Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Dec 1.
Published in final edited form as: Neuropharmacology. 2011 Aug 3;61(8):1256–1264. doi: 10.1016/j.neuropharm.2011.07.025

Chronic Valproate Treatment Blocks D2-like Receptor-Mediated Brain Signaling via Arachidonic Acid in Rats

Epolia Ramadan 1,*, Mireille Basselin 1, Ameer Y Taha 1, Yewon Cheon 1, Lisa Chang 1, Mei Chen 1, Stanley I Rapoport 1
PMCID: PMC3190603  NIHMSID: NIHMS316741  PMID: 21839100

Abstract

Background and Objective

Hyperdopaminergic signaling and an upregulated brain arachidonic acid (AA) cascade may contribute to bipolar disorder (BD). Lithium and carbamazepine, FDA-approved for the treatment of BD, attenuate brain dopaminergic D2-like (D2, D3, and D4) receptor signaling involving AA when given chronically to awake rats. We hypothesized that valproate (VPA), with mood-stabilizing properties, would also reduce the D2-like-mediated signaling via AA.

Methods

An acute dose of quinpirole (1 mg/kg) or saline was administered to unanesthetized rats that had been treated for 30 days with a therapeutically relevant dose of VPA (200 mg/kg/day) or vehicle. Regional brain AA incorporation coefficients, k*, and incorporation rates, Jin, markers of AA signaling and metabolism, were measured by quantitative autoradiography after intravenous [1-14C]AA infusion. Whole brain concentrations of prostaglandin (PG)E2 and thromboxane (TX)B2 also were measured.

Results

Quinpirole compared to saline significantly increased k* in 40 of 83 brain regions, and increased brain concentrations of PGE2 in chronic vehicle-treated rats. VPA treatment by itself reduced concentrations of plasma unesterified AA and whole brain PGE2 and TXB2, and blocked the quinpirole-induced increments in k* and PGE2.

Conclusion

These results further support our hypothesis that similar to lithium and carbamazepine, VPA downregulates brain dopaminergic D2-like receptor-signaling involving AA.

Keywords: arachidonic acid, phospholipase A2, valproate, D2-like receptor, quinpirole, bipolar disorder

1. Introduction

Valproate (2-propylpentanoate, VPA) has a wide clinical spectrum of use in both psychiatric and neurological disorders. It is one of the most frequently used antiepileptic drugs, has mood-stabilizing properties in the treatment of acute mania (Bowden, 2009), and might be effective for the reduction of depressive symptoms of acute bipolar depression (Smith et al., 2010; Wang et al., 2010). Despite more than 40 years of clinical use, the mechanism(s) of action of VPA in bipolar disorder (BD) is still not fully understood. It is well known that VPA exerts multiple pharmacological effects, and has been found to affect glycogen synthase kinase-3 (GSK-3), the Wnt/β-catenin pathway, the extracellular signal-regulated kinase (ERK) pathway, γ-aminobutyric acid (GABA)ergic neurotransmission, N-methyl-D-aspartate (NMDA) glutamatergic signaling, pre- and post-synaptic dopamine (DA) neurotransmission, voltage-gated sodium and T-type calcium channels, histone acetylation, and brain lipids and their metabolism (Basselin et al., 2008a; Montezinho et al., 2006; Phiel et al., 2001; Rapoport et al., 2009; Yatham et al., 2002). In addition, VPA is neuroprotective in several models of neurodegenerative diseases (Monti et al., 2010; Monti et al., 2009).

Hyperdopaminergic neurotransmission is suggested to be involved in the pathophysiology of mania in BD (Berk et al., 2007; Cousins et al., 2009; Diehl and Gershon, 1992; Goetz, 1997). Reports show that administration of drugs that inhibit DAergic transmission (haloperidol, chlorpromazine) has antimanic action whereas drugs that stimulate DA synthesis (levodopa), bind to D2-receptors (bromocriptine) or reduce DA reuptake (amphetamine) often precipitate mania (Anand et al., 2000; Cipriani et al., 2006; Peet and Peters, 1995). In this context, psychotic BD brains show higher D2 receptor expression in the caudate and prefrontal cortex (Feng, 2008; Pearlson et al., 1995) and genetic studies have linked the DA reuptake transporter (DAT) and BD (Greenwood et al., 2001; Greenwood et al., 2006), with a DAT mutation causing inhibition of the transporter cell surface expression being associated with BD (Horschitz et al., 2005). Furthermore, analysis of postmortem cortex from BD patients shows significantly elevated levels of the neuronal calcium sensor-1 (NCS-1), which inhibits D2 desensitization/internalization (Kabbani et al., 2002; Koh et al., 2003), changes in the levels of DA and cyclic adenosine 3’:5’-monophosphate-regulated phosphoprotein of relative molecular mass 32,000 (DARPP-32) (Ishikawa et al., 2007; Zhan et al., 2011), and decreased protein and mRNA levels of DAT (Rao JS and Rapoport SI, unpublished data).

Dopaminergic D2-like (D2, D3, and D4) receptors in brain can be coupled via a Gαi/o-protein to Ca2+-dependent cytosolic phospholipase (cPLA2 , EC 3.1.1.4), which when activated releases arachidonic acid (AA, 20:4n-6) from the stereospecifically numbered (sn)-2 position of synaptic membrane phospholipid (Clark et al., 1995; Nilsson et al., 1998; Ong et al., 1999; Vial and Piomelli, 1995). AA is an important second messenger in brain with multiple effects, and a precursor of bioactive eicosanoids such as prostaglandin E2 (PG)E2 (Rapoport, 2008). Markers of the AA cascade have been reported to be abnormal in BD (Kim et al., 2011; Noponen et al., 1993). The brain AA signaling can be measured in unanesthetized rodents by infusing radiolabeled AA intravenously, quantifying integrated plasma radioactivity, using quantitative autoradiography to determine regional brain radioactivity due to tracer AA incorporated in membrane phospholipid, then applying a mathematical model to calculate AA incorporation coefficients and rates, k* and Jin, respectively (Rapoport et al., 2001; Robinson et al., 1992). Since the AA lost after release and metabolism cannot be synthesized de novo from 2-carbon fragments, or elongated significantly (< 1%) from its shorter chain polyunsaturated precursor, linoleic acid (18:2n-6) (Demar et al., 2006; Holman, 1986), k* and Jin for AA represent net AA consumption following release from phospholipid.

We previously showed using the intravenous infusion method that acute administration to unanesthetized rats of quinpirole (1 mg/kg, D2-like receptor agonist) (Seeman and Van Tol, 1994), amphetamine or apomorphine (D1/D2 receptor agonist), but not the D1-like receptor agonist, SKF-38393, increased k* and Jin for AA in many brain regions rich in D2-like receptors, and that the increases could be blocked by pre-administration of a D2-receptor antagonist (e.g. butaclamol, raclopride) or of each of the two FDA-approved antimanic mood stabilizers, lithium and carbamazepine when given chronically (Basselin et al., 2005; Basselin et al., 2008b; Bhattacharjee et al., 2005; Bhattacharjee et al., 2006, 2008; Hayakawa et al., 2001). Each mood stabilizer downregulated brain AA turnover and/or reduced levels and activities of essential enzymes and metabolites of the brain AA cascade (Bazinet et al., 2006; Bosetti et al., 2002; Chang et al., 1996; Ghelardoni et al., 2004; Rao et al., 2007; Rao et al., 2005).

VPA also is approved as an antimanic mood stabilizer for BD, and when given chronically reduces AA turnover within brain phospholipids and decreases activity and concentrations of cyclooxygenase (COX) and its metabolites, respectively (Bosetti et al., 2003; Chang et al., 2001). We hypothesized that chronic administration of VPA to produce therapeutically relevant plasma levels, also would block the quinpirole-initiated AA signal and other AA cascade markers in rat brain. We applied our established in vivo fatty acid and activity methods, and measured AA incorporation coefficients ,k*, and rates, Jin, in each of 83 brain regions after acutely giving saline or quinpirole (1 mg/kg) to unanesthetized rats that had chronically received VPA (200 mg/kg/day, i.p) or vehicle for 30 days. Whole brain concentrations of prostaglandin PGE2 and thromboxane (TX)B2 were also measured.

2. Material and methods

2.1. Animals and Diets

Two-month-old male Fischer CDF 344 rats (Charles River Laboratories, Wilmington, MA) were acclimated for 1 week in an animal facility with regulated temperature, humidity and light cycle, and had free access to food and water. The diet (Rodent NIH-31 auto 18-4 diet, Zeigler Bros, Gardens, PA) contained (as % of total fatty acid) 20.1% saturated, 22.5% monounsaturated, 47.9% linoleic, 5.1% α-linolenic, 0.02% AA, 2.0% eicosapentaenoic, and 2.3% docosahexaenoic acid (Demar et al., 2006). Experiments were conducted following the “Guide for the Care and Use of Laboratory Animals” (National Institutes of Health Publication No. 86-23) and were approved by the Animal Care and Use Committee of Eunice Kennedy Shriver National Institute of Child Health and Human Development. All efforts were made to reduce the number of animals used and to minimize animal suffering.

2.2. Drugs and Tracers

Radiolabeled [1-14C]AA in ethanol (53 mCi/mmol, >98% pure, Moravek Biochemicals, Brea, CA) was evaporated and resuspended in HEPES buffer, pH 7.4, containing 50 mg/ml fatty acid-free bovine serum albumin (Sigma-Aldrich, St Louis, MO). VPA (sodium salt, Sigma-Aldrich)-treated rats received 200 mg/kg intraperitoneally (i.p) once daily for 30 days. VPA was dissolved in saline (0.9% NaCl, Hospira, Lake Forest, IL) as described previously (Basselin et al., 2008a; Bazinet et al., 2005; Bosetti et al., 2003; Chang et al., 2001). A control group received the same volume of saline (vehicle) under parallel conditions. An acute 1 mg/kg i.v. dose of (−)-quinpirole hydrochloride dissolved in saline (Sigma-Aldrich) was chosen because it produces widespread significant increments in k* for AA in the brain of unanesthetized rats that can be blocked by D2-like receptor antagonists, butaclamol or raclopride, without causing convulsions (Basselin et al., 2005; Bhattacharjee et al., 2005).

2.3. Surgical Procedures and Tracer Infusion

On the morning of day 30, a rat was injected with the last VPA or vehicle dose, and then anesthetized with halothane (2–3% v/v in O2). Polyethylene (PE 50) catheters were surgically inserted into the right femoral artery and vein as described previously (Basselin et al., 2005). The wound was closed with surgical clips and the rat was wrapped loosely, with its upper body remaining free, in a fast-setting plaster cast taped to a wooden block. Surgery lasted 20–25 min. Rats were allowed to recover from anesthesia for 3–4 h in an environment maintained at 25°C. Rectal temperature was maintained at 36.4–37.1°C using a feedback-heating device and rectal thermometer. Arterial blood pressure and heart rate were measured with a blood pressure recorder (CyQ 103/302; Cybersense, Nicholasville, KY). One minute after an i.v. injection of quinpirole or saline, [1-14C]AA (170 µCi/kg, 2 ml) was infused into the femoral vein for 5 min at a rate of 400 µl/min, using an infusion pump (Harvard Apparatus Model 22, Natick, MA). Twenty min after beginning tracer infusion, the rat was euthanized with an overdose of Nembutal® (90 mg/kg, i.v.) and decapitated. The brain was removed (<30 s), frozen in 2-methylbutane maintained at −40°C in dry ice, and stored at −80°C until sectioned.

2.4. Chemical Analysis

Blood samples, collected before, during or after [1-14C]AA infusion, were centrifuged immediately at 18,000 g for 30 s. Total lipids were extracted from plasma (30 µl) using a modified Folch procedure (Folch et al, 1957). One hundred µl of the lower organic phase was used to determine the radiolabeled unesterified plasma AA concentration by liquid scintillation counting. As previously reported (DeGeorge et al, 1989), greater than 95–98% of total plasma and brain radioactivity at 5 min following [1-14C]AA infusion is radiolabeled AA. Concentrations of unlabeled, unesterified fatty acids were determined from frozen/thawed arterial plasma. Total lipids were extracted and separated by thin layer chromatography on 60 silica gel plates (Whatman, Clifton, NJ) using the solvent system heptane: diethylether:glacial acetic acid (60:40:3, v/v/v). Unesterified fatty acids (identified under UV light) were scraped from the plate and methylated with 1% H2SO4 (by vol) in anhydrous methanol (3 h at 70°C), then separated and quantified by gas-liquid chromatography using heptadecanoic acid (17:0) as an internal standard.

2.5. Quantitative Autoradiography

Quantitative autoradiography was performed as described earlier (Basselin et al., 2006a). A total of 83 brain regions from autoradiographs of coronal brain sections were identified from a stereotaxic rat brain atlas (Paxinos and Watson, 1987), and were sampled in both hemispheres. The average of bilateral measurements for each region from three consecutive brain sections was used to calculate regional radioactivity (nCi/g wet brain) by digital quantitative densitometry, using the public domain 1.62 Analysis NIH Image program. Regional brain incorporation coefficients k* (ml plasma/s/g wet brain) of AA were calculated as (Robinson et al, 1992),

k*=cbrain*(20 min)020cplasma*dt (Eq.1)

cbrain* (nCi/g wet brain wt) is brain radioactivity 20 min after beginning infusion, cplasma* (nCi/ml plasma) is arterial labeled unesterified AA, and t (min) is time after beginning [1-14C] AA infusion. Integrated plasma radioactivity (input function) was determined by trapezoidal integration and used to calculate k* for each experiment. The regional rate of incorporation of unesterified AA from plasma into brain phospholipids, Jin (pmol/s/g), was calculated as follows:

Jin=k*cplasma (Eq.2)

where cplasma is the plasma concentration (nmol/ml) of unlabeled unesterified AA.

2.6. Brain PGE2 and TXB2 Concentrations

In a separate experiment and after the last of 30 daily administrations of VPA or vehicle, a rat was injected with quinpirole (1 mg/kg, i.p) or saline. Twenty-one minutes later (Basselin et al., 2008b), it was anesthetized with Nembutal® (45 mg/kg, i.p.), and immediately subjected to head-focused microwave irradiation (5.5 kW, 3.8 s; Cober Electronics, Stamford, CT) to stop postmortem brain lipid metabolism (Farias et al., 2008; Poddubiuk et al., 1982). A half-brain was weighed, homogenized with 18 volumes of hexane:isopropanol (3:2, by volume) using a glass Tenbroeck homogenizer and the homogenates were centrifuged (800 g, 5 min). Tissue residues were rinsed with 3 × 2 volumes of the same solvent. The resultant lipid extracts were concentrated to dryness under N2 and resuspended in the enzyme immunoassay buffer provided by the polyclonal PGE2 and TXB2 immunoassay kits (Oxford Biochemical Research, Oxford, MI).

2.7. Statistical Analyses

A paired t test using GraphPad Prism version 4.0b (GraphPad Software, San Diego, CA) was applied to compare mean physiological parameters in the same animal before and after drug injection. A standard two-way analysis of variance (ANOVA) was performed to compare chronic VPA and vehicle treatment with acute quinpirole vs. saline administration with regard to: integrated arterial plasma radioactivity input functions, plasma unesterified fatty acid concentrations, brain PGE2 and TXB2 concentrations, and regional values of k* and Jin for AA. If interactions between VPA and quinpirole were statistically insignificant, probabilities of effects of VPA and quinpirole were reported. If interactions were statistically significant, probabilities of main effects of VPA and quinpirole were not reported (Tabachnick and Fidell, 2001). Alternatively, a one-way ANOVA with Bonferroni’s post-test was used to compare quinpirole and saline responses between chronic VPA- and vehicle-treated rats, as well as saline responses in VPA-treated compared with vehicle-treated rats. Data are reported as the mean ± SD, with statistical significance taken as p ≤ 0.05.

3. Results

3.1. Physiology, behavior and Arterial Plasma Radioactivity

After 30 days of treatment, the mean body weight of VPA-treated rats was significantly lower than that of vehicle-treated rats (294.1 ± 25.9 g [n = 14] vs. 263.4 ± 21.0 g [n = 14], p = 0.002), as previously reported (Basselin et al., 2008a; Daoud et al., 2004; Hassel et al., 2001). There was no significant difference between rats chronically injected with VPA or saline with regard to rectal temperature, heart rate or arterial blood pressure (Table 1). Acute quinpirole provoked repeated cycles of an “active” period of repetitive head and mouth movements and sniffing, followed by a “calm” period (Horvitz et al., 2001). No significant difference in mean cycling periods was observed in VPA-treated compared to vehicle-treated rats (Table 1).

Table 1.

Physiological parameters following drug administration in unanesthetized rats

Chronic Vehicle
Chronic Valproate
Saline Quinpirole Saline Quinpirole
Before After Before After Before After Before After

Rectal Temperature (°C) 36.8 ± 0.5 36.7 ± 0.4 36.8 ± 0.3 36.9 ± 0.2 36.9 ± 0.4 37.0 ± 0.3 36.8 ± 0.4 37.0 ± 0.2
Heart Rate (beats/min) 414 ± 26 402 ± 48 409 ± 42 430 ± 30 425 ± 37 434 ± 27 441 ± 31 433 ± 53
Arterial blood pressure
     (mmHg)
     Systolic 178 ± 13 166 ± 7 174 ± 10 181 ± 13 175 ± 10 166 ± 8 162 ± 14 162 ± 11
     Diastolic 109 ± 8 101 ± 7 109 ± 7 103 ± 7 109 ± 6 96 ± 11 102 ± 7 100 ± 6
Orofacial activity duraion (s)
     Orofacial activity 10 ± 2 11 ± 3
     Calm period 17 ± 4 15 ± 3

Values are means ± SD (n= 7) measured before drug injection (quinpirole 1mg/kg, i.v.) and 10 minutes after [14C] AA infusion. Paired t-tests were used to compare means in the same animal before and after drug injection.

Neither chronic VPA nor acute quinpirole modified the time course of arterial plasma radioactivity (Eq. 1) following intravenous [14C]AA infusion. The mean integral of radioactivity in the plasma organic fraction (nCi × s)/ml (n = 7), the input function, did not differ significantly among groups: chronic vehicle + saline, 149,317 ± 30,502; chronic vehicle + quinpirole, 152,433 ± 32,473; chronic VPA + saline, 121,565 ± 8,959; chronic VPA + quinpirole, 144,614 ± 24,116.

3.2. Plasma Concentrations of Unlabeled Unesterified Fatty Acids

A two-way ANOVA showed a significant VPA and quinpirole interaction for the plasma concentrations of unesterified stearic and AA but not for unesterified palmitic, oleic, linoleic, α-linolenic, or docosahexaenoic acids (Table 2). A one-way ANOVA with Bonferroni’s post-test showed that chronic VPA compared to vehicle significantly reduced plasma concentrations of stearic acid and AA by 39% and 66%, respectively. Compared with vehicle, chronic VPA had a significant main negative effect (−57% to −70%) on each of the remaining six unesterified fatty acids concentrations, while acute quinpirole had no main effect on any of these concentrations.

Table 2.

Effects of Valproate and Quinpirole on plasma unesterified fatty acid concentrations.

Chronic Vehicle
Chronic VPA
VPA X Quin
Interaction
VPA
Effect
Quin
Effect
Fatty Acid Saline Quin Saline Quin P-value P-value P-value
(nmol/ml plasma)
Palmitic(16:0) 199.9 ± 82.8 164.3 ± 43.1 75.7 ± 20.4 70.6 ± 15.9 0.4133 < 0.0001 0.2768
Stearic(18:0) 49.2 ± 10.1 36.6 ± 14.3 30.0 ± 4.8** 33.7 ± 5.9 0.0334
Oleic(18:1n-9) 124.0 ± 50.8 89.9 ± 42.4 53.2 ± 15.8 45.2 ± 11.1 0.3264 0.0002 0.1192
Linoleic (18:2n-6) 181.9 ± 73.3 146.5 ± 68.1 70.2 ± 19.4 72.0 ± 16.9 0.3505 < 0.0001 0.3977
α-Linolenic (18:3n-3) 11.9 ± 5.3 10.3 ± 4.4 4.1 ± 1.5 3.8 ± 1.5 0.6485 < 0.0001 0.4819
Arachidonic (20:4n-6) 16.0 ± 6.1 12.6 ± 2.9 5.5 ± 1.0*** 7.9 ± 1.9 0.0411
Docosahexaenoic (22:6n-3) 19.5 ± 10.2 15.7 ± 4.8 5.8 ± 1.6 6.4 ± 1.3 0.3199 < 0.0001 0.4665

Values are means ± SD (n=7) measured from arterial plasma collected at 19 min after the beginning of [1-14C] AA infusion.

**

P< 0.01,

***

P < 0.001;

vehicle plus quinpirole vs vehicle plus saline, VPA plus saline vs vehicle plus saline, and VPA plus quinpirole vs VPA plus saline (one-way ANOVA Bonferroni’s tests). VPA, valproate; Quin, quinpirole.

3.3. Regional Brain AA Incorporation Coefficients, k*

Figure 1 presents coronal autoradiographs of brains from rats treated chronically (30 days) with vehicle (control) or VPA, then acutely injected with saline or quinpirole. k* for AA, calculated by Eq. 1, is color-coded. The figure shows no difference in regional values of k* in response to saline between VPA- and vehicle-treated rats. Acute quinpirole increased k* in multiple brain regions of the chronic vehicle- but not of the VPA-treated rats. Data obtained from such autoradiographs are summarized in Table 3.

Figure 1.

Figure 1

Coronal autoradiographs of brain showing effects of quinpirole and valproate on regional arachidonic acid incorporation coefficients k* in rats. Values of k* (ml/s/g wet brain × 10−4) are on a color scale from 4 (blue) to 10 (orange). Acb, nucleus accumbens; CP, caudate putamen; Fr 8, frontal cortex (8); Fr 10, frontal cortex (10); PFr, prefrontal cortex; SN, substantia nigra; VTA, ventral tegmantal area. VPA, valproate; Quin, quinpirole.

Table 3.

Chronic Valproate blocked Quinpirole-induced increments in arachidonic acid incorporation coefficients, k*, in dopaminergic brain regions

Chronic Vehicle
Chronic VPA
VPA × Quin
Interaction
Saline Quin Saline Quin
Brain region P-value
Nigrostiatal system
Caudate putamen
Dorsal 5.65 ± 0.82 7.69 ± 1.30*** 6.37 ± 0.21 5.97 ± 0.90 0.0015
Ventral 5.52 ± 0.76 7.90 ± 1.38*** 6.25 ± 0.28 5.96 ± 0.97 0.0009
Lateral 5.69 ± 0.93 7.83 ± 1.38** 6.42 ± 0.22 6.12 ± 0.95 0.0027
Medial 5.52 ± 0.91 7.83 ± 1.15*** 6.08 ± 0.31 5.56 ± 0.88 0.0002
Globus pallidus 5.01 ± 0.69 7.25 ± 1.15*** 5.55 ± 0.42 4.55 ± 1.06 <0.0001
Subthalamic nucleus 5.87 ± 0.89 7.79 ± 1.37** 6.58 ± 0.48 6.05 ± 1.05 0.0035
Substantia nigra 4.87 ± 0.91 6.88 ± 1.46** 6.16 ± 0.52 5.59 ± 0.96 0.0028
Mesocorticolimbic
       Cortical
Prefrontal cortex
       layer I 4.91 ± 0.45 7.34 ± 1.50*** 5.68 ± 0.35 5.06 ± 0.39 <0.0001
       layer IV 5.55 ± 0.78 7.74 ± 1.79** 6.27 ± 0.21 5.86 ± 0.45 0.0023
Primary olfactory cortex
5.34 ± 0.80 7.23 ± 1.21** 5.67 ± 0.47 5.16 ± 0.74 0.0010
Frontal cortex (10)
       Layer I 5.41 ± 0.81 7.48 ± 1.16*** 5.77 ± 0.25 5.49 ± 0.55 0.0005
       Layer IV 5.94 ± 1.06 8.22 ± 1.53*** 6.30 ± 0.23 6.09 ± 0.66 0.0029
Frontal cortex (8)
       Layer I 5.86 ± 0.94 7.85 ± 1.20*** 6.13 ± 0.24 5.69 ± 0.57 0.0007
       Layer IV 6.64 ± 1.23 8.57 ± 1.42** 6.71 ± 0.35 6.93 ± 1.05 0.0489
Motor cortex
       Layer I 5.46 ± 0.94 7.35 ± 1.09** 5.74 ± 0.38 4.89 ± 1.22 0.0009
       Layer II-III 5.85 ± 1.03 7.65 ± 1.35* 6.20 ± 0.31 5.47 ± 1.32 0.0051
       Layer IV 6.44 ± 0.95 8.51 ± 1.73* 6.70 ± 0.27 6.06 ± 1.32 0.0062
       Layer V 5.76 ± 1.28 7.98 ± 1.53** 6.42 ± 0.35 5.68 ± 1.35 0.0037
       Layer VI 5.50 ± 1.23 7.91 ± 1.58** 6.13 ± 0.31 5.62 ± 1.17 0.0030
Somatosensory cortex
       Layer I 5.78 ± 0.85 7.29 ± 1.20* 5.91 ± 0.63 4.99 ± 1.18 0.0055
       Layer II-III 6.12 ± 0.93 7.92 ± 1.39* 6.35 ± 0.27 5.54 ± 1.19 0.0028
       Layer IV 6.41 ± 0.74 8.56 ± 1.51** 7.07 ± 0.26 6.15 ± 1.17 0.0006
       Layer V 6.35 ± 0.82 8.13 ± 1.55* 6.52 ± 0.27 5.80 ± 1.17 0.0144
       Layer VI 6.15 ± 0.89 8.03 ± 1.51** 6.41 ± 0.31 5.66 ± 1.09 0.0028
Pyriform cortex
4.55 ± 0.54 5.55 ± 1.09* 5.20 ± 0.45 4.82 ± 0.56 0.0163
Anterior cingulate cortex
6.91 ± 1.09 8.42 ± 2.57* 7.33 ± 0.40 6.39 ± 0.77 0.0363
Auditory cortex
       Layer I 5.40 ± 1.00 6.99 ± 1.18* 6.14 ± 0.52 5.78 ± 0.83 0.0095
       Layer IV 5.88 ± 0.88 7.81 ± 1.23** 6.99 ± 0.49 6.90 ± 0.97 0.0084
       Layer VI 5.57 ± 1.06 7.29 ± 1.05* 6.36 ± 0.62 6.32 ± 0.99 0.0216
Visual cortex
       Layer I 4.58 ± 0.61 6.77 ± 1.59** 5.79 ± 0.36* 5.25 ± 0.98 0.0014
       Layer IV 5.43 ± 0.60 7.67 ± 1.35** 6.12 ± 0.73 5.97 ± 0.88 0.0024
       Layer VI 4.86 ± 0.55 7.11 ± 1.39*** 6.04 ± 0.72 5.62 ± 0.89 0.0010
       Subcortical
Bed nucleus stria terminalis
4.49 ± 0.59 6.89 ± 1.16*** 5.14 ± 0.38 4.47 ± 0.62 <0.0001
Amygdala basolateral/medial
4.38 ± 0.89 6.92 ± 0.98**** 5.26 ± 0.71 5.02 ± 1.15 0.0007
Nucleus accumbens 4.99 ± 0.59 7.08 ± 0.90*** 5.74 ± 0.32 4.99 ± 0.74 <0.0001
Ventral tegmental area 4.44 ± 0.93 6.40 ± 1.46** 5.67 ± 0.27 5.16 ± 0.89 0.0028
Tuberoinfundibular
Arcuate nucleus hypothalamus
4.88 ± 0.45 6.22 ± 1.02* 5.39 ± 0.41 5.04 ± 1.03 0.0089
Other dopaminergic
Zona Incerta 6.04 ± 0.79 7.82 ± 1.69** 6.93 ± 0.85 6.42 ± 1.44 0.0237
Ventroposterior
       lateral nucleus 5.28 ± 1.37 7.40 ± 1.46* 6.85 ± 0.84 6.34 ± 1.37 0.0126
       medial nucleus 5.25 ± 1.16 7.61 ± 1.38** 6.51 ± 0.84 6.24 ± 1.41 0.0088

Each k* (ml/s/g wet brain) × 10−4 value is a mean ± SD (n = 7). Quinpirole administration: 1 mg/kg i.v., 1min.

*

P< 0.05;

**

P< 0.01;

***

P< 0.001;

vehicle plus quinpirole vs vehicle plus saline, VPA plus saline vs vehicle plus saline, and VPA plus quinpirole vs VPA plus saline (oneway ANOVA with Bonferroni tests). VPA, valproate; Quin, quinpirole.

Values of the mean AA incorporation coefficients, k*, determined in each of 83 brain regions were subjected to a two-way ANOVA. Statistically significant interactions between VPA and quinpirole were found in 40 regions belonging primarily to the nigrostriatal and mesocorticolimbic systems, which comprise the DAergic circuits of the basal ganglia (Baldessarini and Tarazi, 1996) (Table 3, Fig 1). In all 40 regions, a one-way ANOVA with Bonferroni’s post-test showed that chronic VPA did not significantly change mean baseline (after saline) k* in any region (Table 3). The same one-way ANOVA showed that acute quinpirole compared with saline increased k* by 22% to 58% in chronic vehicle-treated rats. Affected regions included caudate-putamen (36–43%), globus pallidus (45%), subthalamic nucleus (33%), substantia nigra (41%), prefrontal cortex (39–50%), primary olfactory cortex (35%), frontal cortex (29–38%), pyriform and anterior cingulated cortex (22%), motor (31–44%), somatosensory, auditory (29–33%), visual (41–48%) cortical areas (26–39%), bed nucleus of the stria terminalis (53%), amygdala (58%), nucleus accumbens (42%), ventral tegmental area (44%), arcuate nucleus of the hypothalamus (28%), ventroposterior thalamic nuclei (40–45%) and zona incerta (29%). Quinpirole compared to saline did not significantly increase k* in any of the 40 regions in chronic VPA-treated rats.

In the 43 regions where VPA and quinpirole interaction was statistically insignificant, neither VPA nor quinpirole had any significant main effect on k* for AA (data not shown). Thus, chronic VPA blocked each of the 40 quinpirole-induced k* increments that were observed in the chronic vehicle-treated rat.

3.4. Regional Rates of Incorporation of Unlabeled Unesterified AA into Brain

Rates of incorporation of unlabeled unesterified AA from plasma into brain, Jin, (data not shown) were calculated by Eq. 2 from regional k* (Table 3) and cplasma for AA (Table 2). A two-way ANOVA showed no statistically significant interaction between VPA and quinpirole in any of the 83 brain regions examined. Chronic VPA compared with vehicle had a significant main negative effect in each of the 83 brain regions while acute quinpirole had no main effect on any. In vehicle-treated rats Jin ranged from 5.9 pmol/s/g in the internal capsule to 28 pmol/s/g in the choroid plexus, whereas Jin ranged from 2.2 pmol/s/g to 11 pmol/s/g in the respective areas of the VPA-treated rats.

3.5. Brain PGE2 and TXB2 Concentrations

As shown in Table 4, a two-way ANOVA demonstrated a statistically significant interaction between VPA and quinpirole with regard to the brain PGE2 concentration. Consequently, a one-way ANOVA with Bonferroni’s post-test showed that chronic VPA reduced basal brain concentrations of PGE2 by 59 % (p < 0.05). Acute quinpirole significantly increased the PGE2 (p < 0.01) concentration by 1.7-fold in vehicle- but not VPA-treated rats. Chronic VPA reduced TXB2 concentration at baseline and in response to quinpirole (significant VPA main effect) by 42%.

Table 4.

Effects of Quinpirole and Valproate on brain PGE2 and TXB2 concentrations in rats

Chronic Vehicle
Chronic VPA
VPA × Quin
Interaction
VPA
Effect
Quin
Effect
Saline Quin Saline Quin P-value P-value P-value
PGE2 (ng/g brain) 10.3 ± 3.1 17.7 ± 6.8** 4.2 ± 1.1* 5.3 ± 0.8 0.0348
TXB2 (pg/g brain) 41.3 ± 11.9 31.4 ± 9.1 22.2 ± 6.8 20.2 ± 9.0 0.2710 0.0002 0.1034

Each value is a mean ± SD (n = 7). Bonferroni’s multiple comparison tests were performed.

*

P< 0.05;

**

P < 0.01;

vehicle plus quinpirole vs vehicle plus saline, VPA plus saline vs vehicle plus saline, and VPA plus quinpirole vs VPA plus saline. VPA, valproate; Quin, quinpirole.

4. Discussion

In this study, we showed that daily administration of VPA (200 mg/kg, i.p.) to rats for 30 days, at a dose that produces a plasma VPA concentration relevant to BD, prevented the statistically significant increases in AA incorporation coefficients k*, and in whole brain PGE2 concentration, that were produced by an acute dose of quinpirole in chronic vehicle-treated rats. To the extent that DAergic signaling via D2-like receptors and the AA cascade are pathologically upregulated in BD patients, for which evidence exists (see “Introduction”) (Berk et al., 2007; Cousins et al., 2009; Diehl and Gershon, 1992; Goetz, 1997; Kim et al., 2011), these results suggest that the efficacy of VPA in the disease treatment is due in part to its ability to dampen upregulated D2-like signaling involving AA and its downstream metabolites. In agreement, chronic administration to rats of a therapeutically relevant plasma concentration of lithium or carbamazepine also dampens D2-induced elevations in k* for AA and in brain eicosanoids (Basselin et al., 2005; Basselin et al., 2008b; Bosetti et al., 2002; Bosetti et al., 2003). Taken together, reduced D2-like signal involving AA and its metabolites may be common to the therapeutic action of mood stabilizers effective in BD. In contrast, topiramate, which appeared effective in Phase II trials in BD, but later failed Phase III placebo-controlled trials (Kushner et al., 2006), did not change markers of the rat brain AA cascade (Ghelardoni et al., 2005; Lee et al., 2005). Topiramate has not been tested with regard to the D2-like signal.

Similar to lithium and carbamazepine, chronic VPA significantly decreased baseline PGE2 and TXB2 concentrations as previously reported (Basselin et al., 2008a; Bosetti et al., 2002; Bosetti et al., 2003; Ghelardoni et al., 2004). We ascribe this to VPA selectively decreasing the binding activity of the transcription factor NF-κB that regulates neuronal COX-2 gene expression, as well as reducing COX-1 and COX-2 protein levels and whole brain COX activity (Bosetti et al., 2003; Kaltschmidt et al., 2002; Rao et al., 2007).

Acute quinpirole significantly increased k* for AA in 40 brain regions, most of which are rich in D2-like receptors (Levant et al., 1992; Lidow et al., 1989) and are related to the topographical distribution of DAergic innervation in the brain (mesocorticolimbic, nigrostriatal, and tuberoinfundibular pathways). The zona incerta, located in the ventral thalamus, and cerebral cortical areas (layers I to VI) including auditory and visual cortex also contain DA neurons (Berger et al., 1985; Bjorklund and Lindvall, 1975; Lidow et al., 1989; Rivera and Chun, 2008). The globus pallidus, subthalamic nucleus, and ventrobasal thalamus also receive DAergic innervation and express DA receptors (Baldessarini and Tarazi, 1996; Govindaiah et al., 2010).

The mechanisms underlying VPA’s ability to block the D2-like-receptor-induced increases in k* for AA and to reduce PGE2 and TXB2 concentrations in rat brain are not clear. VPA could have acted by reducing COX activity, and COX-1 and COX-2 protein levels (Bosetti et al., 2003). When COX enzymes are pharmacologically inhibited or knocked out in rodent brain, k* responses to drugs acting at cPLA2-coupled neuroreceptors are reduced or lost, as are the increases in brain PGE2 and/or TXB2 concentrations (Basselin et al., 2006b; Basselin et al., 2007b). VPA also may have interfered with the DAergic system and D2-like receptors. Consistent with altered gene expression of histone deacetylases and increased in histone H3 and H4 acetylation in BD patients (Hobara et al., 2010; Sharma et al., 2006), VPA, a direct histone deacetylase inhibitor (Phiel et al., 2001), may modify the transcription of the rate-limiting enzyme in DA biosynthesis, tyrosine hydroxylase (D'Souza et al., 2009), or of Sp1 (Marinova et al., 2009), a transcription factor of the D2 receptor (Yajima et al., 1998), and/or DAT gene acetylation (Wang and Bannon, 2005). Consistent with its property, VPA has been shown to decrease D2 receptor protein in the rat prefrontal cortex (Montezinho et al., 2006), lower presynaptic DA function in the striatum of patients with mania (Yatham et al., 2002), and increase DAT gene expression in rat midbrain DA neurons (Wang et al., 2007), thus decreasing extracellular DA concentration at the synaptic cleft. Although VPA has been reported to inhibit GSK-3 (Chen G et al., 1999), which can be regulated by DA via the Akt signaling pathway (Beaulieu et al., 2004; Beaulieu, 2011), this effect is indirect and has been attributed to inhibition of activation of Akt and inactivation of GSK-3 following inhibition of histone deacetylase (Phiel et al., 2001; De Sarno et al., 2002).

In addition, a therapeutically relevant concentration of VPA has been shown to decrease the activity and protein level of protein kinase C (Chen et al., 1994), which mediates phosphorylation, desensitization and trafficking of the D2 receptor (Namkung and Sibley, 2004). Alternatively, chronic VPA may have indirectly attenuated the D2-mediated AA signaling by (i) enhancing GABAergic transmission, which participates in regulating the activity of DA release and inhibiting DAergic activity (Agmo et al., 1996) and/or by (ii) reducing excitatory neurotransmission and blocking the AA signaling mediated by glutamatergic-NMDA receptors (Basselin et al., 2008a), as D2-like and NMDA receptors are often functionally coupled and colocalized on the same neurons in the brain (Cepeda and Levine, 1998; Wang et al., 2003). Together with our previous reports (Basselin et al., 2005, 2006a; Basselin et al., 2008b; Basselin et al., 2007a), we strongly infer that antimanic mood stabilizers effective in BD suppress AA signaling coupled to both NMDA and D2-like receptors. Combined, these data are consistent with VPA protecting DA neurons in lipopolysaccharide-induced neurotoxicity (Peng et al., 2005). In agreement, VPA was neuroprotective in experimental models of cerebral ischemia, Parkinson’s disease and glutamate-induced excitotoxicity via histone deacetylase inhibition (Monti et al., 2010; Monti et al., 2009; Ren et al., 2004; Wang et al., 2010; Chuang et al., 2009).

VPA increased brain-derived neurotrophic factor (BDNF) (Einat et al., 2003; Yasuda et al., 2009), hippocampal neurotrophin-3 (Walz et al., 2008), anti-apoptotic factor B-cell lymphoma-2 (Bcl-2) (Chen et al., 1999), and restored amphetamine-induced downregulation of BDNF and of neurotrophin-3 in rat brain (Frey et al., 2006; Walz et al., 2008). Given that the brain and serum in BD have reduced BDNF and other neurotrophic factors (Kauer-Sant'Anna et al., 2009; Kim et al., 2010; Knable et al., 2004; Tramontina et al., 2009), these actions may contribute to VPA's neuroprotective effect in BD, a disease characterized by progression and apoptosis (Benes et al., 2006; Kim et al., 2010; Rapoport et al., 2009).

Consistent with our previous studies, chronic VPA significantly decreased the plasma concentration of unlabeled unesterified fatty acids including AA (Bazinet et al., 2005; Chang et al., 2001), indicating a widespread effect on whole body fatty acid metabolism. A similar reduction in plasma unesterified fatty acids has been found with other mood stabilizers and antipsychotics used to treat BD, such as lamotrigine, olanzapine and clozapine (Ramadan et al., 2011; Cheon et al., unpublished observation) suggesting a common peripheral effect of these drugs. The decrease in plasma unesterified fatty acids may be due to (i) reduced liver secretion of lipoprotein-bound esterified fatty acids, the main source of unesterified fatty acids in plasma, or (ii) reduced release of unesterified fatty acids from adipose tissue by lipases. Although the effects of VPA on free fatty acid release have not been investigated, evidence of impaired secretion of esterified fatty acids has been demonstrated with a marked reduction in triglyceride secretion following VPA treatment (Bellringer et al., 1988).

The baseline values of k* and Jin in this study agree with previous reports (Basselin et al., 2005; Basselin et al., 2008a; Bhattacharjee et al., 2005; Bhattacharjee et al., 2006, 2008). Values of baseline k* were not altered by chronic VPA, which is consistent with our previous data (Basselin et al., 2008a), and supports the finding that chronic VPA does not affect basal cPLA2-IV expression (Bosetti et al., 2003). Chronic VPA had a significant main negative effect on all Jin values at baseline compared to vehicle-treated rats, indicating that the regional rate of metabolic AA loss from brain is lower in the VPA-treated animals. We ascribe this to VPA’s significant reduction of the plasma concentration of unlabeled unesterified AA and of brain PGE2 and TXB2 concentrations, and to its selective inhibition of acyl-CoA synthetase 4-mediated activation of AA to AA-CoA (Bazinet et al., 2006a; Shimshoni et al., 2011).

Chronic VPA, like chronic carbamazepine but unlike chronic lithium, did not prevent quinpirole-induced hyperactivity or stereotypy (Basselin et al., 2005; Basselin et al., 2008b; Beaulieu et al., 2004; Shaldubina et al., 2002). As each of the three mood stabilizers downregulates the brain AA cascade, their different effects on quinpirole-induced behaviors suggest that these behaviors do not involve AA signaling, and that the quinpirole-induced activity cycles are not modeling BD. In contrast, VPA attenuated the hyperactivity and preservative locomotor behavior in the DAT knockdown mice (Ralph-Williams et al., 2003).

We investigated the effects only of chronic VPA in this study, mood stabilization properties in BD patients only appears after 10 days of treatment with VPA. An acute injection of VPA (200–300 mg/kg) in rats caused no/very transient change in the brain DA level (Ahmad et al., 2005; Mitsikostas et al., 1993).

In conclusion, chronic VPA pretreatment prevented the statistically significant increases in k* for AA and in PGE2 concentrations that were observed in response to quinpirole in chronic vehicle-treated rats. These and observations in rats administered chronic lithium or carbamazepine support the hypothesis that mood stabilizers commonly downregulate brain AA signaling via D2-like receptors, and are consistent with evidence that some BD symptoms arise from excessive DAergic neurotransmission (Goetz, 1997). It would be worthwhile to see if atypical antipsychotics (e.g. clozapine, olanzapine), which are D2-like receptor antagonists, do so as well, which would suggest a more general receptor action of these agents on cPLA2-mediated AA signaling (Liauw and McIntyre, 2010). Additionally positron emission tomography using [1-11C]AA might be employed in BD patients under or without chronic VPA treatment, before and after drug induced D2-like receptor activation, to see if VPA has a similar transient effect on AA signaling BD (Giovacchini et al., 2004; Goetz, 1997; Hosey et al., 2005).

Highlights.

  • The research identifies VPA’s ability to downregulate dopamine-D2 receptor signaling via AA.

  • Quinpirole increases AA signaling and metabolism in vehicle-treated rats.

  • Chronic VPA blocks increments in AA signaling and metabolism induced by quinpirole.

  • Mood stabilizers attenuate hyperdopaminergic neurotransmission.

  • Possible implication for the efficacy of mood stabilizers against bipolar disorder.

Acknowledgments

This work was supported entirely by the Intramural Research Program of the National Institute on Aging, NIH. We thank the Fellows Editorial Board for editorial assistance.

Abbreviations

AA

arachidonic acid

BD

bipolar disorder

BDNF

brain-derived neurotrophic factor

cPLA2

Ca2+-dependent cytosolic phospholipase A2

COX

cyclooxygenase

DA

dopamine

DAT

dopamine reuptake transporter

GSK-3

glycogen synthase kinase-3

VPA

valproate

PGE2

prostaglandin E2

TXB2

thromboxane B2

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

None of the authors has a financial or other conflict of interest related to this work.

References

  1. Agmo A, Belzung C, Giordano M. Interactions between dopamine and GABA in the control of ambulatory activity. J Neural Transm. 1996;103:925–934. doi: 10.1007/BF01291783. [DOI] [PubMed] [Google Scholar]
  2. Ahmad S, Fowler LJ, Whitton PS. Effects of combined lamotrigine and valproate on basal and stimulated extracellular amino acids and monoamines in the hippocampus of freely moving rats. Naunyn Schmiedebergs Arch Pharmacol. 2005;371:1–8. doi: 10.1007/s00210-004-1008-4. [DOI] [PubMed] [Google Scholar]
  3. Anand A, Verhoeff P, Seneca N, Zoghbi SS, Seibyl JP, Charney DS, Innis RB. Brain SPECT imaging of amphetamine-induced dopamine release in euthymic bipolar disorder patients. Am J Psychiatry. 2000;157:1108–1114. doi: 10.1176/appi.ajp.157.7.1108. [DOI] [PubMed] [Google Scholar]
  4. Baldessarini RJ, Tarazi FI. Brain dopamine receptors: a primer on their current status, basic and clinical. Harv Rev Psychiatry. 1996;3:301–325. doi: 10.3109/10673229609017200. [DOI] [PubMed] [Google Scholar]
  5. Basselin M, Chang L, Bell JM, Rapoport SI. Chronic lithium chloride administration to unanesthetized rats attenuates brain dopamine D2-like receptor-initiated signaling via arachidonic acid. Neuropsychopharmacology. 2005;30:1064–1075. doi: 10.1038/sj.npp.1300671. [DOI] [PubMed] [Google Scholar]
  6. Basselin M, Chang L, Bell JM, Rapoport SI. Chronic lithium chloride administration attenuates brain NMDA receptor-initiated signaling via arachidonic acid in unanesthetized rats. Neuropsychopharmacology. 2006a;31:1659–1674. doi: 10.1038/sj.npp.1300920. [DOI] [PubMed] [Google Scholar]
  7. Basselin M, Chang L, Chen M, Bell JM, Rapoport SI. Chronic administration of valproic acid reduces brain NMDA signaling via arachidonic acid in unanesthetized rats! Neurochem. Res. 2008a;33:2229–2240. doi: 10.1007/s11064-008-9700-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Basselin M, Chang L, Chen M, Bell JM, Rapoport SI. Chronic carbamazepine admistration attenuates dopamine D2-like receptor-initiated signaling via arachidonic acid in rat brain. Neurochem. Res. 2008b;33:1373–1383. doi: 10.1007/s11064-008-9595-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Basselin M, Villacreses NE, Chen M, Bell JM, Rapoport SI. Chronic carbamazepine administration reduces NMDA receptor-initiated signaling via arachidonic acid in rat brain. Biol. Psychiatry. 2007a;62:934–943. doi: 10.1016/j.biopsych.2007.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Basselin M, Villacreses NE, Langenbach R, Ma K, Bell JM, Rapoport SI. Resting and arecoline-stimulated brain metabolism and signaling involving arachidonic acid are altered in the cyclooxygenase-2 knockout mice. J. Neurochem. 2006b;96:669–679. doi: 10.1111/j.1471-4159.2005.03612.x. [DOI] [PubMed] [Google Scholar]
  11. Basselin M, Villacreses NE, Lee H-J, Bell JM, Rapoport SI. Flurbiprofen, a cyclooxygenase inhibitor, reduces the brain arachidonic acid signal in response to the cholinergic muscarinic, arecoline, in awake rats. Neurochem. Res. 2007b;32:1857–1867. doi: 10.1007/s11064-007-9372-3. [DOI] [PubMed] [Google Scholar]
  12. Bazinet RP, Rao JS, Chang L, Rapoport SI, Lee HJ. Chronic valproate does not alter the kinetics of docosahexaenoic acid within brain phospholipids of the unanesthetized rat. Psychopharmacology (Berl) 2005;1:180–185. doi: 10.1007/s00213-005-0059-7. [DOI] [PubMed] [Google Scholar]
  13. Bazinet RP, Weis MT, Rapoport SI, Rosenberger TA. Valproic acid selectively inhibits conversion of arachidonic acid to arachionoyl-CoAby brain microsomal long-chain fatty acyl-CoA synthetases: relevance to bipolar disorder. Psychopharmacology (Berl) 2006a;184:122–129. doi: 10.1007/s00213-005-0272-4. [DOI] [PubMed] [Google Scholar]
  14. Bazinet RP, Rao JS, Chang L, Rapoport SI, Lee HJ. Chronic carbamazepine decreases the incorporation rate and turnover of arachidonic acid but not docosahexaenoic acid in brain phospholipids of the unanesthetized rat: relevance to bipolar disorder. Biol. Psychiatry. 2006b;59:401–407. doi: 10.1016/j.biopsych.2005.07.024. [DOI] [PubMed] [Google Scholar]
  15. Beaulieu JM, Sotnikova TD, Yao WD, Kockeritz L, Woodgett JR, Gainetdinov RR, Caron MG. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc Natl Acad Sci U S A. 2004;101:5099–5104. doi: 10.1073/pnas.0307921101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Beaulieu JM. A role of Akt and glycogen synthse kinase-3 as integrators of dopamine and serotonin neurotransmission in mental health. J. Psychiatry Neurosci. 2011;36:1110011. doi: 10.1503/jpn.110011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bellringer ME, Rahman K, Coleman R. Sodium valproate inhibits the movement of secretory vesicles in rat hepatocytes. Biochem. J. 1988;249:513–519. doi: 10.1042/bj2490513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Benes FM, Matzilevich D, Burke RE, Walsh J. The expression of proapoptosis genes is increased in bipolar disorder, but not in schizophrenia. Mol. Psychiatry. 2006;11:241–251. doi: 10.1038/sj.mp.4001758. [DOI] [PubMed] [Google Scholar]
  19. Berger B, Verney C, Alvarez C, Vigny A, Helle KB. New dopaminergic terminal fields in the motor, visual (area 18b) and retrosplenial cortex in the young and adult rat. Immunocytochemical and catecholamine histochemical analyses. Neuroscience. 1985;15:983–998. doi: 10.1016/0306-4522(85)90248-9. [DOI] [PubMed] [Google Scholar]
  20. Berk M, Dodd S, Kauer-Sant'anna M, Malhi GS, Bourin M, Kapczinski F, Norman T. Dopamine dysregulation syndrome: implications for a dopamine hypothesis of bipolar disorder. Acta Psychiatr. Scand. Suppl. 2007:41–49. doi: 10.1111/j.1600-0447.2007.01058.x. [DOI] [PubMed] [Google Scholar]
  21. Bhattacharjee AK, Chang L, Lee HJ, Bazinet RP, Seemann R, Rapoport SI. D2 but not D1 dopamine receptor stimulation augments brain signaling involving arachidonic acid in unanesthetized rats. Psychopharmacology (Berl) 2005;180:735–742. doi: 10.1007/s00213-005-2208-4. [DOI] [PubMed] [Google Scholar]
  22. Bhattacharjee AK, Chang L, White L, Bazinet RP, Rapoport SI. D-Amphetamine stimulates D2 dopamine receptor-mediated brain signaling involving arachidonic acid in unanesthetized rats. J. Cereb. Blood Flow Metab. 2006;26:1378–1388. doi: 10.1038/sj.jcbfm.9600290. [DOI] [PubMed] [Google Scholar]
  23. Bhattacharjee AK, Chang L, White L, Bazinet RP, Rapoport SI. Imaging apomorphine stimulation of brain arachidonic acid signaling via D2-like receptors in unanesthetized rats. Psychopharmacology (Berl) 2008;197:557–566. doi: 10.1007/s00213-008-1073-3. [DOI] [PubMed] [Google Scholar]
  24. Bjorklund A, Lindvall O. Dopamine in dendrites of substantia nigra neurons: suggestions for a role in dendritic terminals. Brain Res. 1975;83:531–537. doi: 10.1016/0006-8993(75)90849-5. [DOI] [PubMed] [Google Scholar]
  25. Bosetti F, Rintala J, Seemann R, Rosenberger TA, Contreras MA, Rapoport SI, Chang MC. Chronic lithium downregulates cyclooxygenase-2 activity and prostaglandin E2 concentration in rat brain. Mol. Psychiatry. 2002;7:845–850. doi: 10.1038/sj.mp.4001111. [DOI] [PubMed] [Google Scholar]
  26. Bosetti F, Weerasinghe GR, Rosenberger TA, Rapoport SI. Valproic acid down-regulates the conversion of arachidonic acid to eicosanoids via cyclooxygenase-1 and −2 in rat brain. J. Neurochem. 2003;85:690–696. doi: 10.1046/j.1471-4159.2003.01701.x. [DOI] [PubMed] [Google Scholar]
  27. Bowden CL. Anticonvulsants in bipolar disorders: current research and practice and future directions. Bipolar Disord. 2009;11 Suppl 2:20–33. doi: 10.1111/j.1399-5618.2009.00708.x. [DOI] [PubMed] [Google Scholar]
  28. Cepeda C, Levine MS. Dopamine and N-methyl-D-aspartate receptor interactions in the neostriatum. Dev. Neurosci. 1998;20:1–18. doi: 10.1159/000017294. [DOI] [PubMed] [Google Scholar]
  29. Chang MCJ, Contreras MA, Rosenberger TA, Rintala JJ, Bell JM, Rapoport SI. Chronic valproate treatment decreases the in vivo turnover of arachidonic acid in brain phospholipids: a possible common effect of mood stabilizers. J. Neurochem. 2001;77:796–803. doi: 10.1046/j.1471-4159.2001.00311.x. [DOI] [PubMed] [Google Scholar]
  30. Chang MCJ, Grange E, Rabin O, Bell JM, Allen DD, Rapoport SI. Lithium decreases turnover of arachidonate in several brain phospholipids. Neurosci. Lett. 1996;220:171–174. doi: 10.1016/s0304-3940(96)13264-x. Erratum in: Neurosci Lett 1997 1931, 1222:1141. [DOI] [PubMed] [Google Scholar]
  31. Chuang DM, Leng Y, Marinov Z, Kim HJ, Chiu CT. Multiple roles of HDAC inhibition in neurodegenrative conditions. Trends Neurosci. 2009;32:591–601. doi: 10.1016/j.tins.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chen G, Manji HK, Hawver DB, Wright CB, Potter WZ. Chronic sodium valproate selectively decreases protein kinase C alpha and epsilon in vitro. J. Neurochem. 1994;63:2361–2364. doi: 10.1046/j.1471-4159.1994.63062361.x. [DOI] [PubMed] [Google Scholar]
  33. Chen G, Zeng WZ, Yuan PX, Huang D, Jiang YM, Zhao ZH, Manji HK. The mood-stabilizing agents lithium and valproate robustly increase the levels of the neuroprotective protein bcl-2 in the CNS. J. Neurochem. 1999;72:879–882. doi: 10.1046/j.1471-4159.1999.720879.x. [DOI] [PubMed] [Google Scholar]
  34. Chen G, Huang LD, Jiang YM, Manji HK. The mood-stabilizing agent valproate inhibits the activity of glycogen synthase kinase-3. J. Neurochem. 1999;72:1327–1330. doi: 10.1046/j.1471-4159.2000.0721327.x. [DOI] [PubMed] [Google Scholar]
  35. Cipriani A, Rendell JM, Geddes JR. Haloperidol alone or in combination for acute mania. Cochrane Database Syst. Rev. 2006;3:CD004362. doi: 10.1002/14651858.CD004362.pub2. [DOI] [PubMed] [Google Scholar]
  36. Clark JD, Schievella AR, Nalefski EA, Lin LL. Cytosolic phospholipase A2. J. Lipid Mediat. Cell Signal. 1995;12:83–117. doi: 10.1016/0929-7855(95)00012-f. [DOI] [PubMed] [Google Scholar]
  37. Cousins DA, Butts K, Young AH. The role of dopamine in bipolar disorder. Bipolar Disord. 2009;11:787–806. doi: 10.1111/j.1399-5618.2009.00760.x. [DOI] [PubMed] [Google Scholar]
  38. D'Souza A, Onem E, Patel P, La Gamma EF, Nankova BB. Valproic acid regulates catecholaminergic pathways by concentration-dependent threshold effects on TH mRNA synthesis and degradation. Brain Res. 2009;1247:1–10. doi: 10.1016/j.brainres.2008.09.088. [DOI] [PubMed] [Google Scholar]
  39. Daoud AS, Bataineh H, Otoom S, Abdul-Zahra E. The effect of Vigabatrin, Lamotrigine and Gabapentin on the fertility, weights, sex hormones and biochemical profiles of male rats. Neuro Endocrinol. Lett. 2004;25:178–183. [PubMed] [Google Scholar]
  40. DeGeorge JJ, Noronha JG, Bell JM, Robinson P, Rapoport SI. Intravenous injection of [1-14C]arachidonate to examine regional brain lipid metabolism in unanesthetized rats. J. Neurosci. Res. 1989;24:413–423. doi: 10.1002/jnr.490240311. [DOI] [PubMed] [Google Scholar]
  41. Demar JC, Jr, Lee HJ, Ma K, Chang L, Bell JM, Rapoport SI, Bazinet RP. Brain elongation of linoleic acid is a negligible source of the arachidonate in brain phospholipids of adult rats. Biochim. Biophys. Acta. 2006;1761:1050–1059. doi: 10.1016/j.bbalip.2006.06.006. [DOI] [PubMed] [Google Scholar]
  42. De Sarno P, Li X, Jope RS. Regulation of Akt and glycogen synthase kinase-3 beta phophorylation by sodium valproate and lithium. Neuropharmacology. 2002;43:1158–1164. doi: 10.1016/s0028-3908(02)00215-0. [DOI] [PubMed] [Google Scholar]
  43. Diehl DJ, Gershon S. The role of dopamine in mood disorders. Compr. Psychiatry. 1992;33:115–120. doi: 10.1016/0010-440x(92)90007-d. [DOI] [PubMed] [Google Scholar]
  44. Einat H, Yuan P, Gould TD, Li J, Du J, Zhang L, Manji HK, Chen G. The role of the extracellular signal-regulated kinase signaling pathway in mood modulation. J. Neurosci. 2003;23:7311–7316. doi: 10.1523/JNEUROSCI.23-19-07311.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Farias SE, Basselin M, Chang L, Heidenreich KA, Rapoport SI, Murphy RC. Formation of eicosanoids, E2/D2-isoprostanes and docosanoids following decapitation-induced ischemia, measured in high-energy microwaved rat brain. J. Lipid. Res. 2008;49:1990–2000. doi: 10.1194/jlr.M800200-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Feng Y. Convergence and divergence in the etiology of myelin impairement in psychiatric disorders and drug addiction. Neurochem Res. 2008;33:1940–1949. doi: 10.1007/s11064-008-9693-x. [DOI] [PubMed] [Google Scholar]
  47. Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 1957;226:497–509. [PubMed] [Google Scholar]
  48. Frey BN, Andreazza AC, Cereser KM, Martins MR, Valvassori SS, Reus GZ, Quevedo J, Kapczinski F. Effects of mood stabilizers on hippocampus BDNF levels in an animal model of mania. Life Sci. 2006;79:281–286. doi: 10.1016/j.lfs.2006.01.002. [DOI] [PubMed] [Google Scholar]
  49. Ghelardoni S, Tomita YA, Bell JM, Rapoport SI, Bosetti F. Chronic carbamazepine selectively downregulates cytosolic phospholipase A2 expression and cyclooxygenase activity in rat brain. Biol. Psychiatry. 2004;56:248–254. doi: 10.1016/j.biopsych.2004.05.012. [DOI] [PubMed] [Google Scholar]
  50. Ghelardoni S, Bazinet RP, Rapoport SI, Bosetti F. Topiramate does not alter expression in rat brain of enzymes of arachidonic acid metabolism. Psychopharmacology. 2005;180:523–529. doi: 10.1007/s00213-005-2189-3. [DOI] [PubMed] [Google Scholar]
  51. Giovacchini G, Lerner A, Toczek MT, Fraser C, Ma K, DeMar JC, Herscovitch P, Eckelman WC, Rapoport SI, Carson RE. Brain incorporation of [11C]arachidonic acid, blood volume, and blood flow in healthy aging: a study with partial-volume correction. J. Nucl. Med. 2004;45:1471–1479. [PubMed] [Google Scholar]
  52. Goetz CG. New strategies with dopaminergic drugs: modified formulations of levodopa and novel agonists. Exp. Neurol. 1997;144:17–20. doi: 10.1006/exnr.1996.6381. [DOI] [PubMed] [Google Scholar]
  53. Govindaiah G, Wang T, Gillette MU, Crandall SR, Cox CL. Regulation of inhibitory synapses by presynaptic D4 dopamine receptors in thalamus. J. Neurophysiol. 2010;104:2757–2765. doi: 10.1152/jn.00361.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Greenwood TA, Alexander M, Keck PE, McElroy S, Sadovnick AD, Remick RA, Kelsoe JR. Evidence for linkage disequilibrium between the dopamine transporter and bipolar disorder. Am. J. Med. Genet. 2001;105:145–151. doi: 10.1002/1096-8628(2001)9999:9999<::aid-ajmg1161>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  55. Greenwood TA, Schork NJ, Eskin E, Kelsoe JR. Identification of additional variants within the human dopamine transporter gene provides further evidence for an association with bipolar disorder in two independent samples. Mol. Psychiatry. 2006;11:125–133. 115. doi: 10.1038/sj.mp.4001764. [DOI] [PubMed] [Google Scholar]
  56. Hassel B, Tauboll E, Gjerstad L. Chronic lamotrigine treatment increases rat hippocampal GABA shunt activity and elevates cerebral taurine levels. Epilepsy Res. 2001;43:153–163. doi: 10.1016/s0920-1211(00)00196-0. [DOI] [PubMed] [Google Scholar]
  57. Hayakawa T, Chang MC, Rapoport SI, Appel NM. Selective dopamine receptor stimulation differentially affects [3H]arachidonic acid incorporation, a surrogate marker for phospholipase A2-mediated neurotransmitter signal transduction, in a rodent model of Parkinson's disease. J. Pharmacol. Exp. Ther. 2001;296:1074–1084. [PubMed] [Google Scholar]
  58. Hobara T, Uchida S, Otsuki K, Matsubara T, Funato H, Matsuo K, Suetsugi M, Watanabe Y. Atered gene expression of histone deacytelase in mood disorder patients. J. Psychiatr. Res. 2010;44:263–270. doi: 10.1016/j.jpsychires.2009.08.015. [DOI] [PubMed] [Google Scholar]
  59. Holman RT. Control of polyunsaturated acids in tissue lipids. J. Am. Coll. Nutr. 1986;5:183–211. doi: 10.1080/07315724.1986.10720125. [DOI] [PubMed] [Google Scholar]
  60. Horschitz S, Hummerich R, Lau T, Rietschel M, Schloss P. A dopamine transporter mutation associated with bipolar affective disorder causes inhibition of transporter cell surface expression. Mol. Psychiatry. 2005;10:1104–1109. doi: 10.1038/sj.mp.4001730. [DOI] [PubMed] [Google Scholar]
  61. Horvitz JC, Williams G, Joy R. Time-dependent actions of D2 family agonist quinpirole on spontaneous behavior in the rat: dissociation between sniffing and locomotion. Psychopharmacology (Berl) 2001;154:350–355. doi: 10.1007/s002130000677. [DOI] [PubMed] [Google Scholar]
  62. Hosey LA, Thompson JL, Metman LV, van den Munckhof P, Braun AR. Temporal dynamics of cortical and subcortical responses to apomorphine in Parkinson disease: an H2(15)O PET study. Clin. Neuropharmacol. 2005;28:18–27. doi: 10.1097/01.wnf.0000154220.30263.0e. [DOI] [PubMed] [Google Scholar]
  63. Ishikawa M, Mizukami K, Iwakiri M, Asada T. Immunohistochemical and immunoblot analysis of Dopamine and cyclic AMP-regulated phosphoprotein, relative molecular mass 32,000 (DARPP-32) in the prefrontal cortex of subjects with schizophrenia and bipolar disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2007;31:1177–1181. doi: 10.1016/j.pnpbp.2007.04.013. [DOI] [PubMed] [Google Scholar]
  64. Kabbani N, Negyessy L, Lin R, Goldman-Rakic P, Levenson R. Interaction with neuronal calcium sensor NCS-1 mediates desensitization of the D2 dopamine receptor. J. Neurosci. 2002;22:8476–8486. doi: 10.1523/JNEUROSCI.22-19-08476.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kaltschmidt B, Linker RA, Deng J, Kaltschmidt C. Cyclooxygenase-2 is a neuronal target gene of NF-κB. BMC Mol. Biol. 2002;3:16. doi: 10.1186/1471-2199-3-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kauer-Sant'Anna M, Kapczinski F, Andreazza AC, Bond DJ, Lam RW, Young LT, Yatham LN. Brain-derived neurotrophic factor and inflammatory markers in patients with early- vs. late-stage bipolar disorder. Int. J. Neuropsychopharmacol. 2009;12:447–458. doi: 10.1017/S1461145708009310. [DOI] [PubMed] [Google Scholar]
  67. Kim HW, Rapoport SI, Rao JS. Altered arachidonic acid cascade enzymes in postmortem brain from bipolar disorder patients. Mol. Psychiatry. 2011;16:419–428. doi: 10.1038/mp.2009.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kim HW, Rapoport SI, Rao JS. Altered expression of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiol. Dis. 2010;37:596–603. doi: 10.1016/j.nbd.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Knable MB, Barci BM, Webster MJ, Meador-Woodruff J, Torrey EF. Molecular abnormalities of the hippocampus in severe psychiatric illness: postmortem findings from the Stanley Neuropathology Consortium. Mol. Psychiatry. 2004;9:609–620. 544. doi: 10.1038/sj.mp.4001471. [DOI] [PubMed] [Google Scholar]
  70. Koh PO, Bergson C, Undie AS, Goldman-Rakic PS, Lidow MS. Up-regulation of the D1 dopamine receptor-interacting protein, calcyon, in patients with schizophrenia. Arch. Gen. Psychiatry. 2003;60:311–319. doi: 10.1001/archpsyc.60.3.311. [DOI] [PubMed] [Google Scholar]
  71. Kushner SF, Khan A, Lane R, Olson WH. Topiramate monotherapy in the management of acute mania: results of four double-blind placebo-controlled trials. Bipolar Disord. 2006;8:15–27. doi: 10.1111/j.1399-5618.2006.00276.x. [DOI] [PubMed] [Google Scholar]
  72. Lee HJ, Ghelardoni S, Chang L, Bosetti F, Rapoport SI, Bazinet RP. Topiramate does not alter the kinetics of arachidonic or docosahexaenoic acid in brain phospholipids of the unanesthetized rat. Neurochem Res. 2005;30:677–683. doi: 10.1007/s11064-005-2756-3. [DOI] [PubMed] [Google Scholar]
  73. Levant B, Grigoriadis DE, DeSouza EB. Characterization of [3H]quinpirole binding to D2-like dopamine receptors in rat brain. J. Pharmacol. Exp. Ther. 1992;262:929–935. [PubMed] [Google Scholar]
  74. Liauw SS, McIntyre RS. Atypical antipsychotic tolerability and switching strategies in bipolar disorder. Expert Opin Pharmacother. 2010;11:2827–2837. doi: 10.1517/14656566.2010.510835. [DOI] [PubMed] [Google Scholar]
  75. Lidow MS, Goldman-Rakic PS, Rakic P, Innis RB. Dopamine D2 receptors in the cerebral cortex: distribution and pharmacological characterization with [3H]raclopride. Proc. Natl. Acad. Sci. USA. 1989;86:6412–6416. doi: 10.1073/pnas.86.16.6412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Marinova Z, Ren M, Wendland JR, Leng Y, Liang MH, Yasuda S, Leeds P, Chuang DM. Valproic acid induces functional heat-shock protein 70 via Class I histone deacetylase inhibition in cortical neurons: a potential role of Sp1 acetylation. J. Neurochem. 2009;111:976–987. doi: 10.1111/j.1471-4159.2009.06385.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Mitsikostas D, Sfikakis A, Papadopoulou-Daifoti Z, Varonos D. The effects of valproate in brain monoamines of juvenile rats after stress. Prog. Neuropsychopharmacol. Biol Psychiatry. 1993;17:295–310. doi: 10.1016/0278-5846(93)90049-x. [DOI] [PubMed] [Google Scholar]
  78. Montezinho LP, Castro MM, Duarte CB, Penschuck S, Geraldes CF, Mork A. The interaction between dopamine D2-like and beta-adrenergic receptors in the prefrontal cortex is altered by mood-stabilizing agents. J. Neurochem. 2006;96:1336–1348. doi: 10.1111/j.1471-4159.2005.03654.x. [DOI] [PubMed] [Google Scholar]
  79. Monti B, Gatta V, Piretti F, Raffaelli SS, Virgili M, Contestabile A. Valproic acid is neuroprotective in the rotenone rat model of Parkinson's disease: involvement of alpha-synuclein. Neurotox. Res. 2010;17:130–141. doi: 10.1007/s12640-009-9090-5. [DOI] [PubMed] [Google Scholar]
  80. Monti B, Polazzi E, Contestabile A. Biochemical, molecular and epigenetic mechanisms of valproic acid neuroprotection. Curr. Mol. Pharmacol. 2009;2:95–109. doi: 10.2174/1874467210902010095. [DOI] [PubMed] [Google Scholar]
  81. Namkung Y, Sibley DR. Protein kinase C mediates phosphorylation, desensitization, and trafficking of the D2 dopamine receptor. J. Biol. Chem. 2004;279:49533–49541. doi: 10.1074/jbc.M408319200. [DOI] [PubMed] [Google Scholar]
  82. Nilsson CL, Hellstrand M, Ekman A, Eriksson E. Direct dopamine D2-receptor-mediated modulation of arachidonic acid release in transfected CHO cells without the concomitant administration of a Ca2+-mobilizing agent. Br. J. Pharmacol. 1998;124:1651–1658. doi: 10.1038/sj.bjp.0702025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Noponen M, Sanfilipo M, Samanich K, Ryer H, Ko G, Angrist B, Wolkin A, Duncan E, Rotrosen J. Elevated PLA2 activity in schizophrenics and other psychiatric patients. Biol. Psychiatry. 1993;34:641–649. doi: 10.1016/0006-3223(93)90157-9. [DOI] [PubMed] [Google Scholar]
  84. Ong WY, Sandhya TL, Horrocks LA, Farooqui AA. Distribution of cytoplasmic phospholipase A2 in the normal rat brain. J. Hirnforsch. 1999;39:391–400. [PubMed] [Google Scholar]
  85. Paxinos G, Watson C. The rat brain in stereotaxic coordinates. Third ed. New York: Academic Press; 1987. [Google Scholar]
  86. Pearlson GD, Wong DF, Tune LE, Ross CA, Chase GA, Links JM, Dannals RF, Wilson AA, Ravert HT, Wagner HN, Jr, et al. In vivo D2 dopamine receptor density in psychotic and nonpsychotic patients with bipolar disorder. Arch. Gen. Psychiatry. 1995;52:471–477. doi: 10.1001/archpsyc.1995.03950180057008. [DOI] [PubMed] [Google Scholar]
  87. Peet M, Peters S. Drug-induced mania. Drug Saf. 1995;12:146–153. doi: 10.2165/00002018-199512020-00007. [DOI] [PubMed] [Google Scholar]
  88. Peng GS, Li G, Tzeng NS, Chen PS, Chuang DM, Hsu YD, Yang S, Hong JS. Valproate pretreatment protects dopaminergic neurons from LPS-induced neurotoxicity in rat primary midbrain cultures: role of microglia. Brain Res. Mol. Brain Res. 2005;134:162–169. doi: 10.1016/j.molbrainres.2004.10.021. [DOI] [PubMed] [Google Scholar]
  89. Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J. Biol. Chem. 2001;276:36734–36741. doi: 10.1074/jbc.M101287200. [DOI] [PubMed] [Google Scholar]
  90. Poddubiuk ZM, Blumberg JB, Kopin IJ. Brain prostaglandin content in rats sacrificed by decapitation vs focused microwave irradiation. Experientia. 1982;38:987–988. doi: 10.1007/BF01953694. [DOI] [PubMed] [Google Scholar]
  91. Ralph-Williams RJ, Paulus MP, Zhuang X, Hen R, Geyer MA. Valproate attenuates hyperactive and perseverative behaviors in mutant mice with a dysregulated dopamine system. Biol. Psychiatry. 2003;53:352–359. doi: 10.1016/s0006-3223(02)01489-0. [DOI] [PubMed] [Google Scholar]
  92. Ramadan E, Basselin M, Rao JS, Chang L, Chen M, Ma K, Rapoport SI. Lamotrigine blocks NMDA receptro-initiated arachidonic acid signalling in rat brain:implications for its efficacy in bipolar disorder. Int. J. Neuopsychophamramcology. 2011;28:1–13. doi: 10.1017/S1461145711001003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Rao JS, Bazinet RP, Rapoport SI, Lee HJ. Chronic treatment of rats with sodium valproate downregulates frontal cortex NF-kB DNA binding activity and COX-2 mRNA. Bipolar Disord. 2007;9:513–520. doi: 10.1111/j.1399-5618.2007.00361.x. [DOI] [PubMed] [Google Scholar]
  94. Rao JS, Bazinet RP, Rapoport SI, Lee HJ. Chronic administration of carbamazepine downregulates AP-2 DNA binding activity and AP-2a protein expression in rat frontal cortex. Biol. Psychiatry. 2007;61:154–161. doi: 10.1016/j.biopsych.2006.03.029. [DOI] [PubMed] [Google Scholar]
  95. Rao JS, Rapoport SI, Bosetti F. Decrease in the AP-2 DNA-binding activity and in the protein expression of AP-2 alpha and AP-2 beta in frontal cortex of rats treated with lithium for 6 weeks. Neuropsychopharmacology. 2005;30:2006–2013. doi: 10.1038/sj.npp.1300740. [DOI] [PubMed] [Google Scholar]
  96. Rapoport SI. Arachidonic acid and the brain. J. Nutr. 2008;138:2515–2520. doi: 10.1093/jn/138.12.2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Rapoport SI, Basselin M, Kim H-W, Rao JS. Bipolar disorder and mechanism of action of mood stabilizers. Brain Res. Rev. 2009;61:185–209. doi: 10.1016/j.brainresrev.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Rapoport SI, Chang MC, Spector AA. Delivery and turnover of plasma-derived essential PUFAs in mammalian brain. J. Lipid Res. 2001;42:678–685. [PubMed] [Google Scholar]
  99. Ren M, Leng Y, Jeong M, Leeds PR, Chuang DM. Valproic acid reduces brain damage induced by transient focal cerebral ischemia in rats: potential roles of histone deacetylase inhibition and heat shock protein induction. J. Neurochem. 2004;89:1358–1367. doi: 10.1111/j.1471-4159.2004.02406.x. [DOI] [PubMed] [Google Scholar]
  100. Rivera R, Chun J. Biological effects of lysophospholipids. Rev. Physiol. Biochem. Pharmacol. 2008;160:25–46. doi: 10.1007/112_0507. [DOI] [PubMed] [Google Scholar]
  101. Robinson PJ, Noronha J, DeGeorge JJ, Freed LM, Nariai T, Rapoport SI. A quantitative method for measuring regional in vivo fatty-acid incorporation into and turnover within brain phospholipids: Review and critical analysis. Brain Res. Brain Res. Rev. 1992;17:187–214. doi: 10.1016/0165-0173(92)90016-f. [DOI] [PubMed] [Google Scholar]
  102. Seeman P, Van Tol HH. Dopamine receptor pharmacology. Trends Pharmacol Sci. 1994;15:264–270. doi: 10.1016/0165-6147(94)90323-9. [DOI] [PubMed] [Google Scholar]
  103. Shaldubina A, Einat H, Szechtman H, Shimon H, Belmaker RH. Preliminary evaluation of oral anticonvulsant treatment in the quinpirole model of bipolar disorder. J Neural Transm. 2002;109:433–440. doi: 10.1007/s007020200035. [DOI] [PubMed] [Google Scholar]
  104. Sharma RP, Rosen C, Kartan S, Guidotti A, Costa E, Grayson DR, Chase K. Valproic acid and chromatin remodeling in schizophrenia and bipolar disorder:preliminary results from clinical popoulation. Schizophr. Res. 2006;88:227–231. doi: 10.1016/j.schres.2006.07.015. [DOI] [PubMed] [Google Scholar]
  105. Shimshoni JA, Basselin M, Li LO, Coleman RA, Rapoport SI, Modi HR. Valproate uncompetitively inhibits arachidonic acid acylation by rat acyl-coA synthetase 4: Relevance to valproates' efficacy against bipolar disorder. Biochim. Biophys. Acta. 2011;1811:163–169. doi: 10.1016/j.bbalip.2010.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Smith LA, Cornelius VR, Azorin JM, Perugi G, Vieta E, Young AH, Bowden CL. Valproate for the treatment of acute bipolar depression: systematic review and meta-analysis. J. Affect. Disord. 2010;122:1–9. doi: 10.1016/j.jad.2009.10.033. [DOI] [PubMed] [Google Scholar]
  107. Tabachnick BG, Fidell LS. Computer-assisted research design and analysis. Boston: Allyn and Bacon ed; 2001. [Google Scholar]
  108. Tramontina JF, Andreazza AC, Kauer-Sant'anna M, Stertz L, Goi J, Chiarani F, Kapczinski F. Brain-derived neurotrophic factor serum levels before and after treatment for acute mania. Neurosci. Lett. 2009;452:111–113. doi: 10.1016/j.neulet.2009.01.028. [DOI] [PubMed] [Google Scholar]
  109. Vial D, Piomelli D. Dopamine D2 receptors potentiate arachidonate release via activation of cytosolic, arachidonic-specific phospholipase A2. J. Neurochem. 1995;64:2765–2772. doi: 10.1046/j.1471-4159.1995.64062765.x. [DOI] [PubMed] [Google Scholar]
  110. Walz JC, Frey BN, Andreazza AC, Cereser KM, Cacilhas AA, Valvassori SS, Quevedo J, Kapczinski F. Effects of lithium and valproate on serum and hippocampal neurotrophin-3 levels in an animal model of mania. J. Psychiatr. Res. 2008;42:416–421. doi: 10.1016/j.jpsychires.2007.03.005. [DOI] [PubMed] [Google Scholar]
  111. Wang J, Bannon MJ. Sp1 and Sp3 activate transcription of the human dopamine transporter gene. J. Neurochem. 2005;93:474–482. doi: 10.1111/j.1471-4159.2005.03051.x. [DOI] [PubMed] [Google Scholar]
  112. Wang J, Michelhaugh SK, Bannon MJ. Valproate robustly increases Sp transcription factor-mediated expression of the dopamine transporter gene within dopamine cells. Eur. J. Neurosci. 2007;25:1982–1986. doi: 10.1111/j.1460-9568.2007.05460.x. [DOI] [PubMed] [Google Scholar]
  113. Wang PW, Nowakowska C, Chandler RA, Hill SJ, Nam JY, Culver JL, Keller KL, Ketter TA. Divalproex extended-release in acute bipolar II depression. J. Affect. Disord. 2010;124:170–173. doi: 10.1016/j.jad.2009.10.021. [DOI] [PubMed] [Google Scholar]
  114. Wang X, Zhong P, Gu Z, Yan Z. Regulation of NMDA receptors by dopamine D4 signaling in prefrontal cortex. J. Neurosci. 2003;23:9852–9861. doi: 10.1523/JNEUROSCI.23-30-09852.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Yajima S, Lee SH, Minowa T, Mouradian MM. Sp family transcription factors regulate expression of rat D2 dopamine receptor gene. DNA Cell Biol. 1998;17:471–479. doi: 10.1089/dna.1998.17.471. [DOI] [PubMed] [Google Scholar]
  116. Yasuda S, Liang MH, Marinova Z, Yahyavi A, Chuang DM. The mood stabilizers lithium and valproate selectively activate the promoter IV of brain-derived neurotrophic factor in neurons. Mol. Psychiatry. 2009;14:51–59. doi: 10.1038/sj.mp.4002099. [DOI] [PubMed] [Google Scholar]
  117. Yatham LN, Liddle PF, Lam RW, Shiah IS, Lane C, Stoessl AJ, Sossi V, Ruth TJ. PET study of the effects of valproate on dopamine D2 receptors in neuroleptic- and mood-stabilizer-naive patients with nonpsychotic mania. Am. J. Psychiatry. 2002;159:1718–1723. doi: 10.1176/appi.ajp.159.10.1718. [DOI] [PubMed] [Google Scholar]
  118. Zhan L, Kerr JR, Lafuente MJ, Maclean A, Chibalina MV, Liu B, Burke B, Bevan S, Nasir J. Altered expression and coregulation of dopamine signalling genes in schizophrenia and bipolar disorder. Neuropathol. Appl. Neurobiol. 2011;37:206–219. doi: 10.1111/j.1365-2990.2010.01128.x. [DOI] [PubMed] [Google Scholar]

RESOURCES