-
dependent astrocyte modulation of synaptic transmission between cul-
tured hippocampal neurons. Eur J Neurosci 10:2129 2142.
Araque A, Sanzgiri RP, Parpura V, Haydon PG (1998b) Calcium elevation
in astrocytes causes an NMDA receptor-dependent increase in the fre-
quency of miniature synaptic currents in cultured hippocampal neurons.
J Neurosci 18:68226829.
Araque A, Li N, Doyle RT, Haydon PG (2000) SNARE protein-dependent
glutamate release from astrocytes. J Neurosci 20:666 673.
Bai L, Xu H, Collins JF, Ghishan FK (2001) Molecular and functional anal-
ysis of a novel neuronal vesicular glutamate transporter. J Biol Chem
276:36764 36769.
Bellocchio EE, Hu H, Pohorille A, Chan J, Pickel VM, Edwards RH (1998)
The localization of the brain-specific inorganic phosphate transporter
suggests a specific presynaptic role in glutamatergic transmission. J Neu-
rosci 18:8648 8659.
Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Rizzini BL, Pozzan T,
Volterra A (1998) Prostaglandins stimulate calcium-dependent gluta-
mate release in astrocytes. Nature 391:281285.
Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq E, Vescovi A,
Bagetta G, Kollias G, Meldolesi J, Volterra A (2001) CXCR4-activated
astrocyte glutamate release via TNFalpha: amplification by microglia trig-
gers neurotoxicity. Nat Neurosci 4:702710.
Bowman EJ, Siebers A, Altendorf K (1988) Bafilomycins: a class of inhibi-
tors of membrane ATPases from microorganisms, animal cells, and plant
cells. Proc Natl Acad Sci USA 85:79727976.
Coco S, Calegari F, Pravettoni E, Pozzi D, Taverna E, Rosa P, Matteoli M,
Verderio C (2003) Storage and release of ATP from astrocytes in culture.
J Biol Chem 278:1354 1362.
Dani JW, Chernjavsky A, Smith SJ (1992) Neuronal activity triggers calcium
waves in hippocampal astrocyte networks. Neuron 8:429 440.
Dieck ST, Heuer H, Ehrchen J, Otto C, Bauer K (1999) The peptide trans-
porter PepT2 is expressed in rat brain and mediates the accumulation of
the fluorescent dipeptide derivative beta-Ala-Lys-Nepsilon-AMCA in as-
trocytes. Glia 25:10 20.
Fremeau Jr RT, Troyer MD, Pahner I, Nygaard GO, Tran CH, Reimer RJ,
Bellocchio EE, Fortin D, Storm-Mathisen J, Edwards RH (2001) The
expression of vesicular glutamate transporters defines two classes of exci-
tatory synapse. Neuron 31:247260.
Fremeau Jr RT, Burman J, Qureshi T, Tran CH, Proctor J, Johnson J, Zhang
H, Sulzer D, Copenhagen DR, Storm-Mathisen J, Reimer RJ, Chaudhry
FA, Edwards RH (2002) The identification of vesicular glutamate trans-
porter 3 suggests novel modes of signaling by glutamate. Proc Natl Acad
Sci USA 99:14488 14493.
Fujiyama F, Furuta T, Kaneko T (2001) Immunocytochemical localization
of candidates for vesicular glutamate transporters in the rat cerebral cor-
tex. J Comp Neurol 435:379 387.
Gras C, Herzog E, Bellenchi GC, Bernard V, Ravassard P, Pohl M, Gasnier B,
Giros B, El Mestikawy S (2002) A third vesicular glutamate transporter
expressed by cholinergic and serotoninergic neurons. J Neurosci
22:54425451.
Hepp R, Perraut M, Chasserot-Golaz S, Galli T, Aunis D, Langley K, Grant NJ
(1999) Cultured glial cells express the SNAP-25 analogue SNAP-23. Glia
27:181187.
Herzog E, Bellenchi GC, Gras C, Bernard V, Ravassard P, Bedet C, Gasnier B,
Giros B, El Mestikawy S (2001) The existence of a second vesicular glu-
tamate transporter specifies subpopulations of glutamatergic neurons.
J Neurosci 21:RC181(16).
Hisano S, Hoshi K, Ikeda Y, Maruyama D, Kanemoto M, Ichijo H, Kojima I,
Takeda J, Nogami H (2000) Regional expression of a gene encoding a
Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes J. Neurosci., March 17, 2004 24(11):2633–2642 2641
neuron-specific Na()-dependent inorganic phosphate cotransporter
(DNPI) in the rat forebrain. Brain Res Mol Brain Res 83:3443.
Huttner WB, Schiebler W, Greengard P, De Camilli P (1983) Synapsin I
(protein I), a nerve terminal-specific phosphoprotein. III. Its association
with synaptic vesicles studied in a highly purified synaptic vesicle prepa-
ration. J Cell Biol 96:1374 1388.
Innocenti B, Parpura V, Haydon PG (2000) Imaging extracellular waves of
glutamate during calcium signaling in cultured astrocytes. J Neurosci
20:1800 1808.
Jeftinija SD, Jeftinija KV, Stefanovic G (1997) Cultured astrocytes express
proteins involved in vesicular glutamate release. Brain Res 750:4147.
Jeremic A, Jeftinija K, Stevanovic J, Glavaski A, Jeftinija S (2001) ATP stim-
ulates calcium-dependent glutamate release from cultured astrocytes.
J Neurochem 77:664 675.
Kaneko T, Fujiyama F, Hioki H (2002) Immunohistochemical localization
of candidates for vesicular glutamate transporters in the rat brain. J Comp
Neurol 444:39 62.
Kang J, Jiang L, Goldman SA, Nedergaard M (1998) Astrocyte-mediated
potentiation of inhibitory synaptic transmission. Nat Neurosci
1:683692.
Kimelberg HK (2001) Glia-neuronal culture modelsdo we need to
change the paradigms? Trends Neurosci 24:205206.
Kimelberg HK, Schools GP, Cai Z, Zhou M (2000a) Freshly isolated astro-
cyte (FIA) preparations: a useful single cell system for studying astrocyte
properties. J Neurosci Res 61:577587.
Kimelberg HK, Cai Z, Schools G, Zhou M (2000b) Acutely isolated astro-
cytes as models to probe astrocyte functions. Neurochem Int 36:359 367.
Krzan M, Stenovec M, Kreft M, Pangrsic T, Grilc S, Haydon PG, Zorec R
(2003) Calcium-dependent exocytosis of atrial natriuretic peptide from
astrocytes. J Neurosci 23:1580 1583.
Latour I, Hamid J, Beedle AM, Zamponi GW, Macvicar BA (2003) Expres-
sion of voltage-gated Ca2channel subtypes in cultured astrocytes. Glia
41:347353.
Maienschein V, Marxen M, Volknandt W, Zimmermann H (1999) A pleth-
ora of presynaptic proteins associated with ATP-storing organelles in
cultured astrocytes. Glia 26:233244.
McCarthy KD, deVellis J (1980) Preparation of separate astroglial and oli-
godendroglial cell cultures from rat cerebral tissue. J Cell Biol 85:890 902.
Nedergaard M (1994) Direct signaling from astrocytes to neurons in cul-
tures of mammalian brain cells. Science 263:1768 1771.
Ni B, Rosteck Jr PR, Nadi NS, Paul SM (1994) Cloning and expression of a
cDNA encoding a brain-specific Na()-dependent inorganic phosphate
cotransporter. Proc Natl Acad Sci USA 91:56075611.
Ni B, Wu X, Yan GM, Wang J, Paul SM (1995) Regional expression and
cellular localization of the Na()-dependent inorganic phosphate co-
transporter of rat brain. J Neurosci 15:5789 5799.
Ni Y, Sunjara V, Parpura V (2003) Expression of vesicular glutamate trans-
porters in cultured astrocytes. FASEB J [Suppl] 17:A457.
Ogita K, Hirata K, Bole DG, Yoshida S, Tamura Y, Leckenby AM, Ueda T
(2001) Inhibition of vesicular glutamate storage and exocytotic release by
Rose Bengal. J Neurochem 77:34 42.
Parpura V, Haydon PG (2000) Physiological astrocytic calcium levels stim-
ulate glutamate release to modulate adjacent neurons. Proc Natl Acad Sci
USA 97:8629 8634.
Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG (1994)
Glutamate-mediated astrocyte-neuron signalling. Nature 369:744 747.
Parpura V, Liu F, Brethorst S, Jeftinija K, Jeftinija S, Haydon PG (1995a)
Alpha-latrotoxin stimulates glutamate release from cortical astrocytes in
cell culture. FEBS Lett 360:266 270.
Parpura V, Fang Y, Basarsky T, Jahn R, Haydon PG (1995b) Expression of
synaptobrevin II, cellubrevin and syntaxin but not SNAP-25 in cultured
astrocytes. FEBS Lett 377:489 492.
Pasti L, Zonta M, Pozzan T, Vicini S, Carmignoto G (2001) Cytosolic cal-
cium oscillations in astrocytes may regulate exocytotic release of gluta-
mate. J Neurosci 21:477484.
Peters A, Palay SL, Webster HD (1991) The fine structure of the nervous
system, Ed 3. New York: Oxford UP.
Sakata-Haga H, Kanemoto M, Maruyama D, Hoshi K, Mogi K, Narita M,
Okado N, Ikeda Y, Nogami H, Fukui Y, Kojima I, Takeda J, Hisano S
(2001) Differential localization and colocalization of two neuron-types
of sodium-dependent inorganic phosphate cotransporters in rat fore-
brain. Brain Res 902:143155.
Sankaranarayanan S, Ryan TA (2001) Calcium accelerates endocytosis of
vSNAREs at hippocampal synapses. Nat Neurosci 4:129 136.
Sankaranarayanan S, De Angelis D, Rothman JE, Ryan TA (2000) The use of
pHluorins for optical measurements of presynaptic activity. Biophys J
79:2199 2208.
Sanzgiri RP, Araque A, Haydon PG (1999) Prostaglandin E(2) stimulates
glutamate receptor-dependent astrocyte neuromodulation in cultured
hippocampal cells. J Neurobiol 41:221229.
Schafer MK, Varoqui H, Defamie N, Weihe E, Erickson JD (2002) Molecu-
lar cloning and functional identification of mouse vesicular glutamate
transporter 3 and its expression in subsets of novel excitatory neurons.
J Biol Chem 277:50734 50748.
Takamori S, Rhee JS, Rosenmund C, Jahn R (2001) Identification of
differentiation-associated brain-specific phosphate transporter as a sec-
ond vesicular glutamate transporter (VGLUT2). J Neurosci
21:RC182(16).
Varoqui H, Schafer MK, Zhu H, Weihe E, Erickson JD (2002) Identification
of the differentiation-associated Na/PI transporter as a novel vesicular
glutamate transporter expressed in a distinct set of glutamatergic syn-
apses. J Neurosci 22:142155.
Zhou M, Kimelberg HK (2000) Freshly isolated astrocytes from rat hip-
pocampus show two distinct current patterns and different [K()](o)
uptake capabilities. J Neurophysiol 84:2746 2757.
2642 J. Neurosci., March 17, 2004 24(11):2633–2642 Montana et al. VGLUT-Dependent Glutamate Release from Astrocytes
... Calcium-dependent and -independent mechanisms for glutamate release from astrocytes have been proposed. These include (a) exocytosis from vesicles, (b) anion channel opening, (c) glutamate exchange via cystine-glutamate antiporter, (d) release from hemichannels, or (e) ionotropic purinergic receptors (Araque et al., 2000;Montana et al., 2004;Zhang et al., 2004;Malarkey and Parpura, 2008). Vesicular gliotransmitter release of glutamate has been supported by morphological and functional evidence. ...
... Interestingly, the mechanisms involved in glutamate-mediated exocytosis have been highly debated in the last years Chai et al., 2017). Functionally, expression in astrocytes with the light chain of tetanus toxin that selectively cleaves the vesicle-associated SNARE protein potently inhibits the release of glutamate from astrocytes (Montana et al., 2004;Xu et al., 2007;Araque et al., 2014). However, complementary evidence has questioned the exact mechanisms involved in Ca 2+ -dependent glutamate exocytosis Chai et al., 2017). ...
... Glutamate has been shown to be released via a variety of mechanisms. These mechanisms include exocytosis, lysosomes, hemichannels, exchangers, anion channels, antiporters as well as channels such as TREK-1 and Bestropin-1 (BEST-1) (Araque et al., 2000;Montana et al., 2004;Zhang et al., 2004;Xu et al., 2007;Malarkey and Parpura, 2008;Yang et al., 2019;Okada et al., 2021). GABA on the other hand, has been shown to be released via BEST-1, hemichannels, as well as anion channels and transporters (Kozlov et al., 2006;Jiménez-González et al., 2011;Le Meur et al., 2012;Yoon and Lee, 2014;Christensen et al., 2018;Kwak et al., 2020). ...
Article
Full-text available
Glia are as numerous in the brain as neurons and widely known to serve supportive roles such as structural scaffolding, extracellular ionic and neurotransmitter homeostasis, and metabolic support. However, over the past two decades, several lines of evidence indicate that astrocytes, which are a type of glia, play active roles in neural information processing. Astrocytes, although not electrically active, can exhibit a form of excitability by dynamic changes in intracellular calcium levels. They sense synaptic activity and release neuroactive substances, named gliotransmitters, that modulate neuronal activity and synaptic transmission in several brain areas, thus impacting animal behavior. This “dialogue” between astrocytes and neurons is embodied in the concept of the tripartite synapse that includes astrocytes as integral elements of synaptic function. Here, we review the recent work and discuss how astrocytes via calcium-mediated excitability modulate synaptic information processing at various spatial and time scales.
... Astrocytes can influence synaptic activity by the release of glutamate, but they also participate in other key aspects of glutamate processing in the CNS including uptake of released glutamate and de-novo synthesis of glutamate from glucose; the latter two activities are likely exclusive to astrocytes (11,32,33). As discussed above, metabolic state can alter astrocytic ability to take up glutamate. ...
Article
Full-text available
Food intake behavior is under the tight control of the central nervous system. Most studies to date focus on the contribution of neurons to this behavior. However, although previously overlooked, astrocytes have recently been implicated to play a key role in feeding control. Most of the recent literature has focused on astrocytic contribution in the hypothalamus or the dorsal vagal complex. The contribution of astrocytes located in the lateral parabrachial nucleus (lPBN) to feeding behavior control remains poorly understood. Thus, here, we first investigated whether activation of lPBN astrocytes affects feeding behavior in male and female rats using chemogenetic activation. Astrocytic activation in the lPBN led to profound anorexia in both sexes, under both ad-libitum feeding schedule and after a fasting challenge. Astrocytes have a key contribution to glutamate homeostasis and can themselves release glutamate. Moreover, lPBN glutamate signaling is a key contributor to potent anorexia, which can be induced by lPBN activation. Thus, here, we determined whether glutamate signaling is necessary for lPBN astrocyte activation-induced anorexia, and found that pharmacological N-methyl D-aspartate (NMDA) receptor blockade attenuated the food intake reduction resulting from lPBN astrocyte activation. Since astrocytes have been shown to contribute to feeding control by modulating the feeding effect of peripheral feeding signals, we further investigated whether lPBN astrocyte activation is capable of modulating the anorexic effect of the gut/brain hormone, glucagon like peptide -1, as well as the orexigenic effect of the stomach hormone - ghrelin, and found that the feeding effect of both signals is modulated by lPBN astrocytic activation. Lastly, we found that lPBN astrocyte activation-induced anorexia is affected by a diet-induced obesity challenge, in a sex-divergent manner. Collectively, current findings uncover a novel role for lPBN astrocytes in feeding behavior control.
... Meanwhile, astrocytes express synaptotagmin 4, 7, and 11 [29][30][31][32], playing the role of calcium sensors to detect elevated intracellular calcium. Astrocytes also express vesicle-associated membrane protein 2 (VAMP2) [33] or VAMP3 [34], syntaxin 4 [35], and synaptosomalassociated protein 25 (SNAP25) or its analog SNAP23 [36,37], suggesting that they can form soluble NSF attachment protein receptor (SNARE) complexes like neurons in order to induce membrane fusion, ensuring eventual Ca2 + -mediated glutamate cytokinesis. ...
Article
Full-text available
Numerous neurological disorders share a fatal pathologic process known as glutamate excitotoxicity. Among which, ischemic stroke is the major cause of mortality and disability worldwide. For a long time, the main idea of developing anti-excitotoxic neuroprotective agents was to block glutamate receptors. Despite this, there has been little successful clinical translation to date. After decades of “neuron-centered” views, a growing number of studies have recently revealed the importance of non-neuronal cells. Glial cells, cerebral microvascular endothelial cells, blood cells, and so forth are extensively engaged in glutamate synthesis, release, reuptake, and metabolism. They also express functional glutamate receptors and can listen and respond for fast synaptic transmission. This broadens the thoughts of developing excitotoxicity antagonists. In this review, the critical contribution of non-neuronal cells in glutamate excitotoxicity during ischemic stroke will be emphasized in detail, and the latest research progress as well as corresponding therapeutic strategies will be updated at length, aiming to reconceptualize glutamate excitotoxicity in a non-neuronal perspective.
... While out of scope of the present study, of additional interest would be to compare the cytokine release pattern from astrocytes exposed to SWCNT-PEG to that of astrocytes exposed to known astrocytic secretagogues, such as adenosine triphosphate. There is a three-pronged rationale for this endeavor: (i) this nucleotide is increased in the extracellular space in experimental TBI [73]; (ii) it can cause glutamate release from cultured uninjured astrocytes via regulated exocytosis, i.e., vesicular release [74,75]; and SCWNT-PEGs affect vesicular recycling in cultured uninjured astrocytes [76]. ...
Article
Full-text available
We tested the effects of water-soluble single-walled carbon nanotubes, chemically functionalized with polyethylene glycol (SWCNT-PEG), on primary mouse astrocytes exposed to a severe in vitro simulated traumatic brain injury (TBI). The application of SWCNT-PEG in the culture media of injured astrocytes did not affect cell damage levels, when compared to those obtained from injured, functionalization agent (PEG)-treated cells. Furthermore, SWCNT-PEG did not change the levels of oxidatively damaged proteins in astrocytes. However, this nanomaterial prevented the reduction in plasmalemmal glutamate transporter EAAT1 expression caused by the injury, rendering the level of EAAT1 on par with that of control, uninjured PEG-treated astrocytes; in parallel, there was no significant change in the levels of GFAP. Additionally, SWCNT-PEG increased the release of selected cytokines that are generally considered to be involved in recovery processes following injuries. As a loss of EAATs has been implicated as a culprit in the suffering of human patients from TBI, the application of SWCNT-PEG could have valuable effects at the injury site, by preventing the loss of astrocytic EAAT1 and consequently allowing for a much-needed uptake of glutamate from the extracellular space, the accumulation of which leads to unwanted excitotoxicity. Additional potential therapeutic benefits could be reaped from the fact that SWCNT-PEG stimulated the release of selected cytokines from injured astrocytes, which would promote recovery after injury and thus counteract the excess of proinflammatory cytokines present in TBI.
... It is now generally accepted that glial cells are excitable: fast ionic signaling, in addition to slow signaling, occurs in glial cells in response to physiological stimuli (Verkhratsky et al., 2020). Notably, astrocytes have been proven to be equipped with transmitter-loading vesicles and to release gliotransmitters in a vesicular way (see Montana et al., 2004Montana et al., , 2006Parpura and Verkhratsky, 2012;Araque et al., 2014;Goenaga et al., 2023). Activation of neuronal target receptors by gliotransmitters (e.g. ...
Article
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
... A massive loss of neurons in the dorsal striatum, along with astrocyte activation, occurs after ischemia onset. Since it is thought that astrocytes express VGLUT2 [26], their activation might be a possible explanation for this data. ...
Article
Full-text available
Brain ischemia is one of the leading causes of death and long-term disability in the world. Interruption of the blood supply to the brain is a direct stimulus for many pathological events. The massive vesicular release of glutamate (Glu) after ischemia onset induces excitotoxicity, which is a potent stress on neurons. Loading of presynaptic vesicles with Glu is the first step of glutamatergic neurotransmission. Vesicular glutamate transporters 1, 2, and 3 (VGLUT1, 2, and 3) are the main players involved in filling presynaptic vesicles with Glu. VGLUT1 and VGLUT2 are expressed mainly in glutamatergic neurons. Therefore, the possibility of pharmacological modulation to prevent ischemia-related brain damage is attractive. In this study, we aimed to determine the effect of focal cerebral ischemia on the spatiotemporal expression of VGLUT1 and VGLUT2 in rats. Next, we investigated the influence of VGLUT inhibition with Chicago Sky Blue 6B (CSB6B) on Glu release and stroke outcome. The effect of CSB6B pretreatment on infarct volume and neurological deficit was compared with a reference model of ischemic preconditioning. The results of this study indicate that ischemia upregulated the expression of VGLUT1 in the cerebral cortex and in the dorsal striatum 3 days after ischemia onset. The expression of VGLUT2 was elevated in the dorsal striatum and in the cerebral cortex 24 h and 3 days after ischemia, respectively. Microdialysis revealed that pretreatment with CSB6B significantly reduced the extracellular Glu concentration. Altogether, this study shows that inhibition of VGLUTs might be a promising therapeutic strategy for the future.
... On the contrary, Ormel et al. (2012) identified VGLUT1 in astrocytic processes in the rat hippocampus, frontal cortex, and striatum. Likewise, astrocytes from postnatal rat brains express VGLUT1 and VGLUT2 (Montana et al., 2004), as well as cortical cultured astrocytes (Anlauf and Derouiche, 2005). VGLUT3 has been detected in astrocytes end-feet in microcultures of rat ventral tegmental area, substantia nigra pars compacta, and raphe nuclei (Fremeau et al., 2002). ...
Article
Full-text available
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte’s role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Article
Full-text available
In the last two decades, there has been increasing evidence supporting non-neuronal cells as active contributors to neurodegenerative disorders. Among glial cells, astrocytes play a pivotal role in driving amyotrophic lateral sclerosis (ALS) progression, leading the scientific community to focus on the “astrocytic signature” in ALS. Here, we summarized the main pathological mechanisms characterizing astrocyte contribution to MN damage and ALS progression, such as neuroinflammation, mitochondrial dysfunction, oxidative stress, energy metabolism impairment, miRNAs and extracellular vesicles contribution, autophagy dysfunction, protein misfolding, and altered neurotrophic factor release. Since glutamate excitotoxicity is one of the most relevant ALS features, we focused on the specific contribution of ALS astrocytes in this aspect, highlighting the known or potential molecular mechanisms by which astrocytes participate in increasing the extracellular glutamate level in ALS and, conversely, undergo the toxic effect of the excessive glutamate. In this scenario, astrocytes can behave as “producers” and “targets” of the high extracellular glutamate levels, going through changes that can affect themselves and, in turn, the neuronal and non-neuronal surrounding cells, thus actively impacting the ALS course. Moreover, this review aims to point out knowledge gaps that deserve further investigation.
Article
Full-text available
A novel method has been developed for the preparation of nearly pure separate cultures of astrocytes and oligodendrocytes. The method is based on (a) the absence of viable neurons in cultures prepared from postnatal rat cerebra, (b) the stratification of astrocytes and oligodendrocytes in culture, and (c) the selective detachment of the overlying oligodendrocytes when exposed to sheer forces generated by shaking the cultures on an orbital shaker for 15--18 h at 37 degrees C. Preparations appear greater than 98% pure and contain approximately 1-2 x 10(7) viable cells (20--40 mg of cell protein). Three methods were used to characterize these two culture t ypes. First, electron microscopic examination was used to identify the cells in each preparation (mixed and separated cultures of astrocytes and oligodendrocytes) and to assess the purity of each preparation. Second, two oligodendroglial cell markers, 2',3'-cyclic nucleotide 3'-phosphohydrolase (EC 3.1.4.37) and glycerol phosphate dehydrogenase (EC 1.1.1.8) were monitored. Third, the regulation of cyclic AMP accumulation in each culture type was examined. In addition to these studies, we examined the influence of brain extract and dibutyryl cAMP on the gross morphology and ultrastructure of each preparation. These agents induced astroglial process formation without any apparent morphological effect on oligodendrocytes. Collectively, the results indicate that essentially pure cultures of astrocytes and of oligodendrocytes can be prepared and maintained. These preparations should significantly aid in efforts to examine the biochemistry, physiology, and pharmacology of these two major classes of central nervous system cells.
Article
Synapsin I (protein I) is a neuron-specific phosphoprotein, which is a substrate for cAMP-dependent and Ca/calmodulin-dependent protein kinases. In two accompanying studies (De Camilli, P., R. Cameron, and P. Greengard, and De Camilli, P., S. M. Harris, Jr., W. B. Huttner, and P. Greengard, 1983, J. Cell Biol. 96:1337-1354 and 1355-1373) we have shown, by immunocytochemical techniques at the light microscopic and electron microscopic levels, that synapsin I is present in the majority of, and possibly in all, nerve terminals, where it is primarily associated with synaptic vesicles. In the present study we have prepared a highly purified synaptic vesicle fraction from rat brain by a procedure that involves permeation chromatography on controlled-pore glass as a final purification step. Using immunological methods, synapsin I concentrations were determined in various subcellular fractions obtained in the course of vesicle purification. Synapsin I was found to copurify with synaptic vesicles and to represent approximately 6% of the total protein in the highly purified synaptic vesicle fraction. The copurification of synapsin I with synaptic vesicles was dependent on the use of low ionic strength media throughout the purification. Synapsin I was released into the soluble phase by increased ionic strength at neutral pH, but not by nonionic detergents. The highly purified synaptic vesicle fraction contained a calcium-dependent protein kinase that phosphorylated endogenous synapsin I in its collagenase-sensitive tail region. The phosphorylation of this region appeared to facilitate the dissociation of synapsin I from synaptic vesicles under the experimental conditions used.
Article
Brain-specific Na+-dependent inorganic phosphate cotransporter (BNPI) was recently reported to serve as a vesicular glutamate transporter (VGluT), and was renamed VGluT1 (Bellocchio et al. [ 2000] Science 289:957–960; Takamori et al. [2000] Nature 407:189–194). Ahead of these reports, cDNA encoding another brain-specific inorganic phosphate transporter, which showed 82% amino acid identity to VGluT1, was cloned and designated differentiation-associated Na+-dependent inorganic phosphate cotransporter (DNPI; Aihara et al. [2000] J Neurochem 74:2622–2625). In the present study, we produced a specific antibody against a C-terminal portion of DNPI, and studied the immunohistochemical localization of DNPI in the rat cerebral cortex in comparison with that of VGluT1. DNPI immunoreactivity was enriched in neuropil of layers I and IV and to a lesser extent in the upper portion of layer VI of the cerebral neocortex, whereas VGluT1 immunoreactivity was distributed more evenly in neuropil of the neocortex. Electron microscopic observation revealed that both DNPI and VGluT1 immunoreactivities were mainly located on synaptic vesicles in nerve terminals which made asymmetrical contacts in the neocortex. Furthermore, neither DNPI nor VGluT1 immunoreactivity in the neocortex was colocalized with gamma aminobutyric acid (GABA)ergic axon terminal markers, immunoreactivity for glutamic acid decarboxylase or vesicular GABA transporter. Neuronal depletion in the ventrobasal thalamic nuclei produced by the kainic acid injection resulted in a clear reduction of DNPI immunoreactivity in layers I, IV, and VI of the somatosensory cortex. These results indicate that DNPI is located on the membrane of synaptic vesicles in thalamocortical axon terminals, and that it may be a candidate for VGluT of thalamocortical glutamatergic neurons. J. Comp. Neurol. 435:379–387, 2001. © 2001 Wiley-Liss, Inc.
Article
It had been thought that quantal size in synaptic transmission is invariable. Evidence has been emerging, however, that quantal size can be varied under certain conditions. We present evidence that alteration in vesicular [3H]l-glutamate (Glu) content within the synaptosome (a pinched-off nerve ending preparation) leads to a change in the amount of exocytotically released [3H]Glu. We found that Rose Bengal, a polyhalogenated fluorescein derivative, is a quite potent membrane-permeant inhibitor (Ki = 19 nm) of glutamate uptake into isolated synaptic vesicles. This vesicular Glu uptake inhibition was achieved largely without affecting H+-pump ATPase. We show that various degrees of reduction elicited by Rose Bengal in [3H]Glu in synaptic vesicles inside the synaptosome result in a corresponding decrease in the amount of [3H]Glu released in a depolarization- (induced by 4-aminopyridine) and Ca2+-dependent manner. In contrast, fluorescein, the halogen-free analog of Rose Bengal, which is devoid of inhibitory activity on vesicular [3H]Glu uptake, failed to change the amount of exocytotically released [3H]Glu. These observations suggest that glutamate synaptic transmission could be altered by pharmacological intervention of glutamate uptake into synaptic vesicles in the nerve terminal, a new mode of synaptic manipulation for glutamate transmission.
Article
We have analyzed expression of a gene encoding a brain-specific Na+-dependent inorganic phosphate cotransporter (DNPI), which was recently cloned from human brain, in rat forebrain using in situ hybridization. The expression of DNPI mRNA showed a widespread but highly heterogeneous pattern of distribution in the forebrain, where hybridization signals were observed in neurons but not in any other types of cells. Neurons expressing the mRNA were far more numerous in the diencephalon than in the telencephalon. In the thalamus, a number of neurons with high levels of signals were localized to all nuclei of the dorsal thalamus, habenular nuclei and subthalamic nucleus, but not the reticular nucleus and zona incerta. Moderate signal levels were seen in many neurons throughout the hypothalamus, particularly the ventromedial, paraventricular, supraoptic and arcuate nuclei, lateral hypothalamic area and mammillary complex. In contrast, expression of DNPI mRNA in the telencephalon was generally at a low level and occurred locally in some restricted regions within the neocortex, retrosplenial cortex, piriform cortex, olfactory regions, hippocampal formation and medial amygdaloid nucleus. The present results suggest that DNPI functions in heterogeneous neuron populations as a neuron-specific Na+-dependent inorganic phosphate cotransport system predominantly expressed in the diencephalon of the rat.
Article
The recent discovery that the neurotransmitter glutamate can trigger actively propagating Ca2+ waves in the cytoplasm of cultured astrocytes suggests the possibility that synaptically released glutamate may trigger similar Ca2+ waves in brain astrocytes in situ. To explore this possibility, we used confocal microscopy and the Ca2+ indicator fluo-3 to study organotypically cultured slices of rat hippocampus, where astrocytic and neuronal networks are intermingled in their normal tissue relationships. We find that astrocytic Ca2+ waves are present under these circumstances and that these waves can be triggered by the firing of glutamatergic neuronal afferents with latencies as short as 2 s. The Ca2+ waves closely resemble those previously observed in cultured astrocytes: they propagate both within and between astrocytes at velocities of 7-27 microns/s at 21 degrees C. The ability of tissue astrocyte networks to respond to neuronal network activity suggests that astrocytes may have a much more dynamic and active role in brain function than has been generally recognized.
Article
Various membrane ATPases have been tested for their sensitivity to bafilomycin A1, a macrolide antibiotic. F1F0 ATPases from bacteria and mitochondria are not affected by this antibiotic. In contrast, E1E2 ATPases--e.g., the K+-dependent (Kdp) ATPase from Escherichia coli, the Na+,K+-ATPase from ox brain, and the Ca2+-ATPase from sarcoplasmic reticulum--are moderately sensitive to this inhibitor. Finally, membrane ATPases from Neurospora vacuoles, chromaffin granules, and plant vacuoles are extremely sensitive. From this we conclude that bafilomycin A1 is a valuable tool for distinguishing among the three different types of ATPases and represents the first relatively specific potent inhibitor of vacuolar ATPases.