Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 1.
Published in final edited form as: Int J Neuropsychopharmacol. 2011 Jun 28;15(7):931–943. doi: 10.1017/S1461145711001003

Lamotrigine blocks NMDA receptor-initiated arachidonic acid signalling in rat brain: Implications for its efficacy in bipolar disorder

Epolia Ramadan 1,*, Mireille Basselin 1, Jagadeesh S Rao 1, Lisa Chang 1, Mei Chen 1, Kaizong Ma 1, Stanley I Rapoport 1
PMCID: PMC3204186  NIHMSID: NIHMS310309  PMID: 21733229

Abstract

An upregulated brain arachidonic acid (AA) cascade and a hyperglutamatergic state characterize bipolar disorder (BD). Lamotrigine (LTG), a mood stabilizer approved for treating BD, is reported to interfere with glutamatergic neurotransmission involving N-methyl-D-aspartate receptors (NMDARs). NMDARs allow extracellular calcium into the cell, thereby stimulating calcium-dependent cytosolic phospholipase A2 (cPLA2) to release arachidonic acid (AA) from membrane phospholipid. We hypothesized that LTG, like other approved mood stabilizers, would reduce NMDAR-mediated AA signaling in rat brain. An acute subconvulsant dose of NMDA (25 mg/kg) or saline was administered intraperitoneally to unanesthetized rats that had been treated p.o. daily for 42 days with vehicle or a therapeutically relevant dose of LTG (10 mg/kg/.d). Regional brain AA incorporation coefficients k* and rates Jin, AA signals, were measured using quantitative autoradiography after intravenous [1-14C]AA infusion, as were other AA cascade markers. In chronic vehicle-treated rats, acute NMDA compared to saline increased k* and Jin in widespread regions of the brain, as well as prostaglandin (PG)E2 and thromboxane B2 concentrations. Chronic LTG treatment compared to vehicle reduced brain cyclooxygenase (COX) activity, PGE2 concentration, and DNA binding activity of the COX-2 transcription factor, NF-κB. Pretreatment with chronic LTG blocked the acute NMDA effects on AA cascade markers. In summary, chronic LTG like other mood stabilizers blocks NMDA-mediated signaling involving the AA metabolic cascade. Since markers of the AA cascade and of NMDAR signaling are up-regulated in the postmortem BD brain, mood stabilizers generally may be effective in BD by dampening NMDAR signalling and the AA cascade.

Keywords: arachidonic acid, cyclooxygenase, mood stabilizer, NMDA receptor, phospholipase A2

INTRODUCTION

Hyperglutamatergic neurotransmission has been implicated in bipolar disorder (BD) (Cherlyn et al., 2010; Clinton and Meador-Woodruff, 2004; Michael et al., 2003; Zarate et al., 2003). Furthermore, the postmortem BD brain shows fewer glutamatergic N-methyl-D-aspartate receptors (NMDARs), decreased protein and mRNA levels of NMDAR subunits NR1 and NR3A, reduced densities of NMDAR-associated postsynaptic proteins, PSD-95 and SAP102, and increased expression of the vesicular glutamate transporter 1 (Eastwood and Harrison, 2010; Hashimoto et al., 2007; McCullumsmith et al., 2007; Mueller and Meador-Woodruff, 2004; Rao et al., 2010). It has been shown that NMDAR stimulation by glutamate or NMDA decreases NR-1 expression (Gascon et al., 2005), and that the NR3A subunit co-assembles with other subunits (NR1, NR2A or NR2B) to form NMDARs with decreased activity and calcium influx (Ciabarra et al., 1995; Sucher et al., 1995). Further, chronic sub-convulsive administration of NMDA to rats decreased NR-1 and NR3A NMDA receptor subunits along with upregulated arachidonic acid (AA) selective cytosloic phospholipase A2 activity, protein and mRNA levels in rat frontal cortex (Rao et al., 2007a). Decreased NR1 and NR3A expression observed in BD is consistent with up-regulated NMDAR function (Kim et al., 2011; Rao et al., 2010), and with the observation that mice lacking the NR-3A subunit have increased brain NMDAR activity (Das et al., 1998).

When glutamate or NMDA binds to an NMDAR, extracellular Ca2+ enters the cell and activates, among other enzymes, Ca2+-dependent-cytosolic phospholipase A2 type IV (cPLA2-IV), which selectively releases arachidonic acid (AA, 20:4n-6) from cell membrane phospholipids (Basselin et al., 2006a; Basselin et al., 2008; Basselin et al., 2007a; Clark et al., 1991; Dumuis et al., 1988; Ramadan et al., 2010). Consistent with a hyperglutamatergic state, the postmortem BD brain shows up-regulated markers of AA metabolism, including cPLA2, cyclooxygenase (COX)-2, and membrane prostaglandin E synthase, which converts AA to pro-inflammatory prostaglandin (PG)E2 (Kim et al., 2011).

The brain AA signal involving NMDARs has been imaged in unanesthetized rats by infusing [1-14C]AA intravenously and measuring regional brain radioactivity following injection of NMDA (Basselin et al., 2007c). Operational equations, derived with a fatty acid model (Rapoport, 2001; Robinson et al., 1992), were used to identify the signal as increased regional AA incorporation coefficients k* or fluxes Jin. Acute administration of a subconvulsive dose of NMDA (25 mg/kg, i.p.) increased k* and Jin for AA in many brain regions. The increases could be blocked by pretreatment with the specific NMDAR antagonist, MK-801, or with each of three mood stabilizers effective against BD, namely lithium, valproate or carbamazepine (Basselin et al., 2006a; Basselin et al., 2008; Basselin et al., 2007a). We have also found that lithium and carbamazepine, when administered chronically at therapeutically relevant concentrations, reduced mRNA, protein, and activity levels of cPLA2-IV, and each of the three drugs, as well as lamotrigine (LTG), another FDA-approved mood stabilizer (Bowden, 2005; FDA, 2009), decreased protein and mRNA of COX-2 in rat brain (Lee et al., 2008; Rao et al., 2007b; Rao et al., 2007c; Rao et al., 2005; Rapoport et al., 2009).

LTG [3,5-diamino-6-(2,3-dichlorophenyl)-1,2,4-triazine] is thought to act on voltage-dependent Na+ and Ca2+ channels so as to reduce presynaptic neuronal depolarization, and thus glutamate release at the excitatory synapse (Cunningham and Jones, 2000; Sitges et al., 2007a; Sitges et al., 2007b; Xie and Hagan, 1998). Further, LTG can modulate neurotransmission via NMDARs (Anand et al., 2000; Farber et al., 2002; Wang et al., 1996).

In the present study, we hypothesized that LTG, like lithium, valproate and carbamazepine, when given chronically to rats (Basselin et al., 2006a; Basselin et al., 2008; Basselin et al., 2007a), would block NMDAR-initiated AA signaling in rat brain, and dampen other parameters of the AA metabolic cascade stimulated by NMDA. Confirming this would support our hypothesis that a common mechanism for the action of FDA-approved mood stabilizers in BD is down-regulation of the brain AA cascade that involves inhibition of NMDAR-mediated AA signaling (Rapoport et al., 2009; Rapoport and Bosetti, 2002). To test this hypothesis, we used our fatty acid method to measure incorporation coefficients k* and rates Jin for AA in 83 brain regions of unanesthetized rats that were injected acutely with saline or NMDA, and which had received LTG or vehicle daily for 42 days as described previously (Lee et al., 2008). Brain cPLA2-IV and COX activities, nuclear factor (NF)-κB DNA binding activity, and PGE2 and thromboxane (TX)B2 concentrations, also were measured.

MATERIALS AND METHODS

Animals and Diets

Male Fischer-344 rats, aged 2 months (Taconic Farms, USA) were acclimated for 1 wk in an animal facility with regulated temperature, humidity and light cycle, and with free access to food and water. They were fed Rodent NIH-31 Auto 18-4 diet (Zeigler Bros, Gardens, PA), which contained (as % total fatty acid), 20.1% saturated, 22.5% monounsaturated, 47.9% linoleic, 5.1% α-linolenic, 0.02% arachidonic, 2.0% eicosapentaenoic, and 2.3% docosahexaenoic acid (Demar et al., 2005). Experiments followed 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 the Eunice Kennedy Shriver National Institute of Child Health and Human Development.

Drugs and Tracers

Radiolabeled [1-14C]AA in ethanol (53 mCi/mmol, 99.4% pure, Moravek Biochemicals, USA) was evaporated and resuspended in Hepes buffer (pH 7.4), containing 50 mg/ml fatty acid-free bovine serum albumin (Sigma-Aldrich, USA) (DeGeorge et al., 1989). LTG (provided by NIMH Chemical Synthesis and Drug Supply Program, RTI International, USA)-treated rats received 10 mg/kg LTG by gavage once daily for 42 days. LTG was dissolved in a 1:1 volume dimethylsulfoxide (DMSO, ≥ 99% Sigma-Aldrich)/saline (0.9% sodium chloride) solution. This regimen produced a serum LTG concentration of 40 µM (Hassel et al., 2001), at the high end of concentrations reported in humans (20–30 µM) (Doose et al., 2003). Vehicle-treated rats (controls) received an equivalent volume of DMSO/saline.

Surgical Procedures and Tracer Infusion

On the morning of day 42, a rat was gavaged with its appropriate treatment at 1 h before undergoing anesthesia with halothane (2–3% v/v in O2). Polyethylene PE-50 catheters were surgically implanted into its right femoral artery and vein (Basselin et al., 2006a), and the rat was allowed to recover from anesthesia (3–4 h) in a temperature-controlled and sound-dampened box, while body 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. Ten minutes after injecting NMDA or saline, [1-14C]AA (2 ml, 170 µCi/kg) was infused into the femoral vein for 5 min with an infusion pump, at a rate of 400 µl/min. Twenty minutes after starting infusion, the rat was killed with an overdose of Nembutal (90 mg/kg, i.v.) and decapitated. The brain was removed quickly, frozen in 2-methylbutane maintained at −40°C on dry ice, and stored at −80°C for later sectioning.

Chemical Analysis

Blood samples, collected before, during or after [1-14C]AA infusion, were centrifuged 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 microlitres of the lower organic phase of the solution was used to determine the radiolabelled unesterified plasma AA concentration by liquid scintillation counting. As reported previously (DeGeorge et al., 1989), > 95–98% of plasma radioactivity following [1-14C] AA infusion was radiolabelled AA.

Unlabeled, unesterified fatty-acid concentrations also were determined in frozen arterial plasma (100 µl). Total lipids were extracted (Folch et al., 1957) and separated by thin-layer chromatography on 60 silica gel plates with heptane:diethylether:glacial acetic acid 60:40:3 (v/v/v). Unesterified fatty acids were scraped from the plate and converted to methyl ester derivatives (1% H2SO4 in methanol, 3 h, 70°C), which then were analyzed by gas chromatography with flame ionization detection, and quantified relative to the added standard, heptadecanoic acid (17:0).

Quantitative Autoradiography

Quantitative autoradiography was performed as reported previously (Basselin et al., 2006a). Radioactivity (nCi/g wet brain) in 83 bilateral anatomically identified regions (Paxinos and Watson, 1987) was determined by quantitative densitometry (NIH Image 1.62). Regional brain AA incorporation coefficients k* (ml plasma/s/g brain) were calculated as (Robinson et al., 1992),

k*=c*brain(20min)oTc*pldt (1)

where cbrain* (nCi/g wet brain wt) is brain radioactivity 20 min after beginning infusion, cplasma* (nCi/ml plasma) is labelled plasma unesterified AA, and t (min) is time after the beginning of [1-14C] AA infusion. Plasma radioactivity was integrated by trapezoidal integration to calculate k* for each experiment. Regional rates of incorporation of unesterified AA from plasma into brain phospholipids, Jin (nmol/s/g) were calculated as,

Jin=k*cplasma (2)

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

Activity of cPLA2 Type IV

In separate experiments, after the last LTG or vehicle dose was administered, a rat was anesthetized with Nembutal (50 mg/kg i.p.) and decapitated. The brain was removed, frozen in 2-methylbutane maintained at −40°C on dry ice, and stored at −80°C. Half of the brain was homogenized at 4°C using a Tenbroeck tissue grinder, in 2 vol cold buffer containing 10 mM Hepes (pH7.5), 1 mM EDTA, 0.34 M sucrose and protease inhibitor cocktail tablet (Complete, Germany). Homogenates were centrifuged (14,000 g for 20 min, then 100,000 g for1 h, 4°C), and supernatants corresponding to the cytosolic fractions were assayed for cPLA2-IV activity (Yang et al., 1999). Protein concentrations in the cytosolic fraction were determined by the Bradford method (Bradford, 1976).

COX Activity

Half of each brain was homogenized using a Tenbroeck tissue grinder in 3 ml of ice-cold lysate buffer at pH 7.8, containing 10 mM Tris-HCl, 0.15 M NaCl, 1 mM EDTA and 1% Igepal CA-630, and the homogenates centrifuged (14,000 g, 20 min, 4°C). Net COX activity was measured as the rate of PGE2 formation (pg PGE2/min.mg cytosolic protein) in the homogenate cytosolic fractions, as reported previously (Basselin et al., 2009). As a positive control, the specific COX-2 inhibitor Celebrex® (400 mg; Pfizer Inc., USA, Division of Veterinary Medicine, USA) was dissolved in DMSO at a concentration of 0.1% (w/v), and added to the mixture 10 min before adding AA (Basselin et al., 2009).

Nuclear Extracts and NF-κB DNA Binding Activity

Nuclear extracts from the frontal cortex of either the chronic vehicle- or LTG-treated animals, prepared as reported (Lahiri, 1998; Rao et al., 2007b), were analyzed by gel shift assays to quantify DNA binding activity of NF-κB (Kaltschmidt et al., 2002).

Eicosanoid Concentrations

Rat brains were prepared, microwaved and used as reported previously (Basselin et al., 2008; Basselin et al., 2007a). Eicosanoid concentrations were measured with polyclonal PGE2 and TXB2 ELISA assay kits (Oxford Biochemical Research, USA).

Statistical Analysis

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

RESULTS

Physiology and Arterial Plasma Radioactivity

Mean body weights of 42-d LTG-treated rats did not differ significantly from weight of the vehicle-treated rats [267.5 ±18.1 g (n = 16) vs. 265.5 ±18.0 g (n = 16)], as reported (Daoud et al., 2004; Hassel et al., 2001). There also was no significant group difference in mean rectal temperature, heart rate or arterial blood pressure (data not shown). Acute NMDA decreased heart rate by 21–23% (p < 0.0001) in the chronic LTG- and vehicle-treated rats (data not shown), as reported by Basselin et al. (2006a, 2007a, 2008).

Neither chronic LTG nor acute NMDA modified the time course of arterial plasma radioactivity [equation (1)] during i.v. [1-14C]AA infusion. Integrated radioactivity in the plasma organic fraction (nCi × s)/(ml plasma) (n = 7–9), the input function in equation (1), did not differ significantly among groups: chronic vehicle + saline, 147,543 ± 30,055; chronic vehicle + NMDA, 150,018 ± 22,506; chronic LTG + saline, 149,499 ± 43,167; chronic LTG + NMDA, 145,409 ± 32,774.

Plasma Concentrations of Unlabeled Unesterified Fatty Acid

A two-way ANOVA showed a significant main effect of LTG on plasma concentrations of unesterified palmitic, palmitoleic, stearic, oleic, linoleic, α-linolenic, arachidonic and docosahexaenoic acid, which were reduced in chronic LTG-treated rats compared to the chronic vehicle-treated rats (p<0.05). No significant main effect of NMDA, or interaction between LTG and NMDA, was detected (Table 1).

Table 1.

Effects of NMDA and Lamotrigine on Unesterified Plasma Fatty Acid Concentrations

Chronic Vehicle
Chronic LTG
LTG X NMDA
Interaction
(p value)
LTG effect
(p value)
NMDA effect
(p value)
Fatty Acid Saline
(n=7)
NMDA
(n=9)
Saline
(n=8)
NMDA
(n=8)
(nmol/ml plasma)
Palmitic (16:0) 469.5 ± 80.1 415.5 ± 72.0 258.8 ± 64.2 326.6 ± 114.2 0.0610 < 0.0001 0.8266
Palmitoleic (16:1) 73.5 ± 19.9 61.4 ± 19.7 32.8 ± 7.0 43.9 ± 19.1 0.0710 < 0.0001 0.7796
Stearic (18:0) 76.7 ± 16.3 63.9 ± 12.1 52.6 ± 20.0 54.9 ± 14.6 0.2174 0.0099 0.3877
Oleic (18:1 n-9) 265.0 ± 44.4 236.7 ± 44.8 171.9 ± 39.4 220.7 ± 72.7 0.0597 0.0097 0.6058
Linoleic (18:2 n-6) 398.3± 63.9 370.8 ± 68.2 221.3 ± 55.4 308.3 ± 117.9 0.0565 0.0003 0.3100
α-Linolenic (18:3 n-3) 32.0 ± 5.0 29.0 ± 5.4 16.6 ± 3.2 22.3 ± 7.9 0.0763 < 0.0001 0.5724
Arachidonic (20:4 n-6) 29.6 ± 6.9 28.7 ± 9.3 21.4 ± 7.9 23.0 ± 11.3 0.2537 0.0062 0.9254
Docosahexaenoic (22:6 n-3) 62.2 ± 17.9 47.5 ± 15.2 28.0 ± 15.9 36.5 ± 21.3 0.0660 0.0009 0.6131

NMDA, N-methyl-D-aspartic acid, 25 mg/kg, i.p.; LTG, lamotrigine.

Values are means ± SD measured from arterial plasma collected before [1-14C] AA infusion.

Regional Brain AA Incorporation Coefficients, k*

Figure 1 presents representative coronal autoradiographs of brains from rats treated for 42 d with vehicle (control) or with LTG, then acutely injected with saline or NMDA. k* for AA, calculated by equation (1), is colour-coded. The figure shows no evident difference in regional values of k* in response to acute saline between animals treated chronically with LTG compared with vehicle. Acute NMDA evidently increased k* in multiple brain regions of the chronic saline-injected but not LTG-injected rats. Data such as illustrated in Fig. 1 were collated in Table 2.

Figure 1.

Figure 1

Coronal brain autoradiographs showing effects of NMDA and LTG on regional AA incorporation coefficients k* in rats. Values of k* (ml/s/g brain) × 10−4 are given on a color scale from 7 (purple) to 23 (yellow-orange). CPu, caudate putamen; Hipp, hippocampus; LTG, lamotrigine; NMDA, N-methyl-D-aspartic acid, 25 mg/kg, i.p.; Mot, motor cortex; PFr, prefrontal cortex; Som, somatosensory cortex. NMDA, N-methyl-D-aspartate, 25 mg/kg, i.p..

Table 2.

Interactive effects of chronic lamotrigine and acute NMDA on regional arachidonic acid incorporation coefficients in rat brain

Vehicle LTG LTG × NMDA
Interaction
(p value)
Brain region Saline
(n = 7)
NMDA
(n = 9)
Saline
(n = 8)
NMDA
(n = 8)
k*, (ml/s/g) × 10−4
Prefrontal cortex layer I 7.83 ± 1.50 11.53 ± 3.65 9.77 ± 2.42 7.59 ± 2.57 0.005
Prefrontal cortex layer IV 10.16 ± 1.66 14.41 ± 3.89 11.37 ± 3.00 9.96 ± 3.90 0.023
Primary olfactory cortex 9.35 ± 1.49 12.97 ± 4.13 11.25 ± 3.12 9.73 ± 3.85 0.041
Pyriform cortex 6.44 ± 2.01 10.01 ± 3.09* 6.88 ± 1.03 5.62 ± 1.43 0.003
Anterior cingulate cortex 12.54 ± 1.95 18.59 ± 5.49 15.25 ± 3.63 12.19 ± 5.00 0.006
Motor cortex
 Layer I 7.89 ± 1.80 11.82 ± 3.17* 10.19 ± 2.27 7.56 ± 2.67 0.001
 Layer II –III 8.91 ± 1.73 13.49 ± 4.03* 11.7 ± 2.87 8.79 ± 3.15 0.002
 Layer IV 10.40 ± 1.81 15.91 ± 5.09* 13.23 ± 3.22 10.77 ± 3.29 0.005
 Layer V 9.96 ± 1.76 15.01 ± 4.80* 12.45 ± 3.04 9.65 ± 3.74 0.005
 Layer VI 9.29 ± 1.95 13.83 ± 4.07* 11.50 ± 2.38 8.89 ± 3.25 0.003
Somatosensory cortex
 Layer I 8.70 ± 1.59 12.70 ± 3.29* 10.77 ± 2.64 8.68 ± 3.57 0.007
 Layer II – III 9.75 ± 2.25 14.83 ± 3.63* 12.33 ± 3.05 9.68 ± 3.61 0.002
 Layer IV 11.21 ± 2.09 17.82 ± 4.79* 14.24 ± 3.75 11.39 ± 2.47 0.002
 Layer V 10.08 ± 2.00 14.98 ± 4.35* 12.83 ± 3.27 9.71 ± 3.72 0.003
 Layer VI 9.74 ± 1.95 14.36 ± 4.06* 11.72 ± 2.38 9.18 ± 3.27 0.003
Preoptic area (LPO/MPO) 6.70 ± 1.19 10.56 ± 3.62* 8.02±1.53 7.67 ± 3.44 0.040
Globus pallidus 7.56 ± 1.45 10.59 ± 2.96 8.97 ± 1.98 7.18 ± 3.17 0.012
Diagonal band Dorsal 9.32 ± 1.73 13.77 ± 3.99* 11.28 ± 3.40 8.32 ± 3.92 0.008
 Ventral 9.47 ± 2.12 14.06 ± 3.68* 11.35 ± 3.05 9.06 ± 4.32 0.009
Amygdala basolateral/medial 7.54 ± 1.19 11.48 ± 3.52* 8.68 ± 2.56 6.98 ± 2.02 0.004
Hippocampus
 CA1 6.72 ± 1.23 9.75 ± 2.84** 7.95 ± 2.73 6.78 ± 2.34 0.021
 CA2 7.57 ± 1.37 10.61 ± 2.82** 8.44 ± 2.72 6.75 ± 2.05 0.009
 CA3 7.54 ± 1.19 10.44 ± 2.37* 8.44 ± 2.79 7.20 ± 2.13 0.017
 Dentate gyrus 8.01 ± 1.54 11.14 ± 2.76* 9.23 ± 2.73 7.65 ± 2.09 0.009
Nucleus Accumbens 10.09 ± 0.72 14.30 ± 3.92* 10.03 ± 2.07 8.39 ± 1.99 0.003
Caudate putamen
 Dorsal 9.69 ± 1.63 13.65 ± 3.34* 10.97 ± 2.76 8.29 ± 3.63 0.004
 Ventral 10.11 ± 1.84 14.04 ± 3.31* 11.00 ± 2.37 8.39 ± 3.89 0.005
 Lateral 10.03±1.99 14.66 ± 3.49* 11.08 ± 2.69 8.37 ± 3.97 0.003
 Medial 9.69 ± 2.05 13.85 ± 3.05* 11.11 ± 2.44 8.32 ± 4.22 0.004
Septal nu lateral 7.56 ± 1.64 10.05± 3.18 8.82 ± 2.07 6.00 ± 2.15 0.004
Septal nu medial 9.91 ± 2.40 13.17 ± 4.53 11.42 ± 3.06 8.12 ± 3.80 0.016
Habenular nu lateral 14.97 ± 2.57 21.09 ± 4.30* 17.17 ± 5.53 15.06 ± 2.45 0.007
Habenular nu medial 12.81 ± 1.88 16.75 ± 4.30 14.86 ± 4.61 12.49 ± 2.32 0.019
Lateral geniculate nu dorsal 11.98 ± 2.10 16.56 ± 4.21 14.13 ± 4.11 11.86 ± 2.48 0.009
Thalamus
Ventroposterior lateral nu 10.30 ± 1.89 15.08 ± 3.71* 12.69 ± 3.47 11.11 ± 2.86 0.008
Ventroposterior medial nu 10.21 ± 1.74 15.30 ± 4.66* 12.41 ± 3.12 10.64 ± 2.72 0.007
Anteroventral nu 14.85 ± 2.30 21.65 ± 6.25* 16.54 ± 3.99 12.74 ± 3.85 0.001
Reticular nu 11.16 ± 1.89 14.38 ± 2.36* 12.62 ± 2.02 10.00 ± 2.66 0.001
Paraventricular nu 9.80 ± 1.11 12.30 ± 4.13 11.69 ± 2.12 8.88 ± 3.22 0.018
Parafascicular nu 10.56 ± 2.12 16.35 ± 4.10** 12.96 ± 3.65 11.27 ± 2.90 0.003
Subthalamic nu 11.09 ± 1.35 14.75 ± 3.38 13.58 ± 4.58 11.24 ± 2.01 0.012
Hypothalamus
Supraoptic nu 8.09 ± 1.61 11.48 ± 3.09* 9.97 ± 2.48 7.50 ± 2.87 0.004
Lateral 8.09 ± 097 10.08 ± 1.33 9.53 ± 1.84 7.60 ± 2.86 0.007
Anterior 8.53 ± 1.23 12.02 ± 3.49 10.36 ± 1.99 8.89 ± 4.45 0.034
Periventricular 8.23 ± 1.08 12.30 ± 3.12** 9.95 ± 1.78 7.73 ± 3.06 0.001
Arcuate 7.86 ± 1.74 11.80 ± 3.13* 9.10 ± 1.81 7.53 ± 2.85 0.004
Ventromedial 7.94 ± 1.74 11.99 ± 3.61* 9.26 ± 2.05 7.51 ± 2.51 0.005
Posterior 8.92 ± 1.51 12.06 ± 2.58* 10.91 ± 1.68 8.90 ± 2.91 0.003
Zona incerta 10.53 ± 1.57 15.53 ± 3.69** 12.42 ± 3.85 11.35 ± 1.82 0.008
Cerebellar white matter 5.53 ± 1.09 7.60 ± 1.23 6.81 ± 2.08 5.88 ± 1.85 0.016
Non-blood-brain barrier regions
Subfornical organ 7.65 ± 1.87 12.07 ± 4.39* 9.24 ± 3.64 6.68 ± 2.74 0.007
Median eminence 7.89 ± 1.75 10.89 ± 2.32 9.17 ± 2.78 7.62 ± 2.81 0.016

Abbreviations: nu, nucleus; lat, lateral; med, medial; SLM, stratum lacunosum-moleculare; LTG, lamotrigine.

NMDA administration: 25 mg/kg i.p. for 10 min. Mean ± S.D.

*

p < 0.05;

**

p < 0.01; vehicle + NMDA vs. vehicle + saline (one-way ANOVA Bonferroni post-hoc tests).

Mean AA incorporation coefficients k* in each of 83 brain regions were subjected to a two-way ANOVA (Table 2). Statistically significant interactions between LTG and NMDA were found in 52 regions, for each of which a one-way ANOVA with Bonferroni’s post-hoc test showed that chronic LTG did not significantly change mean baseline (after saline) k* (Table 2). In 37 of the 52 regions, the same one-way ANOVA showed that NMDA compared with saline significantly increased k* by 29–59% in chronic vehicle-treated rats. Affected regions included pyriform (55%), motor (49–53%), sensory cortical areas (39–46%), preoptic area (58%), diagonal band (48%), amygdala (52%), hippocampus [CA1, CA2, CA3, dentate gyrus] (31–45%), nucleus accumbens (42%), caudate-putamen (39–46%), lateral habenular nucleus (41%), thalamus [ventroposterior lateral, ventroposterior medial, anteroventral and parafascicular nuclei] (46–50%), hypothalamus [supraoptic, periventricular, arcuate, ventromedial, posterior nuclei] (35–51%), zona incerta (47%), and the subfornical organ (58%). However, NMDA compared to saline did not significantly increase k* in any of the 37 regions in chronic LTG-treated rats.

In none of the 31 regions having a statistically insignificant LTG × NMDA interaction did LTG or NMDA have a significant main effect on k* for AA (data not shown). Thus, chronic LTG prevented each of the 37 significant NMDA-induced k* increments that were observed in the chronic vehicle-treated rats, but did not have a significant effect in naive rats (Table 2 and Fig. 1).

Regional Rates of Incorporation of Unlabeled Unesterified AA into Brain

Baseline (after saline) and NMDA-induced regional values of Jin (products of k* and the unesterified plasma AA concentration) were calculated by equation (2). A two-way ANOVA showed statistically significant interactions between LTG and NMDA in 44 of the 83 regions that were examined (data not shown). In 32 of these, a one-way ANOVA and Bonferroni’s post-hoc test showed that acute NMDA compared with saline significantly increased Jin (by 7–65%) in chronic vehicle- but not chronic LTG-treated rats. In the 39 regions where LTG × NMDA interactions with regard to Jin were statistically insignificant, chronic LTG compared to vehicle had a negative main effect (10 of the 39), largely reflecting the reduced plasma AA concentration following LTG.

Brain Enzyme and NF-κB Binding Activities

Since cPLA2-IV selectively releases AA from membrane phospholipids, we measured the enzymatic activity of this protein. Whole brain cPLA2-IV activity did not differ significantly (p = 0.91) between rats that received chronic LTG compared with vehicle (Fig. 2a). Since a Ca2+ chelator is used to determine cPLA2 activity in vitro (see Methods), we did not measure brain cPLA2-IV activity following acute NMDA injection, because the elevated intracellular Ca2+ concentration associated with NMDA administration in vivo could not be produced reliably by NMDA in vitro (Clark et al., 1991).

Figure 2.

Figure 2

Effects of chronic LTG on cPLA2, COX, and NF-kB DNA binding activities.

(A) Effects of chronic LTG on whole brains: cPLA2-IV activity. Data are means ± SD (n=8) and were analyzed with unpaired t-test.

(B) Effects of chronic LTG on whole brain cyclooxygenase (COX) activity. Data are means ± SD (n=8) and were analyzed with unpaired t-test. *** p <0.001. PGE2, prostaglandin E2.

(C) Effects of chronic LTG on NF-kB DNA binding activity. Values are represented as % control (n = 8). *** p < 0.001.

Our previous study showed that chronic LTG down-regulated protein and mRNA of COX-2 by 50% in rat brain in the absence of a decrease of COX-1 (Lee et al., 2008). We therefore determined whether COX activity was altered after LTG treatment. As shown in Fig. 2b, brain COX activity was decreased significantly by 56% (p < 0.001) in chronic LTG-treated compared with vehicle-treated rats. Compared with vehicle, Celebrex® (100 µM) inhibited COX activity by 70% (25.3 ± 3.0 vs. 84.8 ± 10.5 pg/min.mg protein, n = 8, data not shown), as has been reported (Basselin et al., 2009). To consider the transcriptional mechanism underlying the decreased COX-2 expression, we measured the DNA binding activity of NF-κB, a major transcription factor for COX-2 (Kaltschmidt et al., 2002). Figure 2c illustrates that chronic LTG significantly decreased by 38% (p < 0.001) the binding of nuclear proteins to the NF-κB consensus sequence of NF-κB. In the presence of excess (100 times) unlabelled specific NF-κB oligonucleotides, binding of labelled oligonucleotides was blocked, confirming specificity of the DNA binding activity.

Brain Eicosanoid Concentrations

To examine the effect of reduced COX-2 activity on COX-derived products, concentrations of PGE2 and TXB2 were determined in microwaved brains. A two-way ANOVA demonstrated statistically significant interactions between chronic LTG and acute NMDA with regard to brain PGE2 and TXB2 concentrations (Table 3). A subsequent one-way ANOVA with Bonferroni’s post-hoc test showed that chronic LTG reduced the basal PGE2 concentration by 52 % (p < 0.05), without affecting the TXB2 concentration. Acute NMDA significantly increased brain PGE2 (p < 0.001) and TXB2 (p = 0.007) concentrations by 2.2- and 2.4-fold, respectively, in chronic vehicle-treated rats, but did not affect either concentration in chronic LTG-treated rats.

Table 3.

Effect of NMDA on brain PGE2 and TXB2 concentrations in chronic vehicle- and LTG-treated rats

Chronic Vehicle
Chronic LTG
LTG × NMDA
Interaction
P-value
Saline
(n = 8)
NMDA
(n = 8)
Saline
(n = 6)
NMDA
(n = 6)
PGE2 (ng/g brain) 5.2 ± 1.3 11.5 ± 2.8*** 2.5 ± 0.6** 1.8 ± 0.5 < 0.0001
TXB2 (pg/g brain) 27.3 ± 12.4 65.0 ± 29.0*** 19.2 ± 7.1 16.0 ± 9.4 0.0067

Each value is mean ± SD. Bonferroni’s multiple comparison tests were performed.

**

P < 0.01;

***

P<0.001; vehicle plus NMDA versus vehicle plus saline, LTG plus saline versus vehicle plus saline, and LTG plus NMDA versus LTG plus saline.

NMDA, N-methyl-D-aspartic acid; LTG, lamotrigine.

DISCUSSION

Oral administration of LTG (10 mg/kg) daily for 42 d to rats, sufficient to produce a plasma LTG concentration therapeutically relevant to BD (Doose et al., 2003), blocked increments in k* and Jin for AA and in brain PGE2 and TXB2 concentrations that could be produced by acute NMDA administration (25 mg/kg i.p.) in vehicle-treated rats. Chronic LTG treatment compared with vehicle did not change baseline k* or Jin in any of 83 brain regions examined, nor brain cPLA2-IV activity or TXB2 concentration, but did reduce brain COX activity, NF-κB DNA binding activity and PGE2 concentration. Chronic LTG treatment also had a negative main effect on the plasma concentrations of each of eight measured unesterified fatty acids. These observations support our hypothesis that chronic LTG treatment blocks NMDA-signaling involving AA and its metabolites.

As similar effects on the NMDA-induced AA signal have been reported following administration of MK-801, a specific NMDAR antagonist, or of lithium, valproate or carbamazepine to rats (Basselin et al., 2006a, 2007a, 2008; Rapoport et al., 2009), these results support the hypothesis that mood stabilizers effective against BD commonly down-regulate the brain AA cascade, and provide one plausible mechanism for this effect, blockade of NMDAR-mediated activation of the cascade (Rapoport and Bosetti, 2002; Rapoport et al., 2009).

Chronic LTG’s suppression of the NMDA-induced increases in k* and Jin for AA and in PGE2 and TXB2 concentrations, which were produced in chronic vehicle-treated rats, could have been due to LTG’s reduction of COX-2 expression. Indeed, chronic LTG decreased COX activity, DNA binding activity of NF-κB, PGE2 and TXB2 concentrations, consistent with a report showing down-regulation of mRNA and protein of COX-2 in rat brain (Lee et al., 2008). Pharmacological inhibition or knocking out the COX-2 gene in rodent brain also reduced increments in k* for AA and in PGE2 concentration caused by giving drugs acting at cPLA2-coupled neuroreceptors (Basselin et al., 2006b, 2007b). Similar to LTG, chronic administration of lithium, valproate or carbamazepine to rats decreased brain COX activity and PGE2 concentration (Rapoport et al., 2009). When considering the possible role for inflammatory processes in BD with increased brain interleukin-1®, NF-κB subunits (p50 and p65), COX-2, membrane prostaglandin E synthase, and astroglial and microglial markers (glial fibrillary acidic protein, inducible nitric oxide synthase, and CD11b) (Kim et al., 2011; Kim et al., 2010; Rao et al., 2010), it is important to note that the four mood stabilizers, lithium, valproate, carbamazepine, and LTG suppress brain COX-2, PGE2, and/or NF-κB DNA binding activity which can be viewed as protecting against inflammatory processes (Lee et al., 2008; Rapoport et al., 2009). 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 NMDA signal.

Chronic LTG’s effect on the NMDAR itself is unlikely to have influenced the brain AA signal in response to NMDA. Unlike lithium, carbamazepine or valproic acid (Basselin et al., 2006a, 2007a, 2008), however, LTG does not affect glutamate binding to the NMDAR (Xie and Hagan, 1998), nor protein expression or activity of protein kinase C (Hahn et al., 2004), which can phosphorylate the NMDAR (Leonard and Hell, 1997), suggesting that LTG employs different intracellular mechanisms (e.g. reduction in glutamate release following inhibition of Na+ channels), in addition to the COX pathway (see above), for long-term changes in NMDAR-initiated AA signaling. The BD brain shows increased glutamate signalling associated with up-regulated markers of AA metabolism and of excitotoxicity, reduced brain-derived neurotrophic factor (BDNF), and apoptosis (Kim et al., 2010, 2011; Rao et al., 2010) and there is clinical evidence of hyperglutamatergic function (Cherlyn et al., 2010; Clinton et al., 2004; Michael et al., 2003; Zarate et al., 2003). Thus, down-regulation by LTG of NMDAR-initiated AA signaling and of COX activity, and of formation of pro-inflammatory PGE2, may contribute to LTG’s efficacy in BD and to its reported neuroprotective properties (see below). Recent studies indicated that LTG reduced brain glutamine levels in depressed BD patients (Frye et al., 2007), and that memantine, a NMDAR antagonist, was beneficial in BD patients (Koukopoulos et al., 2010; Teng and Demetrio, 2006). As a rationale for applying agents that modulate the glutamatergic system in treating patients with BD, riluzole, an inhibitor of glutamate release, has been reported to be effective alone (Brennan et al., 2010) or in combination with lithium in open-label trials for the treatment of bipolar depression (Zarate et al., 2005). In healthy subjects, LTG decreased perceptual abnormalities induced by ketamine, an NMDAR antagonist (Anand et al., 2000).

As with lithium and valproate, exposing neuroblastoma or cerebral cortical cells to LTG inhibited glycogen synthase kinase-3β and increased activity of glutathione S-transferase, an antioxidant enzyme (Bakare et al., 2009; Hayes and Strange, 2000; Li et al., 2002; Strange et al., 2001). LTG increased brain mRNA and protein levels of BDNF and of anti-apoptotic factor B-cell lymphoma-2 (Bcl-2), and reversed stress-induced downregulation of BDNF in rat brain (Chang et al., 2009; Li et al., 2010). Given that the BD brain shows reduced expression of BDNF and of other neurotrophic factors, and increased markers of apoptosis (Dwivedi et al., 2003; Kim et al., 2010; Knable et al., 2004), these actions may contribute to neuroprotection by LTG in BD (Ketter et al., 2003; Rapoport et al., 2009), and in experimental models of cerebral ischemia and glutamate-induced excitotoxicity (Bacher and Zornow, 1997; Lees and Leach, 1993; Maj et al., 1998; Rataud et al., 1994).

The statistically insignificant effects of chronic LTG on baseline k* and Jin for AA and on baseline cPLA2-IV activity agree with prior reports that chronic LTG did not alter rat brain cPLA2-IV mRNA or protein levels (Bazinet et al., 2006; Chang et al., 1996; Ghelardoni et al., 2004; Lee et al., 2008; Lee et al., 2007; Rintala et al., 1999). Under basal conditions, LTG does not alter resting membrane potential, neuronal excitability, or low-frequency excitatory synaptic transmission (Xie and Hagan, 1998), or glutamate release in the hippocampus of freely moving rats (Ahmad et al., 2004). In this study, we did not determine effects of an acute LTG dose of 10 mg/kg, since this dose produces plasma LTG concentration around 11–13 µM (Ahmad et al., 2004), far below the therapeutic level (Hassel et al., 2001).

The baseline values of k* and Jin in this study agree with reported values, as do the increments in these parameters and in PGE2 and TXB2 concentrations following acute NMDA (Basselin et al., 2007a, 2008; Okada et al., 2000; Pepicelli et al., 2002). Given that Jin represents incorporation of unesterified plasma AA as compensation for AA metabolic loss in brain (Basselin et al., 2007b; Rapoport, 2003), our data indicate comparable baseline AA loss but reduced NMDA-initiated loss following chronic LTG. Lee et al. (2007) reported that chronic LTG did not significantly change the plasma concentration of any unlabelled unesterified fatty acids, but in our study, this effect was statistically significant (p < 0.05) for the eight fatty acids analyzed. This discrepancy may be due to differences in gavage, fasting time, or quality of the unlabelled fatty-acid standards. The reduced unesterified plasma fatty-acid concentrations caused by LTG may reflect a liver effect of drug, since chronic LTG also reduces plasma levels of cholesterol and triglycerides (Daoud et al., 2004). Circulating unesterified fatty acids are derived by hydrolysis of esterified fatty acids that are secreted by the liver and from adipose tissue (Gao et al., 2009; Gibbons et al., 2004; Purdon et al., 1997).

In summary, 42 d of daily LTG blocked acute NMDA-initiated brain signaling via AA in unanesthetized rats. This observation, taken with reports that lithium, valproate and carbamazepine have a comparable action (Basselin et al. 2006a, 2007a, 2008), support our hypothesis that mood stabilizers effective in BD have a common mechanism of action that involves down-regulation of the brain AA cascade and interference with NMDAR receptor signaling via AA (Rapoport et al., 2009; Rapoport & Bosetti, 2002). Studying the AA cascade and the NMDAR signal in unanesthetized rats, as a common target for these agents, might help to screen new and potentially clinically relevant therapeutic agents for BD.

Acknowledgements

The research was supported by the Intramural Research Program of the National Institute on Aging, National Institutes of Health. We appreciate the editorial assistance of the NIH Fellows Editorial Board and thank Dr. Edmund Reese and Dr. Ameer Taha for proofreading the paper.

Footnotes

Statement of Interest

None.

REFERENCES

  1. Ahmad S, Fowler LJ, Whitton PS. Effects of acute and chronic lamotrigine treatment on basal and stimulated extracellular amino acids in the hippocampus of freely moving rats. Brain Research. 2004;1029:41–47. doi: 10.1016/j.brainres.2004.09.016. [DOI] [PubMed] [Google Scholar]
  2. Anand A, Charney DS, Oren DA, Berman RM, et al. Attenuation of the neuropsychiatric effects of ketamine with lamotrigine: support for hyperglutamatergic effects of N-methyl-D-aspartate receptor antagonists. Archives General Psychiatry. 2000;57:270–276. doi: 10.1001/archpsyc.57.3.270. [DOI] [PubMed] [Google Scholar]
  3. Bacher A, Zornow MH. Lamotrigine inhibits extracellular glutamate accumulation during transient global cerebral ischemia in rabbits. Anesthesiology. 1997;86:459–463. doi: 10.1097/00000542-199702000-00021. [DOI] [PubMed] [Google Scholar]
  4. Bakare A, Shao L, Cui J, Young LT, et al. Mood stabilizing drugs lamotrigine and olanzapine increase expression and activity of glutathione S-transferase in primary cultured rat cerebral cortical cells. Neuroscience Letters. 2009;455:70–73. doi: 10.1016/j.neulet.2009.03.022. [DOI] [PubMed] [Google Scholar]
  5. 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]
  6. Basselin M, Chang L, Chen M, Bell JM, et al. Chronic administration of valproic acid reduces brain NMDA signaling via arachidonic acid in unanesthetized rats. Neurochemical Research. 2008;33:1373–1383. doi: 10.1007/s11064-008-9700-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Basselin M, Fox M, Chang L, Bell JM, et al. Imaging elevated brain arachidonic acid signaling in unanesthetized serotonin transporter (5-HTT)-deficient mice. Neuropsychopharmacology. 2009;34:1695–1709. doi: 10.1038/npp.2008.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Basselin M, Villacreses NE, Chen M, Bell JM, et al. Chronic carbamazepine administration reduces NMDA receptor-initiated signaling via arachidonic acid in rat brain. Biological Psychiatry. 2007a;62:934–943. doi: 10.1016/j.biopsych.2007.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Basselin M, Villacreses NE, Langenbach R, Ma K, et al. Resting and arecoline-stimulated brain metabolism and signaling involving arachidonic acid are altered in the cyclooxygenase-2 knockout mice. Journal of Neurochemistry. 2006b;96:669–679. doi: 10.1111/j.1471-4159.2005.03612.x. [DOI] [PubMed] [Google Scholar]
  10. Basselin M, Villacreses NE, Lee H-J, Bell JM, et al. Flurbiprofen, a cyclooxygenase inhibitor, reduces the brain arachidonic acid signal in response to the cholinergic muscarinic, arecoline, in awake rats. Neurochemical Research. 2007b;32:1857–1867. doi: 10.1007/s11064-007-9372-3. [DOI] [PubMed] [Google Scholar]
  11. Basselin M, Villacreses NE, Lee HJ, Bell JM, et al. Chronic lithium administration attenuates up-regulated brain arachidonic acid metabolism in a rat model of neuroinflammation. Journal Neurochemistry. 2007c;102:761–772. doi: 10.1111/j.1471-4159.2007.04593.x. [DOI] [PubMed] [Google Scholar]
  12. Bazinet RP, Rao JS, Chang L, Rapoport SI, et al. 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. Biological Psychiatry. 2006;59:401–407. doi: 10.1016/j.biopsych.2005.07.024. [DOI] [PubMed] [Google Scholar]
  13. Bowden CL. Treatment options for bipolar depression. Journal of Clinical Psychiatry. 2005;66(Suppl):3–6. [PubMed] [Google Scholar]
  14. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical Biochemistry. 1976;72:248–254. doi: 10.1006/abio.1976.9999. [DOI] [PubMed] [Google Scholar]
  15. Brennan BP, Hudson JI, Jensen JE, McCarthy J, et al. Rapid enhancement of glutamatergic neurotransmission in bipolar depression following treatment with riluzole. Neuropsychopharmacology. 2010;35:834–846. doi: 10.1038/npp.2009.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chang MCJ, Grange E, Rabin O, Bell JM, et al. Lithium decreases turnover of arachidonate in several brain phospholipids. Neuroscience Letters. 1996;220:171–174. doi: 10.1016/s0304-3940(96)13264-x. [DOI] [PubMed] [Google Scholar]
  17. Chang YC, Rapoport SI, Rao JS. Chronic administration of mood stabilizers upregulates BDNF and bcl-2 expression levels in rat frontal cortex. Neurochemical Research. 2009;34:536–541. doi: 10.1007/s11064-008-9817-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cherlyn SY, Woon PS, Liu JJ, Ong WY, et al. Genetic association studies of glutamate, GABA and related genes in schizophrenia and bipolar disorder: a decade of advance. Neuroscience Biobehavoral Reviews. 2010;34:958–977. doi: 10.1016/j.neubiorev.2010.01.002. [DOI] [PubMed] [Google Scholar]
  19. Ciabarra AM, Sullivan JM, Gahn LG, Pecht G, et al. Cloning and characterization of chi-1: a developmentally regulated member of a novel class of the ionotropic glutamate receptor family. Journal of Neuroscience. 1995;15:6498–6508. doi: 10.1523/JNEUROSCI.15-10-06498.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Clark JD, Lin LL, Kriz RW, Ramesha CS, et al. A novel arachidonic acid-selective cytosolic PLA2 contains a Ca(2+)-dependent translocation domain with homology to PKC and GAP. Cell. 1991;65:1043–1051. doi: 10.1016/0092-8674(91)90556-e. [DOI] [PubMed] [Google Scholar]
  21. Clinton SM, Meador-Woodruff JH. Abnormalities of the NMDA Receptor and Associated Intracellular Molecules in the Thalamus in Schizophrenia and Bipolar Disorder. Neuropsychopharmacology. 2004;29:1353–1362. doi: 10.1038/sj.npp.1300451. [DOI] [PubMed] [Google Scholar]
  22. Cunningham MO, Jones RS. The anticonvulsant, lamotrigine decreases spontaneous glutamate release but increases spontaneous GABA release in the rat entorhinal cortex in vitro. Neuropharmacology. 2000;39:2139–2146. doi: 10.1016/s0028-3908(00)00051-4. [DOI] [PubMed] [Google Scholar]
  23. 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 Letters. 2004;25:178–183. [PubMed] [Google Scholar]
  24. Das S, Sasaki YF, Rothe T, Premkumar LS, et al. Increased NMDA current and spine density in mice lacking the NMDA receptor subunit NR3A. Nature. 1998;393:377–381. doi: 10.1038/30748. [DOI] [PubMed] [Google Scholar]
  25. DeGeorge JJ, Noronha JG, Bell JM, Robinson P, et al. Intravenous injection of [1-14C]arachidonate to examine regional brain lipid metabolism in unanesthetized rats. Journal of Neuroscience Research. 1989;24:413–423. doi: 10.1002/jnr.490240311. [DOI] [PubMed] [Google Scholar]
  26. Demar JC, Jr, Ma K, Chang L, Bell JM, et al. alpha-Linolenic acid does not contribute appreciably to docosahexaenoic acid within brain phospholipids of adult rats fed a diet enriched in docosahexaenoic acid. Journal of Neurochemistry. 2005;94:1063–1076. doi: 10.1111/j.1471-4159.2005.03258.x. [DOI] [PubMed] [Google Scholar]
  27. Doose DR, Brodie MJ, Wilson EA, Chadwick D, et al. Topiramate and lamotrigine pharmacokinetics during repetitive monotherapy and combination therapy in epilepsy patients. Epilepsia. 2003;44:917–922. doi: 10.1046/j.1528-1157.2003.64402.x. [DOI] [PubMed] [Google Scholar]
  28. Dumuis A, Sebben M, Haynes L, Pin JP, et al. NMDA receptors activate the arachidonic acid cascade system in striatal neurons. Nature. 1988;336:68–70. doi: 10.1038/336068a0. [DOI] [PubMed] [Google Scholar]
  29. Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, et al. Altered gene expression of brain-derived neurotrophic factor and receptor tyrosine kinase B in postmortem brain of suicide subjects. Archives of General Psychiatry. 2003;60:804–815. doi: 10.1001/archpsyc.60.8.804. [DOI] [PubMed] [Google Scholar]
  30. Eastwood SL, Harrison PJ. Markers of glutamate synaptic transmission and plasticity are increased in the anterior cingulate cortex in bipolar disorder. Biological Psychiatry. 2010;67:1010–1016. doi: 10.1016/j.biopsych.2009.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Farber NB, Jiang XP, Heinkel C, Nemmers B. Antiepileptic drugs and agents that inhibit voltage-gated sodium channels prevent NMDA antagonist neurotoxicity. Molecular Psychiatry. 2002;7:726–733. doi: 10.1038/sj.mp.4001087. [DOI] [PubMed] [Google Scholar]
  32. FDA. FDA approved drug products. 2009 http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm.
  33. Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. Journal of Biological Chemistry. 1957;226:497–509. [PubMed] [Google Scholar]
  34. Frye MA, Watzl J, Banakar S, O'Neill J, et al. Increased anterior cingulate/medial prefrontal cortical glutamate and creatine in bipolar depression. Neuropsychopharmacology. 2007;32:2490–2499. doi: 10.1038/sj.npp.1301387. [DOI] [PubMed] [Google Scholar]
  35. Gao F, Kiesewetter D, Chang L, Ma K, et al. Whole-body synthesis-secretion rates of long-chain n-3 PUFAs from circulating unesterified {alpha}-linolenic acid in unanesthetized rats. Journal of Lipid Research. 2009;50:749–758. doi: 10.1194/jlr.D800056-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  36. Gascon S, Deogracias R, Sobrado M, Roda JM, et al. Transcription of the NR1 subunit of the N-methyl-D-aspartate receptor is down-regulated by excitotoxic stimulation and cerebral ischemia. Journal of Biological Chemistry. 2005;280:35018–35027. doi: 10.1074/jbc.M504108200. [DOI] [PubMed] [Google Scholar]
  37. Ghelardoni S, Bazinet RP, Rapoport SI, Bosetti F. Topiramate does not alter expression in rat brain of enzymes of arachidonic acid metabolism. Psychopharmacology (Berl) 2005;180:523–529. doi: 10.1007/s00213-005-2189-3. [DOI] [PubMed] [Google Scholar]
  38. Ghelardoni S, Tomita YA, Bell JM, Rapoport SI, et al. Chronic carbamazepine selectively downregulates cytosolic phospholipase A(2) expression and cyclooxygenase activity in rat brain. Biological Psychiatry. 2004;56:248–254. doi: 10.1016/j.biopsych.2004.05.012. [DOI] [PubMed] [Google Scholar]
  39. Gibbons GF, Wiggins D, Brown AM, Hebbachi AM. Synthesis and function of hepatic very-low-density lipoprotein. Biochemcal Society Transactions. 2004;32:59–64. doi: 10.1042/bst0320059. [DOI] [PubMed] [Google Scholar]
  40. Hahn CG, Gyulai L, Baldassano CF, Lenox RH. The current understanding of lamotrigine as a mood stabilizer. Journal of Clinical Psychiatry. 2004;65:791–804. doi: 10.4088/jcp.v65n0610. [DOI] [PubMed] [Google Scholar]
  41. Hashimoto K, Sawa A, Iyo M. Increased levels of glutamate in brains from patients with mood disorders. Biological Psychiatry. 2007;62:1310–1316. doi: 10.1016/j.biopsych.2007.03.017. [DOI] [PubMed] [Google Scholar]
  42. Hassel B, Tauboll E, Gjerstad L. Chronic lamotrigine treatment increases rat hippocampal GABA shunt activity and elevates cerebral taurine levels. Epilepsy Research. 2001;43:153–163. doi: 10.1016/s0920-1211(00)00196-0. [DOI] [PubMed] [Google Scholar]
  43. Hayes JD, Strange RC. Glutathione S-transferase polymorphisms and their biological consequences. Pharmacology. 2000;61:154–166. doi: 10.1159/000028396. [DOI] [PubMed] [Google Scholar]
  44. Kaltschmidt B, Linker RA, Deng J, Kaltschmidt C. Cyclooxygenase-2 is a neuronal target gene of NF-κB. BMC Molcular Biology. 2002;3:16. doi: 10.1186/1471-2199-3-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ketter TA, Manji HK, Post RM. Potential mechanisms of action of lamotrigine in the treatment of bipolar disorders. J Clinical Psychopharmacoly. 2003;23:484–495. doi: 10.1097/01.jcp.0000088915.02635.e8. [DOI] [PubMed] [Google Scholar]
  46. Kim HW, Rapoport SI, Rao JS. Altered expression of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiological Disorders. 2010;37:596–603. doi: 10.1016/j.nbd.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kim HW, Rapoport SI, Rao JS. Altered arachidonic acid cascade enzymes in postmortem brain from bipolar disorder patients. Molecular Psychiatry. 2011;16:419–428. doi: 10.1038/mp.2009.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Knable MB, Barci BM, Webster MJ, Meador-Woodruff J, et al. Molecular abnormalities of the hippocampus in severe psychiatric illness: postmortem findings from the Stanley Neuropathology Consortium. Molecular Psychiatry. 2004;9:609–620. doi: 10.1038/sj.mp.4001471. 544. [DOI] [PubMed] [Google Scholar]
  49. Koukopoulos A, Reginaldi D, Serra G, Sani G. Antimanic and mood-stabilizing effect of memantine as an augmenting agent in treatment-resistant bipolar disorder. Bipolar Disorder. 2010;12:348–349. doi: 10.1111/j.1399-5618.2010.00803.x. [DOI] [PubMed] [Google Scholar]
  50. 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 Disorder. 2006;8:15–27. doi: 10.1111/j.1399-5618.2006.00276.x. [DOI] [PubMed] [Google Scholar]
  51. Lahiri DK. An region upstream of the gene promoter for the beta-amyloid precursor protein interacts with proteins from nuclear extracts of the human brain and PC12 cells. Brain Research Moleculat Brain Research. 1998;58:112–122. doi: 10.1016/s0169-328x(98)00115-6. [DOI] [PubMed] [Google Scholar]
  52. Lee HJ, Ertley RN, Rapoport SI, Bazinet RP, et al. Chronic administration of lamotrigine downregulates COX-2 mRNA and protein in rat frontal cortex. Neurochemical Research. 2008;33:861–866. doi: 10.1007/s11064-007-9526-3. [DOI] [PubMed] [Google Scholar]
  53. Lee HJ, Ghelardoni S, Chang L, Bosetti F, et al. Topiramate does not alter the kinetics of arachidonic or docosahexaenoic acid in brain phospholipids of the unanesthetized rat. Neurochemical Research. 2005;30:677–683. doi: 10.1007/s11064-005-2756-3. [DOI] [PubMed] [Google Scholar]
  54. Lee HJ, Rao JS, Chang L, Rapoport SI, et al. Chronic lamotrigine does not alter the turnover of arachidonic acid within brain phospholipids of the unanesthetized rat: implications for the treatment of bipolar disorder. Psychopharmacology (Berl) 2007;193:467–474. doi: 10.1007/s00213-007-0803-2. [DOI] [PubMed] [Google Scholar]
  55. Lees G, Leach MJ. Studies on the mechanism of action of the novel anticonvulsant lamotrigine (Lamictal) using primary neurological cultures from rat cortex. Brain Research. 1993;612:190–199. doi: 10.1016/0006-8993(93)91660-k. [DOI] [PubMed] [Google Scholar]
  56. Leonard AS, Hell JW. Cyclic AMP-dependent protein kinase and protein kinase C phosphorylate N-methyl-D-aspartate receptors at different sites. Journal of Biological Chemistry. 1997;272:12107–12115. doi: 10.1074/jbc.272.18.12107. [DOI] [PubMed] [Google Scholar]
  57. Li N, He X, Qi X, Zhang Y, et al. The mood stabilizer lamotrigine produces antidepressant behavioral effects in rats: role of brain-derived neurotrophic factor. Journal of Psychopharmacol. 2010;24:1772–1778. doi: 10.1177/0269881109359102. [DOI] [PubMed] [Google Scholar]
  58. Li X, Bijur GN, Jope RS. Glycogen synthase kinase-3beta, mood stabilizers, and neuroprotection. Bipolar Disorder. 2002;4:137–144. doi: 10.1034/j.1399-5618.2002.40201.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Maj R, Fariello RG, Ukmar G, Varasi M, et al. PNU-151774E protects against kainate-induced status epilepticus and hippocampal lesions in the rat. European Journal of Pharmacology. 1998;359:27–32. doi: 10.1016/s0014-2999(98)00554-8. [DOI] [PubMed] [Google Scholar]
  60. McCullumsmith RE, Kristiansen LV, Beneyto M, Scarr E, et al. Decreased NR1, NR2A, and SAP102 transcript expression in the hippocampus in bipolar disorder. Brain Research. 2007;1127:108–118. doi: 10.1016/j.brainres.2006.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Michael N, Erfurth A, Ohrmann P, Gossling M, et al. Acute mania is accompanied by elevated glutamate/glutamine levels within the left dorsolateral prefrontal cortex. Psychopharmacology (Berl) 2003;168:344–346. doi: 10.1007/s00213-003-1440-z. [DOI] [PubMed] [Google Scholar]
  62. Mueller HT, Meador-Woodruff JH. NR3A NMDA receptor subunit mRNA expression in schizophrenia, depression and bipolar disorder. Schizophrenia Research. 2004;71:361–370. doi: 10.1016/j.schres.2004.02.016. [DOI] [PubMed] [Google Scholar]
  63. Okada S, Murakami Y, Nishihara M, Yokotani K, et al. Perfusion of the hypothalamic paraventricular nucleus with N-methyl-D-aspartate produces thromboxane A2 and centrally activates adrenomedullary outflow in rats. Neuroscience. 2000;96:585–590. doi: 10.1016/s0306-4522(99)00598-9. [DOI] [PubMed] [Google Scholar]
  64. Paxinos G, Watson C. The rat brain in stereotaxic coordinates. New York: Academic Press; 1987. [DOI] [PubMed] [Google Scholar]
  65. Pepicelli O, Fedele E, Bonanno G, Raiteri M, et al. In vivo activation of N-methyl-D-aspartate receptors in the rat hippocampus increases prostaglandin E(2) extracellular levels and triggers lipid peroxidation through cyclooxygenase-mediated mechanisms. Journal of Neurochemistry. 2002;81:1028–1034. doi: 10.1046/j.1471-4159.2002.00897.x. [DOI] [PubMed] [Google Scholar]
  66. Purdon D, Arai T, Rapoport S. No evidence for direct incorporation of esterified palmitic acid from plasma into brain lipids of awake adult rat. Journal of Lipid Research. 1997;38:526–530. [PubMed] [Google Scholar]
  67. Ramadan E, Rosa AO, Chang L, Chen M, et al. Extracellular-derived calcium does not initiate in vivo neurotransmission involving docosahexaenoic acid. Journal of Lipid Research. 2010;51:2334–2340. doi: 10.1194/jlr.M006262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Rao JS, Ertley RN, Rapoport SI, Bazinet RP, et al. Chronic NMDA administration to rats up-regulates frontal cortex cytosolic phospholipase A2 and its transcription factor, activator protein-2. Journal of Neurochemistry. 2007a;102:1918–1927. doi: 10.1111/j.1471-4159.2007.04648.x. [DOI] [PubMed] [Google Scholar]
  69. 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 Disorders. 2007b;9:513–520. doi: 10.1111/j.1399-5618.2007.00361.x. [DOI] [PubMed] [Google Scholar]
  70. 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. Biological Psychiatry. 2007c;61:154–161. doi: 10.1016/j.biopsych.2006.03.029. [DOI] [PubMed] [Google Scholar]
  71. Rao JS, Harry GJ, Rapoport SI, Kim HW. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Molecular Psychiatry. 2010;15:384–392. doi: 10.1038/mp.2009.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. 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]
  73. Rapoport SI. In vivo fatty acid incorporation into brain phospholipids in relation to plasma availability, signal transduction and membrane remodeling. Journal of Molecular Neuroscience. 2001;16:243–261. doi: 10.1385/JMN:16:2-3:243. [DOI] [PubMed] [Google Scholar]
  74. Rapoport SI. In vivo approaches to quantifying and imaging brain arachidonic and docosahexaenoic acid metabolism. Journal of Pediatrics. 2003;143:S26–S34. doi: 10.1067/s0022-3476(03)00399-8. [DOI] [PubMed] [Google Scholar]
  75. Rapoport SI, Basselin M, Kim HW, Rao JS. Bipolar disorder and mechanisms of action of mood stabilizers. Brain Research Reviews. 2009;61:185–209. doi: 10.1016/j.brainresrev.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Rapoport SI, Bosetti F. Do lithium and anticonvulsants target the brain arachidonic acid cascade in bipolar disorder? Archives of General Psychiatry. 2002;59:592–506. doi: 10.1001/archpsyc.59.7.592. [DOI] [PubMed] [Google Scholar]
  77. Rataud J, Debarnot F, Mary V, Pratt J, et al. Comparative study of voltage-sensitive sodium channel blockers in focal ischaemia and electric convulsions in rodents. Neuroscience Letters. 1994;172:19–23. doi: 10.1016/0304-3940(94)90652-1. [DOI] [PubMed] [Google Scholar]
  78. Rintala J, Seemann R, Chandrasekaran K, Rosenberger TA, et al. 85 kDa cytosolic phospholipase A2 is a target for chronic lithium in rat brain. NeuroReport. 1999;10:3887–3890. doi: 10.1097/00001756-199912160-00030. [DOI] [PubMed] [Google Scholar]
  79. Robinson PJ, Noronha J, DeGeorge JJ, Freed LM, et al. A quantitative method for measuring regional in vivo fatty-acid incorporation into and turnover within brain phospholipids: Review and critical analysis. Brain Research Brain Research Reviews. 1992;17:187–214. doi: 10.1016/0165-0173(92)90016-f. [DOI] [PubMed] [Google Scholar]
  80. Sitges M, Chiu LM, Guarneros A, Nekrassov V. Effects of carbamazepine, phenytoin, lamotrigine, oxcarbazepine, topiramate and vinpocetine on Na+ channel-mediated release of [3H]glutamate in hippocampal nerve endings. Neuropharmacology. 2007a;52:598–605. doi: 10.1016/j.neuropharm.2006.09.002. [DOI] [PubMed] [Google Scholar]
  81. Sitges M, Guarneros A, Nekrassov V. Effects of carbamazepine, phenytoin, valproic acid, oxcarbazepine, lamotrigine, topiramate and vinpocetine on the presynaptic Ca2+ channel-mediated release of [3H]glutamate: comparison with the Na+ channel-mediated release. Neuropharmacology. 2007b;53:854–862. doi: 10.1016/j.neuropharm.2007.08.016. [DOI] [PubMed] [Google Scholar]
  82. Strange RC, Spiteri MA, Ramachandran S, Fryer AA. Glutathione-S-transferase family of enzymes. Mutation Research. 2001;482:21–26. doi: 10.1016/s0027-5107(01)00206-8. [DOI] [PubMed] [Google Scholar]
  83. Sucher NJ, Akbarian S, Chi CL, Leclerc CL, et al. Developmental and regional expression pattern of a novel NMDA receptor-like subunit (NMDAR-L) in the rodent brain. Journal of Neuroscience. 1995;15:6509–6520. doi: 10.1523/JNEUROSCI.15-10-06509.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Tabachnick BG, Fidell LS. Computer-assisted research design and analysis. 2001 [Google Scholar]
  85. Teng CT, Demetrio FN. Memantine may acutely improve cognition and have a mood stabilizing effect in treatment-resistant bipolar disorder. Revista Brasileira Psiquiatria. 2006;28:252–254. doi: 10.1590/s1516-44462006000300020. [DOI] [PubMed] [Google Scholar]
  86. Wang SJ, Huang CC, Hsu KS, Tsai JJ, et al. Presynaptic inhibition of excitatory neurotransmission by lamotrigine in the rat amygdalar neurons. Synapse. 1996;24:248–255. doi: 10.1002/(SICI)1098-2396(199611)24:3<248::AID-SYN7>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  87. Xie X, Hagan RM. Cellular and molecular actions of lamotrigine: Possible mechanisms of efficacy in bipolar disorder. Neuropsychobiology. 1998;38:119–130. doi: 10.1159/000026527. [DOI] [PubMed] [Google Scholar]
  88. Yang HC, Mosior M, Johnson CA, Chen Y, et al. Group-specific assays that distinguish between the four major types of mammalian phospholipase A2. Analytical Biochemistry. 1999;269:278–288. doi: 10.1006/abio.1999.4053. [DOI] [PubMed] [Google Scholar]
  89. Zarate CA, Jr, Du J, Quiroz J, Gray NA, et al. Regulation of cellular plasticity cascades in the pathophysiology and treatment of mood disorders: role of the glutamatergic system. Annals of the New York Academy of Sciences. 2003;1003:273–291. doi: 10.1196/annals.1300.017. [DOI] [PubMed] [Google Scholar]
  90. Zarate CA, Jr, Quiroz JA, Singh JB, Denicoff KD, et al. An open-label trial of the glutamate-modulating agent riluzole in combination with lithium for the treatment of bipolar depression. Biological Psychiatry. 2005;57:430–432. doi: 10.1016/j.biopsych.2004.11.023. [DOI] [PubMed] [Google Scholar]

RESOURCES