SlideShare a Scribd company logo
1 of 10
Download to read offline
Opposed effects of lithium on the MEK-ERK pathway in neural
cells: inhibition in astrocytes and stimulation in neurons by GSK3
independent mechanisms
Rau´l Pardo,* Alberto G. Andreolotti,* Bele´n Ramos,  Fernando Picatoste* and Enrique Claro*
*Institut de Neurocie`ncies and Departament de Bioquı´mica i Biologia Molecular, Universitat Auto`noma de Barcelona
 Departamento de Fisiologı´a, Universidad de Extremadura, Spain
Summary
Lithium is widely used in the treatment of bipolar disorder, but
despite its proven therapeutic efficacy, the molecular mech-
anisms of action are not fully understood. The present study
was undertaken to explore lithium effects of the MEK/ERK
cascade of protein kinases in astrocytes and neurons. In
asynchronously proliferating rat cortical astrocytes, lithium
decreased time- and dose-dependently the phosphorylation of
MEK and ERK, with 1 mM concentrations achieving 60 and
50% inhibition of ERK and MEK, respectively, after a 7-day
exposure. Lithium also inhibited [3
H]thymidine incorporation
into DNA and induced a G2/M cell cycle arrest. In serum-
deprived, quiescent astrocytes, pre-exposure to lithium
resulted in the inhibition of cell cycle re-entry as stimulated by
the mitogen endothelin-1: under this experimental setting,
lithium did not affect the rapid, peak phosphorylation of MEK
taking place after 3–5 min, but was effective in inhibiting the
long-term, sustained phosphorylation of MEK. Lithium inhibi-
tion of the astrocyte MEK/ERK pathway was independent of
inositol depletion. Further, compound SB216763 inhibited Tau
phosphorylation at Ser396 and stabilized cytosolic b-catenin,
consistent with the inhibition of glycogen synthase kinase-3b
(GSK-3b), but failed to reproduce lithium effects on MEK and
ERK phosphorylation and cell cycle arrest. In cerebellar
granule neurons, millimolar concentrations of lithium
enhanced MEK and ERK phosphorylation in a concentration-
dependent manner, again through an inositol and GSK-3b
independent mechanism. These opposing effects in astro-
cytes and neurons make lithium treatment a promising strat-
egy to favour neural repair and reduce reactive gliosis after
traumatic injury.
Keywords: astrocytes, cell cycle arrest, ERK, GSK-3, lith-
ium, MEK.
J. Neurochem. (2003) 87, 417–426.
Lithium has been used in the treatment of bipolar mood
disorder for decades, but despite its therapeutic efficacy, the
molecular mechanisms underlying its actions remain unclear
(Jope 1999; Phiel and Klein 2001). In this regard, based on
the observation that lithium inhibits inositol monophospha-
tase and inositol polyphosphate 1-phosphatase, thereby
blocking inositol 1,4,5-trisphosphate recycling to inositol,
the inositol depletion hypothesis considered that persistent
activation of phosphoinositide phospholipase C in the
presence of lithium would lower the cellular inositol
concentration, leading eventually to the depletion of phos-
phatidylinositol 4,5-bisphosphate and the impairment of
calcium signalling (Berridge et al. 1989; Atack 1996). This
hypothesis has received some support after the finding that
lithium, carbamazepine, and valproic acid, all three drugs
used in the treatment of bipolar disorder, increase growth
cone area in sensory neurons, in a manner that was
counteracted by inositol addition, and mediated probably
by inhibition of prolyl oligopeptidase by an as yet unknown
mechanism (Williams et al. 2002).
In the past few years, however, lithium has emerged as a
remarkable neuroprotective agent: it was first shown to
protect cerebellar granule neurons from undergoing apoptotic
cell death when cultured under non-depolarizing potassium
conditions (D’Mello et al. 1994), and soon thereafter similar
observations were extended to many other apoptotic insults
Received June 4, 2003; accepted July 4, 2003.
Address correspondence and reprint requests to Enrique Claro, Institut
de Neurocie`ncies, Edifici M, Universitat Auto`noma de Barcelona,
E-08193 Bellaterra, Spain. E-mail: Enrique.Claro@uab.es
Abbreviations used: DMEM, Dulbecco’s modified Eagle’s medium;
ET-1, endothelin-1; FCS, fetal calf serum; GSK-3b, glycogen synthase
kinase-3b; RBD, Ras-binding domain of Raf-1.
Journal of Neurochemistry, 2003, 87, 417–426 doi:10.1046/j.1471-4159.2003.02015.x
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426 417
like ischaemia (Nonaka and Chuang 1998), anticonvulsants
(Nonaka et al. 1998), C2-ceramide (Centeno et al. 1998), or
b-amyloid (A´ lvarez et al. 1999), among others, where
lithium effects were independent of inositol depletion
(Centeno et al. 1998; Nonaka et al. 1998). Glycogen
synthase kinase-3b (GSK-3b) is inhibited by lithium (Klein
and Melton 1996; Stambolic et al. 1996), and this discovery
has greatly fuelled the interest in the biochemistry of this
classic therapeutic agent. In many instances, the activation of
GSK-3b taking place after disruption of the phosphatidy-
linositol 3-kinase/Akt pathway appears related to apoptotic
cell death, and it is generally acknowledged today that this
kinase constitutes the primary site of action underlying the
cytoprotective actions of lithium (Grimes and Jope 2001).
Lately, more selective GSK-3b inhibitors have been devel-
oped with potent cytoprotective effects, lending further
support to the involvement of GSK-3b in the regulation of
cell survival and apoptosis (Cross et al. 2001). The relevance
of GSK-3b inhibition in bipolar therapy, however, is still a
matter of controversy, although it is supported by the fact that
valproate, another drug used in the treatment of bipolar
disorder, has been reported to inhibit GSK-3b in vitro (Chen
et al. 1999a) and also in vivo (De Sarno et al. 2002; Hall
et al. 2002). The inhibitory action of lithium and valproate
on GSK-3 is not restricted to neuroprotection against
apoptotic insults: lithium has been shown to mimic the
action of WNT-7a, promoting axonal remodelling and
synaptic differentiation (Hall et al. 2000). This neuroplastic
action of anti-bipolar drugs may be therapeutically relevant,
as there is evidence supporting the occurrence of neuronal
loss and/or atrophy in patients with mood disorder (Drevets
et al. 1997; Ongur et al. 1998).
Inhibition of GSK-3b may not be the sole mechanism
underlying lithium-induced neuroprotection and plasticity:
the ion has been shown to potentiate Akt phosphorylation in
cerebellar neurons (Chalecka-Franaszek and Chuang 1999;
Mora et al. 2001). Also, lithium appears to modulate MEK-
ERK signalling, a pathway whose relevance in the regulation
of neuronal function, synaptic plasticity, and survival is
receiving growing attention (Grewal et al. 1999): it has been
reported recently that lithium induced the activation of MEK
and ERK in cerebellar granule neurons (Kopnisky et al.
2003) and, similarly, valproic acid promoted neurite growth
in neuroblastoma cells by a mechanism that implies activa-
tion of these mitogen-activated protein kinases (Yuan et al.
2001).
Experimental studies on the plasticity and regeneration of
the nervous system often disregard the role of glial cells as
trophic support for neurons or as a source of inflammatory
cytokines, so in the present study we compared the effects of
lithium in astrocytes and neurons. In agreement with
Kopnisky et al. (2003), we found that lithium potentiates
ERK phosphorylation in cerebelar granule neurons, but to
our surprise, it exerts opposite effects in astroglial cultures,
inhibiting sustained MEK and ERK phosphorylation, DNA
synthesis, and inducing cell cycle arrest.
Materials and methods
Materials
Dulbecco’s modified Eagle’s medium (DMEM), fetal calf serum
(FCS), penicillin, streptomycin, and glutamine were obtained from
Gibco (Rockville, MD, USA). Endothelin-1 (ET-1) was from
Alexis Corporation (San Diego, CA, USA). MEK inhibitors U0126
and PD98059, tyrphostin AG1478 and antibody against phospho-
serine 396-Tau were from Calbiochem (San Diego, CA, USA).
Antibodies against total and phosphorylated ERK, MEK, and Akt
were from Cell Signaling Technology (Beverly, MA, USA). Anti-
Ras antibody was from Oncogene (Oncogene-EMD Biosciences,
Darmstadt, Germany), antibody against b-catenin was from BD
Transduction Laboratories (Lexington, KY, USA), and antibody
against b-tubulin was from BD Pharmingen. Secondary peroxi-
dase-conjugated antibodies were from Transduction Laboratories.
[Methyl-3
H]thymidine (87.0 Ci/mmol) was from Amersham Phar-
macia Biotech (Piscataway, NJ, USA). GSK-3 inhibitor SB216763
was kindly donated by GlaxoSmithKline, Stevenage, Herts, UK.
Cell culture
Primary cultures of cerebral cortical astrocytes were prepared from
newborn (< 24 h) Sprague-Dawley rats as previously described
(Servitja et al. 2000). Cells were plated into 176 cm2
dishes
(12.5 · 106
viable cells/dish) in DMEM supplemented with 10%
FCS, 33 mM glucose, 2 mM glutamine, 50 units/mL penicillin, and
50 lg/mL streptomycin. Two hours after plating, the medium was
replaced to remove non-adherent cells. The cultures were incubated
at 37°C in a humidified atmosphere of 5% CO2/95% air. After
24 h, the medium was changed again. Before confluence, cells
were replated into polylysine-coated 6-well plates (2 · 105
cells/
well) for western blot analysis, or into 24-well plates
(2–5 · 104
cells/well) to carry out [3
H]thymidine incorporation
assays. At this time, immunocytochemical staining for glial
fibrillary acidic protein revealed that cultures consisted of 90–
95% astrocytes. In some experiments, cells were serum-starved
before being stimulated with ET-1. Primary cultures of rat cerebelar
granule cells were prepared as previously described (Masgrau et al.
2000). Briefly, cells were seeded at 2 · 106
cells/well in 6-well
plates in DMEM containing 10% FCS, 25 mM KCl, 5 mM glucose,
penicillin (50 units/mL), streptomycin (50 lg/mL), sodium pyru-
vate (0.1 mg/mL), 7 mM p-aminobenzoic acid, and 2 mM gluta-
mine. Twenty-four hours after plating, 10 lM cytosine arabinofur-
anoside was added to prevent growth of glial cells, and neurons
were maintained for 7 days.
Western blot analysis
Cells were washed twice with ice-cold phosphate-buffered saline
prior to scraping and lysis. For ERK, MEK and Akt immunoblots,
lysis buffer consisted of 50 mM Tris-HCl, pH 7.5, containing 0.1%
Triton X-100, 2 mM EDTA, 2 mM EGTA, 50 mM NaF, 10 mM
b-glycerophosphate, 5 mM sodium pyrophosphate, 1 mM sodium
orthovanadate, 0.1% (v/v) b-mercaptoethanol, 250 lM phenyl-
methylsulfonyl fluoride (PMSF), 10 lg/mL aprotinin and leupeptin.
418 Rau´l Pardo et al.
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
For immunoblots of cytoplasmic b-catenin, hypotonic lysis buffer
was 50 mM Tris-HCl, pH 7.5, with 2 mM EDTA, 2 mM EGTA,
0.5 mM NaF, 10 mM b-glycerophosphate, 5 mM sodium pyrophos-
phate, 1 mM sodium orthovanadate and the same protease inhibitors
as above. Cell lysates were centrifuged at 15 000 g for 15 min at
4°C. Equal amounts of protein (10–20 lg) were subjected to sodium
dodecylsulfate – polyacrilamide gel electrophoresis (SDS–PAGE)
and electrophoretically transferred to polyvinylidene difluoride
(PVDF) membranes (Immobilon-P, Millipore Corporation, Bedford,
MA, USA). Blots were blocked in 5%-non-fat dry milk/TBST
buffer (20 mM Tris-HCl, 0.15 M NaCl, 0.1% Tween-20) and
immunoblotted for: total ERK1/2, phosphothreonine 202/phospho-
tyrosine 204-ERK1/2, total MEK1/2, phosphoserine 217/221
MEK1/2, total Akt, phosphoserine 473 Akt, Ras, b-tubulin, or
phosphoserine 396-Tau. Blots were then labelled with anti-mouse
IgG : HRP or anti-rabbit IgG : HRP, incubated for 1 min with ECL
reagent (ECL, Amersham-Pharmacia) according to the supplier’s
instructions, exposed to Hyperfilm ECL and developed. Quantifi-
cation was performed by densitometric scaning of the film using a
GS-700 Imaging Densitometer with Multi-Analyst 1.1 software
(Bio-Rad Laboratories, Hercules, CA, USA).
[3
H]Thymidine incorporation into DNA
As an indication of mitogenic induction in astrocytes, we measured
the incorporation of [methyl-3
H]thymidine into DNA. Astrocytes
were left 3 days proliferating in FCS-containing medium with or
without lithium and/or inhibitors, and pulse-labelled with 1 lCi/mL
of [3
H]thymidine during the last 24 h. At the end of the incubation,
cells were washed with ice-cold Krebs-Henseleit buffer and treated
with ice-cold 1 M HClO4 for 30 min. The acid-precipitated material
was washed with 0.4 M HClO4, then with ethanol, and solubilized
with 0.5 M NaOH. A 100-lL aliquot was collected for liquid
scintillation counting.
Cell cycle analysis
The cell cycle distribution was determined by flow cytometric
analysis of propidium iodide-labelled cells. Briefly, astrocytes were
plated in 6-well plates and maintained in FCS-containing medium
28 h prior to treatment. After trypsinization, cells were collected,
washed twice in ice-cold PBS, and fixed in ice-cold 70% ethanol.
Cells were then washed twice with ice-cold PBS, incubated at 37°C
for 30 min with propidium iodide (20 lg/mL) in a PBS solution
containing 0.1% (v/v) Triton X-100 and 100 units/mL Rnase A, and
analyzed by flow cytometry with a Becton Dickinson FACSCalibur
flow cytometer. The results were analyzed with CellQuest and
Modfit LT Software.
Ras activation
Astrocytes were serum-starved for 2 days, and upon mitogen
stimulation cells were lysed at 4°C in a buffer containing 20 mM
HEPES, pH 7.4, 0.1 M NaCl, 1% Triton X-100, 10 mM EGTA,
40 mM b-glycerophosphate, 20 mM MgCl2, 1 mM Na3VO4, 1 mM
dithiothreitol, 10 lg/mL aprotinin, 10 lg/mL leupeptin, and 1 mM
PMSF. Lysates were incubated for 30 min with 10 lg of a GST
fusion protein including the Ras-binding domain of Raf-1 (RBD)
previously bound to glutathione-Sepharose beads (Upstate Bio-
technology, Lake Placid, NY, USA), followed by three washes
with lysis buffer. Total Ras levels in the cell lysates and GTP-
bound forms of Ras associated with GST-Raf-1-RasBD were
quantified by western blot analysis using a monoclonal anti-Ras
antibody (Oncogene).
Statistics
Statistical analysis was performed with one-way ANOVA followed by
Bonferroni’s multiple comparison test, unless indicated otherwise.
Asterisks denote statistical significance: *p < 0.05, **p < 0.01,
***p < 0.001.
Results
Lithium inhibits DNA synthesis and induces a G2/M cell
cycle arrest in asynchronous astrocytes
Stimulation of ERK in dividing cells is expected to constitute
a mitogenic stimulus (Chang and Karin 2001) and, in fact,
pituitary astrocytes have been shown to proliferate in
response to lithium (Levine et al. 2000). Thus, we decided
to study the effect of lithium on astrocyte DNA synthesis by
measuring [3
H]thymidine incorporation. In our initial experi-
ments, we found no stimulatory effects of lithium in
quiescent, serum-deprived astrocytes (not shown), and
looked for effects in subconfluent, asynchronously prolifer-
ating astrocytes that had been exposed to various LiCl
concentrations for 3 days. Compared with controls, the
amount of [3
H]thymidine incorporation into DNA in 5 mM
lithium-treated astrocytes was reduced by more than 60%,
while with 10 mM the reduction was about 90%, as shown in
Fig. 1(a). As lithium is known to interfere with inositol
1,4,5-trisphosphate recycling to myo-inositol, we checked
whether lithium inhibitory effects on DNA synthesis in
astrocytes could be due to intracellular inositol depletion.
Supplementation of the culture medium with 3 mM myo-
inositol did not affect lithium inhibition of [3
H]thymidine
incorporation, but reverted the potentiation due to 5 mM LiCl
towards the accumulation of [3
H]CDP-diacylglycerol in
[3
H]cytidine-prelabelled, carbachol-stimulated astrocytes, an
effect that mirrors inositol depletion (Godfrey 1989; Claro
et al. 1993) (not shown).
To further characterize the inhibitory effect of lithium on
astrocyte proliferation, we monitored the cell cycle profiles
of subconfluent, asynchronous astrocytes that had been
treated for 3 days with different LiCl concentrations. As
shown in Fig. 1(b), the percentage of tetraploid cells
increased with lithium in a dose-dependent manner. The
greater effect was observed with 10 mM LiCl, with 17.2%
occurrence of cells in the G2/M phase compared with 5.8%
observed under control conditions. The cell cycle phase
distribution was also monitored after a 7-day lithium
treatment and showed similar results (14.3% of cells in
G2/M in 5 mM LiCl treated cells, compared with 4.9% in
control cells, not shown). No signs of subdiploid (apoptotic
cells) were observed in any of the conditions assayed. Also,
Lithium inhibits the astrocyte MEK-ERK cascade 419
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
we found no impairment of cell adhesion or decrease of
MTT-reducing ability (Mosmann 1983) in lithium-treated
cells, indicating that lithium-induced cell cycle arrest
occurred in the absence of cytotoxicity.
Lithium reduces ERK and MEK phosphorylation
in astrocytes
We hypothesized that lithium effects on astrocyte DNA
synthesis and cell cycle could be due to the inhibition of
MEK/ERK signalling. As shown in Fig. 2(a), a 7-day
treatment of asynchronously proliferating astrocytes with
lithium resulted in an effective inhibition of ERK and MEK
phosphorylation. This was already patent at the therapeutic-
ally relevant lithium concentration of 1 mM, which reduced
some 60 and 50% the phosphorylation of ERK and MEK,
respectively, while 10 mM lithium reduced these phosphor-
ylated kinases to barely detectable levels. In contrast, lithium
concentrations up to 5 mM did not affect significantly the
phosphorylation status of Akt, indicating that, rather than
being the outcome of a general collapse of the ATP balance,
the effect of lithium is somehow selective on the MEK-ERK
pathway.
We next investigated whether lithium may preclude
mitogen activation of the MEK-ERK pathway in serum-
deprived, quiescent astrocytes. ET-1 is a well characterized
mitogen to astrocytes whose actions are especially relevant
on reactive gliosis taking place upon central nervous system
damage (Stanimirovic et al. 1995). We considered that this
experimental approach may reflect more closely the reactive
astrogliosis occurring in vivo after traumatic disruption of
the blood–brain barrier. Astrocytes were forced to enter G0
and remain quiescent by switching to FCS-free medium for
24 h, and were further maintained in this medium for 7 days
in the presence of different LiCl concentrations, then
challenged with ET-1 for 3 h. As shown in Fig. 2(b),
lithium prevented ET-1-stimulated phosphorylation of ERK
in a dose-dependent manner. A therapeutically relevant
lithium concentration of 1 mM reduced phospho-ERK levels
by 35%, while at 5 mM some 60% inhibition was achieved.
These effects were also apparent at shorter periods of lithium
exposure (6–24 h), but they required 10 mM lithium con-
centrations, as shown in Fig. 2(c). Taken together, the results
outlined above demonstrate a time- and concentration-
dependent inhibition of MEK and ERK phosphorylation in
astrocytes.
GSK-3 inhibition does not account for lithium effects
on MEK-ERK and cell cycle
To gain insight into the pharmacological target underlying
lithium effects, we tested whether GSK-3 inhibition in
astrocytes could be involved in cell cycle arrest or in the
inhibition of the MEK-ERK pathway. For this purpose, we
took advantage of the recently developed compound
SB216763, which has been shown to inhibit GSK-3 and
protect neurons from apoptosis induced by inhibition of
phosphatidylinositol 3-kinase (Cross et al. 2001). As shown
in Table 1, treatment of quiescent astrocytes with serum or
ET-1 induced a synchronized cell cycle re-entry, taking in
both cases 24 h to increase significantly the per cent
occurrence of tetraploid cells (G2/M phase). In astrocytes
exposed to 10 mM lithium for 24 h before mitogen addition,
there was a decreased occurrence of cells in S phase and also
a delayed appearance of the tetraploid cell population, which
took 48 h to increase. In contrast, SB 216763 did not change
the cell cycle distribution of ET-1-stimulated astrocytes at
any time point (Table 1). Further, it did not alter the
phosphorylation status of MEK and ERK (not shown),
although treatment of astrocytes with this compound under
(a)
(b)
Fig. 1 Lithium inhibits astrocyte DNA synthesis and induces a G2/M
cell cycle arrest. (a) [3H]Thymidine incorporation in asynchronously
growing astrocytes. Subconfluent cultures were treated with LiCl at the
indicated doses for 3 days. The cells were pulse-labelled with 1 lCi/mL
of [3H]thymidine during the last 24 h of treatment. [3H]Thymidine
incorporated into DNA was determined as shown in Materials and
methods. The results are means ± SEM of three independent
experiments. (b) Cell cycle analysis. Asynchronously growing astro-
cytes were treated with the indicated concentrations of LiCl for 3 days.
After trypsinization, the cells were ethanol fixed and labelled with
propidium iodide for quantification of DNA content by flow cytometry.
The percentage of cells in each phase of the cell cycle was determined
using CellQuest and Modfit LT software. The flow cytometric profiles
shown are representative of three independent experiments.
420 Rau´l Pardo et al.
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
the same conditions as above induced cytosolic b-catenin
stabilization and completely prevented Tau phosphorylation
in Ser396 (Fig. 3), consistent with the effective inhibition of
GSK-3b (Hart et al. 1998; Ikeda et al. 1998; Torreilles et al.
2000).
Inhibition of p38 MAPK partially reverts the inhibitory
effects of lithium
We investigated the possible participation of the stress
signalling kinase p38 MAPK by using its selective inhibitor
SB203580. As shown in Fig. 4, this compound did not
significantly affect the incorporation of [3
H]thymidine into
DNA of asynchronously proliferating astrocytes, contrary to
the MEK inhibitors U0126 and PD98059, but was effective
to revert partially the inhibition exerted by lithium at 5 mM
and 10 mM concentrations.
(c)
(b)
(a) Fig. 2 Lithium prevents signalling through the MEK/ERK pathway. (a)
Asynchronously growing secondary astrocytes were incubated for
1 week with the indicated concentrations of LiCl in DMEM containing
10% FCS. Cell extracts were processed as indicated in Materials and
methods, and subjected to western blot analysis with anti-phospho-
threonine 202/tyrosine 204-ERK 1/2, anti-phosphoserine 217/221
MEK1/2, anti-phosphoserine 473 Akt, or with the corresponding anti-
ERK1/2, anti-MEK1/2 or anti-Akt antibodies. The blots shown are
representative of five independent experiments that gave similar
results. Lower panels show the densitometric analysis of the shown
blots. (b) Secondary astrocytes were serum-starved for 24 h in
DMEM, and then incubated in the same medium for 1 week in the
presence or absence of the indicated LiCl concentrations. Upon sti-
mulation with 25 nM ET-1 for 3 h, the cells were lysed and processed
for immunoblot analysis. Ten micrograms of protein were loaded on
each lane of a SDS–PAGE and immunoblotted for phospho-ERK and
total ERK. A densitometric analysis of the western blot shown is
included. (c) Subconfluent astrocytes were serum-starved for 24 h,
pre-treated for the indicated times with 10 mM LiCl and stimulated with
25 nM ET-1 for 3 h. In the 0 h condition, LiCl and ET-1 were added
simultaneously. After stimulation, samples were processed for western
blot analysis. Ten micrograms of protein were loaded on each lane of
an SDS–PAGE and immunoblotted for phosphoERK, phosphoMEK
and the corresponding total proteins.
Table 1 Lithium, but not the GSK-3b inhibitor SB216763, delays cell
cycle re-entry
G0/G1 S G2/M
Control 93.6 ± 3.8 3.1 ± 0.8 4.1 ± 0.4
FCS, 20 h 77.6 ± 3.0 18.6 ± 1.1 3.8 ± 0.6
FCS, 24 h 58.1 ± 2.2 21.6 ± 4.0 20.2 ± 3.4
Li+
+ FCS 24 h 79.3 ± 4.2b
16.1 ± 2.5 4.9 ± 1.6b
ET-1 16 h 84.5 ± 3.8 12.1 ± 1.4 3.5 ± 1.9
ET-1 20 h 81.0 ± 2.5 14.5 ± 2.1 4.5 ± 1.0
ET-1 24 h 79.0 ± 2.8 12.0 ± 1.9 8.8 ± 1.9
ET-1 48 h 94.5 ± 2.6 1.9 ± 1.0 3.5 ± 1.8
Li+
+ ET-1 16 h 91.6 ± 3.0 6.5 ± 0.8a
1.9 ± 0.6
Li+
+ ET-1 20 h 90.8 ± 2.7a
8.0 ± 1.4a
1.2 ± 0.9a
Li+
+ ET-1 24 h 88.5 ± 3.3a
8.9 ± 1.6a
2.4 ± 2.0a
Li+
+ ET-1 48 h 87.1 ± 1.6a
6.1 ± 0.7a
6.8 ± 1.1
SB216763 + ET-1 16 h 84.1 ± 5.2 12.6 ± 2.4 3.8 ± 2.4
SB216763 + ET-1 20 h 80.9 ± 1.3 13.8 ± 2.1 5.6 ± 2.7
SB216763 + ET-1 24 h 80.7 ± 3.2 10.5 ± 2.5 8.8 ± 3.0
SB216763 + ET-1 48 h 96.2 ± 4.1 1.7 ± 1.3 2.1 ± 1.2
Astrocytes were deprived from serum for 3 days and treated with
10 mM LiCl or 3 lM SB216763 for 24 h. Then, cell cycle re-entry in
synchronized astrocytes was initiated by addition of 10% FCS or
25 nM ET-1, and cell cycle phase distribution was analyzed at different
times after stimulation. Results are reported as per cent occurrence of
cells in G0/G1 (diploid), G2/M (tetraploid), or S phase and are
means ± SD of two independent experiments. a
Significantly different
from ET-1-treated cells at the same time point (two-tailed Student’s
t-test, p < 0.05);b
significantly different from FCS-treated cells at the
same time point (two-tailed Student’s t-test, p < 0.05).
Lithium inhibits the astrocyte MEK-ERK cascade 421
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
Lithium pre-treatment prevents sustained but not acute
activation of MEK and does not affect activation of Ras
We next monitored lithium effects on the time course of MEK
activation in serum-deprived, quiescent astrocytes challenged
with ET-1 (Fig. 5a). 25 nM ET-1 induced a robust phosphory-
lation of MEK, reaching a peak at 3–5 min and slowly
declining thereafter. Enhanced phosphorylation of MEK could
be detected up to 6 h after ET-1 addition, which was the
longest time examined. In cells pre-treated for 24 h with
10 mM LiCl before ET-1 stimulation, the rapid, peak activation
of MEK remained unaffected, but its long-lasting phosphory-
lation was significantly reduced (Fig. 5a). We next monitored
the activation of Ras at the time points of peak (3 min) and
sustained (3 h) phosphorylation of MEK. As shown in
Fig. 5(b), ET-1 (and also EGF) stimulation resulted in a robust
increase in GTP-bound Ras at 3 min, while after 3 h the effect
C
N
L
U
0126PD
98059SB
203580
Li5m
M
Li5m
M
+SB
203580Li10m
M
Li10m
M
+SB
203580
0
10000
20000
30000
[3H]Thymidine
incorporaion(dpm)
Fig. 4 Effect of MEK and p38 MAPK inhibitors on astrocyte DNA
synthesis. Astrocytes growing on a 10% FCS-containing DMEM were
treated for 3 days with the MEK inhibitors U0126 (10 lM) and
PD98059 (50 lM) or the p38 MAPK inhibitor SB203580 (10 lM). The
cells were pulsed labelled with [3H]thymidine for the last 24 h of
treatment and its incorporation into DNA was determined as indicated
in Materials and methods. The results are means ± SEM of three
independent experiments performed in triplicate. Asterisks denote
statistical significance (p < 0.05) between each lithium condition in the
presence or absence of SB203580.
Fig. 3 SB216763 stabilizes b-catenin and prevents phosphorylation of
Tau on Ser396. Astrocytes growing on a 10% FCS-containing DMEM
medium were either left untreated or treated for 3 h with 3 lM
SB216763. Cells were hypotonically lysed and the levels of cytosolic
b-catenin were determined, or alternatively, cells were lysed in a 0.1%
TritonX-100 containing lysis buffer to determine phosphorylation of
Tau on Ser396 using a phospho-specific antibody. An antibody against
b-tubulin was used in the same western blot as a loading control. This
experiment was performed three times with identical results.
(a)
(b)
Fig. 5 Lithium inhibits sustained phosphorylation of MEK by the
mitogen ET-1 without affecting the activation of Ras. (a) Astrocytes
were serum deprived for 48 h, then stimulated with ET-1 for the indi-
cated times and processed for western blot analysis for phospho and
total MEK. A subset of cells was incubated for 24 h with 10 mM LiCl
prior to stimulation with ET-1. A densitometric analysis of the shown
western blot is included. The same experiment was repeated once
obtaining essentially the same results. Asterisks denote significance
(p < 0.05) as compared with control cells at the same time point, ac-
cording to a two-tailed Student’s t-test. (b) Two snapshots of the time
course of MEK phosphorylation by ET-1 are shown (3 min and 3 h).
The same batch of cells were processed to monitor activation of Ras
GTPase. Serum-starved astrocytes were either left untreated or pre-
treated for 24 h with 10 mM LiCl, and then stimulated for 3 min or 3 h
with either 25 nM ET-1 or 25 ng/mL EGF. To measure active Ras, cells
were lysed and incubated with a GST fusion protein including the Ras
binding domain of Raf-1 previously bound to glutathione-Sepharose
beads. Equal quantities of cell extracts were analyzed to ensure equal
amounts of total Ras in the assay. As an internal negative control,
some cells were also treated for 10 min with an inhibitor of the kinase
activity of the EGR receptor (AG1478, 1 lM) prior to the addition of
EGF.
422 Rau´l Pardo et al.
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
was smaller but still clear. Interestingly, lithium had no effect
on the initial (3 min) or in the late (3 h) phosphorylation of
Ras. In contrast, the EGF receptor kinase inhibitor tyrphostin
AG1478 completely prevented the EGF stimulated GTP
loading of Ras. Taken together, these results indicate that
lithium exerts its inhibitory effects on the sustained phos-
phorylation of MEK at a level downstream of Ras.
Lithium stimulates ERK phosphorylation in neurons
In striking contrast to what was found in astrocytes, treatment
of cerebelar granule neurons with lithium (1–10 mM) for
24 h resulted in a concentration-dependent increase of the
phosphorylation status of ERK and MEK (Fig. 6a). In our
conditions, lithium did not affect Akt phosphorylation, which
is not in conflict with results from Chalecka-Franaszek and
Chuang (1999), as they demonstrated that lithium-stimulated
phosphorylation of Akt returns to basal levels after 3 h.
Again, stimulation of the neuronal MEK-ERK pathway was
neither counteracted by inositol supplementation (not shown)
nor mimicked by SB216763, which was as effective as
lithium to stabilize neuronal b-catenin (Fig. 6b), arguing
against phosphoinositol phosphatases or GSK-3b being
involved in these effects.
Discussion
The present study shows that lithium exerts opposite effects
on the MEK-ERK pathway in cortical astrocytes and in
cerebelar granule neurons. In cells able to proliferate, MEK-
ERK signalling is clearly involved in the transduction of
mitogenic stimuli, and therefore it is not surprising that
lithium inhibition of MEK-ERK in astrocytes was mirrored
by a decreased DNA synthesis, an effect mimicked by MEK
inhibitors, U0126 and PD98059, and also by effects on cell
cycle distribution. In asynchronously proliferating astrocytes,
lithium induced the accumulation of tetraploid cells, consis-
tent with a G2/M cycle arrest. Cell cycle effects were clearer
in synchronized astrocytes, a model that may reflect more
closely the situation in vivo, where astrocytes are quiescent
cells and do not proliferate actively unless serum mitogenic
factors intrude into the brain parenchima. In serum-deprived
cells, lithium inhibited and delayed cell cycle re-entry after
stimulation with ET-1. As for the effects on MEK-ERK
signalling, a 1-week exposure to lithium at concentrations
within the range of therapeutic doses was effective to down-
regulate the phosphorylation of these kinases in asynchro-
nous, proliferating astrocytes. Similarly, in quiescent
astrocytes, lithium inhibited MEK and ERK phosphorylation
as induced by ET-1. A more detailed analysis revealed that
ET-1 induced a rapid phosphorylation of MEK which peaked
after 3–5 min and declined thereafter, but remained elevated
up to 6 h. Interestingly, lithium did not affect the initial peak
but was effective in reducing the sustained phosphorylation
of the kinase, which is relevant for mitogenesis. Mitogens
typically induce a sustained phosphorylation of MEK that
permits entry and progresion through G1 phase of the cell
cycle (Kahan et al. 1992; Meloche et al. 1992) after the
induction of cyclin D1, whose association with Cdk4 is
considered rate-limiting for cell growth (Whitmarsh and
Davis 2000). In contrast, non-mitogens able to stimulate the
MEK/ERK cascade elicit a typically transient phosphoryla-
tion which is over in less than 15 min (Kahan et al. 1992).
Ras activation by ET-1 followed a similar time course to that
of MEK phosphorylation, with a very robust loading of Ras
with GTP at 3 min and a sustained, long lasting activation at
3 h. However, lithium did not affect either the initial or the
long-term activation of Ras by ET-1 or EGF. These results
demonstrate that the mechanisms responsible for lithium
actions on MEK/ERK operate downstream of Ras.
Lithium effects on the cell cycle have been reported before
in bovine aortic endothelial cells, where the ion was shown to
induce cell cycle arrest by a mechanism involving stabiliza-
tion of p53 (Mao et al. 2001). This anti-tumour transcription
factor is stabilized by different mechanisms, including the
Akt-GSK-3-b-catenin signalling pathway. GSK-3-b-medi-
ated mechanisms, however, do not account for our results:
we show that the GSK-3 inhibitor SB216763 (Cross et al.
2001) failed to arrest the astrocyte cell cycle or to alter MEK
(a)
(b)
Fig. 6 Lithium induces MEK and ERK phosphorylation in cerebelar
granule neurons by a mechanism unrelated to GSK-3b inhibition.
(a) Primary cultures of cerebelar granule neurons were treated for
24 h with the indicated LiCl concentrations or with the specific GSK3
inhibitor SB216763 (3 lM), and then cells were lysed and processed
for western blot analysis. Twenty micrograms of protein were loaded
on each lane. The densitometric analysis of phospho-ERK isoforms,
phospho-MEK, and phospho-Akt is included. Open bars represent the
effect of SB216763. The results shown are from a significant experi-
ment that was repeated twice with similar results. (b) Neuronal lysates
treated as above were processed for western blot analysis against
b-catenin.
Lithium inhibits the astrocyte MEK-ERK cascade 423
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
and ERK phosphorylation, while effectively stabilized
b-catenin and inhibited Tau phosphorylation. However, p53
may be involved in lithium-induced astrocyte cell cycle
arrest: it has been reported that ERK inhibition contributes
with the activation of p38 MAP kinase in the stabilization of
p53 induced by nitric oxide (Kim et al. 2002). In fact,
lithium has been reported to stimulate p38 in intestinal
epithelial cells (Nemeth et al. 2002), although a recent study
showed that lithium alone did not cause significant activation
of p38 kinase in cerebellar granule neurons (Chen et al.
2003). This discrepancy may well arise from cell-specific
mechanisms and/or different lithium treatments, which are
typically time and concentration dependent. We have not
attempted to measure lithium effects on the astrocyte p38.
However, we provide some indirect evidence for the
involvement of p38 in lithium-induced inhibition of DNA
synthesis in astrocytes, as the effectc is partly reversed by
SB203580, suggesting that both ERK inhibition and p38
activation could be contributing to cell cycle arrest.
Besides GSK-3, another well-documented pharmacologi-
cal target of lithium are the phosphoinositol phosphatases. In
this regard, we have observed that addition of inositol to the
culture medium does not revert lithium effects on the
astrocyte MEK-ERK pathway, and therefore this mechanism
does not seem to account for our results either. Inositol
supplementation may seem a rather indirect way to test for
lithium actions mediated by phosphoinositol phosphatase
inhibition. However, it is fully effective to revert lithium-
dependent, agonist-stimulated accumulation of CDP-diacyl-
glycerol, a metabolite whose accumulation correlates with
restricted inositol availavility (Godfrey 1989; Claro et al.
1993; Centeno et al. 1998), and its use is justified by the
absence of selective inositol monophosphatase inhibitors
other than the poorly permeable L-690 330 (Atack et al.
1993).
Regarding lithium stimulatory effects on the MEK/ERK
cascade in cerebelar granule neurons, our results confirm
those of Kopnisky et al. (2003), and rule out the possible
implication of GSK-3 and phosphoinositol phosphatases. A
similar effect was found in SH-SY5Y neuroblastoma cells
treated with valproate (Yuan et al. 2001). In that case,
stimulatory effects of valproate were shown to take place
well upstream of the pathway, as the drug was ineffective in
dominant negative Raf and Ras mutants. This illustrates that,
if lithium and valproate effects in neurons are mediated by a
common mechanism, this is not simply the opposite to that
taking place in astrocytes. It is well known that both lithium
and valproate stimulate AP-1 binding activities (Ozaki and
Chuang 1997; Asghari et al. 1998; Chen et al. 1999b). Thus,
as ERK is involved in the activation of AP-1 (Frost et al.
1994), stimulation of neuronal ERK could contribute
substantially to the modulation of gene expression and
plasticity by anti-bipolar drugs. Interestingly, lithium has
been shown recently to stimulate the proliferation of
cerebelar and cortical neuroblasts (Hashimoto et al. 2003).
Although the molecular mechanism remains to be estab-
lished, it is tempting to speculate that, conversely to the
effects of lithium in astrocytes, ERK stimulation may
underlie neuroblast mitogenesis.
To our knowledge, the report from Levine et al. (2000) is
the only one dealing with lithium effects in vivo on glial cell
proliferation and, in that case, lithium treatment resulted in an
estimulated proliferation of pituicytes, or astrocytes from the
neural lobe of the pituitary. We just do not know the reason
for our apparently conflicting results that might arise simply
from the fact that the pituitary and the cerebral cortex are
quite different neural areas. Besides the astrocyte in vitro is a
model rather different from that in vivo. That is why it would
be worth testing whether lithium, a drug with decades of
therapeutical history which favours neuronal survival and
plasticity and, as we show here, also inhibits astrocyte
proliferation in vitro, would be also effective in vivo. This
might represent a potential treatment of CNS traumatic
injuries: these result in a rapid proliferative response from
resident astrocytes, a process known as reactive astrogliosis
or glial scarring, that has been suggested to be an attempt
made by the CNS to restore homeostasis through isolation of
the damaged region (Fitch and Silver 1997). Upon CNS
damage involving destruction of the brain–blood barrier,
endothelium-derived ET-1 and plasma components intrude
into parenchyma and initiate astrogliosis. The subsequent
formation of a glial scar may interfere with any subsequent
neural repair (McGraw et al. 2001). Current experimental
approaches to attenuate gliotic reaction, however, do not
constitute viable therapeutic interventions. In this context,
lithium therapy could contribute to neurological recovery
owing to its cytoprotective and neuroplastic action, which
has been recently demonstrated in an ischaemia/reperfusion
model (Ren et al. 2003), and the inhibition of reactive
astrogliosis.
Acknowledgements
RP is recipient of a pre-doctoral fellowship from Universitat
Auto`noma de Barcelona. AG and BR are recipients of pre-doctoral
fellowships from the Spanish Ministry of Science and Technology
(SMST). This work has been supported by Grant BFI 2001–2017
from the SMST. We appreciate the generous gift of compound
SB216763 by GlaxoSmithKline, Stevenage, Herts, UK, the help of
Dr Joan-Marc Servitja with setting up the assay of GTP-bound Ras,
and the excellent technical assistance of Cristina Gutie´rrez with cell
culture and Manuela Costa with the FACS facility.
References
A´ lvarez G., Mun˜oz-Montano J. R., Satru´stegui J., A´ vila J., Bogo´nez E.
and Dı´az-Nido J. (1999) Lithium protects cultured neurons against
beta-amyloid-induced neurodegeneration. FEBS Lett. 453, 260–
264.
424 Rau´l Pardo et al.
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
Asghari V., Wang J. F., Reiach J. S. and Young L. T. (1998) Differential
effects of mood stabilizers on Fos/Jun proteins and AP-1 DNA
binding activity in human neuroblastoma SH-SY5Y cells. Mol.
Brain Res. 58, 95–102.
Atack J. R. (1996) Inositol monophosphatase, the putative therapeutic
target for lithium. Brain Res. Rev. 22, 183–190.
Atack J. R., Cook S. M., Watt A. P., Fletcher S. R. and Ragan C. I.
(1993) In vitro and in vivo inhibition of inositol monophosphatase
by the bisphosphonate L-690,330. J. Neurochem. 60, 652–658.
Berridge M. J., Downes C. P. and Hanley M. R. (1989) Neural and
developmental actions of lithium: a unifying hypothesis. Cell 59,
411–419.
Centeno F., Mora A., Fuentes J. M., Soler G. and Claro E. (1998) Partial
lithium-associated protection against apoptosis induced by C2-cer-
amide in cerebellar granule neurons. Neuroreport 9, 4199–4203.
Chalecka-Franaszek E. and Chuang D. M. (1999) Lithium activates the
serine/threonine kinase Akt-1 and suppresses glutamate-induced
inhibition of Akt-1 activity in neurons. Proc. Natl Acad. Sci. USA
96, 8745–8750.
Chang L. and Karin M. (2001) Mammalian MAP kinase signalling
cascades. Nature 410, 37–40.
Chen G., Huang L. D., Jiang Y. M. and Manji H. K. (1999a) The mood-
stabilizing agent valproate inhibits the activity of glycogen syn-
thase kinase-3. J. Neurochem. 72, 1327–1330.
Chen G., Yuan P. X., Jiang Y. M., Huang L. D. and Manji H. K. (1999b)
Valproate robustly enhances AP-1 mediated gene expression. Mol.
Brain Res. 64, 52–58.
Chen R. W., Qin Z. H., Ren M., Kanai H., Chalecka-Franaszek E., Leeds
P. and Chuang D. M. (2003) Regulation of c-Jun N-terminal kin-
ase, p38 kinase and AP-1 DNA binding in cultured brain neurons:
roles in glutamate excitotoxicity and lithium neuroprotection.
J. Neurochem. 84, 566–575.
Claro E., Fain J. N. and Picatoste F. (1993) Noradrenaline stimulation
unbalances the phosphoinositide cycle in rat cerebral cortical sli-
ces. J. Neurochem. 60, 2078–2086.
Cross D. A., Culbert A. A., Chalmers K. A., Facci L., Skaper S. D. and
Reith A. D. (2001) Selective small-molecule inhibitors of glycogen
synthase kinase-3 activity protect primary neurones from death.
J. Neurochem. 77, 94–102.
D’Mello S. R., Anelli R. and Calissano P. (1994) Lithium induces
apoptosis in immature cerebellar granule cells but promotes sur-
vival of mature neurons. Exp. Cell Res. 211, 332–338.
De Sarno P., Li X. and Jope R. S. (2002) Regulation of Akt and glycogen
synthase kinase-3beta phosphorylation by sodium valproate and
lithium. Neuropharmacology 43, 1158–1164.
Drevets W. C., Price J. L., Simpson J. R. Jr, Todd R. D., Reich T.,
Vannier M. and Raichle M. E. (1997) Subgenual prefrontal cortex
abnormalities in mood disorders. Nature 386, 824–827.
Fitch M. T. and Silver J. (1997) Glial cell extracellular matrix: bound-
aries for axon growth in development and regeneration. Cell Tissue
Res. 290, 379–384.
Frost J. A., Geppert T. D., Cobb M. H. and Feramisco J. R. (1994) A
requirement for extracellular signal-regulated kinase (ERK) func-
tion in the activation of AP-1 by Ha-Ras, phorbol 12-myristate
13-acetate, and serum. Proc. Natl Acad. Sci. USA 91, 3844–3848.
Godfrey P. P. (1989) Potentiation by lithium of CMP-phosphatidate
formation in carbachol-stimulated rat cerebral-cortical slices and its
reversal by myo-inositol. Biochem. J. 258, 621–624.
Grewal S. S., York R. D. and Stork P. J. S. (1999) Extracellular-signal-
regulated kinase signalling in neurons. Curr. Opin. Neurobiol. 9,
544–553.
Grimes C. A. and Jope R. S. (2001) The multifaceted roles of glycogen
synthase kinase 3 beta in cellular signaling. Prog. Neurobiol. 65,
391–426.
Hall A. C., Lucas F. R. and Salinas P. C. (2000) Axonal remodeling and
synaptic differentiation in the cerebellum is regulated by WNT-7a
signaling. Cell 100, 525–535.
Hall A. C., Brennan A., Goold R. G., Cleverley K., Lucas F. R., Gordon-
Weeks P. R. and Salinas P. C. (2002) Valproate regulates GSK-
3-mediated axonal remodeling and synapsin I clustering in
developing neurons. Mol. Cell Neurosci. 20, 257–270.
Hart M. J., de los Santos R., Albert I. N., Rubinfeld B. and Polakis P.
(1998) Downregulation of beta-catenin by human Axin and its
association with the APC tumor suppressor, beta-catenin and
GSK3 beta. Curr. Biol. 8, 573–581.
Hashimoto R., Senatorov V., Kanai H., Leeds P. and Chuang D. M.
(2003) Lithium stimulates progenitor proliferation in cultured brain
neurons. Neuroscience 117, 55–61.
Ikeda S., Kishida S., Yamamoto H., Murai S., Koyama S. and Kikuchi
A. (1998) Axin, a negative regulator of the Wnt signaling pathway,
forms a complex with GSK-3beta and beta-catenin and promotes
GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J.
17, 1371–1384.
Jope R. S. (1999) Anti-bipolar therapy: mechanism of action of lithium.
Mol. Psychiatry 4, 117–128.
Kahan C., Seuwen K., Meloche S. and Poysse´gur J. (1992) Coordinate
biphasic activation of p44 mitogen activated protein kinase and S6
kinase by growth factors in hamster fibroblasts. J. Biol. Chem. 267,
13369–13375.
Kim S. J., Ju J. W., Oh C. D., Yoon Y. M., Song W. K., Kim J. H., Yoo Y.
J., Bang O. S., Kang S. S. and Chun J. S. (2002) ERK-1/2 and p38
kinase oppositely regulate nitric oxide-induced apoptosis of
chondrocytes in association with p53, caspase-3, and differenti-
ation status. J. Biol. Chem. 277, 1332–1339.
Klein P. S. and Melton D. A. (1996) A molecular mechanism for the
effect of lithium on development. Proc. Natl Acad. Sci. USA 93,
8455–8459.
Kopnisky K. L., Chalecka-Franaszek E., Gonzalez-Zulueta M. and
Chuang D. M. (2003) Chronic lithium treatment antagonizes glu-
tamate-induced decrease of phosphorylated CREB in neurons via
reducing protein phosphatase 1 and increasing MEK activities.
Neuroscience 116, 425–435.
Levine S., Saltzman A. and Klein A. W. (2000) Proliferation of glial
cells in vivo induced in the neural lobe of the rat pituitary by
lithium. Cell. Prolif. 33, 203–207.
Mao C. D., Hoang P. and DiCorleto P. E. (2001) Lithium inhibits cell
cycle progression and induces stabilization of p53 in bovine aortic
endothelial cells. J. Biol. Chem. 276, 26180–26188.
Masgrau R., Servitja J. M., Sarri E., Young K. W., Nahorski S. R. and
Picatoste F. (2000) Intracellular Ca2+ stores regulate muscarinic
receptor stimulation of phospholipase C in cerebellar granule cells.
J. Neurochem. 74, 818–826.
McGraw J., Hiebert G. W. and Steeves J. D. (2001) Modulating astro-
gliosis after neurotrauma. J. Neurosci. Res. 63, 109–115.
Meloche S., Seuwen K., Pages G. and Poysse´gur J. (1992) Biphasic and
synergistic activation of p44 MAPK (ERK1) by growth factors:
correlation between late phase activation. Mol. Endocrinol. 6, 845–
854.
Mora A., Sabio G., Gonza´lez-Polo R. A., Cuenda A., Alessi D. R.,
Alonso J. C., Fuentes J. M., Soler G. and Centeno F. (2001)
Lithium inhibits caspase 3 activation and dephosphorylation of
PKB and GSK3 induced by K+ deprivation in cerebellar granule
cells. J. Neurochem. 78, 199–206.
Mosmann T. (1983) Rapid colorimetric assay for cellular growth and
survival: application to proliferation and cytotoxicity assays.
J. Immunol. Meth 65, 55–63.
Nemeth Z. H., Deitch E. A., Szabo C., Fekete Z., Hauser C. J. and Hasko
G. (2002) Lithium induces NF-kappa B activation and interleukin-
Lithium inhibits the astrocyte MEK-ERK cascade 425
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
8 production in human intestinal epithelial cells. J. Biol. Chem.
277, 7713–7719.
Nonaka S. and Chuang D. M. (1998) Neuroprotective effects of chro-
nic lithium on focal cerebral ischemia in rats. Neuroreport 9, 2081–
2084.
Nonaka S., Katsube N. and Chuang D. M. (1998) Lithium protects rat
cerebellar granule cells against apoptosis induced by anticonvul-
sants, phenytoin and carbamazepine. J. Pharmacol. Exp. Ther. 286,
539–547.
Ongur D., Drevets W. C. and Price J. L. (1998) Glial reduction in the
subgenual prefrontal cortex in mood disorders. Proc. Natl Acad.
Sci. USA 95, 13290–13295.
Ozaki N. and Chuang D. M. (1997) Lithium increases transcription
factor binding to AP-1 and cyclic AMP-responsive element in
cultured neurons and rat brain. J. Neurochem. 69, 2336–2344.
Phiel C. J. and Klein P. S. (2001) Molecular targets of lithium action.
Annu. Rev. Pharmacol. Toxicol. 41, 789–813.
Ren M., Senatorov V. V., Chen R. W. and Chuang D. M. (2003) Post-
insult treatment with lithium reduces rat brain damage and facili-
tates neurological recovery in a rat ischemia/reperfusion model.
Proc. Natl Acad. Sci. USA 100, 6210–6215.
Servitja J. M., Masgrau R., Sarri E. and Picatoste F. (2000) Effects of
oxidative stress on phospholipid signaling in rat cultured astrocytes
and brain slices. J. Neurochem. 75, 788–794.
Stambolic V., Ruel L. and Woodgett J. R. (1996) Lithium inhibits gly-
cogen synthase kinase-3 activity and mimics wingless signalling in
intact cells. Curr. Biol. 6, 1664–1668.
Stanimirovic D. B., Ball R., Mealing G., Morley P. and Durkin J. P. (1995)
The role of intracellular calcium and protein kinase C in endothelin-
stimulated proliferation of rat type I astrocytes. Glia 15, 119–130.
Torreilles F., Roquet F., Granier C., Pau B. and Mourton-Gilles C. (2000)
Binding specificity of monoclonal antibody AD2: influence of the
phosphorylation state of tau. Mol. Brain Res. 78, 181–185.
Whitmarsh A. J. and Davis R. J. (2000) A central control for cell growth.
Nature 403, 255–256.
Williams R. S., Cheng L., Mudge A. W. and Harwood A. J. (2002) A
common mechanism of action for three mood-stabilizing drugs.
Nature 417, 292–295.
Yuan P.-X., Huang L.-D., Jiang Y.-M., Gutkind J. S., Manji H. K. and
Chen G. (2001) The mood stabilizer valproic acid activates mito-
gen-activated protein kinases and promotes neurite growth. J. Biol.
Chem. 276, 31674–31683.
426 Rau´l Pardo et al.
Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426

More Related Content

What's hot

A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...
A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...
A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...johnohab
 
Looking downstream review 2015
Looking downstream review 2015Looking downstream review 2015
Looking downstream review 2015Mustafa Siddiq
 
Efficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentEfficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentjanelle_leggere
 
Reversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeReversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeJosé Luis Moreno Garvayo
 
KDM5 epigenetic modifiers as a focus for drug discovery
KDM5 epigenetic modifiers as a focus for drug discoveryKDM5 epigenetic modifiers as a focus for drug discovery
KDM5 epigenetic modifiers as a focus for drug discoveryChristopher Wynder
 
Epigenetics
Epigenetics Epigenetics
Epigenetics mpattani
 
Final Dissertation
Final DissertationFinal Dissertation
Final DissertationRachel Li
 
2014 IP3K J Cell Biol-2014-Heller-1219-36
2014 IP3K    J Cell Biol-2014-Heller-1219-362014 IP3K    J Cell Biol-2014-Heller-1219-36
2014 IP3K J Cell Biol-2014-Heller-1219-36Monica Ghidinelli
 
Shapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentShapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentHannah Shapero
 
CVA Biology I - B10vrv4133
CVA Biology I - B10vrv4133CVA Biology I - B10vrv4133
CVA Biology I - B10vrv4133ClayVirtual
 
Biosynthesis of enediyne antitumor antibiotics
Biosynthesis of enediyne antitumor antibioticsBiosynthesis of enediyne antitumor antibiotics
Biosynthesis of enediyne antitumor antibioticsDr Robert Craig PhD
 
Effect Of Ethanol In Oligodendrocytes, research miami
Effect Of Ethanol In Oligodendrocytes, research miamiEffect Of Ethanol In Oligodendrocytes, research miami
Effect Of Ethanol In Oligodendrocytes, research miamiLydia Cortes
 

What's hot (20)

A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...
A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...
A Critical Role of Erythropoietin Receptor in Neurogenesis and Post-Stroke Re...
 
Looking downstream review 2015
Looking downstream review 2015Looking downstream review 2015
Looking downstream review 2015
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Alzheimer’s disease
Alzheimer’s disease Alzheimer’s disease
Alzheimer’s disease
 
Efficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentEfficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatment
 
Reversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeReversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndrome
 
KDM5 epigenetic modifiers as a focus for drug discovery
KDM5 epigenetic modifiers as a focus for drug discoveryKDM5 epigenetic modifiers as a focus for drug discovery
KDM5 epigenetic modifiers as a focus for drug discovery
 
Epigenetics
Epigenetics Epigenetics
Epigenetics
 
Articulo electro y celulas
Articulo  electro y celulasArticulo  electro y celulas
Articulo electro y celulas
 
Final Dissertation
Final DissertationFinal Dissertation
Final Dissertation
 
2014 IP3K J Cell Biol-2014-Heller-1219-36
2014 IP3K    J Cell Biol-2014-Heller-1219-362014 IP3K    J Cell Biol-2014-Heller-1219-36
2014 IP3K J Cell Biol-2014-Heller-1219-36
 
Shapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocumentShapero.ResearchProject.FinalDocument
Shapero.ResearchProject.FinalDocument
 
Definition of epigenetics
Definition of epigeneticsDefinition of epigenetics
Definition of epigenetics
 
Mutation
MutationMutation
Mutation
 
CVA Biology I - B10vrv4133
CVA Biology I - B10vrv4133CVA Biology I - B10vrv4133
CVA Biology I - B10vrv4133
 
4 Real Role
4 Real Role4 Real Role
4 Real Role
 
In press
In pressIn press
In press
 
Epigenetics diet and cancer
Epigenetics diet and cancerEpigenetics diet and cancer
Epigenetics diet and cancer
 
Biosynthesis of enediyne antitumor antibiotics
Biosynthesis of enediyne antitumor antibioticsBiosynthesis of enediyne antitumor antibiotics
Biosynthesis of enediyne antitumor antibiotics
 
Effect Of Ethanol In Oligodendrocytes, research miami
Effect Of Ethanol In Oligodendrocytes, research miamiEffect Of Ethanol In Oligodendrocytes, research miami
Effect Of Ethanol In Oligodendrocytes, research miami
 

Viewers also liked

Alzheimers - Tideglusib presentation
Alzheimers - Tideglusib presentationAlzheimers - Tideglusib presentation
Alzheimers - Tideglusib presentationHelen Turner
 
Mol Brain 2010
Mol Brain 2010Mol Brain 2010
Mol Brain 2010Raul Pardo
 
Recent advances in the treatment of alzheimer's disease
Recent advances in the treatment of alzheimer's diseaseRecent advances in the treatment of alzheimer's disease
Recent advances in the treatment of alzheimer's diseaseDr. Mohit Kulmi
 
J Neurosci 2006
J Neurosci 2006J Neurosci 2006
J Neurosci 2006Raul Pardo
 
Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Dr Durga Gahlot
 

Viewers also liked (7)

930710.v2
930710.v2930710.v2
930710.v2
 
Alzheimers - Tideglusib presentation
Alzheimers - Tideglusib presentationAlzheimers - Tideglusib presentation
Alzheimers - Tideglusib presentation
 
Mol Brain 2010
Mol Brain 2010Mol Brain 2010
Mol Brain 2010
 
Recent advances in the treatment of alzheimer's disease
Recent advances in the treatment of alzheimer's diseaseRecent advances in the treatment of alzheimer's disease
Recent advances in the treatment of alzheimer's disease
 
J Neurosci 2006
J Neurosci 2006J Neurosci 2006
J Neurosci 2006
 
signal transduction
signal transductionsignal transduction
signal transduction
 
Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.
 

Similar to J Neurochem 2003

Book role of plants, environmental toxins and physical neurotoxicological fac...
Book role of plants, environmental toxins and physical neurotoxicological fac...Book role of plants, environmental toxins and physical neurotoxicological fac...
Book role of plants, environmental toxins and physical neurotoxicological fac...M. Luisetto Pharm.D.Spec. Pharmacology
 
Nutrição e Inflamação, Maio, 2013
Nutrição e Inflamação, Maio, 2013Nutrição e Inflamação, Maio, 2013
Nutrição e Inflamação, Maio, 2013Pedro Bastos
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methileneJorge Parodi
 
hemichannel makes it a major contributor toionic dysregula
hemichannel makes it a major contributor toionic dysregulahemichannel makes it a major contributor toionic dysregula
hemichannel makes it a major contributor toionic dysregulaSusanaFurman449
 
The Two Component Stressors
The Two Component StressorsThe Two Component Stressors
The Two Component StressorsPatricia Viljoen
 
2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTERSudeep Pisipaty
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeGanesh Murthi
 
GSK3 in neurodegeneration
GSK3 in neurodegenerationGSK3 in neurodegeneration
GSK3 in neurodegenerationSaeed Aghdam
 
PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)Marco Garza
 
2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTERSudeep Pisipaty
 
Florence Molinari - INMED-INSERM U901. Marseille.
Florence Molinari - INMED-INSERM U901. Marseille. Florence Molinari - INMED-INSERM U901. Marseille.
Florence Molinari - INMED-INSERM U901. Marseille. Fundación Ramón Areces
 
Florence Molinari INMED-INSERM U901. Marseille.
Florence Molinari   INMED-INSERM U901. Marseille. Florence Molinari   INMED-INSERM U901. Marseille.
Florence Molinari INMED-INSERM U901. Marseille. Fundación Ramón Areces
 
MAPK Pathway in Skeletal Muscle Diseases
MAPK Pathway in Skeletal Muscle DiseasesMAPK Pathway in Skeletal Muscle Diseases
MAPK Pathway in Skeletal Muscle DiseasesAnnex Publishers
 

Similar to J Neurochem 2003 (20)

Book role of plants, environmental toxins and physical neurotoxicological fac...
Book role of plants, environmental toxins and physical neurotoxicological fac...Book role of plants, environmental toxins and physical neurotoxicological fac...
Book role of plants, environmental toxins and physical neurotoxicological fac...
 
Nutrição e Inflamação, Maio, 2013
Nutrição e Inflamação, Maio, 2013Nutrição e Inflamação, Maio, 2013
Nutrição e Inflamação, Maio, 2013
 
AGEP:SRI research project
AGEP:SRI research projectAGEP:SRI research project
AGEP:SRI research project
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methilene
 
Edgardo Arroyo CV
Edgardo Arroyo CVEdgardo Arroyo CV
Edgardo Arroyo CV
 
hemichannel makes it a major contributor toionic dysregula
hemichannel makes it a major contributor toionic dysregulahemichannel makes it a major contributor toionic dysregula
hemichannel makes it a major contributor toionic dysregula
 
Herrmann STAT3 2008
Herrmann STAT3 2008Herrmann STAT3 2008
Herrmann STAT3 2008
 
The Two Component Stressors
The Two Component StressorsThe Two Component Stressors
The Two Component Stressors
 
Genes,brain & behavior1
Genes,brain & behavior1Genes,brain & behavior1
Genes,brain & behavior1
 
jbc1998
jbc1998jbc1998
jbc1998
 
JC paper-2009
JC paper-2009JC paper-2009
JC paper-2009
 
2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-like
 
GSK3 in neurodegeneration
GSK3 in neurodegenerationGSK3 in neurodegeneration
GSK3 in neurodegeneration
 
PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)
 
2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER2014 DURF CONFERENCE POSTER
2014 DURF CONFERENCE POSTER
 
Anxiety paper 2014
Anxiety paper 2014Anxiety paper 2014
Anxiety paper 2014
 
Florence Molinari - INMED-INSERM U901. Marseille.
Florence Molinari - INMED-INSERM U901. Marseille. Florence Molinari - INMED-INSERM U901. Marseille.
Florence Molinari - INMED-INSERM U901. Marseille.
 
Florence Molinari INMED-INSERM U901. Marseille.
Florence Molinari   INMED-INSERM U901. Marseille. Florence Molinari   INMED-INSERM U901. Marseille.
Florence Molinari INMED-INSERM U901. Marseille.
 
MAPK Pathway in Skeletal Muscle Diseases
MAPK Pathway in Skeletal Muscle DiseasesMAPK Pathway in Skeletal Muscle Diseases
MAPK Pathway in Skeletal Muscle Diseases
 

J Neurochem 2003

  • 1. Opposed effects of lithium on the MEK-ERK pathway in neural cells: inhibition in astrocytes and stimulation in neurons by GSK3 independent mechanisms Rau´l Pardo,* Alberto G. Andreolotti,* Bele´n Ramos,  Fernando Picatoste* and Enrique Claro* *Institut de Neurocie`ncies and Departament de Bioquı´mica i Biologia Molecular, Universitat Auto`noma de Barcelona  Departamento de Fisiologı´a, Universidad de Extremadura, Spain Summary Lithium is widely used in the treatment of bipolar disorder, but despite its proven therapeutic efficacy, the molecular mech- anisms of action are not fully understood. The present study was undertaken to explore lithium effects of the MEK/ERK cascade of protein kinases in astrocytes and neurons. In asynchronously proliferating rat cortical astrocytes, lithium decreased time- and dose-dependently the phosphorylation of MEK and ERK, with 1 mM concentrations achieving 60 and 50% inhibition of ERK and MEK, respectively, after a 7-day exposure. Lithium also inhibited [3 H]thymidine incorporation into DNA and induced a G2/M cell cycle arrest. In serum- deprived, quiescent astrocytes, pre-exposure to lithium resulted in the inhibition of cell cycle re-entry as stimulated by the mitogen endothelin-1: under this experimental setting, lithium did not affect the rapid, peak phosphorylation of MEK taking place after 3–5 min, but was effective in inhibiting the long-term, sustained phosphorylation of MEK. Lithium inhibi- tion of the astrocyte MEK/ERK pathway was independent of inositol depletion. Further, compound SB216763 inhibited Tau phosphorylation at Ser396 and stabilized cytosolic b-catenin, consistent with the inhibition of glycogen synthase kinase-3b (GSK-3b), but failed to reproduce lithium effects on MEK and ERK phosphorylation and cell cycle arrest. In cerebellar granule neurons, millimolar concentrations of lithium enhanced MEK and ERK phosphorylation in a concentration- dependent manner, again through an inositol and GSK-3b independent mechanism. These opposing effects in astro- cytes and neurons make lithium treatment a promising strat- egy to favour neural repair and reduce reactive gliosis after traumatic injury. Keywords: astrocytes, cell cycle arrest, ERK, GSK-3, lith- ium, MEK. J. Neurochem. (2003) 87, 417–426. Lithium has been used in the treatment of bipolar mood disorder for decades, but despite its therapeutic efficacy, the molecular mechanisms underlying its actions remain unclear (Jope 1999; Phiel and Klein 2001). In this regard, based on the observation that lithium inhibits inositol monophospha- tase and inositol polyphosphate 1-phosphatase, thereby blocking inositol 1,4,5-trisphosphate recycling to inositol, the inositol depletion hypothesis considered that persistent activation of phosphoinositide phospholipase C in the presence of lithium would lower the cellular inositol concentration, leading eventually to the depletion of phos- phatidylinositol 4,5-bisphosphate and the impairment of calcium signalling (Berridge et al. 1989; Atack 1996). This hypothesis has received some support after the finding that lithium, carbamazepine, and valproic acid, all three drugs used in the treatment of bipolar disorder, increase growth cone area in sensory neurons, in a manner that was counteracted by inositol addition, and mediated probably by inhibition of prolyl oligopeptidase by an as yet unknown mechanism (Williams et al. 2002). In the past few years, however, lithium has emerged as a remarkable neuroprotective agent: it was first shown to protect cerebellar granule neurons from undergoing apoptotic cell death when cultured under non-depolarizing potassium conditions (D’Mello et al. 1994), and soon thereafter similar observations were extended to many other apoptotic insults Received June 4, 2003; accepted July 4, 2003. Address correspondence and reprint requests to Enrique Claro, Institut de Neurocie`ncies, Edifici M, Universitat Auto`noma de Barcelona, E-08193 Bellaterra, Spain. E-mail: Enrique.Claro@uab.es Abbreviations used: DMEM, Dulbecco’s modified Eagle’s medium; ET-1, endothelin-1; FCS, fetal calf serum; GSK-3b, glycogen synthase kinase-3b; RBD, Ras-binding domain of Raf-1. Journal of Neurochemistry, 2003, 87, 417–426 doi:10.1046/j.1471-4159.2003.02015.x Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426 417
  • 2. like ischaemia (Nonaka and Chuang 1998), anticonvulsants (Nonaka et al. 1998), C2-ceramide (Centeno et al. 1998), or b-amyloid (A´ lvarez et al. 1999), among others, where lithium effects were independent of inositol depletion (Centeno et al. 1998; Nonaka et al. 1998). Glycogen synthase kinase-3b (GSK-3b) is inhibited by lithium (Klein and Melton 1996; Stambolic et al. 1996), and this discovery has greatly fuelled the interest in the biochemistry of this classic therapeutic agent. In many instances, the activation of GSK-3b taking place after disruption of the phosphatidy- linositol 3-kinase/Akt pathway appears related to apoptotic cell death, and it is generally acknowledged today that this kinase constitutes the primary site of action underlying the cytoprotective actions of lithium (Grimes and Jope 2001). Lately, more selective GSK-3b inhibitors have been devel- oped with potent cytoprotective effects, lending further support to the involvement of GSK-3b in the regulation of cell survival and apoptosis (Cross et al. 2001). The relevance of GSK-3b inhibition in bipolar therapy, however, is still a matter of controversy, although it is supported by the fact that valproate, another drug used in the treatment of bipolar disorder, has been reported to inhibit GSK-3b in vitro (Chen et al. 1999a) and also in vivo (De Sarno et al. 2002; Hall et al. 2002). The inhibitory action of lithium and valproate on GSK-3 is not restricted to neuroprotection against apoptotic insults: lithium has been shown to mimic the action of WNT-7a, promoting axonal remodelling and synaptic differentiation (Hall et al. 2000). This neuroplastic action of anti-bipolar drugs may be therapeutically relevant, as there is evidence supporting the occurrence of neuronal loss and/or atrophy in patients with mood disorder (Drevets et al. 1997; Ongur et al. 1998). Inhibition of GSK-3b may not be the sole mechanism underlying lithium-induced neuroprotection and plasticity: the ion has been shown to potentiate Akt phosphorylation in cerebellar neurons (Chalecka-Franaszek and Chuang 1999; Mora et al. 2001). Also, lithium appears to modulate MEK- ERK signalling, a pathway whose relevance in the regulation of neuronal function, synaptic plasticity, and survival is receiving growing attention (Grewal et al. 1999): it has been reported recently that lithium induced the activation of MEK and ERK in cerebellar granule neurons (Kopnisky et al. 2003) and, similarly, valproic acid promoted neurite growth in neuroblastoma cells by a mechanism that implies activa- tion of these mitogen-activated protein kinases (Yuan et al. 2001). Experimental studies on the plasticity and regeneration of the nervous system often disregard the role of glial cells as trophic support for neurons or as a source of inflammatory cytokines, so in the present study we compared the effects of lithium in astrocytes and neurons. In agreement with Kopnisky et al. (2003), we found that lithium potentiates ERK phosphorylation in cerebelar granule neurons, but to our surprise, it exerts opposite effects in astroglial cultures, inhibiting sustained MEK and ERK phosphorylation, DNA synthesis, and inducing cell cycle arrest. Materials and methods Materials Dulbecco’s modified Eagle’s medium (DMEM), fetal calf serum (FCS), penicillin, streptomycin, and glutamine were obtained from Gibco (Rockville, MD, USA). Endothelin-1 (ET-1) was from Alexis Corporation (San Diego, CA, USA). MEK inhibitors U0126 and PD98059, tyrphostin AG1478 and antibody against phospho- serine 396-Tau were from Calbiochem (San Diego, CA, USA). Antibodies against total and phosphorylated ERK, MEK, and Akt were from Cell Signaling Technology (Beverly, MA, USA). Anti- Ras antibody was from Oncogene (Oncogene-EMD Biosciences, Darmstadt, Germany), antibody against b-catenin was from BD Transduction Laboratories (Lexington, KY, USA), and antibody against b-tubulin was from BD Pharmingen. Secondary peroxi- dase-conjugated antibodies were from Transduction Laboratories. [Methyl-3 H]thymidine (87.0 Ci/mmol) was from Amersham Phar- macia Biotech (Piscataway, NJ, USA). GSK-3 inhibitor SB216763 was kindly donated by GlaxoSmithKline, Stevenage, Herts, UK. Cell culture Primary cultures of cerebral cortical astrocytes were prepared from newborn (< 24 h) Sprague-Dawley rats as previously described (Servitja et al. 2000). Cells were plated into 176 cm2 dishes (12.5 · 106 viable cells/dish) in DMEM supplemented with 10% FCS, 33 mM glucose, 2 mM glutamine, 50 units/mL penicillin, and 50 lg/mL streptomycin. Two hours after plating, the medium was replaced to remove non-adherent cells. The cultures were incubated at 37°C in a humidified atmosphere of 5% CO2/95% air. After 24 h, the medium was changed again. Before confluence, cells were replated into polylysine-coated 6-well plates (2 · 105 cells/ well) for western blot analysis, or into 24-well plates (2–5 · 104 cells/well) to carry out [3 H]thymidine incorporation assays. At this time, immunocytochemical staining for glial fibrillary acidic protein revealed that cultures consisted of 90– 95% astrocytes. In some experiments, cells were serum-starved before being stimulated with ET-1. Primary cultures of rat cerebelar granule cells were prepared as previously described (Masgrau et al. 2000). Briefly, cells were seeded at 2 · 106 cells/well in 6-well plates in DMEM containing 10% FCS, 25 mM KCl, 5 mM glucose, penicillin (50 units/mL), streptomycin (50 lg/mL), sodium pyru- vate (0.1 mg/mL), 7 mM p-aminobenzoic acid, and 2 mM gluta- mine. Twenty-four hours after plating, 10 lM cytosine arabinofur- anoside was added to prevent growth of glial cells, and neurons were maintained for 7 days. Western blot analysis Cells were washed twice with ice-cold phosphate-buffered saline prior to scraping and lysis. For ERK, MEK and Akt immunoblots, lysis buffer consisted of 50 mM Tris-HCl, pH 7.5, containing 0.1% Triton X-100, 2 mM EDTA, 2 mM EGTA, 50 mM NaF, 10 mM b-glycerophosphate, 5 mM sodium pyrophosphate, 1 mM sodium orthovanadate, 0.1% (v/v) b-mercaptoethanol, 250 lM phenyl- methylsulfonyl fluoride (PMSF), 10 lg/mL aprotinin and leupeptin. 418 Rau´l Pardo et al. Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 3. For immunoblots of cytoplasmic b-catenin, hypotonic lysis buffer was 50 mM Tris-HCl, pH 7.5, with 2 mM EDTA, 2 mM EGTA, 0.5 mM NaF, 10 mM b-glycerophosphate, 5 mM sodium pyrophos- phate, 1 mM sodium orthovanadate and the same protease inhibitors as above. Cell lysates were centrifuged at 15 000 g for 15 min at 4°C. Equal amounts of protein (10–20 lg) were subjected to sodium dodecylsulfate – polyacrilamide gel electrophoresis (SDS–PAGE) and electrophoretically transferred to polyvinylidene difluoride (PVDF) membranes (Immobilon-P, Millipore Corporation, Bedford, MA, USA). Blots were blocked in 5%-non-fat dry milk/TBST buffer (20 mM Tris-HCl, 0.15 M NaCl, 0.1% Tween-20) and immunoblotted for: total ERK1/2, phosphothreonine 202/phospho- tyrosine 204-ERK1/2, total MEK1/2, phosphoserine 217/221 MEK1/2, total Akt, phosphoserine 473 Akt, Ras, b-tubulin, or phosphoserine 396-Tau. Blots were then labelled with anti-mouse IgG : HRP or anti-rabbit IgG : HRP, incubated for 1 min with ECL reagent (ECL, Amersham-Pharmacia) according to the supplier’s instructions, exposed to Hyperfilm ECL and developed. Quantifi- cation was performed by densitometric scaning of the film using a GS-700 Imaging Densitometer with Multi-Analyst 1.1 software (Bio-Rad Laboratories, Hercules, CA, USA). [3 H]Thymidine incorporation into DNA As an indication of mitogenic induction in astrocytes, we measured the incorporation of [methyl-3 H]thymidine into DNA. Astrocytes were left 3 days proliferating in FCS-containing medium with or without lithium and/or inhibitors, and pulse-labelled with 1 lCi/mL of [3 H]thymidine during the last 24 h. At the end of the incubation, cells were washed with ice-cold Krebs-Henseleit buffer and treated with ice-cold 1 M HClO4 for 30 min. The acid-precipitated material was washed with 0.4 M HClO4, then with ethanol, and solubilized with 0.5 M NaOH. A 100-lL aliquot was collected for liquid scintillation counting. Cell cycle analysis The cell cycle distribution was determined by flow cytometric analysis of propidium iodide-labelled cells. Briefly, astrocytes were plated in 6-well plates and maintained in FCS-containing medium 28 h prior to treatment. After trypsinization, cells were collected, washed twice in ice-cold PBS, and fixed in ice-cold 70% ethanol. Cells were then washed twice with ice-cold PBS, incubated at 37°C for 30 min with propidium iodide (20 lg/mL) in a PBS solution containing 0.1% (v/v) Triton X-100 and 100 units/mL Rnase A, and analyzed by flow cytometry with a Becton Dickinson FACSCalibur flow cytometer. The results were analyzed with CellQuest and Modfit LT Software. Ras activation Astrocytes were serum-starved for 2 days, and upon mitogen stimulation cells were lysed at 4°C in a buffer containing 20 mM HEPES, pH 7.4, 0.1 M NaCl, 1% Triton X-100, 10 mM EGTA, 40 mM b-glycerophosphate, 20 mM MgCl2, 1 mM Na3VO4, 1 mM dithiothreitol, 10 lg/mL aprotinin, 10 lg/mL leupeptin, and 1 mM PMSF. Lysates were incubated for 30 min with 10 lg of a GST fusion protein including the Ras-binding domain of Raf-1 (RBD) previously bound to glutathione-Sepharose beads (Upstate Bio- technology, Lake Placid, NY, USA), followed by three washes with lysis buffer. Total Ras levels in the cell lysates and GTP- bound forms of Ras associated with GST-Raf-1-RasBD were quantified by western blot analysis using a monoclonal anti-Ras antibody (Oncogene). Statistics Statistical analysis was performed with one-way ANOVA followed by Bonferroni’s multiple comparison test, unless indicated otherwise. Asterisks denote statistical significance: *p < 0.05, **p < 0.01, ***p < 0.001. Results Lithium inhibits DNA synthesis and induces a G2/M cell cycle arrest in asynchronous astrocytes Stimulation of ERK in dividing cells is expected to constitute a mitogenic stimulus (Chang and Karin 2001) and, in fact, pituitary astrocytes have been shown to proliferate in response to lithium (Levine et al. 2000). Thus, we decided to study the effect of lithium on astrocyte DNA synthesis by measuring [3 H]thymidine incorporation. In our initial experi- ments, we found no stimulatory effects of lithium in quiescent, serum-deprived astrocytes (not shown), and looked for effects in subconfluent, asynchronously prolifer- ating astrocytes that had been exposed to various LiCl concentrations for 3 days. Compared with controls, the amount of [3 H]thymidine incorporation into DNA in 5 mM lithium-treated astrocytes was reduced by more than 60%, while with 10 mM the reduction was about 90%, as shown in Fig. 1(a). As lithium is known to interfere with inositol 1,4,5-trisphosphate recycling to myo-inositol, we checked whether lithium inhibitory effects on DNA synthesis in astrocytes could be due to intracellular inositol depletion. Supplementation of the culture medium with 3 mM myo- inositol did not affect lithium inhibition of [3 H]thymidine incorporation, but reverted the potentiation due to 5 mM LiCl towards the accumulation of [3 H]CDP-diacylglycerol in [3 H]cytidine-prelabelled, carbachol-stimulated astrocytes, an effect that mirrors inositol depletion (Godfrey 1989; Claro et al. 1993) (not shown). To further characterize the inhibitory effect of lithium on astrocyte proliferation, we monitored the cell cycle profiles of subconfluent, asynchronous astrocytes that had been treated for 3 days with different LiCl concentrations. As shown in Fig. 1(b), the percentage of tetraploid cells increased with lithium in a dose-dependent manner. The greater effect was observed with 10 mM LiCl, with 17.2% occurrence of cells in the G2/M phase compared with 5.8% observed under control conditions. The cell cycle phase distribution was also monitored after a 7-day lithium treatment and showed similar results (14.3% of cells in G2/M in 5 mM LiCl treated cells, compared with 4.9% in control cells, not shown). No signs of subdiploid (apoptotic cells) were observed in any of the conditions assayed. Also, Lithium inhibits the astrocyte MEK-ERK cascade 419 Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 4. we found no impairment of cell adhesion or decrease of MTT-reducing ability (Mosmann 1983) in lithium-treated cells, indicating that lithium-induced cell cycle arrest occurred in the absence of cytotoxicity. Lithium reduces ERK and MEK phosphorylation in astrocytes We hypothesized that lithium effects on astrocyte DNA synthesis and cell cycle could be due to the inhibition of MEK/ERK signalling. As shown in Fig. 2(a), a 7-day treatment of asynchronously proliferating astrocytes with lithium resulted in an effective inhibition of ERK and MEK phosphorylation. This was already patent at the therapeutic- ally relevant lithium concentration of 1 mM, which reduced some 60 and 50% the phosphorylation of ERK and MEK, respectively, while 10 mM lithium reduced these phosphor- ylated kinases to barely detectable levels. In contrast, lithium concentrations up to 5 mM did not affect significantly the phosphorylation status of Akt, indicating that, rather than being the outcome of a general collapse of the ATP balance, the effect of lithium is somehow selective on the MEK-ERK pathway. We next investigated whether lithium may preclude mitogen activation of the MEK-ERK pathway in serum- deprived, quiescent astrocytes. ET-1 is a well characterized mitogen to astrocytes whose actions are especially relevant on reactive gliosis taking place upon central nervous system damage (Stanimirovic et al. 1995). We considered that this experimental approach may reflect more closely the reactive astrogliosis occurring in vivo after traumatic disruption of the blood–brain barrier. Astrocytes were forced to enter G0 and remain quiescent by switching to FCS-free medium for 24 h, and were further maintained in this medium for 7 days in the presence of different LiCl concentrations, then challenged with ET-1 for 3 h. As shown in Fig. 2(b), lithium prevented ET-1-stimulated phosphorylation of ERK in a dose-dependent manner. A therapeutically relevant lithium concentration of 1 mM reduced phospho-ERK levels by 35%, while at 5 mM some 60% inhibition was achieved. These effects were also apparent at shorter periods of lithium exposure (6–24 h), but they required 10 mM lithium con- centrations, as shown in Fig. 2(c). Taken together, the results outlined above demonstrate a time- and concentration- dependent inhibition of MEK and ERK phosphorylation in astrocytes. GSK-3 inhibition does not account for lithium effects on MEK-ERK and cell cycle To gain insight into the pharmacological target underlying lithium effects, we tested whether GSK-3 inhibition in astrocytes could be involved in cell cycle arrest or in the inhibition of the MEK-ERK pathway. For this purpose, we took advantage of the recently developed compound SB216763, which has been shown to inhibit GSK-3 and protect neurons from apoptosis induced by inhibition of phosphatidylinositol 3-kinase (Cross et al. 2001). As shown in Table 1, treatment of quiescent astrocytes with serum or ET-1 induced a synchronized cell cycle re-entry, taking in both cases 24 h to increase significantly the per cent occurrence of tetraploid cells (G2/M phase). In astrocytes exposed to 10 mM lithium for 24 h before mitogen addition, there was a decreased occurrence of cells in S phase and also a delayed appearance of the tetraploid cell population, which took 48 h to increase. In contrast, SB 216763 did not change the cell cycle distribution of ET-1-stimulated astrocytes at any time point (Table 1). Further, it did not alter the phosphorylation status of MEK and ERK (not shown), although treatment of astrocytes with this compound under (a) (b) Fig. 1 Lithium inhibits astrocyte DNA synthesis and induces a G2/M cell cycle arrest. (a) [3H]Thymidine incorporation in asynchronously growing astrocytes. Subconfluent cultures were treated with LiCl at the indicated doses for 3 days. The cells were pulse-labelled with 1 lCi/mL of [3H]thymidine during the last 24 h of treatment. [3H]Thymidine incorporated into DNA was determined as shown in Materials and methods. The results are means ± SEM of three independent experiments. (b) Cell cycle analysis. Asynchronously growing astro- cytes were treated with the indicated concentrations of LiCl for 3 days. After trypsinization, the cells were ethanol fixed and labelled with propidium iodide for quantification of DNA content by flow cytometry. The percentage of cells in each phase of the cell cycle was determined using CellQuest and Modfit LT software. The flow cytometric profiles shown are representative of three independent experiments. 420 Rau´l Pardo et al. Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 5. the same conditions as above induced cytosolic b-catenin stabilization and completely prevented Tau phosphorylation in Ser396 (Fig. 3), consistent with the effective inhibition of GSK-3b (Hart et al. 1998; Ikeda et al. 1998; Torreilles et al. 2000). Inhibition of p38 MAPK partially reverts the inhibitory effects of lithium We investigated the possible participation of the stress signalling kinase p38 MAPK by using its selective inhibitor SB203580. As shown in Fig. 4, this compound did not significantly affect the incorporation of [3 H]thymidine into DNA of asynchronously proliferating astrocytes, contrary to the MEK inhibitors U0126 and PD98059, but was effective to revert partially the inhibition exerted by lithium at 5 mM and 10 mM concentrations. (c) (b) (a) Fig. 2 Lithium prevents signalling through the MEK/ERK pathway. (a) Asynchronously growing secondary astrocytes were incubated for 1 week with the indicated concentrations of LiCl in DMEM containing 10% FCS. Cell extracts were processed as indicated in Materials and methods, and subjected to western blot analysis with anti-phospho- threonine 202/tyrosine 204-ERK 1/2, anti-phosphoserine 217/221 MEK1/2, anti-phosphoserine 473 Akt, or with the corresponding anti- ERK1/2, anti-MEK1/2 or anti-Akt antibodies. The blots shown are representative of five independent experiments that gave similar results. Lower panels show the densitometric analysis of the shown blots. (b) Secondary astrocytes were serum-starved for 24 h in DMEM, and then incubated in the same medium for 1 week in the presence or absence of the indicated LiCl concentrations. Upon sti- mulation with 25 nM ET-1 for 3 h, the cells were lysed and processed for immunoblot analysis. Ten micrograms of protein were loaded on each lane of a SDS–PAGE and immunoblotted for phospho-ERK and total ERK. A densitometric analysis of the western blot shown is included. (c) Subconfluent astrocytes were serum-starved for 24 h, pre-treated for the indicated times with 10 mM LiCl and stimulated with 25 nM ET-1 for 3 h. In the 0 h condition, LiCl and ET-1 were added simultaneously. After stimulation, samples were processed for western blot analysis. Ten micrograms of protein were loaded on each lane of an SDS–PAGE and immunoblotted for phosphoERK, phosphoMEK and the corresponding total proteins. Table 1 Lithium, but not the GSK-3b inhibitor SB216763, delays cell cycle re-entry G0/G1 S G2/M Control 93.6 ± 3.8 3.1 ± 0.8 4.1 ± 0.4 FCS, 20 h 77.6 ± 3.0 18.6 ± 1.1 3.8 ± 0.6 FCS, 24 h 58.1 ± 2.2 21.6 ± 4.0 20.2 ± 3.4 Li+ + FCS 24 h 79.3 ± 4.2b 16.1 ± 2.5 4.9 ± 1.6b ET-1 16 h 84.5 ± 3.8 12.1 ± 1.4 3.5 ± 1.9 ET-1 20 h 81.0 ± 2.5 14.5 ± 2.1 4.5 ± 1.0 ET-1 24 h 79.0 ± 2.8 12.0 ± 1.9 8.8 ± 1.9 ET-1 48 h 94.5 ± 2.6 1.9 ± 1.0 3.5 ± 1.8 Li+ + ET-1 16 h 91.6 ± 3.0 6.5 ± 0.8a 1.9 ± 0.6 Li+ + ET-1 20 h 90.8 ± 2.7a 8.0 ± 1.4a 1.2 ± 0.9a Li+ + ET-1 24 h 88.5 ± 3.3a 8.9 ± 1.6a 2.4 ± 2.0a Li+ + ET-1 48 h 87.1 ± 1.6a 6.1 ± 0.7a 6.8 ± 1.1 SB216763 + ET-1 16 h 84.1 ± 5.2 12.6 ± 2.4 3.8 ± 2.4 SB216763 + ET-1 20 h 80.9 ± 1.3 13.8 ± 2.1 5.6 ± 2.7 SB216763 + ET-1 24 h 80.7 ± 3.2 10.5 ± 2.5 8.8 ± 3.0 SB216763 + ET-1 48 h 96.2 ± 4.1 1.7 ± 1.3 2.1 ± 1.2 Astrocytes were deprived from serum for 3 days and treated with 10 mM LiCl or 3 lM SB216763 for 24 h. Then, cell cycle re-entry in synchronized astrocytes was initiated by addition of 10% FCS or 25 nM ET-1, and cell cycle phase distribution was analyzed at different times after stimulation. Results are reported as per cent occurrence of cells in G0/G1 (diploid), G2/M (tetraploid), or S phase and are means ± SD of two independent experiments. a Significantly different from ET-1-treated cells at the same time point (two-tailed Student’s t-test, p < 0.05);b significantly different from FCS-treated cells at the same time point (two-tailed Student’s t-test, p < 0.05). Lithium inhibits the astrocyte MEK-ERK cascade 421 Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 6. Lithium pre-treatment prevents sustained but not acute activation of MEK and does not affect activation of Ras We next monitored lithium effects on the time course of MEK activation in serum-deprived, quiescent astrocytes challenged with ET-1 (Fig. 5a). 25 nM ET-1 induced a robust phosphory- lation of MEK, reaching a peak at 3–5 min and slowly declining thereafter. Enhanced phosphorylation of MEK could be detected up to 6 h after ET-1 addition, which was the longest time examined. In cells pre-treated for 24 h with 10 mM LiCl before ET-1 stimulation, the rapid, peak activation of MEK remained unaffected, but its long-lasting phosphory- lation was significantly reduced (Fig. 5a). We next monitored the activation of Ras at the time points of peak (3 min) and sustained (3 h) phosphorylation of MEK. As shown in Fig. 5(b), ET-1 (and also EGF) stimulation resulted in a robust increase in GTP-bound Ras at 3 min, while after 3 h the effect C N L U 0126PD 98059SB 203580 Li5m M Li5m M +SB 203580Li10m M Li10m M +SB 203580 0 10000 20000 30000 [3H]Thymidine incorporaion(dpm) Fig. 4 Effect of MEK and p38 MAPK inhibitors on astrocyte DNA synthesis. Astrocytes growing on a 10% FCS-containing DMEM were treated for 3 days with the MEK inhibitors U0126 (10 lM) and PD98059 (50 lM) or the p38 MAPK inhibitor SB203580 (10 lM). The cells were pulsed labelled with [3H]thymidine for the last 24 h of treatment and its incorporation into DNA was determined as indicated in Materials and methods. The results are means ± SEM of three independent experiments performed in triplicate. Asterisks denote statistical significance (p < 0.05) between each lithium condition in the presence or absence of SB203580. Fig. 3 SB216763 stabilizes b-catenin and prevents phosphorylation of Tau on Ser396. Astrocytes growing on a 10% FCS-containing DMEM medium were either left untreated or treated for 3 h with 3 lM SB216763. Cells were hypotonically lysed and the levels of cytosolic b-catenin were determined, or alternatively, cells were lysed in a 0.1% TritonX-100 containing lysis buffer to determine phosphorylation of Tau on Ser396 using a phospho-specific antibody. An antibody against b-tubulin was used in the same western blot as a loading control. This experiment was performed three times with identical results. (a) (b) Fig. 5 Lithium inhibits sustained phosphorylation of MEK by the mitogen ET-1 without affecting the activation of Ras. (a) Astrocytes were serum deprived for 48 h, then stimulated with ET-1 for the indi- cated times and processed for western blot analysis for phospho and total MEK. A subset of cells was incubated for 24 h with 10 mM LiCl prior to stimulation with ET-1. A densitometric analysis of the shown western blot is included. The same experiment was repeated once obtaining essentially the same results. Asterisks denote significance (p < 0.05) as compared with control cells at the same time point, ac- cording to a two-tailed Student’s t-test. (b) Two snapshots of the time course of MEK phosphorylation by ET-1 are shown (3 min and 3 h). The same batch of cells were processed to monitor activation of Ras GTPase. Serum-starved astrocytes were either left untreated or pre- treated for 24 h with 10 mM LiCl, and then stimulated for 3 min or 3 h with either 25 nM ET-1 or 25 ng/mL EGF. To measure active Ras, cells were lysed and incubated with a GST fusion protein including the Ras binding domain of Raf-1 previously bound to glutathione-Sepharose beads. Equal quantities of cell extracts were analyzed to ensure equal amounts of total Ras in the assay. As an internal negative control, some cells were also treated for 10 min with an inhibitor of the kinase activity of the EGR receptor (AG1478, 1 lM) prior to the addition of EGF. 422 Rau´l Pardo et al. Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 7. was smaller but still clear. Interestingly, lithium had no effect on the initial (3 min) or in the late (3 h) phosphorylation of Ras. In contrast, the EGF receptor kinase inhibitor tyrphostin AG1478 completely prevented the EGF stimulated GTP loading of Ras. Taken together, these results indicate that lithium exerts its inhibitory effects on the sustained phos- phorylation of MEK at a level downstream of Ras. Lithium stimulates ERK phosphorylation in neurons In striking contrast to what was found in astrocytes, treatment of cerebelar granule neurons with lithium (1–10 mM) for 24 h resulted in a concentration-dependent increase of the phosphorylation status of ERK and MEK (Fig. 6a). In our conditions, lithium did not affect Akt phosphorylation, which is not in conflict with results from Chalecka-Franaszek and Chuang (1999), as they demonstrated that lithium-stimulated phosphorylation of Akt returns to basal levels after 3 h. Again, stimulation of the neuronal MEK-ERK pathway was neither counteracted by inositol supplementation (not shown) nor mimicked by SB216763, which was as effective as lithium to stabilize neuronal b-catenin (Fig. 6b), arguing against phosphoinositol phosphatases or GSK-3b being involved in these effects. Discussion The present study shows that lithium exerts opposite effects on the MEK-ERK pathway in cortical astrocytes and in cerebelar granule neurons. In cells able to proliferate, MEK- ERK signalling is clearly involved in the transduction of mitogenic stimuli, and therefore it is not surprising that lithium inhibition of MEK-ERK in astrocytes was mirrored by a decreased DNA synthesis, an effect mimicked by MEK inhibitors, U0126 and PD98059, and also by effects on cell cycle distribution. In asynchronously proliferating astrocytes, lithium induced the accumulation of tetraploid cells, consis- tent with a G2/M cycle arrest. Cell cycle effects were clearer in synchronized astrocytes, a model that may reflect more closely the situation in vivo, where astrocytes are quiescent cells and do not proliferate actively unless serum mitogenic factors intrude into the brain parenchima. In serum-deprived cells, lithium inhibited and delayed cell cycle re-entry after stimulation with ET-1. As for the effects on MEK-ERK signalling, a 1-week exposure to lithium at concentrations within the range of therapeutic doses was effective to down- regulate the phosphorylation of these kinases in asynchro- nous, proliferating astrocytes. Similarly, in quiescent astrocytes, lithium inhibited MEK and ERK phosphorylation as induced by ET-1. A more detailed analysis revealed that ET-1 induced a rapid phosphorylation of MEK which peaked after 3–5 min and declined thereafter, but remained elevated up to 6 h. Interestingly, lithium did not affect the initial peak but was effective in reducing the sustained phosphorylation of the kinase, which is relevant for mitogenesis. Mitogens typically induce a sustained phosphorylation of MEK that permits entry and progresion through G1 phase of the cell cycle (Kahan et al. 1992; Meloche et al. 1992) after the induction of cyclin D1, whose association with Cdk4 is considered rate-limiting for cell growth (Whitmarsh and Davis 2000). In contrast, non-mitogens able to stimulate the MEK/ERK cascade elicit a typically transient phosphoryla- tion which is over in less than 15 min (Kahan et al. 1992). Ras activation by ET-1 followed a similar time course to that of MEK phosphorylation, with a very robust loading of Ras with GTP at 3 min and a sustained, long lasting activation at 3 h. However, lithium did not affect either the initial or the long-term activation of Ras by ET-1 or EGF. These results demonstrate that the mechanisms responsible for lithium actions on MEK/ERK operate downstream of Ras. Lithium effects on the cell cycle have been reported before in bovine aortic endothelial cells, where the ion was shown to induce cell cycle arrest by a mechanism involving stabiliza- tion of p53 (Mao et al. 2001). This anti-tumour transcription factor is stabilized by different mechanisms, including the Akt-GSK-3-b-catenin signalling pathway. GSK-3-b-medi- ated mechanisms, however, do not account for our results: we show that the GSK-3 inhibitor SB216763 (Cross et al. 2001) failed to arrest the astrocyte cell cycle or to alter MEK (a) (b) Fig. 6 Lithium induces MEK and ERK phosphorylation in cerebelar granule neurons by a mechanism unrelated to GSK-3b inhibition. (a) Primary cultures of cerebelar granule neurons were treated for 24 h with the indicated LiCl concentrations or with the specific GSK3 inhibitor SB216763 (3 lM), and then cells were lysed and processed for western blot analysis. Twenty micrograms of protein were loaded on each lane. The densitometric analysis of phospho-ERK isoforms, phospho-MEK, and phospho-Akt is included. Open bars represent the effect of SB216763. The results shown are from a significant experi- ment that was repeated twice with similar results. (b) Neuronal lysates treated as above were processed for western blot analysis against b-catenin. Lithium inhibits the astrocyte MEK-ERK cascade 423 Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 8. and ERK phosphorylation, while effectively stabilized b-catenin and inhibited Tau phosphorylation. However, p53 may be involved in lithium-induced astrocyte cell cycle arrest: it has been reported that ERK inhibition contributes with the activation of p38 MAP kinase in the stabilization of p53 induced by nitric oxide (Kim et al. 2002). In fact, lithium has been reported to stimulate p38 in intestinal epithelial cells (Nemeth et al. 2002), although a recent study showed that lithium alone did not cause significant activation of p38 kinase in cerebellar granule neurons (Chen et al. 2003). This discrepancy may well arise from cell-specific mechanisms and/or different lithium treatments, which are typically time and concentration dependent. We have not attempted to measure lithium effects on the astrocyte p38. However, we provide some indirect evidence for the involvement of p38 in lithium-induced inhibition of DNA synthesis in astrocytes, as the effectc is partly reversed by SB203580, suggesting that both ERK inhibition and p38 activation could be contributing to cell cycle arrest. Besides GSK-3, another well-documented pharmacologi- cal target of lithium are the phosphoinositol phosphatases. In this regard, we have observed that addition of inositol to the culture medium does not revert lithium effects on the astrocyte MEK-ERK pathway, and therefore this mechanism does not seem to account for our results either. Inositol supplementation may seem a rather indirect way to test for lithium actions mediated by phosphoinositol phosphatase inhibition. However, it is fully effective to revert lithium- dependent, agonist-stimulated accumulation of CDP-diacyl- glycerol, a metabolite whose accumulation correlates with restricted inositol availavility (Godfrey 1989; Claro et al. 1993; Centeno et al. 1998), and its use is justified by the absence of selective inositol monophosphatase inhibitors other than the poorly permeable L-690 330 (Atack et al. 1993). Regarding lithium stimulatory effects on the MEK/ERK cascade in cerebelar granule neurons, our results confirm those of Kopnisky et al. (2003), and rule out the possible implication of GSK-3 and phosphoinositol phosphatases. A similar effect was found in SH-SY5Y neuroblastoma cells treated with valproate (Yuan et al. 2001). In that case, stimulatory effects of valproate were shown to take place well upstream of the pathway, as the drug was ineffective in dominant negative Raf and Ras mutants. This illustrates that, if lithium and valproate effects in neurons are mediated by a common mechanism, this is not simply the opposite to that taking place in astrocytes. It is well known that both lithium and valproate stimulate AP-1 binding activities (Ozaki and Chuang 1997; Asghari et al. 1998; Chen et al. 1999b). Thus, as ERK is involved in the activation of AP-1 (Frost et al. 1994), stimulation of neuronal ERK could contribute substantially to the modulation of gene expression and plasticity by anti-bipolar drugs. Interestingly, lithium has been shown recently to stimulate the proliferation of cerebelar and cortical neuroblasts (Hashimoto et al. 2003). Although the molecular mechanism remains to be estab- lished, it is tempting to speculate that, conversely to the effects of lithium in astrocytes, ERK stimulation may underlie neuroblast mitogenesis. To our knowledge, the report from Levine et al. (2000) is the only one dealing with lithium effects in vivo on glial cell proliferation and, in that case, lithium treatment resulted in an estimulated proliferation of pituicytes, or astrocytes from the neural lobe of the pituitary. We just do not know the reason for our apparently conflicting results that might arise simply from the fact that the pituitary and the cerebral cortex are quite different neural areas. Besides the astrocyte in vitro is a model rather different from that in vivo. That is why it would be worth testing whether lithium, a drug with decades of therapeutical history which favours neuronal survival and plasticity and, as we show here, also inhibits astrocyte proliferation in vitro, would be also effective in vivo. This might represent a potential treatment of CNS traumatic injuries: these result in a rapid proliferative response from resident astrocytes, a process known as reactive astrogliosis or glial scarring, that has been suggested to be an attempt made by the CNS to restore homeostasis through isolation of the damaged region (Fitch and Silver 1997). Upon CNS damage involving destruction of the brain–blood barrier, endothelium-derived ET-1 and plasma components intrude into parenchyma and initiate astrogliosis. The subsequent formation of a glial scar may interfere with any subsequent neural repair (McGraw et al. 2001). Current experimental approaches to attenuate gliotic reaction, however, do not constitute viable therapeutic interventions. In this context, lithium therapy could contribute to neurological recovery owing to its cytoprotective and neuroplastic action, which has been recently demonstrated in an ischaemia/reperfusion model (Ren et al. 2003), and the inhibition of reactive astrogliosis. Acknowledgements RP is recipient of a pre-doctoral fellowship from Universitat Auto`noma de Barcelona. AG and BR are recipients of pre-doctoral fellowships from the Spanish Ministry of Science and Technology (SMST). This work has been supported by Grant BFI 2001–2017 from the SMST. We appreciate the generous gift of compound SB216763 by GlaxoSmithKline, Stevenage, Herts, UK, the help of Dr Joan-Marc Servitja with setting up the assay of GTP-bound Ras, and the excellent technical assistance of Cristina Gutie´rrez with cell culture and Manuela Costa with the FACS facility. References A´ lvarez G., Mun˜oz-Montano J. R., Satru´stegui J., A´ vila J., Bogo´nez E. and Dı´az-Nido J. (1999) Lithium protects cultured neurons against beta-amyloid-induced neurodegeneration. FEBS Lett. 453, 260– 264. 424 Rau´l Pardo et al. Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 9. Asghari V., Wang J. F., Reiach J. S. and Young L. T. (1998) Differential effects of mood stabilizers on Fos/Jun proteins and AP-1 DNA binding activity in human neuroblastoma SH-SY5Y cells. Mol. Brain Res. 58, 95–102. Atack J. R. (1996) Inositol monophosphatase, the putative therapeutic target for lithium. Brain Res. Rev. 22, 183–190. Atack J. R., Cook S. M., Watt A. P., Fletcher S. R. and Ragan C. I. (1993) In vitro and in vivo inhibition of inositol monophosphatase by the bisphosphonate L-690,330. J. Neurochem. 60, 652–658. Berridge M. J., Downes C. P. and Hanley M. R. (1989) Neural and developmental actions of lithium: a unifying hypothesis. Cell 59, 411–419. Centeno F., Mora A., Fuentes J. M., Soler G. and Claro E. (1998) Partial lithium-associated protection against apoptosis induced by C2-cer- amide in cerebellar granule neurons. Neuroreport 9, 4199–4203. Chalecka-Franaszek E. and Chuang D. M. (1999) Lithium activates the serine/threonine kinase Akt-1 and suppresses glutamate-induced inhibition of Akt-1 activity in neurons. Proc. Natl Acad. Sci. USA 96, 8745–8750. Chang L. and Karin M. (2001) Mammalian MAP kinase signalling cascades. Nature 410, 37–40. Chen G., Huang L. D., Jiang Y. M. and Manji H. K. (1999a) The mood- stabilizing agent valproate inhibits the activity of glycogen syn- thase kinase-3. J. Neurochem. 72, 1327–1330. Chen G., Yuan P. X., Jiang Y. M., Huang L. D. and Manji H. K. (1999b) Valproate robustly enhances AP-1 mediated gene expression. Mol. Brain Res. 64, 52–58. Chen R. W., Qin Z. H., Ren M., Kanai H., Chalecka-Franaszek E., Leeds P. and Chuang D. M. (2003) Regulation of c-Jun N-terminal kin- ase, p38 kinase and AP-1 DNA binding in cultured brain neurons: roles in glutamate excitotoxicity and lithium neuroprotection. J. Neurochem. 84, 566–575. Claro E., Fain J. N. and Picatoste F. (1993) Noradrenaline stimulation unbalances the phosphoinositide cycle in rat cerebral cortical sli- ces. J. Neurochem. 60, 2078–2086. Cross D. A., Culbert A. A., Chalmers K. A., Facci L., Skaper S. D. and Reith A. D. (2001) Selective small-molecule inhibitors of glycogen synthase kinase-3 activity protect primary neurones from death. J. Neurochem. 77, 94–102. D’Mello S. R., Anelli R. and Calissano P. (1994) Lithium induces apoptosis in immature cerebellar granule cells but promotes sur- vival of mature neurons. Exp. Cell Res. 211, 332–338. De Sarno P., Li X. and Jope R. S. (2002) Regulation of Akt and glycogen synthase kinase-3beta phosphorylation by sodium valproate and lithium. Neuropharmacology 43, 1158–1164. Drevets W. C., Price J. L., Simpson J. R. Jr, Todd R. D., Reich T., Vannier M. and Raichle M. E. (1997) Subgenual prefrontal cortex abnormalities in mood disorders. Nature 386, 824–827. Fitch M. T. and Silver J. (1997) Glial cell extracellular matrix: bound- aries for axon growth in development and regeneration. Cell Tissue Res. 290, 379–384. Frost J. A., Geppert T. D., Cobb M. H. and Feramisco J. R. (1994) A requirement for extracellular signal-regulated kinase (ERK) func- tion in the activation of AP-1 by Ha-Ras, phorbol 12-myristate 13-acetate, and serum. Proc. Natl Acad. Sci. USA 91, 3844–3848. Godfrey P. P. (1989) Potentiation by lithium of CMP-phosphatidate formation in carbachol-stimulated rat cerebral-cortical slices and its reversal by myo-inositol. Biochem. J. 258, 621–624. Grewal S. S., York R. D. and Stork P. J. S. (1999) Extracellular-signal- regulated kinase signalling in neurons. Curr. Opin. Neurobiol. 9, 544–553. Grimes C. A. and Jope R. S. (2001) The multifaceted roles of glycogen synthase kinase 3 beta in cellular signaling. Prog. Neurobiol. 65, 391–426. Hall A. C., Lucas F. R. and Salinas P. C. (2000) Axonal remodeling and synaptic differentiation in the cerebellum is regulated by WNT-7a signaling. Cell 100, 525–535. Hall A. C., Brennan A., Goold R. G., Cleverley K., Lucas F. R., Gordon- Weeks P. R. and Salinas P. C. (2002) Valproate regulates GSK- 3-mediated axonal remodeling and synapsin I clustering in developing neurons. Mol. Cell Neurosci. 20, 257–270. Hart M. J., de los Santos R., Albert I. N., Rubinfeld B. and Polakis P. (1998) Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr. Biol. 8, 573–581. Hashimoto R., Senatorov V., Kanai H., Leeds P. and Chuang D. M. (2003) Lithium stimulates progenitor proliferation in cultured brain neurons. Neuroscience 117, 55–61. Ikeda S., Kishida S., Yamamoto H., Murai S., Koyama S. and Kikuchi A. (1998) Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J. 17, 1371–1384. Jope R. S. (1999) Anti-bipolar therapy: mechanism of action of lithium. Mol. Psychiatry 4, 117–128. Kahan C., Seuwen K., Meloche S. and Poysse´gur J. (1992) Coordinate biphasic activation of p44 mitogen activated protein kinase and S6 kinase by growth factors in hamster fibroblasts. J. Biol. Chem. 267, 13369–13375. Kim S. J., Ju J. W., Oh C. D., Yoon Y. M., Song W. K., Kim J. H., Yoo Y. J., Bang O. S., Kang S. S. and Chun J. S. (2002) ERK-1/2 and p38 kinase oppositely regulate nitric oxide-induced apoptosis of chondrocytes in association with p53, caspase-3, and differenti- ation status. J. Biol. Chem. 277, 1332–1339. Klein P. S. and Melton D. A. (1996) A molecular mechanism for the effect of lithium on development. Proc. Natl Acad. Sci. USA 93, 8455–8459. Kopnisky K. L., Chalecka-Franaszek E., Gonzalez-Zulueta M. and Chuang D. M. (2003) Chronic lithium treatment antagonizes glu- tamate-induced decrease of phosphorylated CREB in neurons via reducing protein phosphatase 1 and increasing MEK activities. Neuroscience 116, 425–435. Levine S., Saltzman A. and Klein A. W. (2000) Proliferation of glial cells in vivo induced in the neural lobe of the rat pituitary by lithium. Cell. Prolif. 33, 203–207. Mao C. D., Hoang P. and DiCorleto P. E. (2001) Lithium inhibits cell cycle progression and induces stabilization of p53 in bovine aortic endothelial cells. J. Biol. Chem. 276, 26180–26188. Masgrau R., Servitja J. M., Sarri E., Young K. W., Nahorski S. R. and Picatoste F. (2000) Intracellular Ca2+ stores regulate muscarinic receptor stimulation of phospholipase C in cerebellar granule cells. J. Neurochem. 74, 818–826. McGraw J., Hiebert G. W. and Steeves J. D. (2001) Modulating astro- gliosis after neurotrauma. J. Neurosci. Res. 63, 109–115. Meloche S., Seuwen K., Pages G. and Poysse´gur J. (1992) Biphasic and synergistic activation of p44 MAPK (ERK1) by growth factors: correlation between late phase activation. Mol. Endocrinol. 6, 845– 854. Mora A., Sabio G., Gonza´lez-Polo R. A., Cuenda A., Alessi D. R., Alonso J. C., Fuentes J. M., Soler G. and Centeno F. (2001) Lithium inhibits caspase 3 activation and dephosphorylation of PKB and GSK3 induced by K+ deprivation in cerebellar granule cells. J. Neurochem. 78, 199–206. Mosmann T. (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Meth 65, 55–63. Nemeth Z. H., Deitch E. A., Szabo C., Fekete Z., Hauser C. J. and Hasko G. (2002) Lithium induces NF-kappa B activation and interleukin- Lithium inhibits the astrocyte MEK-ERK cascade 425 Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426
  • 10. 8 production in human intestinal epithelial cells. J. Biol. Chem. 277, 7713–7719. Nonaka S. and Chuang D. M. (1998) Neuroprotective effects of chro- nic lithium on focal cerebral ischemia in rats. Neuroreport 9, 2081– 2084. Nonaka S., Katsube N. and Chuang D. M. (1998) Lithium protects rat cerebellar granule cells against apoptosis induced by anticonvul- sants, phenytoin and carbamazepine. J. Pharmacol. Exp. Ther. 286, 539–547. Ongur D., Drevets W. C. and Price J. L. (1998) Glial reduction in the subgenual prefrontal cortex in mood disorders. Proc. Natl Acad. Sci. USA 95, 13290–13295. Ozaki N. and Chuang D. M. (1997) Lithium increases transcription factor binding to AP-1 and cyclic AMP-responsive element in cultured neurons and rat brain. J. Neurochem. 69, 2336–2344. Phiel C. J. and Klein P. S. (2001) Molecular targets of lithium action. Annu. Rev. Pharmacol. Toxicol. 41, 789–813. Ren M., Senatorov V. V., Chen R. W. and Chuang D. M. (2003) Post- insult treatment with lithium reduces rat brain damage and facili- tates neurological recovery in a rat ischemia/reperfusion model. Proc. Natl Acad. Sci. USA 100, 6210–6215. Servitja J. M., Masgrau R., Sarri E. and Picatoste F. (2000) Effects of oxidative stress on phospholipid signaling in rat cultured astrocytes and brain slices. J. Neurochem. 75, 788–794. Stambolic V., Ruel L. and Woodgett J. R. (1996) Lithium inhibits gly- cogen synthase kinase-3 activity and mimics wingless signalling in intact cells. Curr. Biol. 6, 1664–1668. Stanimirovic D. B., Ball R., Mealing G., Morley P. and Durkin J. P. (1995) The role of intracellular calcium and protein kinase C in endothelin- stimulated proliferation of rat type I astrocytes. Glia 15, 119–130. Torreilles F., Roquet F., Granier C., Pau B. and Mourton-Gilles C. (2000) Binding specificity of monoclonal antibody AD2: influence of the phosphorylation state of tau. Mol. Brain Res. 78, 181–185. Whitmarsh A. J. and Davis R. J. (2000) A central control for cell growth. Nature 403, 255–256. Williams R. S., Cheng L., Mudge A. W. and Harwood A. J. (2002) A common mechanism of action for three mood-stabilizing drugs. Nature 417, 292–295. Yuan P.-X., Huang L.-D., Jiang Y.-M., Gutkind J. S., Manji H. K. and Chen G. (2001) The mood stabilizer valproic acid activates mito- gen-activated protein kinases and promotes neurite growth. J. Biol. Chem. 276, 31674–31683. 426 Rau´l Pardo et al. Ó 2003 International Society for Neurochemistry, J. Neurochem. (2003) 87, 417–426