ArticlePDF Available

Molecular Signalling Mediating the Protective Effect of A1 Adenosine and mGlu3 Metabotropic Glutamate Receptor Activation against Apoptosis by Oxygen/Glucose Deprivation in Cultured Astrocytes

Authors:

Abstract and Figures

Astrocyte death may occur in neurodegenerative disorders and complicates the outcome of brain ischemia, a condition associated with high extracellular levels of adenosine and glutamate. We show that pharmacological activation of A(1) adenosine and mGlu3 metabotropic glutamate receptors with N(6)-chlorocyclopentyladenosine (CCPA) and (-)2-oxa-4-aminocyclo-[3.1.0]hexane-4,6-dicarboxylic acid (LY379268), respectively, protects cultured astrocytes against apoptosis induced by a 3-h exposure to oxygen/glucose deprivation (OGD). Protection by CCPA and LY379268 was less than additive and was abrogated by receptor blockade with selective competitive antagonists or pertussis toxin. Both in control astrocytes and in astrocytes exposed to OGD, CCPA and LY379268 induced a rapid activation of the phosphatidylinositol-3-kinase (PI3K) and extracellular signal-regulated kinases 1 and 2 (ERK1/2)/mitogen-activated protein kinase (MAPK) pathways, which are known to support cell survival. In cultures exposed to OGD, CCPA and LY379268 reduced the activation of c-Jun N-terminal kinase and p38/MAPK, reduced the levels of the proapoptotic protein Bad, increased the levels of the antiapoptotic protein Bcl-X(L), and were highly protective against apoptotic death, as shown by nuclear 4'-6-diamidino-2-phenylindole staining and measurements of caspase-3 activity. All of these effects were attenuated by treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) and 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002), which inhibit the MAPK and the PI3K pathways, respectively. These data suggest that pharmacological activation of A(1) and mGlu3 receptors protects astrocytes against hypoxic/ischemic damage by stimulating the PI3K and ERK1/2 MAPK pathways.
Content may be subject to copyright.
Molecular Signalling Mediating the Protective Effect of A
1
Adenosine and mGlu3 Metabotropic Glutamate Receptor
Activation against Apoptosis by Oxygen/Glucose Deprivation in
Cultured Astrocytes
Renata Ciccarelli, Iolanda D’Alimonte, Patrizia Ballerini, Mariagrazia D’Auro, Eleonora Nargi,
Silvana Buccella, Patrizia Di Iorio, Valeria Bruno, Ferdinando Nicoletti, and Francesco Caciagli
Department. of Biomedical Sciences, “G. D’Annunzio” University, Chieti, Italy (R.C., I.D.A., P.B., M.D.A., E.N., S.B., P.D.I.,
F.C.); and Department of Human Physiology and Pharmacology, University of Rome “La Sapienza,” Rome, Italy, and
Department of Neuropharmacology, I.N.M. Neuromed, Pozzilli, Italy (V.B., F.N.)
Received October 11, 2006; accepted February 9, 2007
ABSTRACT
Astrocyte death may occur in neurodegenerative disorders and
complicates the outcome of brain ischemia, a condition as-
sociated with high extracellular levels of adenosine and glu-
tamate. We show that pharmacological activation of A
1
adenosine and mGlu3 metabotropic glutamate receptors
with N
6
-chlorocyclopentyladenosine (CCPA) and ()2-oxa-
4-aminocyclo-[3.1.0]hexane-4,6-dicarboxylic acid (LY379268),
respectively, protects cultured astrocytes against apoptosis
induced by a 3-h exposure to oxygen/glucose deprivation
(OGD). Protection by CCPA and LY379268 was less than ad-
ditive and was abrogated by receptor blockade with selective
competitive antagonists or pertussis toxin. Both in control as-
trocytes and in astrocytes exposed to OGD, CCPA and
LY379268 induced a rapid activation of the phosphatidylinosi-
tol-3-kinase (PI3K) and extracellular signal-regulated kinases 1
and 2 (ERK1/2)/mitogen-activated protein kinase (MAPK) path-
ways, which are known to support cell survival. In cultures ex-
posed to OGD, CCPA and LY379268 reduced the activation of
c-Jun N-terminal kinase and p38/MAPK, reduced the levels of the
proapoptotic protein Bad, increased the levels of the antiapoptotic
protein Bcl-X
L
, and were highly protective against apoptotic
death, as shown by nuclear 4-6-diamidino-2-phenylindole stain-
ing and measurements of caspase-3 activity. All of these effects
were attenuated by treatment with 1,4-diamino-2,3-dicyano-1,4-
bis(methylthio)butadiene (U0126) and 2-(4-morpholinyl)-8-phenyl-
1(4H)-benzopyran-4-one hydrochloride (LY294002), which inhibit
the MAPK and the PI3K pathways, respectively. These data sug-
gest that pharmacological activation of A
1
and mGlu3 receptors
protects astrocytes against hypoxic/ischemic damage by stimu-
lating the PI3K and ERK1/2 MAPK pathways.
Astrocytes, the most abundant glial cell types in the brain,
provide metabolic and trophic support to neurons by several
mechanisms that include the clearance of ions and environ-
mental toxins, the supply of energy substrates, and the pro-
duction of trophic factors, and modulate synaptic activity
(Volterra and Meldolesi, 2005). Impairments in these func-
tions critically affect neuronal survival.
Recent studies have shown that ischemic and inflamma-
tory insults induce astrocyte apoptotic death, and this con-
tributes to the pathophysiology of short- and long-term neu-
rodegenerative disorders (Takuma et al., 2004). Apoptotic
astrocytes are found in Alzheimer’s disease (Kobayashi et al.,
This research was supported by funds (to R.C. and F.C.) from the Centre of
Excellence on Aging of the University of Chieti and the Italian Ministry of
Education, University and Research.
R.C. and I.D.A. contributed equally to this article.
Article, publication date, and citation information can be found at
http://molpharm.aspetjournals.org.
doi:10.1124/mol.106.031617.
ABBREVIATIONS: CCPA, N
6
-chlorocyclopentyladenosine; ASK1, apoptosis-signal-regulating kinase 1; DAPI, 4-6-diamidino-2-phenylindole;
DMEM, Dulbecco’s modified Eagle’s medium; DPCPX, 1,3-dipropyl-8-cyclopentyl xanthine; ERK1/2, extracellular signal-regulated kinases 1 and
2; HRP, horseradish peroxidase; JNK, c-Jun N-terminal kinase; LY294002, 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride;
LY341495, (2S,1S,2S)-2(9-xanthylmethyl)-2-(2-carboxycyclo-propyl)glycine; LY379268, ()2-oxa-4-aminocyclo-[3.1.0] hexane-4,6-dicarboxylic
acid; MAPK, mitogen-activated protein kinase; mGluR, metabotropic glutamate receptor; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-
phenyl)-2-(4-sulfophenyl)-2H-tetrazolium; PBS, phosphate-buffered saline; PI3K, phosphatidylinositol-3 kinase; OGD, oxygen glucose depriva-
tion; PTX, pertussis toxin; PKB, protein kinase B; U0126, 1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)-butadiene; ANOVA, analysis of
variance; RFLU, relative fluorescence units; MEK, mitogen-activated protein kinase kinase; solvent A, KH
2
PO
4
and tetrabutylammonium phos-
phate; solvent B, methanol plus KH
2
PO
4
and tetrabutylammonium phosphate.
0026-895X/07/7105-1369–1380$20.00
M
OLECULAR PHARMACOLOGY Vol. 71, No. 5
Copyright © 2007 The American Society for Pharmacology and Experimental Therapeutics 31617/3200018
Mol Pharmacol 71:1369–1380, 2007 Printed in U.S.A.
1369
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
2002), ischemic demyelinating lesions in vascular dementia
(Tomimoto et al., 1997), and in the gray matter of frontotem-
poral dementia (Martin et al., 2000). Moreover, astrocyte
damage precedes neuronal death in the spinal cord of mice
carrying mutations of type-1 superoxide dismutase that are
typically found in amyotrophic lateral sclerosis (Bruijn et al.,
1997). Hence, drugs with potential for use in the treatment of
neurodegenerative disorders may target membrane receptors
that support glial cell survival. We have focused on some
adenosine and glutamate receptors that are known to be
expressed by astrocytes and regulate several functions of
glial cells, including the production of trophic factors (Bruno
et al., 1998; Ciccarelli et al., 1999). Activation of A
1
adenosine
receptors produces neuroprotective effects (Ribeiro et al.,
2002) and protects human vascular endothelial cells against
apoptosis induced by low concentrations of ethanol (Liu et al.,
2002); however, whether these receptors regulate processes
of death/survival in glial cells survival is unknown. In con-
trast, deleterious effects are induced by the activation of
other adenosine receptor subtypes, and induction of astrocyte
apoptosis by adenosine is mediated by A
3
receptors (Appel et
al., 2001; Di Iorio et al., 2002).
Glutamate activates both ionotropic (
-amino-3-hydroxy-5-
methyl-4-isoxazolepropionic acid, N-methyl-
D-aspartate, and
kainate) and metabotropic (mGlu1 to mGlu8) receptors. As-
trocytes mainly express
-amino-3-hydroxy-5-methyl-4-isox-
azolepropionic acid/kainate, mGlu3, and mGlu5 receptors,
although other subtypes are occasionally found. Activation of
glial mGlu3 receptors protects neighbor neurons in cultured
cortical cells through a paracrine mechanism mediated by
the production of transforming growth-factor
(Bruno et al.,
1998), and the presence of astrocytes is required for the
neuroprotective effects of mGlu2/3 receptor agonists
(D’Onofrio et al., 2001). The role of mGlu3 receptors in the
regulation of glial cell survival is also unknown. It is note-
worthy that A
1
and mGlu2/3 receptors are both linked to a G
i
protein and functionally interact in modulating glutamate
release from nerve endings (Di Iorio et al., 1996).
The study of these two receptors is now facilitated by the
availability of potent and selective ligands. A
1
receptors are
activated by N
6
-chlorocyclopentyl-ADO (CCPA) and antago
-
nized by 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) (Jacob-
son and Gao, 2006), whereas mGlu3 receptors are activated
by LY379268 and antagonized by LY341495 (Schoepp et al.,
1999). Using these drugs, we now report that activation of
both receptors protects cultured astrocytes against apoptotic
death and that this effect is mediated by the activation of the
phosphatidylinositol-3 kinase (PI3K) and extracellular sig-
nal-regulated kinase (ERK) 1/2/mitogen-activated protein ki-
nase (MAPK) pathways.
Materials and Methods
Materials. Poly(D-lysine), L-leucine methyl ester, and pertussis
toxin (PTX) were supplied by Sigma (Sigma-Aldrich, Milan, Italy),
whereas LY294002, LY341495, and DPCPX were from Tocris (Bris-
tol, UK). U0126 was purchased from Calbiochem (San Diego, CA),
and CCPA was from Research Biochemical Incorporated (Sigma-
Aldrich). LY379268 was kindly provided by Eli Lilly (Indianapolis,
IN). Disposable materials for tissue cultures were supplied from
Nalge Nunc (Mascia Brunelli, Milan, Italy). Culture medium, anti-
biotics, and serum were from Invitrogen (Milan, Italy). All other
chemicals were of analytical grade or were the best commercially
available.
Cell Culture and Treatments. Primary cultures of rat astro-
cytes were prepared from neonatal rats 2 to 4 days after birth as
described previously (Di Iorio et al., 2004). Cerebral cortices were
collected in growth medium [high-glucose Dulbecco modified Eagle’s
medium (DMEM) supplemented with 10% fetal calf serum and 1%
penicillin/streptomycin (10,000 U/ml penicillin G sodium and 10,000
g/ml streptomycin sulfate in 0.85% saline)]. Then the tissue was
washed in phosphate-buffered saline (PBS), cut in small fragments,
and digested with 0.025% trypsin/0.04% EDTA solution in PBS for
20 min at 37°C. The cells were then dissociated in 0.01% DNase
solution in growth medium for 10 min at 37°C and centrifuged at
1000 rpm for 10 min. The pellet was resuspended in growth medium
containing 5 mM
L-leucine methyl ester to constrain microglia con-
tamination. Cells, seeded on poly(
D-lysine)-coated T75 flasks, were
grown in this medium for the first 24 h and were then maintained in
an identical medium without leucine methyl ester. The medium was
replaced every 3 to 4 days. At the 7th and then at the 13th day in
vitro, cells were shaken for3hat80rpmonaplate shaker to
minimize attachment and hence microglial contamination of the
cultures. Astrocytes were detached from the culture flasks by treat-
ment (5–10 min at 37°C) with 0.025% trypsin/0.04% EDTA. Astro-
cytes were replated onto poly(
D-lysine)-coated 100-mm dishes at a
concentration of 2 10
6
cells/dish for Western blot analyses or they
were plated onto poly(
D-lysine)-coated round-glass coverslips 13
mm) at a concentration of 3 10
4
cells/coverslip and 96-multiwell
plate at a concentration of 1 10
4
cells/well for the apoptosis/
viability experiments. In experiments in which enzyme inhibitors or
receptor antagonists were tested, astrocytes were pretreated with
various agents for 30 min before the addition of CCPA or LY379268,
except for the ADP-ribosylating factor of the inhibitory guanosine
nucleotide binding protein (G
i
), PTX, which was added overnight (16
h). These treatments included the following: selective inhibitors of
PI3K, LY294002, and MAPK kinase U0126; and the selective antag-
onists of the A
1
adenosine receptor, DPCPX, and of the mGlu2/3
receptor, LY341495.
Oxygen Glucose Deprivation Protocol. Apoptosis was induced
in cultured astrocytes by exposing cells to a combined deprivation of
oxygen and glucose (OGD). Twenty-four hours after replating, cells
were serum-starved for a further 24 h. Then a glucose-free bicarbon-
ate-buffered DMEM (Sigma-Aldrich) was added to the cultures after
a gentle cell washing with the same buffer. This medium was bub-
bled previously with 95% N
2
/5% CO
2
at 3 l/min for 5 min and
prewarmed at 37°C. Hypoxia was induced by placing cells in a
humidified, sealed chamber (Billups-Rothenberg, Del Mar, CA) at
37°C, which was flushed with 95% N
2
/5% CO
2
for 5 min. In this
condition, all but 0.3% oxygen tension could be removed, as indicated
byapO
2
meter (oxygen analyzer OM-11; Beckman Coulter, Milan,
Italy). At the end of the OGD period (3 h), cultures were returned to
standard condition for the indicated periods. In each experiment,
cultures exposed to OGD were always compared with normoxic con-
trols, supplied with DMEM containing glucose, and maintained in
standard incubation condition.
Evaluation of Apoptosis. DNA fragmentation was evaluated
histochemically by 4-6-diamidino-2-phenylindole (DAPI) staining
(Roche Molecular Biochemicals, Mannheim, Germany) and fluores-
cence microscopy. Cells were seeded onto poly(
D-lysine)-coated glass
coverslips, and part of them was exposed to OGD after 24-h serum
starvation in the presence or absence of CCPA or LY379268, for 3 h
in a glucose-free DMEM without fetal calf serum. Cells were then
maintained in normal DMEM without serum, like control cells (not
subjected to OGD), and then astrocytes were fixed with 3.7% para-
formaldehyde in PBS for 25 min at room temperature and then
incubated with 70% ethanol for 15 min at room temperature. DAPI
Antifade ES solution (0.125
g/ml) was added for 5 min at room
temperature to fixed astrocytes. Observations were carried out using
a fluorescence microscope (Leica DMRXA2) (excitation, 358 nm;
1370 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
emission, 461 nm). The number of apoptotic (i.e., showing frag-
mented nuclei with condensed chromatin) and viable astrocytes was
counted in five fixed fields/coverslip of up to five separate cultures.
The percentage of apoptotic cells was calculated as follows: percent-
age of apoptotic cells (total number of cells with apoptotic nuclei/
total number of counted cells) 100.
MTS Assay. The number of viable cells was determined using the
CellTiter 96 AQ
ueous
One Solution Cell Proliferation Assay (Promega
Corporation, Madison, WI) according to the manufacturer’s instruc-
tions. In brief, cell cultures (1 10
4
cells/well) were added with 20
l
of CellTiter 96 AQ
ueous
One Solution Reagent containing a tetrazo
-
lium compound, 3-(4,5–dimethylthiazol-2-yl)-5-(3-carboxymethoxy-
phenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS), and an electron-
coupling reagent (phenazine ethosulfate). The plate was incubated at
37°C for2hinahumidified atmosphere. The reduction of MTS in the
presence of cellular dehydrogenases yielded formazan crystals at the
bottom of the plate. The absorbance was measured at 490 nm using
a microtiter plate reader (Spectracount; PerkinElmer Life and Ana-
lytical Sciences, Boston, MA).
Western Blot Analysis. Western blot analysis was used to detect
phosphorylated extracellular signal-regulated kinases 1 and 2
(ERK1/2), p38 MAPK, c-Jun N-terminal kinase (JNK), Akt/protein
kinase B (PKB), apoptosis signal-regulating kinase 1 (ASK1), and
Bad protein as well as procaspase 3, Bad, and Bcl-X
L
protein content.
Cultured astrocytes were serum-deprived for 24 h before pharmaco-
logical treatments (as reported in the figures). At the end of drug
incubation, astrocytes were washed twice with ice-cold PBS and then
harvested at 4°C in a lysis buffer (25 mM Tris buffer, pH 7.4,
containing 150 mM NaCl, 100
M sodium orthovanadate, 1.5 mM
MgCl
2
, 1.0 mM EDTA, 1.0 mM EGTA, 1% Nonidet P-40, 10% glyc
-
erol, 1 mM phenylmethylsulfonyl fluoride, 5
g/ml leupeptin, and 5
g/ml aprotinin). Cells were disrupted by sonication and then were
centrifuged at 14,000 rpm for 5 min at 4°C. Aliquots (20
l) were
removed from the supernatants for the determination of protein
concentration by the Bio-Rad method. Samples were diluted in SDS-
bromphenol blue buffer and boiled for 5 min. Cell lysates were
separated on 12% SDS-polyacrylamide gels and electrophoretically
transferred to a polyvinylidene fluoride membrane (Bio-Rad Labora-
tories, Milan, Italy).
Membranes were incubated overnight at 4°C with specific primary
antibodies [polyclonal rabbit phospho-ERK1/2, phospho-Akt, phos-
pho-p38, phospho-JNK, phospho-ASK1 (Ser83), and phospho-Bad
(Ser112 or Ser136)] from Cell Signaling Technology (Danvers, MA)
diluted 1:1000, or with polyclonal rabbit anticaspase-3, anti-Bad, or
anti-Bcl-X
L
(final dilution 1:200; Santa Cruz Biotechnologies, Santa
Cruz, CA). Membranes were then exposed to a secondary antibody
for1hatroom temperature [donkey anti-rabbit horseradish perox-
idase (HRP)-conjugated; GE Healthcare, Milan, Italy]. To confirm
that equal amounts of protein were loaded in each lane, the mem-
branes were incubated in stripping buffer (62.5 mM Tris-HCl, pH
6.7, 2% SDS, and 100 mM
-mercaptoethanol) at 50°C for 30 min to
remove the primary/secondary antibody complex. The blots were
then reprobed with nonphosphorylated form of the antibodies men-
tioned above (dilution, 1:1000; Cell Signaling) or with goat polyclonal
anti-
actin (dilution 1:100; Santa Cruz Biotechnologies) (incubation,
1 h at room temperature). Membranes were then exposed to a
secondary antibody for1hatroom temperature (donkey anti-
rabbit HRP-conjugated from GE Healthcare, or donkey anti-goat
HRP-conjugated from Santa Cruz Biotechnologies, respectively, both
diluted 1:2500), according to the manufacturer’s instructions. Immu-
nocomplexes were visualized using the enhancing chemilumines-
cence detection system (GE Healthcare). Densitometric analysis was
performed for the quantification of the immunoblots using the
Molecular Analyst System (Bio-Rad Laboratories) program.
Assay of Caspase-3 Activity. The activity of caspase-3 was
determined using the Apo-ONE Homogenous Caspase 3/7 Assay
according to manufacturer’s instruction (Promega). Cell cultures
(1 10
4
cell/well) were added with Homogeneous Caspase Reagent
containing the fluorescent caspase substrate rhodamine 110, bis(N-
CBZ-
L-aspartyl-L-glutamyl-L-valyl-L-aspartic acid amide and were
incubated for 4 h. After shaking at 300 rpm for 30 s, the plate was
incubated at room temperature for 30 min to 18 h. The fluorescence
at different time points was measured at an excitation wavelength of
485 20 nm and an emission wavelength of 530 25 nm using a
Fig. 1. Evaluation of OGD-induced
apoptosis in rat cultured astrocytes.
Astrocytes were cultured in serum-
free medium for 24 h and then ex-
posed to OGD for 3 h. Apoptosis was
assessed by DAPI nuclear staining
(A), the MTS assay (B), and caspase-3
activity (C). Values are means
S.E.M. of three independent experi-
ments performed in duplicate. ⴱⴱ, p
0.01 versus controls (unpaired Stu-
dent’s t test). D, immunoblot analysis
of procaspase 3 in control astrocytes
and in astrocytes exposed to OGD. At
the end of OGD, cells were main-
tained in DMEM without serum for
further 9 h. Densitometric values are
means S.E.M. from three indepen-
dent experiments. ⴱⴱⴱ, p 0.001 ver-
sus controls (unpaired Student’s t
test).
A
1
and mGlu3 Receptor Antiapoptotic Effect in Astrocytes 1371
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
microtiter plate reader (Fluorocount; PerkinElmer Life and Analyt-
ical Sciences). The values were expressed as the relative fluorescence
units (RFLU) measured at each time point.
Measurements of Extracellular Adenosine and Glutamate.
To evaluate the endogenous adenosine and glutamate released from
astrocytes, culture medium was replaced with Krebs’ solution con-
taining 118.5 mM NaCl, 4.7 mM KCl, 2.5 mM CaCl
2
, 1.2 mM MgSO
4
,
1.2 mM KH
2
PO
4
, 10 mM glucose, and 25 mM NaHCO
3
equilibrated
with 95% air/5% CO
2
at 37°C (pH adjusted at 7.3–7.4). The cultures
were maintained in Krebs’ solution for 60 min to evaluate the purine
and glutamate release in a steady-state condition, and then this
medium was renewed for a further 60 min. Then, some cultures were
gently washed with glucose-free bicarbonate-buffered Krebs, placed
in the apparatus described above, and incubated for 1 h under
hypoxic-hypoglycemic conditions. The extracellular adenosine and
glutamate levels in medium samples from cells under basal condi-
tions and 1 h after OGD exposure were measured by high-perfor-
mance liquid chromatography as described previously (Di Iorio et al.,
1996). Adenine purine separation was carried out with a reverse-
phase analytical column (LiChrospher 100 RP-18 5
m; Merck,
Darmstadt, Germany). Elution was performed by applying a linear
gradient from 100% solvent A (60 mM KH
2
PO
4
and 5 mM tetrabu
-
tylammonium phosphate, pH 6.0) to 100% solvent B (30% methanol
plus 70% solvent A) for 15 min at a flow rate of 1.5 ml/min. Adenosine
Fig. 2. Molecular pathways activated
by OGD in cultured astrocytes. Cul-
tures were grown in serum-free me-
dium for 24 h and then exposed to
OGD for different times. Western blot
analyses of phosphorylated (p-)
ERK1/2 (A), p-Akt (B), p-ASK1-ser83
(B), p-JNK (C), p-p38 (D), p-Bad-
ser112 and -ser136 (F), total Bad, and
Bcl-X
L
(E) are shown. Densitometric
values are means S.E.M. from three
independent experiments. , p 0.05;
ⴱⴱ, p 0.01; ⴱⴱⴱ, p 0.001 (one-way
ANOVA plus Dunnett’s test) versus
control astrocytes.
1372 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
was revealed by absorbance at 254 nm. To evaluate extracellular
glutamate levels, samples were subjected to precolumn derivatiza-
tion with o-phthaldialdehyde/2-mercaptoethanol reagent. Glutamate
was separated on a Waters Pico/TAG column with a linear gradient
from 100% 100 mM potassium acetate, pH 7.1/methanol (80:20)
(solvent A) to 100% methanol/100 mM potassium acetate, pH 7.1
(80:20) (solvent B) and detected fluorometrically.
Statistical Analysis. All data are presented as the means
S.E.M. for a series of n experiments. Statistical analyses were per-
formed by Prism software version 3 (GraphPad Software Inc., San
Diego, CA), using unpaired unpaired Student’s t test or one-way
analysis of variance (ANOVA) followed by Dunnett’s post hoc com-
parison test. Group differences with P 0.05 were considered sta-
tistically significant. Dose- or time-response curves were calculated
by using nonlinear regression (Prism software).
Results
Apoptosis Induced by OGD in Cultured Astrocytes
Cultured astrocytes that had been maintained in serum-free
medium for 24 h were exposed to OGD for 3 h. This treatment
caused apoptotic cell death, which was detectable after 3 to 6 h
by combining microscopic analysis after DAPI nuclear staining
(Fig. 1A), the MTS assay (Fig. 1B), and measurements of
caspase 3 activity (Fig. 1C) and procaspase 3 levels by immu-
noblotting (Fig. 1D). The extent of apoptotic death reached a
plateau between 12 and 24 h after OGD exposure, when 50%
of cell nuclei bore chromatin condensation or pyknosis, MTS
activity decreased by 50%, caspase 3 activity increased by
approximately 3-fold, and procaspase 3 levels decreased by
80% (Fig. 1). OGD induced a sustained activation of the JNK
and p38/MAPK pathways, which peaked between 30 min and
2 h and could still be detected after 3 h (Fig. 2, A and B). In
contrast, OGD had no effect on the ERK1/2 MAPK pathway and
caused only a small and delayed stimulation of the PI3K path-
way, as assessed by immunoblot analysis of phosphorylated-
ERK1/2 and phosphorylated-Akt, respectively (Fig. 2, C and D).
Stimulation of PI3K was associated to a transient increase in
the levels of phosphorylated-(Ser83)/inactivated ASK1, which is
a MAP kinase kinase kinase upstream to JNK and p38 MAPK
in the death cascade (Sumbayev and Yasinska, 2005) and is
under the inhibitory control of the PI3K pathway (Kim et al.,
2001a). Finally, OGD altered the balance between pro- and
antiapoptotic members of the Bcl-2 family, causing a remark-
able increase in the Bcl-2-associated death protein, BAD, with-
out affecting the levels of the antiapoptotic factor Bcl-X
L
(Fig.
2F).
Extracellular adenosine levels were 23 6 nM under basal
conditions and 62 5 nM after 1-h exposure to OGD. Glu-
tamate levels were 0.35 0.07
M under basal conditions
and 1.5 0.22
M after 1-h exposure to OGD (means
S.E.M., n 4).
Pharmacological Activation of A
1
or mGlu3 Receptors
Protects Astrocytes against Apoptosis by
Oxygen/Glucose Deprivation
The A
1
adenosine receptor agonist CCPA (2.5–75 nM)
and/or the mGlu2/3 receptor agonist LY379268 (0.25–7.5
M) was added to the cultures 1 h before OGD exposure and
was maintained in the medium throughout OGD exposure
(i.e., for the following 3 h). Both drugs reduced the percentage
of apoptotic cells in a concentration-dependent manner (Fig.
3A), with maximal antiapoptotic effect at 30 nM CCPA and 5
M LY379268. The calculated EC
50
values were 15.4 2.3
nM for CCPA and 2.23 0.7
M for LY379268. Similar
results were obtained using the MTS assay (data not shown).
Maximal concentrations of CCPA or LY379268 reduced the
increase in caspase 3 activity by approximately 50%, as as-
Fig. 3. Protective effect of CCPA and LY379268 against astrocyte apo-
ptosis induced by OGD. In A, different concentrations of CCPA (2.5–75
nM) or LY379268 (0.25–7.5
M) were applied to the cultures 1 h before
OGD exposure. In B and C, cells were pretreated with DPCPX or
LY341495 30 min before the addition of CCPA or LY379268, respectively.
Pretreatment with PTX was carried out for 16 h. In D, concentrations of
CCPA or LY379268 corresponding to their IC
25
and IC
50
values were
applied alone or in combination 1 h before OGD. All drugs were main-
tained during the3hofOGDexposure. Apoptosis was assessed 24 h after
the beginning of the experiment by DAPI staining. Results are expressed
as the percentage of apoptotic cells and are the means S.E.M. of eight
values from four independent experiments. ⴱⴱⴱ, p 0.001 (one-way
ANOVA plus Dunnett’s test) versus control astrocytes.
A
1
and mGlu3 Receptor Antiapoptotic Effect in Astrocytes 1373
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
sessed 12 h after OGD exposure (caspase 3 activity expressed
as RFLU 10
3
70 8.1 in untreated cultures, 35 4.7 in
cultures treated with 30 nM CCPA, and 33 5.1 in cultures
treated with 5
M LY379268, respectively). The antiapo-
ptotic effect of CCPA was largely attenuated by the A
1
recep
-
tor antagonist DPCPX (100 nM) and by a pretreatment with
Fig. 4. Pharmacological activation of A
1
or mGlu3 receptors stimulates the PI3K
pathway and induces ASK1 phosphory-
lation in cultured astrocytes. Astrocytes
were starved for 24 h and then exposed
to increasing concentrations of CCPA or
LY379268 for 30 min (A) or to 30 nM
CCPA or 5
M LY379268 for different
times (B). In C, astrocytes were treated
with either the PI3K inhibitor
LY294002 or the receptor antagonists
DPCPX or LY341495 30 min before the
addition of CCPA or LY379268, whereas
pretreatment with PTX was carried out
for 16 h. Levels of phosphorylated Akt/
PKB (pAkt) and ASK1 at serine 83
(pASK1-Ser83) were determined by
Western blot analysis (60
g of proteins
was loaded per lane). Densitometric val-
ues are means S.E.M., from three in-
dependent experiments. , p 0.05; ⴱⴱ,
p 0.01; ⴱⴱⴱ, p 0.001 (one-way
ANOVA plus Dunnett’s test) versus con-
trol astrocytes.
1374 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
PTX (200 ng/ml). The antiapoptotic effect of LY379268 was
instead sensitive to the preferential mGlu2/3 receptor antag-
onist LY341495 (1
M) and to PTX (Fig. 3, B and C). We also
combined CCPA and LY379268 at concentrations close to the
IC
25
or the IC
50
values in cultures exposed to OGD. Protec
-
tion was lower than that expected if the antiapoptotic effects
of the two drugs were additive (Fig. 3D). This suggests that
the activation of A
1
and mGlu2/3 receptors converges into a
common, G
i
-mediated intracellular pathway, which is ulti
-
mately responsible for the antiapoptotic effect in cultured
astrocytes challenged with OGD.
Signaling Pathways and Downstream Effector Molecules
Mediating the Antiapoptotic Effect of A
1
and mGlu3
Receptors in Cultured Astrocytes
Effect of A
1
and mGlu3 Receptor Activation in Con
-
trol Cultures. In control cultures, addition of CCPA (5–60
nM) induced a concentration-dependent activation of the
PI3K pathway, which showed a rapid and long-lasting kinet-
ics. The increase in Akt phosphorylation was detectable at 5
min and reached a long plateau between 30 min and 4 h (Fig.
4, A and B). Stimulation was abrogated by DPCPX, PTX, and
the PI3K inhibitor LY294002 (Fig. 4C). Stimulation of the
PI3K pathway was associated with a parallel increase in
ASK1 phosphorylation, which also peaked after 30 min (Fig.
4D). CCPA also stimulated ERK1/2 phosphorylation with a
slightly different kinetics. Stimulation peaked after 5 min,
remained stable up to 4 h (Fig. 5, A and B), and was sensitive
to DPCPX, PTX, and the MEK inhibitor U0126 (Fig. 5C). It is
interesting that stimulation of the ERK1/2 pathway by CCPA
was also reduced by LY294002 (Fig. 5C), suggesting that the
PI3K pathway precedes the MAPK pathway in the cascade of
reactions triggered by A
1
receptors in astrocytes. Similar
results were obtained with the mGlu2/3 receptor agonist
Fig. 5. Activation of A
1
or mGlu3 re
-
ceptors stimulates the ERK1/2 MAPK
pathway in cultured astrocytes. As-
trocytes were starved for 24 h and
then exposed to increasing concentra-
tions of CCPA or LY379268 for 30 min
(A) or to 30 nM CCPA or 5
M
LY379268 for different times (B). In
C, astrocytes were treated with either
the MEK inhibitor U0126 or the re-
ceptor antagonists DPCPX or
LY341495 30 min before the addition
of CCPA or LY379268, whereas pre-
treatment with PTX was carried out
for 16 h. Levels of phosphorylated
ERK1/2 (pERK1/2) and total ERK1/2
were determined by Western blot
analysis (10
g of proteins was loaded
per lane). Densitometric values are
means S.E.M. from three indepen-
dent experiments. , p 0.05; ⴱⴱ, p
0.01; ⴱⴱⴱ, p 0.001 (one-way ANOVA
plus Dunnett’s test) versus control as-
trocytes.
A
1
and mGlu3 Receptor Antiapoptotic Effect in Astrocytes 1375
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
LY379268, which, however, stimulated both pathways with a
different kinetics compared with CCPA. Phosphorylation of
Akt/PI3K and ERK1/2 MAPK peaked after 60 min of expo-
sure to LY379268, whereas phosphorylation of ASK1 in-
creased linearly from 30 min to 3 h. The action of LY379268
was sensitive to the mGlu2/3 receptor antagonist LY341495
(Figs. 4 and 5). Neither CCPA nor LY379268 had any effect
on the JNK and the p38 MAPK pathways in control cultures
(data not shown). However, both drugs increased the levels of
the phosphorylated/inactivated forms of the proapoptotic pro-
tein Bad (Fig. 6, A and B) at serine residues 136 and 112,
which are phosphorylated by PI3K and ERK1/2, respectively
(Datta et al., 1997; Scheid et al., 1999). As expected, phos-
phorylation at serine 136 was prevented by the PI3K inhib-
itor LY294002, whereas phosphorylation at serine 122 was
prevented by the MEK inhibitor U0126 (Fig. 6, A and B).
CCPA and LY379268 also increased the levels of the anti-
apoptotic protein Bcl-X
L
, as assessed by Western blotting.
This effect was abolished by LY294002 and attenuated by
U0126 (Fig. 6C).
Effect of A
1
or mGlu3 Receptor Activation in Cul
-
tures Exposed to OGD. In cultures challenged with OGD,
the two receptor agonists CCPA and LY379268 retained the
ability to stimulate the MAPK and PI3K pathways and at-
tenuated the pathological activation of the JNK and p38
MAPK pathways (Fig. 7, A–D). The two drugs could still
activate Bad phosphorylation (Fig. 8, A and B), thus reducing
the increase in Bad levels caused by OGD (Fig. 8C). CCPA
and LY379268 could still enhance Bcl-X
L
levels in cultures
exposed to OGD (Fig. 8D). Both effects were largely attenu-
ated by the PI3K inhibitor LY294002 and the MEK inhibitor
U0126 (Fig. 8, C and D). Finally, U0126 and LY294002
Fig. 6. Pharmacological activation of A
1
or
mGlu3 receptors promotes Bad phosphorylation
and increases the cytosolic content of the anti-
apoptotic protein Bcl-X
L
. Astrocytes were
starved for 24 h and then treated with 30 nM
CCPA or 5
M LY379268 for 1 (A and B) or 4 h
(C). At the end of the treatment, cells were
switched into fresh serum-free medium for addi-
tional 2 h. In some experiments, astrocytes were
treated with either LY294002 or U0126 30 min
before the addition of CCPA or LY379268. Levels
of phosphorylated Bad and levels of Bcl-X
L
were
examined by Western blotting (50
g of proteins
was loaded per lane). Values are means S.E.M.
from three independent experiments. ⴱⴱ, p
0.01; ⴱⴱⴱ, p 0.001(one-way ANOVA plus Dun-
nett’s test) versus control astrocytes.
1376 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
prevented the antiapoptotic effect of CCPA and LY379268 in
cultures exposed to OGD, as assessed by DAPI staining and
caspase 3 activity (Fig. 9, A and B).
Discussion
Apoptosis is a phenotype of death common to virtually all
cell types during development, senescence, and a variety of
pathological conditions. In the central nervous system, most
studies have focused on mechanisms regulating apoptotic
death in neurons as potential targets for protective agents in
neurodegenerative disorders. We switched the attention to
astrocytes because astrocyte death also occurs in neurode-
generative disorders and may have a profound impact on
synaptic transmission and neuronal viability (Takuma et al.,
2004). We challenged cultured astrocytes by OGD in an at-
tempt to mimic cell damage occurring under hypoxic/isch-
emic conditions (Giffard and Swanson, 2005). A 3-h exposure
to OGD induced approximately 50% of cell death under our
conditions. This extent of death allowed a reliable assess-
ment of the underlying mechanisms and, at the same time,
was considered optimal for the identification of protective
strategies, which could have been obscured by a more severe
insult.
OGD induced a number of processes that are causally
related to apoptotic death, such as the activation of the p38
MAPK and JNK pathways and an increase in the levels of
Bad (Chen et al., 2005; Sumbayev and Yasinska, 2005). The
increase in Bad levels was not apparently related to a reduc-
tion of protein phosphorylation. We rather observed a tran-
sient increase in Bad phosphorylation in cultures exposed to
OGD, which may represent a compensatory mechanism
aimed at avoiding excessive increases in Bad levels.
Our major finding was that pharmacological activation of
A
1
or group II mGlu receptors was highly protective against
astrocyte death. Both types of receptors have an established
protective function against neuronal damage in a variety of
in vitro and in vivo models of neurodegenerative disorders
(Bruno et al., 2001; Ribeiro et al., 2002; Chong et al., 2003).
At least a component of neuroprotection is mediated by A
1
and mGlu3 receptors present in astrocytes, which control the
production of neurotrophic factors such as transforming
growth factor-
and nerve growth factor (Bruno et al., 1998;
Ciccarelli et al., 1999; D’Onofrio et al., 2001). Thus, pharma-
cological activation of A
1
and mGlu3 receptors may provide a
dual strategy of protection limiting the death of neurons and
astrocytes at the same time.
CCPA and LY379268 have nanomolar affinity for A
1
and
mGlu3 receptors (Schoepp et al., 1999; Jacobson and Gao,
2006), respectively, but their intrinsic efficacy is not greater
than that of adenosine and glutamate. This suggests that A
1
and mGlu3 receptors present in astrocytes were not satu-
rated by the relatively high amounts of extracellular adeno-
sine and glutamate found during OGD. Whether glial recep-
tors are also responsive to pharmacological agonists in vivo is
Fig. 7. Pharmacological activation of A
1
or mGlu3 receptors stimulates the PI3K
and ERK1/2 MAPK pathways and re-
duces the stimulation of the p38MAPK
and the JNK pathways in cultured astro-
cytes exposed to OGD. Astrocytes were
starved for 24 h and then pretreated
with 30 nM CCPA or 5
M LY379268 1 h
before exposure to OGD in the absence or
presence of LY294002 (30
M) or U0126
(10
M) (both added 30 min before recep-
tor agonists). Levels of p-ERK1/2 (A), p-
Akt (B), p-JNK (C), and p-p38 (D) were
evaluated by Western blot analysis (20 or
50
g of proteins was loaded per lane in
A or in B–D, respectively). Values are
means S.E.M. from three independent
experiments. ⴱⴱⴱ, p 0.01 versus basal
values (unpaired Student’s t test); E, p
0.05; EE, p 0.01; EEE, p 0.001 (one-
way ANOVA plus Dunnett’s test) versus
OGD (control) values.
A
1
and mGlu3 Receptor Antiapoptotic Effect in Astrocytes 1377
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
unknown, and one should take into account that neurons
release high amounts of purines and glutamate under hy-
poxic/ischemic conditions (Franceschini et al., 2006; Franke
et al., 2006). However, it is noteworthy that at least glial
mGlu3 receptors are not present on the surface of astrocytes
facing the synaptic cleft (Shigemoto et al., 1999) and might
not be accessible to synaptic glutamate.
A combined treatment with CCPA and LY379268 at the
respective EC
25
or EC
50
values was protective to a lower
extent than that predicted if the two drugs were additive.
Colocalization and interactions between A
1
receptors and
mGlu or other neurotransmitter receptors have been de-
scribed previously (Ciruela et al., 2001; Torvinen et al.,
2002). Because group II mGlu receptor agonists to do not
influence [
3
H]CCPA binding to A
1
receptors on astrocyte
membranes (R. Ciccarelli, F. Nicoletti, and F. Caciagli, un-
published observations), it is possible that group-II mGlu and
A
1
receptors converge in activating mechanisms that are
relevant to cytoprotection. They might recruit the same pool
of G
i
proteins or converge in the activation of the MAPK or
the PI3K pathways (see below). Thus, the combination be-
tween A
1
and mGlu2/3 receptor agonists may not be partic
-
ularly helpful in in vivo models.
Searching for intracellular mechanisms that mediate the
antiapoptotic effect of A
1
and mGlu3 receptors in cultured
astrocytes, we focused on the PI3K/Akt and the ERK/MAPK
pathways. Akt/PKB is a multifunctional mediator of PI3K-
dependent signaling that promotes cell survival and exerts in
models of neuronal or astrocyte death (Dudek et al., 1997; Di
Iorio et al., 2004). ERK1/2 activation can also produce neu-
roprotection (Xia et al., 1995). Both intracellular pathways
are activated by A
1
receptor stimulation in non-neuronal
(Germack and Dickenson, 2000) and neuronal cells (Angulo
et al., 2003) and in brain tissue (Gervitz et al., 2002). Acti-
vation of mGlu2/3 receptors can also stimulate Akt/PKB and
ERK1/2 in astrocytes and in brain tissue (D’Onofrio et al.,
2001). The following observations demonstrate that A
1
and
mGlu3 receptors protect cultured astrocytes through the ac-
tivation of PI3K/Akt and ERK/MAPK pathways: 1) CPPA
and LY379268 activated both pathways in a dose- and time-
dependent manner; 2) activation persisted in cultures ex-
posed to OGD; and 3) selective inhibitors of the two pathways
largely reduced the protective activity of A
1
or mGlu3 recep
-
tor agonists in astrocytes challenged with OGD.
Akt or ERK1/2 can phosphorylate several proapoptotic pro-
teins, leading to suppression of death signals. One of these is
ASK1, which triggers an apoptogenic kinase cascade causing
the phosphorylation/activation of JNK and p38 MAPK (Sum-
bayev and Yasinska, 2005). CCPA or LY379268 induced an
early and sustained phosphorylation/inhibition of ASK1 at
serine 83, a site that is phosphorylated by PI3K/Akt (Kim et
al., 2001a). As expected, both drugs partially reduced the
activation of the stress-related JNK and p38 MAPK path-
ways in cultures challenged with OGD. This might be one of
the PI3K/Akt-dependent mechanisms whereby activation of
A
1
and mGlu3 receptors protect astrocytes against apoptosis.
In addition, CCPA and LY379268 changed the balance be-
tween the proapoptotic protein Bad and the antiapoptotic
protein Bcl-X
L
. Bad is the only member of the Bcl-2 family
whose expression is up-regulated significantly during the
Fig. 8. Pharmacological activation of
A
1
or mGlu3 receptors reduces Bad
and increases Bcl-X
L
levels in cultured
astrocytes exposed to OGD. Astrocytes
were starved for 24 h and then treated
with 30 nM CCPA or 5
M LY379268
1 h before OGD exposure. When
present, LY294002 (30
M) or U0126
(10
M) was added 30 min before re-
ceptor agonists. Levels of Bad (A),
Bcl-X
L
(B), and phosphorylated Bad at
serine 112 or 136 (C and D) were eval-
uated by Western blotting. Values are
means S.E.M. from three indepen-
dent experiments. ⴱⴱⴱ, p 0.01 versus
basal values (unpaired Student’s t
test); E, p 0.05; EE, p 0.01; EEE,
p 0.001 (one-way ANOVA plus Dun-
nett’s test) versus OGD (control) val-
ues.
1378 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
early stages of an ischemic insult in astrocytes (Chen et al.,
2005). Bad associates with Bcl-X
L
preventing Bcl-X
L
from
exerting antiapoptotic effects in ischemic astrocytes (Chen et
al., 2005). CCPA or LY379268 increased the expression of
Bcl-X
L
and reduced the OGD-induced increase in Bad levels
by phosphorylating Bad at sites targeted by PI3K/Akt and
ERK1/2. Phosphorylated Bad is inactivated and is no longer
able to counteract Bcl-X
L
antiapoptotic activity.
It is interesting that activation of the ERK1/2 MAPK path-
way by CCPA or LY379268 was greater in cultures exposed
to OGD than in control cultures, and this is noteworthy
because the ERK1/2 MAPK is considered one of the major
protective pathways in ischemic astrocytes (Chen et al.,
2005). Why OGD amplifies the activation of the ERK1/2
MAPK pathway mediated by A
1
or mGlu3 receptors is un
-
known. Part of this activation was sensitive to LY294002,
suggesting that activation of PI3K is upstream to ERK1/2
MAPK. Other mechanisms are likely to be involved. Activa-
tion of the PI3K and ERK1/2 pathways may be triggered by
the
␤␥
subunits of the G
i
protein, as reported for other G
protein-coupled receptors (Marinissen and Gutkind, 2001).
Inhibition of cAMP formation (i.e., the canonical transduc-
tion pathway coupled to A
1
and mGlu3 receptors) may also be
involved because cAMP limits membrane localization of
phosphoinositide-dependent kinase-1 (Kim et al., 2001b),
which is a direct effector kinase of Akt.
In conclusion, our data suggest that activation of A
1
and
mGlu3 receptors produces a strong prosurvival effect in as-
trocytes degenerating in response to “ischemic-like” condi-
tions. At least in culture, these receptors effectively respond
to pharmacological activation despite the large amounts of
adenosine and glutamate released during OGD. A
1
or mGlu3
receptor agonists cater the potential to exert a variety of
beneficial effects during ischemia. These drugs can protect
astrocytes and neurons at the same time and can also stim-
ulate astrocytes to produce neurotrophic/neuroprotective fac-
tors (Bruno et al., 1998; Ciccarelli et al., 1999; D’Onofrio et
al., 2001). LY379268 is a member of a growing list of brain-
permeant and highly potent mGlu2/3 receptor agonists that
are under preclinical development for the treatment of anx-
iety, drugs addiction, and other central nervous system dis-
orders (Schiefer et al., 2004; Kim et al., 2005). These drugs
might be highly effective in limiting astrocyte death in neu-
rodegenerative disorders. A systemic use of A
1
receptor ago
-
nists has long been precluded because of the occurrence of
peripheral side effects. However, innovative delivery systems
are now available that may allow a systemic use of A
1
recep
-
tor agonists in neurodegenerative disorders (Dalpiaz et al.,
2005).
References
Angulo E, Casado V, Mallol J, Canela EI, Vin˜als F, Ferrer I, Lluis C, and Franco R
(2003) A
1
adenosine receptors accumulate in neurodegenerative structures in
Alzheimer’s disease and mediate both amyloid precursor protein processing and
tau phosphorylation and translocation. Brain Pathol 13:440 451.
Appel E, Kazimirsky G, Ashkenazi E, Kim SG, Jacobson KA, and Brodie C (2001)
Roles of BCL-2 and caspase 3 in the adenosine A3 receptor-induced apoptosis. J
Mol Neurosci 17:285–292.
Bruijn LI, Becher MW, Lee MK, Anderson KL, Jenkins NA, Copeland NG, Sisodia
SS, Rothstein JD, Borchelt DR, Price DL, et al. (1997) ALS-linked SOD1 mutant
G85R mediates damage to astrocytes and promote rapidly progressive disease with
SOD1-containing inclusions. Neuron 18:327–338.
Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, and Nicoletti F (1998)
Neuroprotection by glial metabotropic glutamate receptors is mediated by trans-
forming growth factor-beta. J Neurosci 18:9594 –9600.
Bruno V, Battaglia G, Copani A, D’Onofrio M, Di Iorio P, De Blasi A, Melchiorri D,
Flor PJ, and Nicoletti F (2001) Metabotropic glutamate receptor subtypes as
targets for neuroprotective drugs. J Cereb Blood Flow Metab 21:1013–1033.
Chen XQ, Lau LT, Fung Y-WW, and Yu ACH (2005) Inactivation of Bad by site-
specific phosphorylation: the checkpoint for ischemic astrocytes to initiate or resist
to apoptosis. J Neurosci Res 79:798 808.
Chong ZZ, Kang JQ, and Maiese K (2003) Metabotropic glutamate receptors promote
neuronal and vascular plasticity through novel intracellular pathways. Histol
Histopathol 18:173–189.
Ciccarelli R, Di Iorio P, Bruno V, Battaglia G, D’Alimonte I, D’Onofrio M, Nicoletti F,
and Caciagli F (1999) Activation of A
1
adenosine or mGlu3 metabotropic glutamate
receptors enhances the release of nerve growth factor and S-100beta protein from
cultured astrocytes. Glia 27:275–281.
Ciruela F, Escriche M, Burgueno J, Angulo E, Casado V, Soloviev MM, Canela EI,
Mallol J, Chan WY, Lluis C, et al. (2001) Metabotropic glutamate 1
and adenosine
A
1
receptors assemble into functionally interacting complexes. J Biol Chem 276:
18345–18351.
Dalpiaz A, Leo E, Vitali F, Pavan B, Scatturin A, Bortolotti F, Manfredini S, Durini
E, Forni F, Brina B, et al. (2005) Development and characterization of biodegrad-
able nanospheres as delivery systems of anti-ischemic adenosine derivatives.
Biomaterials 26:1299 –1306.
Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, and Greenberg ME (1997) Akt
phosphorylation of BAD couples survival signals to the cell-intrinsic death ma-
chinery. Cell 91:231–241.
Di Iorio P, Ballerini P, Traversa U, Nicoletti F, D’Alimonte I, Kleywegt S, Werstiuk
ES, Rathbone MP, Caciagli F, and Ciccarelli R (2004) The anti-apoptotic effect of
guanosine is mediated by the activation of the PI3-kinase/Akt/PKB pathway in
cultured rat astrocytes. Glia 46:356 –368.
Di Iorio P, Battaglia G, Ciccarelli R, Ballerini P, Giuliani P, Poli A, Nicoletti F, and
Caciagli F (1996) Interaction between A1 adenosine and class II metabotropic
glutamate receptors in the regulation of purine and glutamate release from rat
hippocampal slices. J Neurochem 67:302–309.
Di Iorio P, Kleywegt S, Ciccarelli R, Traversa U, Andrew CM, Crocker CE, Werstiuk
ES, and Rathbone MP (2002) Mechanisms of apoptosis induced by purine nucleo-
sides in astrocytes. Glia 38:179 –190.
D’Onofrio M, Cuomo L, Battaglia G, Ngomba RT, Storto M, Kingston AE, Orzi F, De
Blasi A, Di Iorio P, Nicoletti F, et al. (2001) Neuroprotection mediated by glial
group-II metabotropic glutamate receptors requires the activation of the MAP
kinase and the phosphatidylinositol-3-kinase pathways. J Neurochem 78:435–445.
Dudek H, Datta SR, Franke TF, Birnbaum MJ, Yao R, Cooper GM, Segal RA, Kaplan
Fig. 9. Pharmacological inhibition of the PI3K/Akt and ERK1/2 MAPK
pathways with LY294002 and U0126 attenuates the protective activity of
CCPA and LY379268 against apoptosis in cultured astrocytes exposed to
OGD. CCPA (30 nM) and LY379268 (5
M) were applied to the cultures
1 h before OGD. When present, LY294002 (30
M) or U0126 (10
M) was
added 30 min before receptor agonists. Apoptosis was assessed 24 h later
by either DAPI staining (A) or measurements of caspase-3 activity (B).
Values are mean S.E.M. from four independent experiments. ⴱⴱⴱ, p
0.001 versus basal values (no treatment) (unpaired Student’s t test); EE,
p 0.01 versus OGD control values (one-way ANOVA plus Dunnett’s
test).
A
1
and mGlu3 Receptor Antiapoptotic Effect in Astrocytes 1379
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
DR, and Greenberg ME (1997) Regulation of neuronal survival by the serine-
threonine protein kinase Akt. Science (Wash DC) 275:661–665.
Franceschini D, Giusti P, and Skaper SD (2006) MEK inhibition exacerbates isch-
emic calcium imbalance and neuronal cell death in rat cortical cultures. Eur
J Pharmacol 553:18–27.
Franke H, Krugel U, and Illes P (2006) P2 receptors and neuronal injury. Pflueg Arch
Eur J Physiol 452:622– 644.
Germack R and Dickenson JM (2000) Activation of protein kinase B by the A
1
-
adenosine receptor in DDT
1
MF-2 cells. Br J Pharmacol 130:867– 874.
Gervitz LM, Nalbant D, Williams SC, and Fowler JC (2002) Adenosine-mediated
activation of Akt/protein kinase B in the rat hippocampus in vitro and in vivo.
Neurosci Lett 328:175–179.
Giffard RG and Swanson R (2005) Ischemia-induced programmed cell death in
astrocytes. Glia 50:299–306.
Jacobson KA and Gao Z-G (2006) Adenosine receptors as therapeutic targets. Nat
Rev Drug Discov 5: 247–264.
Kim AH, Khursigara G, Sun X, Franke TF, and Chao MV (2001a) Akt phosphory-
lates and negatively regulates apoptosis signal-regulating kinase 1. Mol Cell Biol
21:893–901.
Kim JH, Austin JD, Tanabe L, Creekmore E, and Vezina P (2005) Activation of group
II mGlu receptors blocks the enhanced drug taking induced by previous exposure
to amphetamine. Eur J Neurosci 21:295–300.
Kim S, Jee K, Kim D, Koh H, and Chung J (2001b) Cyclic AMP inhibits Akt activity
by blocking the membrane localization of PDK1. J Biol Chem 276:12864 –12870.
Kobayashi K, Hayashi M, Nakano H, Fukutani Y, Sasaki K, Shimizaki M, and
Koshino Y (2002) Apoptosis of astrocytes with enhanced lysosomal activity and
oligodendrocytes in white matter lesions in Alzheimer’s disease. Neuropathol Appl
Neurobiol 28:238 –251.
Liu J, Tian Z, Gao B, and Kunos G (2002) Dose-dependent activation of antiapoptotic
and proapoptotic pathways by ethanol treatment in human vascular endothelial
cells. Differential involvement of adenosine. J Biol Chem 277:20927–20933.
Marinissen MJ and Gutkind JS (2001) G-protein-coupled receptors and signalling
networks: emerging paradigms. Trends Pharmacol Sci 22: 368–376.
Martin JA, Craft DK, Su JH, Kim RC, and Cotman CW (2000) Astrocytes degenerate
in frontotemporal dementia: possible relation to hypoperfusion. Neurobiol Aging
22:195–207.
Ribeiro JA, Sebastia˜o AM, and de Mendonc¸a A (2002) Adenosine receptors in the
nervous system: pathophysiological implications. Prog Neurobiol (Oxford) 68:377–
392.
Scheid MP, Schubert KM, and Duronio V (1999) Regulation of BAD phosphorylation
and association with Bcl-x
L
by the MAPK/Erk kinase. J Biol Chem 274:31108
31113.
Schiefer J, Sprinken A, Puls C, Luesse HG, Milkereit A, Milkereit E, Johann V, and
Kosinski CM (2004) The metabotropic glutamate receptor 5 antagonist MPEP and
the mGluR2 agonist LY379268 modify disease progression in a transgenic mouse
model of Huntington’s disease. Brain Res 1019:246 –254.
Schoepp DD, Jane DE, and Monn JA (1999) Pharmacological agents acting at
subtypes of metabotropic glutamate receptors. Neuropharmacology 38:1431–1476.
Shigemoto R, Masugi M, and Fujimoto K (1999) Assembly-disassembly of metabo-
tropic glutamate receptor 3 and water channel aquaporin 4 in astrocyte cell
membrane. Neuropharmacology 38:A42.
Sumbayev VV and Yasinska IM (2005) Regulation of MAP kinase-dependent apo-
ptotic pathway: implication of reactive oxygen and nitrogen species. Arch Biochem
Biophys 436:406 412.
Takuma K, Baba A, and Matsuda T (2004) Astrocyte apoptosis: implications for
neuroprotection. Prog Neurobiol (Oxford) 72:111–127.
Tomimoto H, Akiguchi I, Wakita H, Suenaga T, Nakamura S, and Kimura J (1997)
Regressive changes of astroglia in white matter lesions in cerebrovascular disease
and Alzheimer’s disease patients. Acta Neuropathol 94:146 –152.
Torvinen M, Gines S, Hillion J, Latini S, Canals M, Ciruela F, Bordoni F, Staines W,
Pedata F, Agnati LF, et al. (2002) Interactions among adenosine deaminase,
adenosine A
1
receptors and dopamine D
1
receptors in stably cotransfected fibro
-
blast cell and neurons. Neuroscience 113:709 –719.
Volterra A and Meldolesi J (2005) Astrocytes, from brain glue to communication
elements: the revolution continues. Nat Rev Neurosci 6:626640.
Xia Z, Dickens M, Raingeaud J, Davis RJ, and Greenberg ME (1995) Opposing effects
of ERK and JNK-p38 MAP kinases on apoptosis. Science (Wash DC) 270:1326
1331.
Address correspondence to: Dr. Renata Ciccarelli, Department of Biomed-
ical Sciences, Section of Pharmacology, University of Chieti, Medical School,
Via dei Vestini 29, pal. B, 66013 Chieti, Italy. E-mail: r.ciccarelli@dsb.unich.it
1380 Ciccarelli et al.
at ASPET Journals on November 6, 2015molpharm.aspetjournals.orgDownloaded from
... Although, there is no information on the role of Bergmann glia in EAE, it is known that these cells have specific transcriptional signature compared to spinal cord astrocytes under physiological conditions and this dichotomy is also present in the induction of EAE [67]. Moreover, Bergmann glia is essential for neuronal metabolic function and its role in neuroprotection has also been demonstrated [68,69]. On the other hand, enhancement of A 1 R-mediated signaling is known to suppress the release of proinflammatory cytokines by astrocytes and microglia, reduce the number and chemotaxis of CD4 + T cells [70], and reduce apoptotic cell death [71][72][73]. ...
Article
Full-text available
Experimental autoimmune encephalomyelitis (EAE) is widely used animal model of multiple sclerosis (MS). The disease is characterized by demyelination and neurodegeneration triggered by infiltrated autoimmune cells and their interaction with astrocytes and microglia. While neuroinflammation is most common in the spinal cord and brainstem, it is less prevalent in the cerebellum, where it predisposes to rapid disease progression. Because the induction and progression of EAE are tightly regulated by adenosinergic signaling, in the present study we compared the adenosine-producing and -degrading enzymes, ecto-5'-nucleotidase (eN/CD73) and adenosine deaminase (ADA), as well as the expression levels of adenosine receptors A1R and A2AR subtypes in nearby areas around the fourth cerebral ventricle—the pontine tegmentum, the choroid plexus (CP), and the cerebellum. Significant differences in histopathological findings were observed between pontine tegmentum and cerebellum on the same horizontal section level. Reactive astrogliosis and massive infiltration of CD4 + cells and macrophages in CP and pontine tegmentum resulted in local demyelination. In cerebellum, there was no evidence of infiltrates, microgliosis and neuroinflammation at the same sectional level. In addition, Bergman glia showed no signs of reactive gliosis. As for adenosinergic signaling, significant upregulation of eN/CD73 was observed in all areas studied, but in association with different adenosine receptor subtypes. In CP and pons, overexpression of eN/CD73 was coupled with induction of A2AR, whereas in cerebellum, a modest increase in eN/CD73 in resident Bergman glia was accompanied by a strong induction of A1R in the same type of astrocytes. Thus, the presence of specialized astroglia and intrinsic differences in adenosinergic signaling may play a critical role in the differential regional susceptibility to EAE inflammation.
... Rights reserved. extracellular ADO signaling and soluble factors derived from microglia [108]. ...
Article
Full-text available
Neuroplasticity refers to the nervous system’s ability to adapt and reorganize its cell structures and neuronal networks in response to internal and external stimuli. In adults, this process involves neurogenesis, synaptogenesis, and synaptic and neurochemical plasticity. Several studies have reported the significant impact of the purinergic system on neuroplasticity modulation. And, there is considerable evidence supporting the role of purine nucleosides, such as adenosine, inosine, and guanosine, in this process. This review presents extensive research on how these nucleosides enhance the neuroplasticity of the adult central nervous system, particularly in response to damage. The mechanisms through which these nucleosides exert their effects involve complex interactions with various receptors and signaling pathways. Adenosine’s influence on neurogenesis involves interactions with adenosine receptors, specifically A1R and A2AR. A1R activation appears to inhibit neuronal differentiation and promote astrogliogenesis, while A2AR activation supports neurogenesis, neuritogenesis, and synaptic plasticity. Inosine and guanosine positively impact cell proliferation, neurogenesis, and neuritogenesis. Inosine seems to modulate extracellular adenosine levels, and guanosine might act through interactions between purinergic and glutamatergic systems. Additionally, the review discusses the potential therapeutic implications of purinergic signaling in neurodegenerative and neuropsychiatric diseases, emphasizing the importance of these nucleosides in the neuroplasticity of brain function and recovery.
... The study of "metamodulation" or more generically of the "receptorreceptor" interaction preferentially focused on the colocalization and functional cross-talk of metabotropic receptors Borroto-Escuela et al., 2011;Ciruela, 2012;Ferré et al., 2022;Ferraro et al., 2012;Fiorentini et al., 2008;Nicoletti et al., 2011;Torvinen et al., 2005) belonging to different groups or even different families (Ciccarelli et al., 2007;Pelassa et al., 2019). Metabotropic receptors were reported to associate in receptor heteromers, with high levels of complexity, typified by pharmacological profiles and functional activities that are qualitatively and quantitatively different from those of the single receptor (Baki et al., 2016;Dale et al., 2022;Fuxe et al., 2010;Gomes et al., 2016;González-Maeso, 2014;Maggio and Millan, 2010;Yin et al., 2014); see for the proposed nomenclature (Ferré et al., 2009). ...
Article
Full-text available
Metamodulation shifted the scenario of the central neuromodulation from a simplified unimodal model to a multimodal one. It involves different receptors/membrane proteins physically associated or merely colocalized that act in concert to control the neuronal functions influencing each other. Defects or maladaptation of metamodulation would subserve neuropsychiatric disorders or even synaptic adaptations relevant to drug dependence. Therefore, this "vulnerability" represents a main issue to be deeply analyzed to predict its aetiopathogenesis, but also to propose targeted pharmaceutical interventions. The review focusses on presynaptic release-regulating NMDA receptors and on some of the mechanisms of their metamodulation described in the literature. Attention is paid to the interactors, including both ionotropic and metabotropic receptors, transporters and intracellular proteins, which metamodulate their responsiveness in physiological conditions but also undergo adaptation that are relevant to neurological dysfunctions. All these structures are attracting more and more the interest as promising druggable targets for the treatment of NMDAR-related central diseases: these substances would not exert on-off control of the colocalized NMDA receptors (as usually observed with NMDAR full agonists/antagonists), but rather modulate their functions, with the promise of limiting side effects that would favor their translation from preclinic to clinic.
... A1ARs are highly expressed throughout brain tissue and have a high affinity for adenosine [54]. In cultured astrocytes exposed to oxygen/glucose deprivation (OGD), pharmacological activation of A1ARs protected astrocytes against hypoxic/ischemic damage [55], suggesting that activation of A1ARs play a protective role in the acute phase of IS. ...
Article
Full-text available
Pathogenesis of ischemic stroke is mainly characterized by thrombosis and neuroinflammation. Purinergic signaling pathway constitutes adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP), and adenosine (ADO). ATP is hydrolyzed to ADP and then to AMP by extracellular nucleotidase CD39; AMP is subsequently converted to adenosine by CD73. All these nucleotides and nucleosides act on purinergic receptors protecting against thrombosis and inhibit inflammation. In addition, many physical methods have been found to play a neuroprotective role through purinergic signaling. This review mainly introduces the role and potential mechanism of purinergic signalings in the treatment of ischemic stroke, so as to provide reference for seeking new treatment methods for stroke.
... However, activation of mGlu3 receptors did not boost mGlu5-mediated PI hydrolysis in the thalamus (present data), suggesting that other signaling pathways might be involved. The MAP kinase (MAPK) and phosphatidyilinositol-3-kinase (PI3K) pathways are potential candidates because both pathways are stimulated by mGlu3 receptor activation, at least in cultured astrocytes [55]. ...
Article
Full-text available
Background: Previous studies suggest that different metabotropic glutamate (mGlu) receptor subtypes are potential drug targets for treating absence epilepsy. However, no information is available on mGlu3 receptors. Objective: To examine whether (i) changes of mGlu3 receptor expression/signaling are found in the somatosensory cortex and thalamus of WAG/Rij rats developing spontaneous absence seizures; (ii) selective activation of mGlu3 receptors with LY2794193 affects the number and duration of spike-wave discharges (SWDs) in WAG/Rij rats; and (iii) a genetic variant of GRM3 (encoding themGlu3 receptor) is associated with absence epilepsy. Methods: Animals: immunoblot analysis of mGlu3 receptors, GAT-1, GLAST, and GLT-1; realtime PCR analysis of mGlu3 mRNA levels; assessment of mGlu3 receptor signaling; EEG analysis of SWDs; assessment of depressive-like behavior. Humans: search for GRM3 and GRM5 missense variants in 196 patients with absence epilepsy or other Idiopathic Generalized Epilepsy (IGE)/ Genetic Generalized Epilepsy (GGE) and 125,748 controls. Results: mGlu3 protein levels and mGlu3-mediated inhibition of cAMP formation were reduced in the thalamus and somatosensory cortex of pre-symptomatic (25-27 days old) and symptomatic (6-7 months old) WAG/Rij rats compared to age-matched controls. Treatment with LY2794193 (1 or 10 mg/kg, i.p.) reduced absence seizures and depressive-like behavior in WAG/Rij rats. LY2794193 also enhanced GAT1, GLAST, and GLT-1 protein levels in the thalamus and somatosensory cortex. GRM3 and GRM5 gene variants did not differ between epileptic patients and controls. Conclusion: We suggest that mGlu3 receptors modulate the activity of the cortico-thalamo-cortical circuit underlying SWDs and that selective mGlu3 receptor agonists are promising candidate drugs for absence epilepsy treatment.
... However, activation of mGlu3 receptors did not boost mGlu5-mediated PI hydrolysis in the thalamus (present data), suggesting that other signaling pathways might be involved. The MAP kinase (MAPK) and phosphatidyilinositol-3-kinase (PI3K) pathways are potential candidates because both pathways are stimulated by mGlu3 receptor activation, at least in cultured astrocytes [55]. ...
Article
Background Previous studies suggest that different metabotropic glutamate (mGlu) receptor subtypes are potential drug targets for the treatment of absence epilepsy. However, no information is available on mGlu3 receptors. Objective To examine whether (i) abnormalities changes of mGlu3 receptor expression/signaling are found in the somatosensory cortex and thalamus of WAG/Rij rats developing spontaneous absence seizures; (ii) selective activation of mGlu3 receptors with LY2794193 affects the number and duration of spike-wave discharges (SWDs) in WAG/Rij rats; and (iii) a genetic variant of GRM3 (encoding the mGlu3 receptor) is associated with absence epilepsy. Methods Animals: immunoblot analysis of mGlu3 receptors, GAT-1, GLAST, and GLT-1; real-time PCR analysis of mGlu3 mRNA levels; assessment of mGlu3 receptor signaling; EEG analysis of SWDs; assessment of depressive-like behavior. Humans: search for GRM3 and GRM5 missense variants in 196 patients with absence epilepsy or other IGE/GGE Idiopathic Generalized Epilepsy (IGE)/ Genetic Generalized Epilepsy (GGE) and 125,748 controls. Results mGlu3 protein levels and mGlu3-mediated inhibition of cAMP formation were reduced in the thalamus and somatosensory cortex of pre-symptomatic (25-27 days old) and symptomatic (6-7 months old) WAG/Rij rats compared to age-matched controls. Treatment with LY2794193 (1 or 10 mg/kg, i.p.) reduced absence seizures and depressive-like behavior in WAG/Rij rats. LY2794193 also enhanced GAT1, GLAST, and GLT-1 protein levels in the thalamus and somatosensory cortex. GRM3 and GRM5 gene variants did not differ between epileptic patients and controls. Conclusions We suggest that mGlu3 receptors modulate the activity of the cortico-thalamo-cortical circuit underlying SWDs and that selective mGlu3 receptor agonists are promising candidate drugs for absence epilepsy treatment.
... mGlu3R can control extracellular glutamate by increasing the expression of astroglial glutamate transporters (Aronica, Gorter, Ijlst-Keizers, et al., 2003a;Gegelashvili et al., 2000;Spampinato et al., 2018;Yao et al., 2005;Zhou et al., 2006). mGlu3R activation also protects brain from oxygen-glucose deprivation (Ciccarelli et al., 2007) and ischemia (Mastroiacovo et al., 2021) and induces the release of glial-derived neurotrophic factor (GDNF) and transforming growth factor β (TGFβ) from neurons and astrocytes, respectively (reviewed in (Spampinato et al., 2018). Our group demonstrated that astroglial mGlu3R activation by the synthetic agonist LY379268 promotes the non-amyloidogenic cleavage of amyloid precursor protein (APP) by increasing α-secretase levels and sAPPα release and reducing β-secretase expression (Durand et al., 2014). ...
Article
Full-text available
Subtype 3 metabotropic glutamate receptor (mGlu3R) displays a broad range of neuroprotective effects. We previously demonstrated that mGlu3R activation in astrocytes protects hippocampal neurons from Aβ neurotoxicity through stimulation of both neurotrophin release and Aβ uptake. Alternative‐spliced variants of mGlu3R were found in human brains. The most prevalent variant, mGlu3Δ4, lacks exon 4 encoding the transmembrane domain and can inhibit ligand binding to mGlu3R. To date, neither its role in neurodegenerative disorders nor its endogenous expression in CNS cells has been addressed. The present paper describes for the first time an association between altered hippocampal expression of mGlu3Δ4 and Alzheimer's disease (AD) in the preclinical murine model PDAPP‐J20, as well as a deleterious effect of mGlu3Δ4 in astrocytes. As assessed by western blot, hippocampal mGlu3R levels progressively decreased with age in PDAPP‐J20 mice. On the contrary, mGlu3Δ4 levels were drastically increased with aging in nontransgenic mice, but prematurely over‐expressed in 5‐month‐old PDAPP‐J20‐derived hippocampi, prior to massive senile plaque deposition. Also, we found that mGlu3Δ4 co‐precipitated with mGlu3R mainly in 5‐month‐old PDAPP‐J20 mice. We further showed by western blot that primary cultured astrocytes and neurons expressed mGlu3Δ4, whose levels were reduced by Aβ, thereby discouraging a causal effect of Aβ on mGlu3Δ4 induction. However, heterologous expression of mGlu3Δ4 in astrocytes induced cell death, inhibited mGlu3R expression, and prevented mGlu3R‐dependent Aβ glial uptake. Indeed, mGlu3Δ4 promoted neurodegeneration in neuron–glia co‐cultures. These results provide evidence of an inhibitory role of mGlu3Δ4 in mGlu3R‐mediated glial neuroprotective pathways, which may lie behind AD onset. image
... In fact, an increase in intracellular levels of cAMP is essential to initiate the differentiation process, while it can be harmful if present in the subsequent period [40], when insulin, whose pro-adipogenic activity is mainly linked to the activation of the PI3K pathway, becomes more important, being crucial mainly in the late part of the process leading to a complete adipogenic differentiation of ASCs [23]. On the other hand, it is known that the stimulation of A 1 R is generally linked to the inhibition of cAMP formation through the interaction with an alpha subunit of the Gi protein, whereas the activity of the beta and gamma subunits of the Gi protein coupled to A 1 R could cause the activation of other molecular pathways, including that of PI3K leading to Akt phosphorylation [41][42][43]. Thus, the full agonist CCPA, causing the activation of Gi proteins, would inhibit the formation of cAMP, at the same time stimulating the PI3K pathway. ...
Article
Full-text available
Adenosine A1 receptor (A1R) activation, stimulating lipogenesis and decreasing insulin resistance, could be useful for metabolic syndrome management in obese subjects. Since full A1R agonists induce harmful side-effects, while partial agonists show a better pharmacological profile, we investigated the influence of two derivatives of the full A1R agonist 2-chloro-N6-cyclopentyladenosine (CCPA), C1 and C2 behaving as A1R partial agonists in animal models, on the adipogenic differentiation of stromal/stem cells (ASCs) from human subcutaneous adipose tissue, which mainly contribute to increase fat mass in obesity. The ASCs from normal-weight subjects showed increased proliferation and A1R expression but reduced adipogenic differentiation compared to obese individual-derived ASCs. Cell exposure to CCPA, C1, C2 or DPCPX, an A1R antagonist, did not affect ASC proliferation, while mainly C2 and DPCPX significantly decreased adipogenic differentiation of both ASC types, reducing the activity of glycerol-3-phosphate dehydrogenase and the expression of PPARγ and FABP-4, all adipogenic markers, and phosphorylation of Akt in the phosphatidylinositol-3-kinase pathway, which plays a key-role in adipogenesis. While requiring confirmation in in vivo models, our results suggest that A1R partial agonists or antagonists, by limiting ASC differentiation into adipocytes and, thereby, fat mass expansion, could favor development/worsening of metabolic syndrome in obese subjects without a dietary control.
Article
Full-text available
It is commonly accepted that the role of astrocytes exceeds far beyond neuronal scaffold and energy supply. Their unique morphological and functional features have recently brough much attention as it became evident that they play a fundamental role in neurotransmission and interact with synapses. Synaptic transmission is a highly orchestrated process, which triggers local and transient elevations in intracellular Ca2+, a phenomenon with specific temporal and spatial properties. Presynaptic activation of Ca2+-dependent adenylyl cyclases represents an important mechanism of synaptic transmission modulation. This involves activation of the cAMP-PKA pathway to regulate neurotransmitter synthesis, release and storage, and to increase neuroprotection. This aspect is of paramount importance for the preservation of neuronal survival and functionality in several pathological states occurring with progressive neuronal loss. Hence, the aim of this review is to discuss mutual relationships between cAMP and Ca2+ signaling and emphasize those alterations at the Ca2+/cAMP crosstalk that have been identified in neurodegenerative disorders, such as Alzheimer's and Parkinson's disease.
Article
Full-text available
Alzheimer’s disease (AD) was first identified more than 100 years ago and, yet, aspects pertaining its origin as well as the mechanisms underlying disease progression are not well known. To this date, there is no therapeutic approach or disease modifying drug that could halt or at least delay disease progression. Until recently, glial cells were seen as secondary actors in brain homeostasis. Although this view was gradually refuted and the relevance of glial cells for the most diverse brain functions such as synaptic plasticity and neurotransmission was vastly proved, many aspects of its functioning as well as its role in pathological conditions remain poorly understood. Metabotropic glutamate receptors (mGluRs) in glial cells were shown to be involved in neuroinflammation and neurotoxicity. Besides its relevance for glial function, glutamatergic receptors are also central in the pathology of AD and recent studies have shown that glial mGluRs play a role in the establishment and progression of AD. Glial mGluRs influence AD-related alterations in Ca2+ signalling, APP processing and Aβ burden, as well as AD-related neurodegeneration. However, different types of mGluRs play different roles, depending on the cell type and brain region that is being analysed. Therefore, in this review we focus on the current understanding of glial mGluRs and their implication in AD, providing an insight for future therapeutics and identifying existing research gaps worth investigating.
Article
Full-text available
Recently, evidence has emerged that seven transmembrane G protein-coupled receptors may be present as homo- and heteromers in the plasma membrane. Here we describe a new molecular and functional interaction between two functionally unrelated types of G protein-coupled receptors, namely the metabotropic glutamate type 1α (mGlu1α receptor) and the adenosine A1 receptors in cerebellum, primary cortical neurons, and heterologous transfected cells. Co-immunoprecipitation experiments showed a close and subtype-specific interaction between mGlu1α and A1 receptors in both rat cerebellar synaptosomes and co-transfected HEK-293 cells. By using transiently transfected HEK-293 cells a synergy between mGlu1α and A1 receptors in receptor-evoked [Ca2+]i signaling has been shown. In primary cultures of cortical neurons we observed a high degree of co-localization of the two receptors, and excitotoxicity experiments in these cultures also indicate that mGlu1α and A1 receptors are functionally related. Our results provide a molecular basis for adenosine/glutamate receptors cross-talk and open new perspectives for the development of novel agents to treat neuropsychiatric disorders in which abnormal glutamatergic neurotransmission is involved.
Article
Pharmacological activation of A1 adenosine receptor with 2-chloro-N6-cyclopentyladenosine (CCPA) or mGlu3 metabotropic glutamate receptors with (2S,2′R,3′R)-2-(2′,3′-dicarboxycyclopropyl)glycine (DCG-IV) or aminopyrrolidine-2R,4R-dicarboxylate (2R,4R-APDC) enhanced the release of nerve growth factor (NGF) or S-100β protein from rat cultured astrocytes. Stimulation of release by CCPA and DCG-IV or 2R,4R-APDC was inhibited by the A1 adenosine receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine and by the mGlu2/3 receptor antagonist (2S,1′S,2′S,3′R)-2-(2′-carboxy-3′-phenylcyclopropyl)glycine (PCCG-4), respectively. Time-course studies revealed a profound difference between the release of S-100β protein and the release of NGF in response to extracellular signals. Stimulation of S-100β protein exhibited rapid kinetics, peaking after 1 h of drug treatment, whereas the enhancement of NGF release was much slower, requiring at least 6 h of A1 adenosine or mGlu3 receptor activation. In addition, stimulation of NGF but not S-100β release was substantially reduced in cultures treated with the protein synthesis inhibitor cycloheximide. In addition, a 6–8 h treatment of cultured astrocytes with A1 or mGlu3 receptor agonists increased the levels of both NGF mRNA and NGF-like immunoreactive proteins, including NGF prohormone. We conclude that activation of A1 adenosine or mGlu3 receptors produces pleiotropic effects in astrocytes, stimulating the synthesis and/or the release of protein factors. Astrocytes may therefore become targets for drugs that stimulate the local production of neurotrophic factors in the CNS, and this may provide the basis for a novel therapeutic strategy in chronic neurodegenerative disorders. GLIA 27:275–281, 1999. © 1999 Wiley-Liss, Inc.
Article
Redox processes occurring during the CO–O2 reaction and catalytic properties for CO oxidation have been examined for CuO/CeO2 and CuO/ZrCeO4 catalysts using catalytic activity tests, in situ DRIFTS and EPR spectroscopies, and CO-TPD. EPR results suggest synergetic effects in the reduction–oxidation of these systems, both copper and the support being involved in these redox processes. Both EPR and CO-TPD experiments indicate a more facile reduction by CO of the CuO/CeO2 system, mainly due to the greater reducibility of sites at the interface between the copper oxide clusters and the support for this catalyst. A greater CO oxidation activity is exhibited by the CuO/CeO2 catalyst. A comparison of both catalysts and analysis of the redox properties of the different copper entities present in the catalysts indicate that the active copper sites for CO oxidation are located on the copper oxide clusters. On the basis of the results obtained by the different techniques, it is proposed that CO oxidation in these systems follows a redox mechanism in which both the support and copper oxide clusters are simultaneously reduced or oxidized following interaction with CO or O2, respectively. The limiting step in the reaction is related to the oxidation process.
Article
The mGlu2/3 receptor agonists 4-carboxy-3-hydroxyphenylglycine (4C3HPG) and LY379268 attenuated NMDA toxicity in primary cultures containing both neurons and astrocytes. Neuroprotection was abrogated by PD98059 and LY294002, which inhibit the mitogen activated protein kinase (MAPK) and the phosphatidylinositol-3-kinase (PI-3-K) pathways, respectively. Cultured astrocytes lost the ability to produce transforming growth factor-β1 (TGF-β1) in response to mGlu2/3 receptor agonists when co-incubated with PD98059 or LY294002. As a result, the glial medium was no longer protective against NMDA toxicity. Activation of the MAPK and PI-3-K pathways in cultured astrocytes treated with 4C3HPG or LY379268 was directly demonstrated by an increase in the phosphorylated forms of ERK-1/2 and Akt. Similarly to that observed in the culture, intracerebral or systemic injections of mGlu2/3 receptor agonists enhanced TGF-β1 formation in the rat or mouse caudate nucleus, and this effect was reduced by PD98059. PD98059 also reduced the ability of LY379268 to protect striatal neurons against NMDA toxicity. These results suggest that activation of glial mGlu2/3 receptors induces neuroprotection through the activation of the MAPK and PI-3-K pathways leading to the induction of TGF-β.
Article
Apoptosis plays an important role during neuronal development, and defects in apoptosis may underlie various neurodegenerative disorders. To characterize molecular mechanisms that regulate neuronal apoptosis, the contributions to cell death of mitogen-activated protein (MAP) kinase family members, including ERK (extracellular signal-regulated kinase), JNK (c-JUN NH2-terminal protein kinase), and p38, were examined after withdrawal of nerve growth factor (NGF) from rat PC-12 pheochromocytoma cells. NGF withdrawal led to sustained activation of the JNK and p38 enzymes and inhibition of ERKs. The effects of dominant-interfering or constitutively activated forms of various components of the JNK-p38 and ERK signaling pathways demonstrated that activation of JNK and p38 and concurrent inhibition of ERK are critical for induction of apoptosis in these cells. Therefore, the dynamic balance between growth factor-activated ERK and stress-activated JNK-p38 pathways may be important in determining whether a cell survives or undergoes apoptosis.
Article
The pathogenesis of white matter lesions, which are frequently found in ischemic cerebrovascular disease and Alzheimer’s disease, remains unclear. Using light and electron microscopic immunohistochemistry for glial fibrillary acidic protein (GFAP) as a marker, the present study focused on the role of astroglia which show characteristic morphological alterations. Of 29 brains of patients with cerebrovascular disease and Alzheimer’s disease, 4 brains showed extensive swelling and vacuolation of white matter astroglia with their processes disintegrated and beaded (termed clasmatodendrosis). No such cells were observed in 6 control patients. Clasmatodendritic astroglia were not intensely eosinophilic using hematoxylin and eosin staining and included large lipophilic granules in their perikarya. These astroglia were immunoreactive for serum proteins such as immunoglobulins, fibrinogen and complement C3, C1q and C3d, as well as for proteins which are known to increase in reactive astroglia, such as vimentin, α-B crystallin, apolipoprotein-E and laminin. Double labeling for GFAP and microglial cell markers indicated that these cells were of astroglial lineage. Immunoelectron microscopy for GFAP revealed that clasmatodendritic astroglia had condensed chromatin, lysosomes and large membrane-bound osmiophilic cytoplasmic inclusions, which corresponded to the lipophilic granules observed with light microscopy. These cytochemical features collectively suggest that clasmatodendritic astroglia incorporate edema fluid and phagocytose cellular debris, and eventually degenerate as a result of cerebral edema.
Article
Selective A3 adenosine receptor agonists have been shown to induce apoptosis in a variety of cell types. In this study we examined the effects of adenosine receptor agonists selective for A1, A2A, or A3 receptors on the induction of apoptosis in primary cultures of rat astrocytes and in C6 glial cells. Treatment of the cells with the A3 receptor agonist Cl-IB-MECA (10 µM) induced apoptosis in both cell types. The effects of Cl-IB-MECA were partially antagonized by the A3 receptor-selective antagonist MRS 1191. In contrast, the A1 and A2A receptor agonists, CPA and CGS 21680, respectively, did not have significant effects on apoptosis in these cells. Cl-IB-MECA reduced the expression of endogenous Bcl-2, whereas it did not affect the expression of Bax. Overexpression of Bcl-2 in C6 cells abrogated the induction of apoptosis induced by the A3 agonist. Cl-IB-MECA also induced an increase in caspase 3 activity and caspase inhibitors decreased the apoptosis induced by the A3 agonist. These findings suggest that intense activation of the A3 receptor is pro-apoptotic in glial cells via bcl2 and caspase-3 dependent pathways.