You are on page 1of 12

J Mol Neurosci

DOI 10.1007/s12031-012-9884-4

α-Lipoic Acid Interaction with Dopamine D2


Receptor-Dependent Activation of the Akt/GSK-3β Signaling
Pathway Induced by Antipsychotics: Potential Relevance
for the Treatment of Schizophrenia
Jessica Deslauriers & Christian Desmarais &
Philippe Sarret & Sylvain Grignon

Received: 1 August 2012 / Accepted: 3 September 2012


# Springer Science+Business Media, LLC 2012

Abstract Chronic administration of antipsychotics has interaction. Regarding Akt/GSK-3β downstream targets, anti-
been associated with dopamine D2 receptor (D2R) upregu- psychotics increased β-catenin levels, whereas (±)-α-lipoic
lation and tardive dyskinesia. We have previously shown acid induced an elevation of mTOR activation. These results
that haloperidol, a first-generation antipsychotic (FGA), suggest (1) that the effect of antipsychotics on the Akt/GSK-
exerted an increase in D2R expression and oxidative stress 3β pathway in SH-SY5Y cells is reminiscent of their in vivo
and that (±)-α-lipoic acid reversed its effect. Previous stud- action, (2) that (±)-α-lipoic acid partially synergizes with
ies have implicated the Akt/glycogen synthase kinase-3β antipsychotic drugs (APDs) on the same pathway, and (3) that
(GSK-3β) signaling pathway in antipsychotic action. These the Akt/GSK-3β signaling cascade is not involved in the
findings led us to examine whether the Akt/GSK-3β path- preventive effect of (±)-α-lipoic acid on antipsychotics-
way was involved in D2R upregulation and oxidative induced D2R upregulation.
stress elicited by antipsychotics and, in (±)-α-lipoic
acid-induced reversal of these phenomena, in SH- Keywords Haloperidol . (±)-α-Lipoic acid . Dopamine D2
SY5Y cells. Antipsychotics increased phosphorylation receptor . Oxidative stress . Akt . Glycogen synthase kinase-3β
of Akt and GSK-3β, and additive effects were observed with
(±)-α-lipoic acid. GSK-3β inhibitors reversed haloperidol-
induced overexpression of D2R mRNA levels but did not Introduction
affect haloperidol-induced oxidative stress. Sustained antipsy-
chotic treatment increased β-arrestin-2 and D2R receptor Antipsychotic drugs (APDs) are dopamine D2 receptor
(D2R) antagonists or partial agonists and represent the
mainstay of the pharmacological treatment of schizophrenia.
J. Deslauriers : C. Desmarais : P. Sarret : S. Grignon APDs are classified as either typical antipsychotic (first-
Department of Physiology and Biophysics, Faculty of Medicine generation antipsychotic, FGA) or atypical antipsychotic,
and Health Sciences, Université de Sherbrooke, depending on their receptor affinity profiles and their
12e avenue Nord,
related-side effects (Richelson 1999). Atypical antipsy-
Sherbrooke, QC J1H 5N4, Canada
chotics include second-generation antipsychotics (SGAs)
S. Grignon and, recently, third-generation antipsychotics (partial D2R
Department of Psychiatry, agonists) (Lieberman 2004; Mailman and Murthy 2010).
Centre Hospitalier Universitaire de Sherbrooke,
Chronic administration of APDs, mainly FGAs, has been
580 Bowen Sud,
Sherbrooke, QC J1G 2E8, Canada associated with tardive dyskinesia, a serious neurological
syndrome characterized by motor disorders, most common-
S. Grignon (*) ly affecting the orofacial region (Jeste et al. 1999). Despite a
Department of Psychiatry, CHUS Hôtel-Dieu,
high incidence (20–40 %) in patients treated chronically
580 rue Bowen Sud,
Sherbrooke, QC J1G 2E8, Canada with FGAs (Morgenstern and Glazer 1993), the pathophys-
e-mail: Sylvain.Grignon@USherbrooke.ca iology of tardive dyskinesia is not well understood (Paulson
J Mol Neurosci

2005), with current pathogenic hypotheses pointing to D2R GSK-3β signaling pathway was involved in D2R upregula-
hypersensitivity and striatal toxicity possibly associated tion elicited by APD application and in its reversal by (±)-α-
with oxidative stress (Harrison 1999). lipoic acid.
Chronic treatment with haloperidol, an FGA with high-
affinity for D2R, has been associated in preclinical models
with D2R upregulation and hypersensitivity (Jenner et al. Material and Methods
1985). Imaging studies in clinical populations suggest that
chronic treatment (Laruelle et al. 1992) with antipsychotics Materials
is also associated with an increase in striatal D2R binding
potential, which appears to correlate with poor treatment Mouse monoclonal GSK-3β (1F7), rabbit polyclonal
response and occurrence of neurological side effects phospho-GSK-3β (Ser9), mouse monoclonal DRD2 (B-
(Schroder et al. 1998; Silvestri et al. 2000). Tardive dyski- 10), rabbit polyclonal β-arrestin-2 (H-47), goat anti-
nesia has also been linked to oxidative status on the basis of mouse IgG-HRP, and goat anti-rabbit IgG-HRP antibodies
clinical studies both in the cerebrospinal fluid (Lohr et al. were purchased from Santa Cruz Biotechnology (Santa
1990; Peet et al. 1993; Tsai et al. 1998) and in the systemic Cruz, CA, USA). Rabbit polyclonal Akt, rabbit polyclonal
compartment (Lohr et al. 2003); this relationship has been phospho-Akt (Ser473), rabbit monoclonal phospho-mTOR
amply replicated in animal models where interventional (Ser2448), and rabbit polyclonal mTOR antibodies were
studies, including lipoic acid (see below), aimed at normal- purchased from Cell Signaling Technology (Pickering,
izing oxidative status have generally improved dyskinetic ON, Canada). Mouse monoclonal β-actin antibody, haloper-
phenomena (Kulkarni and Naidu 2001; Shamir et al. 2001; idol, amisulpride, (±)-α-lipoic acid, lithium chloride, AR-
Abilio et al. 2003; Naidu et al. 2003; Burger et al. 2005; 014418, and dihydroethidium (DHE) were all acquired from
Bishnoi et al. 2008; Thaakur and Himabindhu 2009). Of Sigma-Aldrich (Oakville, ON, Canada).
note, FGAs appear to be less disruptive vis-à-vis the oxida-
tive status than SGAs, which parallels their respective pro- Cell Culture and Treatment
pensity to induce tardive dyskinesia (Dakhale et al. 2004;
Kropp et al. 2005; Pillai et al. 2007; Correll and Schenk As described previously (Deslauriers et al. 2011), an undif-
2008). Although the evidence is conflicting, interventional ferentiated human SH-SY5Y neuroblastoma cell line
studies suggest that the antioxidant vitamin E (α-tocopher- (American Type Culture Collection, Manassas, VA, USA)
ol) improves tardive dyskinesia, at least in its early phase was cultured in Dulbecco’s Modified Eagle’s Media
(Soares and McGrath 1999). Altogether, these findings sug- (DMEM) with 4.5 mg/L D-glucose, 2 mM L-glutamine,
gest that oxidative stress has a place in the pathogenesis of 110 mg/mL sodium pyruvate (Invitrogen, Burlington, ON,
tardive dyskinesia. Canada) and supplemented with 10 % fetal bovine serum,
In cultured undifferentiated human SH-SY5Y neuroblasto- 100 U/mL penicillin, 100 μg/mL streptomycin, and MEM
ma, we have previously reported that haloperidol, an FGA nonessential amino acids (all media supplements were pur-
prone to increase oxidative stress, elicited a more robust chased from Wisent Bioproducts, St-Bruno, QC, Canada).
increase in D2R expression and in oxidative stress biomarker Haloperidol and amisulpride were dissolved in 100 % di-
levels than amisulpride, an SGA, which does not significantly methyl sulfoxide (DMSO; Sigma-Aldrich). Lithium chlo-
influence oxidative status. We also demonstrated that pretreat- ride and AR-A014418, two GSK-3β inhibitors, were also
ment with (±)-α-lipoic acid reversed the effects of APDs on dissolved in 100 % DMSO. (±)-α-Lipoic acid was dissolved
D2R and oxidative stress levels (Deslauriers et al. 2011). in 100 % ethanol. Cells were pretreated for 24 h with
Interestingly (±)-α-lipoic acid, an antioxidant, has shown 200 μM (±)-α-lipoic acid (Jia et al. 2008). For GSK-3β
protective effects in a rat model of tardive dyskinesia inhibition studies, cells were pretreated for 1 h with 1 mM
(Thaakur and Himabindhu 2009). However, the mechanisms lithium chloride (Beaulieu et al. 2009) or 1 μM AR-
by which it prevents haloperidol-induced D2R upregulation A014418 (Bhat et al. 2003). Then, cells were treated with
and, more generally, how it interacts with transduction events 10 nM haloperidol or 100 nM amisulpride (3 h for mRNA
elicited by APDs are currently unknown. analysis or 6 days for protein level determination).
Previous studies have implicated the Akt/glycogen syn-
thase kinase-3β (GSK-3β) downstream signaling pathway Real-Time Polymerase Chain Reaction
of D2R as a key event in the action of APDs (Emamian et al.
2004; Emamian 2012). Indeed, antagonism of D2R by typ- As performed previously (Deslauriers et al. 2011), RNA from
ical and atypical APDs increases the phosphorylation of SH-SY5Y cells was extracted with TRIzol reagent
Akt, thus preventing activation of GSK-3β (Beaulieu et al. (Invitrogen) and quantified using the NanoDrop®
2009). These findings led us to examine whether the Akt/ (NanoDrop Technologies, Wilmington, DE, USA). Reverse
J Mol Neurosci

transcription was done as follows: RNA (1 μg) was incubated for 15 min and supernatant was kept and stored at −20 °C
with 0.02 % (w/v) random primer (Roche Applied Science, until use. Coomassie Plus (Bradford) Protein Assay reagent
Laval, QC, Canada) for 5 min at 70 °C followed by an (Thermo Fisher Scientific, Rockford, IL, USA) was used for
incubation at 42 °C for 60 min with 1 mM dNTPs (Roche measuring protein concentrations, using bovine serum albu-
Applied Science), 20 U RNAse inhibitor (Invitrogen), and min (BSA) as a standard. For western blotting, migration of
30 U AMV reverse transcriptase (Roche Applied Science). soluble proteins (10–30 μg) was done by SDS-PAGE elec-
Amplification of cDNA (1 μL) was done with 12.5 μL SYBR trophoresis (10 % sodium dodecyl sulfate polyacrylamide
Green (Qiagen, Toronto, ON, Canada), 10.5 μL water, and gel) for 90 min at 120 V and proteins were transferred onto
0.5 μL of each primer. For DRD2 amplification, the primers immobilon-P polyvinylidene fluoride (PVDF) transfer
were 5′-TCGTCATCGCTGTCATCGTC-3′ (forward) and 5′- membrane (Millipore, Billerica, MA, USA) for 55 min at
CAGCTGTGTACCTGTCGATG-3′ (reverse) and for 120 mA. Blockade of the membranes was done in 8 % (w/v)
GAPDH amplification, the primers were 5′-CCAAAGTTGT milk powder in Tris-buffered saline (TBS) supplemented
CATGGATGAC-3′ (forward) and 5′-GTGAAGGTCG with 0.2 % (v/v) Tween-20 (TBST) for 90 min at room
GTGTGAACCG-3′ (reverse) (Deslauriers et al. 2011). The temperature. Membranes were incubated with primary anti-
cycling parameters were 95 °C 10 min followed by 40 cycles bodies, diluted in blocking solution, phospho-Akt (Ser473)
(95 °C 15 s, 60 °C 30 s, and 72 °C 45 s). GAPDH was used as (1:500), Akt (1:500), phospho-GSK-3β (Ser9) (1:500),
reference gene. GSK-3β (1:500), β-arrestin-2 (H-47) (1:200), β-catenin
(E-5) (1:500), phospho-mTOR (Ser2448) (1:500), mTOR
Cellular Superoxide Anion Assay (1:500), and β-actin (1:10,000) at 4 °C overnight. After
three washes of 10 min each with TBST, incubation with
Blue fluorescent dye DHE is oxidized by superoxide anion secondary antibody goat anti-mouse IgG-HRP or goat anti-
(O2−) to ethidium, which stains the cytoplasm and nucleus, rabbit IgG-HRP (1:10,000) was done for 90 min at room
and produces a red nuclear fluorescence (Deslauriers et al. temperature. After three additional washes, membranes were
2011). DHE was dissolved in 100 % DMSO for a final developed with Western Lightning Chemiluminescence
concentration of 5 % (w/v) and stocked in −20 °C. SH- Reagent Plus (PerkinElmer, Waltham, MA, USA) and
SY5Y cells were grown on micro cover glasses (22 × Hyperfilm ECL (Amersham Biosciences, Baie d’Urfe, QC,
22 mm) in six-well plate and treated with DHE for a final Canada). NIH ImageJ software was used for densitometric
concentration of 10 μM, 20 min before the end of APDs analysis. β-actin was used as normalization control.
treatment. Then, micro cover glasses were mounted on
microslides (25×75×1 mm) with ProLong® Gold antifade Immunoprecipitation
reagent with DAPI (Invitrogen), following the manufac-
turer’s instructions. The red staining by ethidium and the To study interactions between D2R and β-arrestin-2, we used
nuclear blue staining with DAPI were measured by a con- immunoprecipitation protocol as previously described (Chen
focal microscopy (Olympus Fluoview FV1000) at excitation et al. 2002). Total protein lysate (500 μg) was precleared with
wavelength 543 nm and 405 nm, respectively. Confocal 50 μL protein A agarose (Roche Applied Science) for 1 h at
microscope settings were maintained constant for each se- 4 °C. After centrifugation at 13,000g for 30 min, supernatant
ries of assays and the fluorescence intensity was evaluated was kept. Primary antibody DRD2 (B-10) (10 μL; 2 μg for
using MetaMorph software. Ethidium staining was normal- 500 μg of proteins lysate) was added to supernatant overnight
ized with DAPI fluorescence. at 4 °C. For immunoprecipitation, 50 μL of protein A agarose
was added for 3 h at 4 °C. Immunoprecipitated proteins were
Western Blotting then centrifuged at 500g for 5 min. The pellet was washed
three times with 800 μL RIPA buffer and suspended with
Total protein extracts were prepared from SH-SY5Y cells, 60 μL loading buffer. The proteins was migrated and trans-
as previously described (Deslauriers et al. 2011). Cells were ferred as described above (see “Western Blotting”). After the
washed with D-PBS (Invitrogen), scraped, and lysed on ice blockade, the membranes were incubated overnight at 4 °C
for 15 min with RIPA buffer: 20 mM Tris pH 7.5, 150 mM with either β-arrestin-2 (H-47) (1:200) or DRD2 (B-10)
NaCl, 1 mM EDTA, 1.0 % (v/v) Nonidet-P40, 0.5 % (w/v) (1:500) primary antibody diluted in blocking solution. After
Na-deoxycholate, and 0.1 % (w/v) SDS, supplemented with three washes of 10 min each with TBST, incubation with
SIGMAFAST™ protease inhibitor cocktail 1× (containing 4- secondary antibody goat anti-mouse IgG-HRP or goat anti-
(2-aminoethyl)benzenesulfonyl fluoride hydrochloride rabbit IgG-HRP (1:10,000) was done for 90 min at room
(AEBSF), bestatin hydrochroride, leupeptin, E-64, aproti- temperature. Following three other washes, development
nin, pepstatin A, and phosphoramidon disodium salt) was done as described above. For analysis, ratio of β-
(Sigma-Aldrich). The lysate was centrifuged at 13,000g arrestin-2 blot and DRD2 blot of immunoprecipitation with
J Mol Neurosci

DRD2 was used and normalized with β-arrestin-2 blot. β- GSK-3β signaling pathway was involved in APDs and (±)-α-
actin was used as reference. lipoic effects on D2R mRNA. Accordingly, we assessed the
effects of applying APDs to SH-SY5Y cells on Akt phosphor-
Statistical Analysis ylation at serine 473. In line with their effect in other systems,
treatment for 6 days with either haloperidol or amisulpride
Data are presented as mean±SD. Data were analyzed using increased Akt phosphorylation (+146 %; **P < 0.01
Kruskal–Wallis analysis of variance (ANOVA) followed by and +127 %; ***P<0.001, respectively, compared to control)
post hoc Tukey test. (Fig. 2). We next evaluated the effect of (±)-α-lipoic acid
pretreatment on APDs-induced Akt activation. Preincubation
of SH-SY5Y cells for 24 h with (±)-α-lipoic acid induced a
strong increase in Akt phosphorylation (+149 %; ***P<0.001
Results
compared to control) and increased haloperidol-induced Akt
phosphorylation (+299 %; **P<0.01 and +62 %; #P<0.05
Effects of Antipsychotics on D2R Expression
compared to control and haloperidol alone, respectively)
(Fig. 2). However, (±)-α-lipoic acid did not significantly affect
In agreement with our previously published results, a 3-
the level of Akt phosphorylation achieved by amisulpride
h incubation of the SH-SY5Y human neuroblastoma cell line
(+34 %; P00.46 compared to amisulpride alone) but increased
with haloperidol or amisulpride at saturation, yet clinically
Akt phosphorylation as compared to control +204 %;
relevant concentrations (Kapur et al. 2002; Marchese et al.
**P<0.01 (Fig. 2).
2002; Muller et al. 2007), induced an increase in D2R mRNA
levels, a phenomenon which we have previously shown to be
predictive of D2 receptor (protein) upregulation at 6 days Effects of Antipsychotics and (±)-α-Lipoic Acid on GSK-3β
(Deslauriers et al. 2011). Ten nanomolars of haloperidol for Phosphorylation
3 h induced a more robust increase in D2R mRNA levels
(+33 %; ***P<0.001) (Fig. 1) than treatment with 100 nM Along the same pathway, it has been shown that Akt phos-
amisulpride (+11 %; *P<0.05) (Fig. 1). This pattern was not phorylation inhibits GSK-3β activation by its phosphorylation
altered by higher amisulpride concentrations (1 μM and
10 μM; Fig. 1): therefore, it likely reflects intrinsic properties
of the molecules rather than differences in receptor occupancy.

Effects of Antipsychotics and (±)-α-Lipoic Acid on Akt


Phosphorylation

It has been previously reported that the Akt signaling pathway


participates in the therapeutic effects of APDs (Beaulieu et al.
2009). We, therefore, set out to investigate whether the Akt/

Fig. 2 Effects of haloperidol (HAL), amisulpride (AMI), and (±)-α-


lipoic acid (LA) on Akt phosphorylation at Ser473 and total Akt ratio
Fig. 1 Effects of haloperidol (HAL) and different doses of amisulpride as compared to untreated SH-SY5Y cells (**P<0.01; ***P<0.001)
(AMI) on dopamine D2 receptor (D2R) mRNA level, as compared to and to haloperidol alone (#P<0.05) (n010–11). Representative immu-
untreated SH-SY5Y neuroblastoma cells (CTL) (**P<0.01) (n06) noblots of phospho-Akt (Ser473), Akt, and β-actin are shown
J Mol Neurosci

on serine 9 (Cross et al. 1995; Doble and Woodgett 2003). We GSK-3β activation, or with 1 μM AR-A014418, a selective
then evaluated the effects of APDs and (±)-α-lipoic acid on GSK-3β inhibitor (Bhat et al. 2003). Pretreatment with
GSK-3β phosphorylation at serine 9. Treatment of SH-SY5Y lithium chloride or AR-A014418 significantly reduced the
cells for 6 days with haloperidol or amisulpride increased D2R mRNA increase induced by haloperidol application
GSK-3β phosphorylation (+66 %; *P<0.05 and +78 %; (−19 %; #P < 0.05 and −28 %; ##P < 0.01 compared to
***P<0.001, respectively) (Fig. 3). (±)-α-Lipoic acid alone haloperidol alone, respectively) (Fig. 4). Furthermore, AR-
had no significant effect on the level of GSK-3β phosphory- A014418 reversed amisulpride-induced increase in D2R
lation (Fig. 3). Furthermore, pretreatment for 24 h with (±)-α- mRNA levels (−26 %; ##P<0.01 compared to amisulpride
lipoic acid did not affect the level of GSK-3β phosphorylation alone), whereas lithium chloride had no significant effect
induced by haloperidol or amisulpride. Therefore, it appeared (Fig. 4). When applied alone, GSK-3β inhibitors did not
that (±)-α-lipoic acid exerted a partial additive effect with change D2R mRNA levels. Altogether, these results
APDs on Akt phosphorylation, which did not reflect on revealed that the Akt/GSK-3β signaling pathway is more
GSK-3β phosphorylation. Based on these results, it seems that specifically involved in the regulation of D2R expression
(±)-α-lipoic acid exerts its beneficial effects on antipsychotic- induced by APDs.
induced tardive dyskinesia through an Akt/GSK-3β-indepen-
dent signaling pathway.
Lithium Effect is Not Mediated by Oxidative Status
Normalization
Effects of GSK-3β Inhibition on D2R Expression
Although GSK-3β inhibition is a common property of lith-
We next investigated whether inhibition of the Akt/GSK-3β
ium and AR-A014418, it is also known that subacute (Cui et
signaling pathway attenuated haloperidol-induced D2R
al. 2007) or chronic (Nciri et al. 2012) lithium treatment
overexpression. The functional involvement of GSK-3β
increases antioxidant capacities in this cell line. Given the
was assessed by pretreating cells (1 h before APD treatment)
significant impact of oxidative status on the D2R upregula-
with 1 mM lithium chloride, which is known to inhibit
tion induced by APDs (Deslauriers et al. 2011), we assessed
the effect of lithium chloride on oxidative status by measur-
ing superoxide anion production by SH-SY5Y cells using a
DHE-derived fluorescence assay. Acute treatment for 3 h
with 100 nM haloperidol increased superoxide anion pro-
duction (+70 %; **P < 0.01), whereas incubation with
100 nM amisulpride had no significant effect (Fig. 5).

Fig. 3 Effects of haloperidol (HAL), amisulpride (AMI), and (±)-α- Fig. 4 Effects of haloperidol (HAL), amisulpride (AMI), lithium
lipoic acid (LA) on GSK-3β phosphorylation at Ser9 and total GSK- chloride (LiCl), and AR-A014418 (AR) on dopamine D2 receptor
3β ratio as compared to untreated SY-SY5Y cells (*P<0.05; ***P< (D2R) mRNA levels, as compared to untreated SH-SY5Y human
0.001) (n010–11). Representative immunoblots of phospho-GSK-3β neuroblastoma cells (*P<0.05; ***P<0.001) and to haloperidol or
(Ser9), GSK-3β, and β-actin are shown amisulpride alone (#P<0.05; ##P<0.01) (n010–12)
J Mol Neurosci

Pretreatment with 1 mM lithium chloride for 1 h before β-Arrestin 2-Dependent Effects of Antipsychotics
APD application was not able to reverse haloperidol- and (±)-α-Lipoic Acid
induced superoxide anion production (+84 %; *P<0.05
compared to control) and even potentiated amisulpride- Upstream of the previous events, D2R stimulation inhibits
induced effect (+27 %; *P<0.05 compared to amisulpr- Akt signaling through dephosphorylation via a G protein-
ide alone) (Fig. 5). Furthermore, lithium chloride ap- independent mechanism implying the β-arrestin-2/phospha-
plied alone to SH-SY5Y cells did not change the basal tase PP2A complex (Beaulieu et al. 2004; Beaulieu et al.
level of superoxide anion. These data, thus, revealed 2005, 2006). We, thus, investigated whether APDs led to
that the effects of APDs on oxidative stress are GSK- changes in D2R–β-arrestin-2 interactions. As compared
3β-independent. Consequently, the effects of lithium on with control, treatment for 6 days with haloperidol increased
D2R levels do not depend on the normalization of the interaction between β-arrestin-2 and D2R (+35 %; *P<
oxidative status. 0.05) (Fig. 6). Preincubation for 24 h with (±)-α-lipoic acid,

Fig. 5 Effects of haloperidol


(HAL), amisulpride (AMI), and
lithium chloride (LiCl) on su-
peroxide anion production as
compared to untreated SH-
SY5Y cells (*P<0.05; **P<
0.01). DAPI stains the nucleus
in blue and ethidium, the prod-
uct of the oxidation of dihy-
droethidium (DHE) by
superoxide anion, produces a
red fluorescence (n08)
J Mol Neurosci

Fig. 7 Effects of haloperidol (HAL), amisulpride (AMI), and (±)-α-


lipoic acid (LA) on β-catenin protein level as compared to untreated
SH-SY5Y cells (*P < 0.05; **P < 0.01) (n 09–10). Representative
Fig. 6 Effects of haloperidol (HAL) and (±)-α-lipoic acid (LA) on β- immunoblots of β-catenin and β-actin are shown
arrestin-2 and dopamine D2 receptor (D2R) interaction, as compared to
untreated SH-SY5Y cells (*P<0.05) (n08). Representative blots of β-
arrestin-2 blot (IP DRD2 IB β-arr) and DRD2 blot (IP DRD2 IB mTOR level at serine residue 2448 to total mTOR level was
DRD2) on immunoprecipitated DRD2 and blot of β-arrestin (IB β- used to evaluate mTOR activation. As compared to control,
arr) are shown
haloperidol and amisulpride did not induce any significant
effect on mTOR activation (Fig. 8). (±)-α-Lipoic acid, com-
either alone or in association with haloperidol, did not bined to haloperidol, increased mTOR phosphorylation
significantly modify the interaction between β-arrestin-2 (+110 %; *P<0.05 compared to control, and +133 %; #P<
and D2R (Fig. 6). 0.05 compared to haloperidol alone) (Fig. 8). Yet, (±)-α-lipoic
acid alone or combined to amisulpride was devoid of any
Effects of Antipsychotics and (±)-α-Lipoic Acid effect (Fig. 8). It should be noted, however, that haloperidol
on the Downstream Substrates of the Akt/GSK-3β Pathway and (±)-α-lipoic acid combination elicited an elevation of total
mTOR protein level (+147 %; *P<0.05 compared to control)
The effects of APDs and (±)-α-lipoic acid on Akt/GSK-3β as well as an increase of phosphorylated mTOR level (+50 %;
signaling prompted us to investigate some of its downstream *P<0.05 compared to control) (data not shown).
targets. β-Catenin, a crucial transcription factor, which is
inhibited by GSK-3β activity (Grimes and Jope 2001) is of
particular interest, since it has also been implicated in Discussion
schizophrenia and APD action (Alimohamad et al. 2005a,
b). We, thus, examined the effect of D2R antagonist APDs We previously demonstrated that haloperidol, an FGA prone
on the levels of β-catenin. After a 6 days of treatment, both to increase oxidative stress, exerted a more prominent effect
haloperidol and amisulpride induced an increase in β- on D2R expression than amisulpride, a SGA with no signi-
catenin protein levels, as compared to control (+49 %; ficant effect on oxidative status (Deslauriers et al. 2011).
**P <0.01 and +61 %; *P <0.05, respectively) (Fig. 7). Those effects could contribute to the higher occurrence of
(±)-α-Lipoic acid did not exert any significant effect on β- side effects, like tardive dyskinesia, observed with the use of
catenin protein levels, either alone or combined with anti- FGAs (Kropp et al. 2005). It has been reported that seroto-
psychotics (Fig. 7). We also investigated the effects of APDs nin 5-HT2A receptor antagonism, a common property of
and (±)-α-lipoic acid treatment on the activation of mam- SGAs (Meltzer 1995), decreased dopamine release, reduced
malian target of rapamycin (mTOR), a serine/threonine ki- the binding of SGA to D2R receptor, and, thus, explained
nase, which participates in L-DOPA-induced dyskinesia the lower incidence of SGAs-induced side effects (Seeman
(Murer and Moratalla 2011). The ratio of phosphorylated et al. 1997). This mechanism is unlikely in our former
J Mol Neurosci

patients, a decrease in Akt activation has been reported


(Zhao et al. 2008). Also, in dopamine transporter-knockout
(DAT-KO) mice, an animal model of schizophrenia (Giros et
al. 1996; Gainetdinov et al. 1999), an inactivation of Akt
and an activation of GSK-3β have been observed. These
changes were normalized by a decrease of dopamine syn-
thesis by D2R blockade and by administration of lithium, a
GSK-3β inhibitor (Beaulieu et al. 2004). Acute or chronic
in vivo administration of FGAs or SGAs normalized Akt/
GSK-3β pathway, through D2R regulation, by increasing
Akt phosphorylation and consequently, inhibiting GSK-3β
activation (Li and Xu 2007). Thus, the Akt/GSK-3β path-
way constitutes an acknowledged therapeutic target for psy-
chiatric disorders (Beaulieu et al. 2004, 2005, 2007;
Emamian et al. 2004; Li and Xu 2007). In line with their
effects in animal models (Beaulieu et al. 2009), haloperidol
and amisulpride increased the phosphorylation of Akt
(Fig. 9). In a similar fashion, the phosphorylation of GSK-
3β, which has been repeatedly implicated in the effect of
APDs as well as other psychotropic drugs, was also in-
creased by incubation with APDs (Fig. 9). To assess the
significance of GSK-3β inhibition vis-à-vis our parameter
of interest, D2R mRNA increase, we used two different
Fig. 8 Effects of haloperidol (HAL), amisulpride (AMI), and (±)-α- inhibitors which prevented the D2R transcriptional activa-
lipoic acid (LA) on mTOR phosphorylation at Ser2448 and total tion elicited by APDs but were devoid of effect on basic
mTOR ratio as compared to untreated SY-SY5Y cells (*P<0.05) or
to haloperidol alone (#P<0.05) (n09–10). Representative immuno- levels of D2R mRNA (Fig. 4). This result is consistent with
blots of phospho-mTOR (Ser473), mTOR, and β-actin are shown clinical observations demonstrating that lithium prevents the
development of tolerance in dopamine metabolism and
(Deslauriers et al. 2011) or present set of results, since we functional dopamine supersensitivity caused by chronic hal-
used antipsychotics with little (haloperidol) or no significant operidol administration (Sternberg et al. 1983). Lithium
(amisulpride) serotonin 5-HT2A antagonism (Schoemaker et effect was not dependent on the normalization of oxidative
al. 1997; Vernaleken et al. 2004). In our previous study, we status, at odds with (±)-α-lipoic acid (Fig. 5 and Deslauriers
reported the preventive effect with respect to haloperidol- et al. 2011). Indeed, it has been shown that long-term
induced D2R upregulation, of (±)-α-lipoic acid, a natural exposure to lithium was required to exert neuroprotection
compound endowed with antioxidant properties through the against oxidative stress, whereas we used a short-term treat-
scavenging of reactive oxygen species (ROS) and the mod- ment (Allagui et al. 2009). Whatsoever, the effects of two
ulation of cell signaling networks that regulate endogenous structurally different GSK-3β inhibitors (lithium chloride
antioxidant potential (Packer and Tritschler 1996). This and AR-A014418) on D2R regulation suggest that the
effect of (±)-α-lipoic acid prompted us to investigate its Akt/GSK-3β pathway may be involved in APD-induced
impact on signal transduction events, specifically the Akt/ D2R upregulation, as it is in other aspects of APD action
glycogen synthase-3β (GSK-3β) pathway, elicited by (Beaulieu et al. 2009).
APDs, using the same in vitro model. Regarding Akt/GSK-3β downstream targets, we report
here that both APDs increase β-catenin levels (Figs. 7, 9).
Again, this is in line with in vivo effects of APDs: β-
Signaling Events Induced by APDs in SH-SY5Y catenin, which transduces activation of the canonical Wnt
Are Mostly Similar to Those Observed In Vivo pathway has been associated with CNS disorders like
schizophrenia (Kozlovsky et al. 2002); its level is increased
Along this line, the first step was to assess whether signaling by APDs (Alimohamad et al. 2005a, b) and conversely
events triggered by APDs in our cell culture model are, to decreased following amphetamine treatment, as well as in
some extent, reminiscent of those observed in vivo. animal models of schizophrenia (Kozlovsky et al. 2000;
Phosphorylation and activation of Akt is a well-known Beasley et al. 2001; Nadri et al. 2003; Beaulieu et al. 2004).
downstream event of D2R activation (Kihara et al. 2002; Therefore, the effects of APDs on the Akt/GSK-3β/β-
Nair and Sealfon 2003; Beom et al. 2004). In schizophrenia catenin cascade in SH-SY5Y were overall consistent with
J Mol Neurosci

Fig. 9 Schematic
representation of antipsychotics
and (±)-α-lipoic acid actions on
dopamine D2 receptor (DRD2)
regulation of Akt/glycogen
synthase kinase-3β (GSK-3β)
signaling pathway

their in vivo actions, with the notable exception of D2R–β- reporting an activation of this pathway by α-lipoic acid
arrestin-2 interaction. It has been reported that D2R signal- (Zhang et al. 2007a, b). However, this additive effect did not
ing by a G protein-independent mechanism, notably with extend downstream to GSK-3β Ser9 phosphorylation or β-
respect to the Akt/GSK-3β pathway, implicates β-arrestin- catenin levels, which were not modified by (±)-α-lipoic alone
2, which is associated with GPCR desensitization (Beaulieu or in combination (Fig. 9). These data suggest that the pre-
et al. 2004, 2005, 2006). However, apart from their role in ventive effect of (±)-α-lipoic acid towards the regulation of
GPCR desensitization, β-arrestins can also scaffold signal- D2R level (Deslauriers et al. 2011) does not involve a reversal
ing protein complexes and, therefore, initiate new intracel- of APD effects on the Akt/GSK-3β pathway. Besides, it is
lular signaling cascades (Lefkowitz and Shenoy 2005). interesting that (±)-α-lipoic acid displays additive or neutral,
Here, we found that haloperidol alone or coincubated with rather than antagonist, effects towards one of the pathways
(±)-α-lipoic acid induced an increase in β-arrestin-2–D2R implicated in APD action, given its clinical potential in the
interaction (Fig. 9), as evidenced by immunoprecipitation management of neurological (Thaakur and Himabindhu 2009)
(Fig. 6). This finding is at odds with previous results and metabolic (Kim et al. 2008) side effects of these drugs.
(Beaulieu et al. 2005; Masri et al. 2008), which could be mTOR is another downstream target of the Akt/GSK-3β
explained by technical differences: we measured endoge- signaling pathway. Indeed, Akt and GSK-3β phosphoryla-
nous D2R receptor expression, as opposed to transfected tions induce mTOR phosphorylation and this effect is
cells, and we did not use agonist stimulation; furthermore, blocked by wortmannin (which inhibits Akt through
we used immunoprecipitation instead of bioluminescence phosphatidylinositol-3 kinase inhibition), but not by rapa-
resonance energy transfer (BRET) to assess the interaction mycin, suggesting that mTOR phosphorylation is mediated
between the D2R and β-arrestin-2. Finally, a more pro- by Akt (Nave et al. 1999). APDs were devoid of any direct
tracted incubation with antipsychotics (6 days) was used effect, as previously reported in a glioblastoma cell line
here in contrast to shorter durations (less than 24 h) in (Zhang et al. 2007a, b). However (±)-α-lipoic acid com-
BRET assays (Masri et al. 2008). Further work will be bined with haloperidol induced a sustained increase in
necessary to elucidate if long-term in vivo APD treatment mTOR activation (Fig. 8) in contrast with the transient
leads to a similar increase in D2R/β-arrestin-2 interaction. increase previously described (Wang et al. 2010). This in-
crease was the result of an elevation of mTOR phoshoryla-
(±)-α-Lipoic Acid Effects on the Akt/GSK-3β Pathway tion at Ser2448 as well as total protein overexpression, as
are Partly Additive with Those of APDs shown in other systems (Kruck et al. 2010). The lack of
effect of haloperidol and amisulpride on mTOR level or
We show here that the combined application of APDs and (±)- phosphorylation status is consistent with the available liter-
α-lipoic acid exerted additive effects on the initial step of the ature and suggests that the mTOR signaling pathway is not
Akt/GSK-3β signaling pathway, namely, Akt Ser473 phos- involved in D2R mRNA increase or other signaling events
phorylation (Fig. 2), which is in line with other studies elicited by APDs in this model (Fig. 9).
J Mol Neurosci

Study Limitations GSK3-β pathway involved in the therapeutic effects of


these drugs, but the latter pathway does not seem to sub-
Undifferentiated SH-SY5Y cells are a valid model to study serve the normalization of D2R expression by (±)-α-lipoic
the dynamics of D2 receptor regulation (Shaul et al. 2003) acid. It has been suggested that targeting multiple signaling
as well as its cellular effects (Iizuka et al. 2007), but they do pathways can have greater therapeutic effect than monother-
not recapitulate the complex synaptic organization of striatal apy in CNS disorder treatment, to avoid the development of
dopamine transmission, and this should be kept in mind in tolerance (Mi et al. 2009). Further work will be required to
the interpretation of the present results. Besides, while the determine which signaling pathways may be implicated in
drug concentrations used here have been chosen to achieve the effect of (±)-α-lipoic acid on tardive dyskinesia-
saturation of D2R while remaining clinically relevant, associated phenomena, such as D2R upregulation and oxi-
effects at receptors other than the D2R cannot completely dative stress, and tolerance with chronic APD treatment.
be ruled out. SH-SY5Y cells are endowed with low
(Presgraves et al. 2004), but functionally significant (Pan Acknowledgments This work was supported by an unrestricted ed-
ucational grant from Novartis Pharma Canada to the Department of
et al. 2005), levels of D3 receptor (D3R); amisulpride has a
Psychiatry, Université de Sherbrooke and by the Centre des Neuro-
higher affinity for D3 than D2 receptors, and haloperidol sciences de Sherbrooke. Philippe Sarret, PhD is a CIHR investigator.
would be expected to saturate at least 50 % of D3R at the Philippe Sarret, PhD and Sylvain Grignon, MD, PhD are members of
concentration used here. The contribution of D3 antago- the FRSQ-funded Centre de Recherche Clinique Étienne Lebel.
nism, if any, is not prominent, since this would predict that
amisulpride would elicit larger responses than haloperidol,
References
while the reverse was observed. In a similar fashion, 10 nM
haloperidol could induce some occupancy of dopamine D4
(D4R) and adrenergic α1 receptors. The precise functional Abilio VC, Araujo CC, Bergamo M, Calvente PR, D’Almeida V,
Ribeiro RA, Frussa-Filho R (2003) Vitamin E attenuates
consequences, however, are difficult to assess: to our knowl-
reserpine-induced oral dyskinesia and striatal oxidized glutathi-
edge, no data exist on the expression and functional status of one/reduced glutathione ratio (GSSG/GSH) enhancement in rats.
D4R in undifferentiated, untransfected SH-SY5Y cells. Prog Neuropsychopharmacol Biol Psychiatry 27:109–114
Regarding adrenergic receptors, their functional status is Alimohamad H, Rajakumar N, Seah YH, Rushlow W (2005a) Anti-
psychotics alter the protein expression levels of beta-catenin and
also unclear, since noradrenaline fails to increase GTPγS
GSK-3 in the rat medial prefrontal cortex and striatum. Biol
binding at concentrations up to 1 μM on the same model Psychiatry 57:533–542
(Bundey and Nahorski 2001). Haloperidol Ki at cloned 5- Alimohamad H, Sutton L, Mouyal J, Rajakumar N, Rushlow WJ
HT2A human receptors is in the 70–200 nM range, which (2005b) The effects of antipsychotics on beta-catenin, glycogen
synthase kinase-3 and dishevelled in the ventral midbrain of rats. J
makes any significant effect at the concentrations used here
Neurochem 95:513–525
unlikely (Schoemaker et al. 1997). Haloperidol is also a Allagui MS, Nciri R, Rouhaud MF, Murat JC, El FA, Croute F, Vincent
high affinity sigma-1 (σ1) antagonist (Cobos et al. 2007) C (2009) Long-term exposure to low lithium concentrations stim-
and this effect is potentially relevant to the present data. ulates proliferation, modifies stress protein expression pattern and
enhances resistance to oxidative stress in SH-SY5Y cells. Neuro-
Indeed, σ1 inhibition has been shown to increase oxidative
chem Res 34:453–462
stress (Pal et al. 2012), which could be one pertinent mech- Beasley C, Cotter D, Khan N, Pollard C, Sheppard P, Varndell I,
anism by which haloperidol modulates oxidative status. The Lovestone S, Anderton B, Everall I (2001) Glycogen synthase
potential impact of σ1 inhibition on the downstream signal- kinase-3beta immunoreactivity is reduced in the prefrontal cortex
in schizophrenia. Neurosci Lett 302:117–120
ing events described here is less clear: in non-neuronal
Beaulieu JM, Sotnikova TD, Yao WD, Kockeritz L, Woodgett JR,
models, σ1 agonism is associated with an increase in Akt Gainetdinov RR, Caron MG (2004) Lithium antagonizes
phosphorylation, and σ1 antagonism by haloperidol would, dopamine-dependent behaviors mediated by an AKT/glycogen
therefore, be expected to decrease Akt phosphorylation, at synthase kinase 3 signaling cascade. Proc Natl Acad Sci USA
101:5099–5104
odds with its effects in this and other models.
Beaulieu JM, Sotnikova TD, Marion S, Lefkowitz RJ, Gainetdinov
In conclusion, (±)-α-lipoic acid appeared to act additive- RR, Caron MG (2005) An Akt/beta-arrestin 2/PP2A signaling
ly with APDs on the Akt pathway, but not on GSK-3β status complex mediates dopaminergic neurotransmission and behavior.
and β-catenin levels. Conversely, under our conditions, (±)- Cell 122:261–273
Beaulieu JM, Sotnikova TD, Gainetdinov RR, Caron MG (2006)
α-lipoic acid, combined with haloperidol, appeared to in-
Paradoxical striatal cellular signaling responses to psychostimu-
crease mTOR level and phosphorylation, an effect that was lants in hyperactive mice. J Biol Chem 281:32072–32080
not shared by APD drugs. (±)-α-Lipoic acid seems to be a Beaulieu JM, Gainetdinov RR, Caron MG (2007) The Akt-GSK-3
promising compound with respect to metabolic and neuro- signaling cascade in the actions of dopamine. Trends Pharmacol
Sci 28:166–172
logical side effects of APD drugs. Pending clinical confir-
Beaulieu JM, Gainetdinov RR, Caron MG (2009) Akt/GSK3 signaling
mation, (±)-α-lipoic acid also seems to display an in the action of psychotropic drugs. Annu Rev Pharmacol Toxicol
acceptable profile with respect to some steps of the Akt/ 49:327–347
J Mol Neurosci

Beom S, Cheong D, Torres G, Caron MG, Kim KM (2004) Compar- internalization and signaling but not D1 internalization. J Neuro-
ative studies of molecular mechanisms of dopamine D2 and D3 sci 27:12390–12395
receptors for the activation of extracellular signal-regulated ki- Jenner P, Rupniak NM, Marsden CD (1985) Differential alteration of
nase. J Biol Chem 279:28304–28314 striatal D-1 and D-2 receptors induced by the long-term adminis-
Bhat R, Xue Y, Berg S, Hellberg S, Ormo M, Nilsson Y, Radesater AC, tration of haloperidol, sulpiride or clozapine to rats. Psychophar-
Jerning E, Markgren PO, Borgegard T, Nylof M, Gimenez- macology Suppl 2:174–181
Cassina A, Hernandez F, Lucas JJ, Diaz-Nido J, Avila J (2003) Jeste DV, Lacro JP, Bailey A, Rockwell E, Harris MJ, Caligiuri MP
Structural insights and biological effects of glycogen synthase (1999) Lower incidence of tardive dyskinesia with risperidone
kinase 3-specific inhibitor AR-A014418. J Biol Chem compared with haloperidol in older patients. J Am Geriatr Soc
278:45937–45945 47:716–719
Bishnoi M, Chopra K, Kulkarni SK (2008) Protective effect of Curcu- Jia Z, Hallur S, Zhu H, Li Y, Misra HP (2008) Potent upregulation of
min, the active principle of turmeric (Curcuma longa) in glutathione and NAD(P)H:quinone oxidoreductase 1 by alpha-
haloperidol-induced orofacial dyskinesia and associated behav- lipoic acid in human neuroblastoma SH-SY5Y cells: protection
ioural, biochemical and neurochemical changes in rat brain. Phar- against neurotoxicant-elicited cytotoxicity. Neurochem Res
macol Biochem Behav 88:511–522 33:790–800
Bundey RA, Nahorski SR (2001) Homologous and heterologous Kapur S, Langlois X, Vinken P, Megens AA, De Coster R, Andrews JS
uncoupling of muscarinic M(3) and alpha(1B) adrenoceptors to (2002) The differential effects of atypical antipsychotics on pro-
Galpha(q/11) in SH-SY5Y human neuroblastoma cells. Br J lactin elevation are explained by their differential blood–brain
Pharmacol 134:257–264 disposition: a pharmacological analysis in rats. J Pharmacol Exp
Burger ME, Fachinetto R, Zeni G, Rocha JB (2005) Ebselen attenuates Ther 302:1129–1134
haloperidol-induced orofacial dyskinesia and oxidative stress in Kihara T, Shimohama S, Sawada H, Honda K, Nakamizo T, Kanki R,
rat brain. Pharmacol Biochem Behav 81:608–615 Yamashita H, Akaike A (2002) Protective effect of dopamine D2
Chen Z, Dupre DJ, Le Gouill C, Rola-Pleszczynski M, Stankova J agonists in cortical neurons via the phosphatidylinositol 3 kinase
(2002) Agonist-induced internalization of the platelet-activating cascade. J Neurosci Res 70:274–282
factor receptor is dependent on arrestins but independent of G- Kim E, Park DW, Choi SH, Kim JJ, Cho HS (2008) A preliminary
protein activation. Role of the C terminus and the (D/N)PXXY investigation of alpha-lipoic acid treatment of antipsychotic drug-
motif. J Biol Chem 277:7356–7362 induced weight gain in patients with schizophrenia. J Clin Psy-
Cobos EJ, del Pozo E, Baeyens JM (2007) Irreversible blockade of chopharmacol 28:138–146
sigma-1 receptors by haloperidol and its metabolites in guinea pig Kozlovsky N, Belmaker RH, Agam G (2000) Low GSK-3beta immu-
brain and SH-SY5Y human neuroblastoma cells. J Neurochem noreactivity in postmortem frontal cortex of schizophrenic
102:812–825 patients. Am J Psychiatry 157:831–833
Correll CU, Schenk EM (2008) Tardive dyskinesia and new antipsy- Kozlovsky N, Belmaker RH, Agam G (2002) GSK-3 and the neuro-
chotics. Curr Opin Psychiatry 21:151–156 developmental hypothesis of schizophrenia. Eur Neuropsycho-
Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA pharmacol 12:13–25
(1995) Inhibition of glycogen synthase kinase-3 by insulin medi- Kropp S, Kern V, Lange K, Degner D, Hajak G, Kornhuber J, Ruther
ated by protein kinase B. Nature 378:785–789 E, Emrich HM, Schneider U, Bleich S (2005) Oxidative stress
Cui J, Shao L, Young LT, Wang JF (2007) Role of glutathione in during treatment with first- and second-generation antipsychotics.
neuroprotective effects of mood stabilizing drugs lithium and J Neuropsychiatry Clin Neurosci 17:227–231
valproate. Neuroscience 144:1447–1453 Kruck S, Bedke J, Hennenlotter J, Ohneseit PA, Kuehs U, Senger E,
Dakhale G, Khanzode S, Saoji A, Khobragade L, Turankar A (2004) Sievert KD, Stenzl A (2010) Activation of mTOR in renal cell
Oxidative damage and schizophrenia: the potential benefit by carcinoma is due to increased phosphorylation rather than protein
atypical antipsychotics. Neuropsychobiology 49:205–209 overexpression. Oncol Rep 23:159–163
Deslauriers J, Lefrancois M, Larouche A, Sarret P, Grignon S (2011) Kulkarni SK, Naidu PS (2001) Animal models of tardive dyskinesia—
Antipsychotic-induced DRD2 upregulation and its prevention by a review. Indian J Physiol Pharmacol 45:148–160
alpha-lipoic acid in SH-SY5Y neuroblastoma cells. Synapse Laruelle M, Jaskiw GE, Lipska BK, Kolachana B, Casanova MF,
65:321–331 Kleinman JE, Weinberger DR (1992) D1 and D2 receptor modu-
Doble BW, Woodgett JR (2003) GSK-3: tricks of the trade for a multi- lation in rat striatum and nucleus accumbens after subchronic and
tasking kinase. J Cell Sci 116:1175–1186 chronic haloperidol treatment. Brain Res 575:47–56
Emamian ES (2012) AKT/GSK3 signaling pathway and schizophre- Lefkowitz RJ, Shenoy SK (2005) Transduction of receptor signals by
nia. Front Mol Neurosci 5:33 beta-arrestins. Science 308:512–517
Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA (2004) Li XM, Xu H (2007) Evidence for neuroprotective effects of antipsy-
Convergent evidence for impaired AKT1-GSK3beta signaling in chotic drugs: implications for the pathophysiology and treatment
schizophrenia. Nat Genet 36:131–137 of schizophrenia. Int Rev Neurobiol 77:107–142
Gainetdinov RR, Jones SR, Caron MG (1999) Functional hyperdopa- Lieberman JA (2004) Dopamine partial agonists: a new class of anti-
minergia in dopamine transporter knock-out mice. Biol Psychiatry psychotic. CNS Drugs 18:251–267
46:303–311 Lohr JB, Kuczenski R, Bracha HS, Moir M, Jeste DV (1990) Increased
Giros B, Jaber M, Jones SR, Wightman RM, Caron MG (1996) Hyper- indices of free radical activity in the cerebrospinal fluid of patients
locomotion and indifference to cocaine and amphetamine in mice with tardive dyskinesia. Biol Psychiatry 28:535–539
lacking the dopamine transporter. Nature 379:606–612 Lohr JB, Kuczenski R, Niculescu AB (2003) Oxidative mechanisms
Grimes CA, Jope RS (2001) The multifaceted roles of glycogen syn- and tardive dyskinesia. CNS Drugs 17:47–62
thase kinase 3beta in cellular signaling. Prog Neurobiol 65:391– Mailman RB, Murthy V (2010) Third generation antipsychotic drugs:
426 partial agonism or receptor functional selectivity? Curr Pharm
Harrison PJ (1999) The neuropathological effects of antipsychotic Des 16:488–501
drugs. Schizophr Res 40:87–99 Marchese G, Bartholini F, Ruiu S, Casti P, Saba P, Gessa G, Pani L
Iizuka Y, Sei Y, Weinberger DR, Straub RE (2007) Evidence that the (2002) Effect of the amisulpride isomers on rat catalepsy. Eur J
BLOC-1 protein dysbindin modulates dopamine D2 receptor Pharmacol 444:69–74
J Mol Neurosci

Masri B, Salahpour A, Didriksen M, Ghisi V, Beaulieu JM, Richelson E (1999) Receptor pharmacology of neuroleptics: relation to
Gainetdinov RR, Caron MG (2008) Antagonism of dopamine clinical effects. J Clin Psychiatry 60(Suppl 10):5–14
D2 receptor/beta-arrestin 2 interaction is a common property Schoemaker H, Claustre Y, Fage D, Rouquier L, Chergui K, Curet O,
of clinically effective antipsychotics. Proc Natl Acad Sci Oblin A, Gonon F, Carter C, Benavides J, Scatton B (1997)
USA 105:13656–13661 Neurochemical characteristics of amisulpride, an atypical dopa-
Meltzer HY (1995) The role of serotonin in schizophrenia and the mine D2/D3 receptor antagonist with both presynaptic and limbic
place of serotonin–dopamine antagonist antipsychotics. J Clin selectivity. J Pharmacol Exp Ther 280:83–97
Psychopharmacol 15:2S–3S Schroder J, Silvestri S, Bubeck B, Karr M, Demisch S, Scherrer S,
Mi R, Ma J, Zhang D, Li L, Zhang H (2009) Efficacy of combined Geider FJ, Sauer H (1998) D2 dopamine receptor up-regulation,
inhibition of mTOR and ERK/MAPK pathways in treating a treatment response, neurological soft signs, and extrapyramidal
tuberous sclerosis complex cell model. J Genet Genomics side effects in schizophrenia: a follow-up study with 123I-
36:355–361 iodobenzamide single photon emission computed tomography in
Morgenstern H, Glazer WM (1993) Identifying risk factors for tardive the drug-naive state and after neuroleptic treatment. Biol Psychi-
dyskinesia among long-term outpatients maintained with neuro- atry 43:660–665
leptic medications. Results of the Yale Tardive Dyskinesia Study. Seeman P, Corbett R, Van Tol HH (1997) Atypical neuroleptics have low
Arch Gen Psychiatry 50:723–733 affinity for dopamine D2 receptors or are selective for D4 receptors.
Muller MJ, Regenbogen B, Hartter S, Eich FX, Hiemke C (2007) Neuropsychopharmacology 16:93–110, discussion 111-35
Therapeutic drug monitoring for optimizing amisulpride therapy Shamir E, Barak Y, Shalman I, Laudon M, Zisapel N, Tarrasch R,
in patients with schizophrenia. J Psychiatr Res 41:673–679 Elizur A, Weizman R (2001) Melatonin treatment for tardive
Murer MG, Moratalla R (2011) Striatal signaling in L-DOPA- dyskinesia: a double-blind, placebo-controlled, crossover study.
induced dyskinesia: common mechanisms with drug abuse Arch Gen Psychiatry 58:1049–1052
and long term memory involving D1 dopamine receptor Shaul U, Ben-Shachar D, Karry R, Klein E (2003) Modulation of
stimulation. Front Neuroanat 5:51 frequency and duration of repetitive magnetic stimulation affects
Nadri C, Lipska BK, Kozlovsky N, Weinberger DR, Belmaker RH, catecholamine levels and tyrosine hydroxylase activity in human
Agam G (2003) Glycogen synthase kinase (GSK)-3beta levels neuroblastoma cells: implication for the antidepressant effect of
and activity in a neurodevelopmental rat model of schizophrenia. rTMS. Int J Neuropsychopharmacol 6:233–241
Brain Res Dev Brain Res 141:33–37 Silvestri S, Seeman MV, Negrete JC, Houle S, Shammi CM, Remington
Naidu PS, Singh A, Kulkarni SK (2003) Quercetin, a bioflavonoid, GJ, Kapur S, Zipursky RB, Wilson AA, Christensen BK, Seeman P
attenuates haloperidol-induced orofacial dyskinesia. Neurophar- (2000) Increased dopamine D2 receptor binding after long-term
macology 44:1100–1106 treatment with antipsychotics in humans: a clinical PET study.
Nair VD, Sealfon SC (2003) Agonist-specific transactivation of phos- Psychopharmacology (Berl) 152:174–180
phoinositide 3-kinase signaling pathway mediated by the dopa- Soares KV, McGrath JJ (1999) The treatment of tardive dyskinesia—a
mine D2 receptor. J Biol Chem 278:47053–47061 systematic review and meta-analysis. Schizophr Res 39:1–16,
Nave BT, Ouwens M, Withers DJ, Alessi DR, Shepherd PR (1999) discussion 17-8
Mammalian target of rapamycin is a direct target for protein Sternberg DE, Bowers MB Jr, Heninger GR, Charney DS (1983)
kinase B: identification of a convergence point for opposing Lithium prevents adaptation of brain dopamine systems to halo-
effects of insulin and amino-acid deficiency on protein translation. peridol in schizophrenic patients. Psychiatry Res 10:79–86
Biochem J 344(Pt 2):427–431 Thaakur S, Himabindhu G (2009) Effect of alpha lipoic acid on the
Nciri R, Desmoulin F, Allagui MS, Murat JC, Feki AE, Vincent C, tardive dyskinesia and oxidative stress induced by haloperidol in
Croute F (2012) Neuroprotective effects of chronic exposure of rats. J Neural Transm 116:807–814
SH-SY5Y to low lithium concentration involve glycolysis stimu- Tsai G, Goff DC, Chang RW, Flood J, Baer L, Coyle JT (1998) Markers
lation, extracellular pyruvate accumulation and resistance to oxi- of glutamatergic neurotransmission and oxidative stress associated
dative stress. Int J Neuropsychopharmacol 1–12 with tardive dyskinesia. Am J Psychiatry 155:1207–1213
Packer L, Tritschler HJ (1996) Alpha-lipoic acid: the metabolic anti- Vernaleken I, Siessmeier T, Buchholz HG, Hartter S, Hiemke C,
oxidant. Free Radic Biol Med 20:625–626 Stoeter P, Rosch F, Bartenstein P, Grunder G (2004) High striatal
Pal A, Fontanilla D, Gopalakrishnan A, Chae YK, Markley JL, Ruoho occupancy of D2-like dopamine receptors by amisulpride in the
AE (2012) The sigma-1 receptor protects against cellular oxida- brain of patients with schizophrenia. Int J Neuropsychopharmacol
tive stress and activates antioxidant response elements. Eur J 7:421–430
Pharmacol 682:12–20 Wang Y, Li X, Guo Y, Chan L, Guan X (2010) Alpha-lipoic acid increases
Pan T, Xie W, Jankovic J, Le W (2005) Biological effects of prami- energy expenditure by enhancing adenosine monophosphate-
pexole on dopaminergic neuron-associated genes: relevance to activated protein kinase-peroxisome proliferator-activated receptor-
neuroprotection. Neurosci Lett 377:106–109 gamma coactivator-1alpha signaling in the skeletal muscle of aged
Paulson GW (2005) Historical comments on tardive dyskinesia: a mice. Metabolism 59:967–976
neurologist’s perspective. J Clin Psychiatry 66:260–264 Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, Zhu H, Yu AD, Xie X, Ma
Peet M, Laugharne J, Rangarajan N, Reynolds GP (1993) Tardive D, Yuan J (2007a) Small molecule regulators of autophagy iden-
dyskinesia, lipid peroxidation, and sustained amelioration with tified by an image-based high-throughput screen. Proc Natl Acad
vitamin E treatment. Int Clin Psychopharmacol 8:151–153 Sci USA 104:19023–19028
Pillai A, Parikh V, Terry AV Jr, Mahadik SP (2007) Long-term anti- Zhang WJ, Wei H, Hagen T, Frei B (2007b) Alpha-lipoic acid attenu-
psychotic treatments and crossover studies in rats: differential ates LPS-induced inflammatory responses by activating the phos-
effects of typical and atypical agents on the expression of antiox- phoinositide 3-kinase/Akt signaling pathway. Proc Natl Acad Sci
idant enzymes and membrane lipid peroxidation in rat brain. J USA 104:4077–4082
Psychiatr Res 41:372–386 Zhao Z, Robinson RG, Barnett SF, Defeo-Jones D, Jones RE, Hartman
Presgraves SP, Ahmed T, Borwege S, Joyce JN (2004) Terminally GD, Huber HE, Duggan ME, Lindsley CW (2008) Development
differentiated SH-SY5Y cells provide a model system for study- of potent, allosteric dual Akt1 and Akt2 inhibitors with improved
ing neuroprotective effects of dopamine agonists. Neurotox Res physical properties and cell activity. Bioorg Med Chem Lett
5:579–598 18:49–53

You might also like