Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Oct 15.
Published in final edited form as: Biol Psychiatry. 2007 Jul 12;62(8):934–943. doi: 10.1016/j.biopsych.2007.04.021

Chronic carbamazepine administration reduces NMDA receptor-initiated signaling via arachidonic acid in rat brain

Mireille Basselin 1,*, Nelly E Villacreses 1, Mei Chen 1, Jane M Bell 1, Stanley I Rapoport 1
PMCID: PMC2131715  NIHMSID: NIHMS32542  PMID: 17628508

Abstract

Background

Lithium and carbamazepine (CBZ) are used to treat mania in bipolar disorder. When given chronically to rats, both agents reduce brain arachidonic acid (AA) turnover in brain phospholipids and downstream AA metabolism. Lithium administration to rats also attenuates N-methyl-D-aspartic acid receptor (NMDAR) signaling via AA.

Hypothesis

Chronic CBZ administration to rats, like chronic lithium, will reduce NMDAR-mediated signaling via AA.

Methods

We used our fatty acid method with quantitative autoradiography to image the regional brain incorporation coefficient k* of AA, a marker of AA signaling, in unanesthetized rats that had been given 25 mg/kg/day i.p. CBZ or vehicle for 30 days, then injected with NMDA (25 mg/kg i.p.) or saline. We also measured brain concentration of two AA metabolites, prostaglandin E2 (PGE2) and thromboxane B2 (TXB2).

Results

In chronic vehicle-treated rats, NMDA compared with saline increased k* significantly in 69 of 82 brain regions examined, but did not change k* significantly in any region in the CBZ-treated rats. In vehicle- but not CBZ-treated rats, NMDA also increased brain concentration of PGE2 and TXB2.

Conclusions

Chronic CBZ administration to rats blocks the brain NMDAR-mediated AA signal k* and the increments in PGE2 and TXB2 that are seen in vehicle-treated rats. The clinical action of antimanic drugs may involve inhibition of brain NMDAR-mediated signaling involving AA and its metabolites.

Keywords: carbamazepine, NMDA, arachidonic acid, bipolar disorder, phospholipase A2, prostaglandin E2

Introduction

Glutamatergic neurotransmission is thought to play a role in the pathophysiology and treatment of bipolar disorder (BD) (1,2). An elevated brain glutamate/glutamine ratio has been reported using magnetic resonance spectroscopy in children and adults with BD (3,4), and gene variants of the NR1 and NR2 subunits of the N-methyl-D-aspartate receptor (NMDAR) have been linked to the disease (57). Furthermore, NMDAR density and levels of NR1, NR2A and NR3A mRNA were decreased in post-mortem BD brain, as were densities of the NMDAR-associated post-synaptic proteins, PSD-95 and SAP102 (8,9). It has been shown that NMDAR stimulation by glutamate or NMDA decreases NR-1 expression (10), and that the NR3A subunit co-assembles with other subunits (NR1, NR2A or NR2B) to form NMDAR with decreased activity and calcium influx (11, 12). Decreased NR1 and NR3A expression observed in BD is consistent with upregulated NMDAR function, and with the observation that mice lacking the NR-3A subunit have increased brain NMDAR activity (13).

Binding of glutamate or NMDA to NMDAR will allow extracellular Ca2+ into the cell, thereby activating a number of Ca2+-dependent enzymes. One of these, cytosolic phospholipase A2 (cPLA2), selectively releases arachidonic acid (AA, 20:4n-6) from synaptic membrane phospholipid, to initiate the AA signaling cascade (1418). A fraction of the released AA may be converted by cyclooxygenase (COX), lipoxygenase and cytochrome P-450 enzymes to eicosanoids, or lost via other metabolic pathways, whereas the remainder will be reincorporated into brain phospholipid (1921). Markers of the AA cascade have been reported to be abnormal in BD (2224).

Three chemically disparate agents, carbamazepine (CBZ), lithium and valproic acid, have proven effective against acute mania and mixed episodes in BD (2528). When administered chronically to rats to produce clinically relevant plasma concentrations, each was reported to downregulate the brain AA cascade -- AA turnover in brain phospholipids, cPLA2 or acyl-CoA synthetase activity as well as COX activity, and the concentration of prostaglandin E2 (PGE2), an AA metabolite produced preferentially via COX-2 (18,2937).

In addition, chronic administration of lithium to rats and pretreatment with the NMDAR antagonist MK-801 reduced the brain AA signal following acute NMDA administration (38). This signal was measured using quantitative autoradiography as a regional AA incorporation coefficient k* (brain radioactivity/integrated plasma radioactivity) following the intravenous injection of radiolabeled AA. It represents AA lost by metabolism following its release from synaptic membrane phospholipid by cPLA2 activation, due to Ca2+ entry into the cell at NMDARs (17,20,38). Suppression by chronic lithium of the NMDA-initiated AA signal suggested that the lithium acts in BD by reducing this signal, consistent with evidence for upregulated glutamatergic transmission in the disease (1,2).

In the present study we hypothesized that effective mood stabilizers might generally interfere with NMDA-initiated signaling via AA, and tested this hypothesis by measuring effects of chronic administration of CBZ to rats on the AA signal, and on the concentrations of AA metabolites, PGE2 and thromboxane B2 (TXB2). In this regard, CBZ has been reported to block NMDA-induced currents in cultured spinal cord neurons (39), to inhibit NMDA-induced depolarization in cortical wedges (40), to prevent NMDA-initiated convulsions in mice (41), inhibit NMDA-evoked Ca2+ influx in cultured neurons (42,43), and protect against NMDA-mediated neurotoxicity (44,45).

In the present study, we measured regional k* responses and brain concentrations of PGE2 and TXB2 after giving 25 mg/kg i.p. NMDA or saline to unanesthetized rats that were injected daily for 30 days with 25 mg/k.g. i.p. CBZ or vehicle. This NMDA dose does not produce convulsions, but causes widespread increments in k* that can be prevented by chronic LiCl feeding or pretreatment with MK-801 (38). The CBZ regimen, which we have used before, produces a mean plasma concentration of 54 μM, in the high end of concentrations reported in CBZ-treated BD patients (51 μM) (31,34,46,47).

Methods and Materials

Animals and Diets

The National Institutes of Health (NIH) Animal Care and Use Committee approved the study in accordance with NIH guidelines on the care and use of laboratory animals. Two-month-old male Fischer CDF (F-344)/CrlBR rats (Charles River Laboratories, Wilmington, MA, USA) were acclimatized for 1 week in an animal facility with regulated temperature, humidity and light cycle, and had ad libitum access to food (NIH-31 diet, Zeigler, Gardners, PA, USA) and water. The diet contained (as percent of total fatty acids) 20.1% saturated, 22.5% monounsaturated, 47.9% linoleic, 5.1% linolenic, 0.02% AA, 2.0% eicosapentaenoic, and 2.3% docosahexaenoic acid.

Drugs

[1-14C]AA in ethanol (53 mCi/mmol, >98% pure, Moravek Biochemicals, Brea, CA, USA) 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, USA). CBZ-treated rats received 25 mg/kg intraperitoneally once daily for 30 days (Sigma-Aldrich). The CBZ was dissolved in a 50:50 (v/v) dimethyl sulfoxide (DMSO, ≥99.9% Sigma-Aldrich): saline (0.9% NaCl) mixture and kept at 37°C (31,34). A control group received the same volume of DMSO: saline (vehicle) under parallel conditions.

Surgical Procedures and Tracer Infusion

On the morning following the 30th CBZ or vehicle injection, a rat was anesthetized with 2–3% halothane in O2, and PE 50 polyethylene catheters were inserted into the right femoral artery and vein as described previously (38). The wound was closed with surgical clips and the hindquarters of the rat were wrapped loosely, with its upper body remaining free, in a fast-setting plaster cast taped to a wooden block. The rat was allowed to recover from anesthesia for 3–4 hours in an environment maintained at 25°C. Body temperature was kept 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, Inc., Nicholasville, KY, USA). Arterial blood pH, pO2 and pCO2 were measured with a blood gas analyzer (Model 248, Bayer Health Care, Norwood, MA, USA).

Ten min after injecting NMDA or saline (about 4 hours after the last of the chronic CBZ or saline dose), 2 ml [1-14C]AA (170 μCi/kg) 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, USA). Twenty min after beginning infusion, the rat was euthanized with an overdose of Nembutal® (100 mg/kg, intravenously) and decapitated. The brain was rapidly removed, frozen in 2-methylbutane maintained at −40°C with dry ice, and stored at −80°C until sectioned.

Chemical Analysis

Thirteen arterial blood samples collected before, during and after [1-14C]AA infusion were centrifuged immediately (30 s at 18,000 g). Total lipids were extracted from 30 μl of plasma with 3 ml chloroform:methanol (2:1, by vol) and 1.5 ml 0.1 M KCl (48). As reported, following [14C]AA infusion, greater than 97% of plasma radioactivity was radiolabeled AA at 5 min (49). Concentrations of unesterified fatty acids were determined in 100–150 μl of frozen arterial plasma. Total lipids were extracted (48) and separated by thin layer chromatography on silica gel 60 plates (Whatman, Clifton, NJ, USA) using the solvent system heptane:diethylether:glacial acetic acid (60:40:3, by vol). Unesterified fatty acids were scraped from the plate and methylated with 1% H2SO4 in anhydrous methanol for 3 hours at 70°C (50). Fatty acid methyl esters were separated and quantified by gas chromatography using heptadecanoic acid (17:0) as an internal standard (31).

Quantitative Autoradiography

Frozen brains were cut in serial 20-μm thick coronal sections in a cryostat at −20°C. The sections were placed for 5 weeks with calibrated [14C]methylmethacrylate standards on Kodak Ektascan C/RA film (Eastman Kodak Company, Rochester, NY, USA). Eighty-two brain regions from autoradiographs were identified from a stereotaxic brain atlas (51), 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 of brain) by digital quantitative densitometry, using a Macintosh computer and the public domain NIH Image program 1.62 (developed at the U.S. National Institutes of Health and available on the Internet at http://rsb.info.nih.gov/nih-image/). Regional incorporation coefficients k* (ml plasma/sec/g brain) of AA were calculated as (21),

k=cbrain(20min)020cplasmadt (Eq. 1)

c*plasma equals plasma radioactivity determined by scintillation counting (nCi/ml), c*brain(20 min) equals brain radioactivity (nCi/g of brain) at 20 min after staring infusion, and t equals time (min) after beginning [1-14C]AA infusion. Rates of incorporation of unesterified AA from plasma into brain, Jin (nmol/sec/g), were calculated as,

Jin=kcplasma (Eq. 2)

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

Brain prostaglandin E2 and thromboxane B2 concentrations

In separate experiments, vehicle- and CBZ-treated rats were given i.p. NMDA (25 mg/kg) or saline. Ten min later, they were anesthetized with Nembutal® (100 mg/kg, i.p.) and subjected to head-focused microwave irradiation (5.5 kW, 3.4 s then 1.4 s; Cober Electronics, Stamford, CT, USA) to stop post-mortem changes, such as release of fatty acids and formation of prostaglandins (52). Extraction was performed by Radin’s method (53). Briefly, half-brains were weighed, then extracted in 18 volumes of hexane:isopropanol (3:2, by vol) using a glass Tenbroeck homogenizer. The extract was concentrated to dryness under nitrogen and resuspended in an enzyme immunoassay buffer provided with a polyclonal PGE2 or TXB2 assay kit (Oxford Biochemical Research, Oxford, MI, USA).

Statistical Analyses

An unpaired two-tailed t-test was used to compare mean physiological parameters in CBZ- and vehicle-treated rats, using GraphPad Prism version 4.0b (GraphPad Software, San Diego, CA, www.graphpad.com). A standard two-way ANOVA, comparing CBZ administration (CBZ vs. vehicle) with drug (NMDA vs. saline) was performed with regard to arterial plasma radioactivity, plasma unesterified fatty acid concentrations, brain PGE2 and TXB2 concentrations, and regional values of k* using SPSS 11.0 (SPSS Inc., Chicago, IL, USA, http://www.spss.com). Where interactions between CBZ and NMDA were statistically significant, probabilities of main effects of CBZ and NMDA were not reported as they cannot be interpreted (54,55). Instead, unpaired two-tailed t-tests were used to compare NMDA and saline responses between vehicle and CBZ-treated rats, as well as saline responses in CBZ- compared with vehicle-treated rats. Other comparisons were not considered relevant. A post-hoc test was not used to avoid a correction for multiple comparisons. However, when a Bonferroni’s post-hoc test with correction for three comparisons was performed, statistical significances of differences were not changed. Data are reported as means ± SD, with statistical significance taken as p ≤ 0.05.

Results

Physiology, behavior and arterial plasma radioactivity

After 30 days of treatment, body weight in the chronic CBZ- and vehicle-treated rats did not differ significantly [276.4 ± 14.6 (n = 15) vs. 276.1 ± 16.1 (n = 16)]. Compared with acute saline, acute NMDA did not significantly affect arterial pH, pCO2, pO2 or blood pressure, but significantly decreased heart rate by 20–29% in both groups (Table 1), as reported (38). NMDA produced repeated cycles of head weaving and body movements following by a resting period. Cycling continued and then ceased after 90–110 sec (Table 1). The mean cycling period did not differ between CBZ- and vehicle-treated rats.

Table 1.

Effects of chronic carbamazepine and acute NMDA on physiological parameters

Vehicle
Carbamazepine (CBZ)
Saline NMDA Saline NMDA
Before After Before After Before After Before After
Body weight (g) 277 ± 14 - 275 ± 19 - 275 ± 13 - 278 ± 18 -
Body temperature (°C) 36.5 ± 0.3 36.7 ± 0.2 36.4 ± 0.3 36.6 ± 0.3 36.6 ± 0.3 36.8 ± 0.4 36.3 ± 0.5 36.6 ± 0.2
Heart rate (beats/min) 461 ± 40 451 ± 33 461 ± 24 326 ± 35*** 442 ± 38 436 ± 42 446 ± 32 356 ± 21***
Arterial blood pressure (mm Hg)
 Systolic 158 ± 15 148 ± 13 149 ± 11 139 ± 7 153 ± 7 152 ± 5 159 ± 6 159 ± 6
 Diastolic 104 ± 9 107 ± 7 104 ± 9 103 ± 6 94 ± 6 96 ± 6 94 ± 4 97 ± 4
pH 7.460 ± 0.010 7.463 ± 0.021 7.440 ± 0.039 7.453 ± 0.021 7.475 ± 0.019 7.461 ± 0.027 7.450 ± 0.020 7.443 ± 0.027
pO2 (mm Hg) 83.6 ± 6.2 85.5 ± 3.0 81.7 ± 7.3 85.3 ± 1.1 87.6 ± 4.1 88.6 ± 5.6 91.3 ± 5.8 92.0 ± 3.2
pCO2 (mm Hg) 33.2 ± 2.8 36.1 ± 3.2 36.7 ± 2.7 36.0 ± 1.0 36.2 ± 3.2 40.0 ± 4.9 37.8 ± 2.7 40.0 ± 1.3
Behavior duration (s)
 Cycle
  Activity 5 ± 1 4 ± 1
  Calm 10 ± 2 9 ± 1
  Net cycling 90 ± 20 110 ± 10

Values are means ± SD (n = 7–8) measured before surgery (body weight) or before [1-14C]AA infusion.

***

p < 0.001, significant effect of 25 mg/kg i.p. NMDA in chronic vehicle- or CBZ-treated rats; -, not measured

Neither CBZ nor NMDA modified the time course of arterial plasma radioactivity, the input function in Eq. 1, following [1-14C]AA infusion. Mean integrated radioactivity in the plasma organic fraction, in units of (nCi x sec)/ml (n = 7–8), did not differ significantly between groups: vehicle plus saline, 183981 ± 38623; vehicle plus NMDA, 190501 ± 38459; CBZ plus saline, 194895 ± 24409; CBZ plus NMDA, 202102 ± 17701.

Plasma unesterified fatty acid concentrations

A two-way ANOVA did not show a significant interaction between CBZ and NMDA in any measured plasma unesterified fatty acid concentration (Table 2). CBZ compared with vehicle had no significant main effect on any concentration, whereas NMDA compared with saline significantly increased concentrations of unlabeled unesterified palmitic, palmitoleic, stearic, oleic, linoleic, α-linolenic, AA and docosahexaenoic acid in both vehicle- and CBZ-treated rats (Table 2).

Table 2.

Effects of NMDA and carbamazepine on plasma unesterified fatty acid concentrations

Vehicle CBZ CBZ x NMDA interaction CBZ effect NMDA effect
Fatty Acid saline (n = 8) NMDA (n = 8) saline (n = 7) NMDA (n = 7) p value p value p value
Concentration, nmol/ml plasma
Palmitic (16:0) 183.5 ± 33.7 375.7 ± 234.5 175.5 ± 41.7 399.8 ± 167.5 0.769 0.884 0.001
Palmitoleic (16:1) 21.9 ± 5.0 39.9 ± 24.5 18.9 ± 5.1 37.6 ± 13.6 0.950 0.629 0.002
Stearic (18:0) 52.3 ± 6.9 60.7 ± 16.0 54.4 ± 11.4 67.3 ± 16.8 0.648 0.381 0.039
Oleic (18:1 n-9) 130.2 ± 34.7 313.7 ± 207.7 134.4 ± 42.2 394.6 ± 167.9 0.453 0.405 < 0.001
Linoleic (18:2 n-6) 134.7 ± 31.6 363.6 ± 262.7 134.0 ± 44.4 394.6 ± 168.0 0.789 0.798 < 0.001
α-Linolenic (18:3 n-3) 8.5 ± 2.6 19.8 ± 13.0 7.8 ± 3.1 20.5 ± 9.1 0.814 0.988 0.001
Arachidonic (20:4 n-6) 12.8 ± 3.6 27.1 ± 18.6 13.1 ± 4.7 32.3 ± 10.7 0.602 0.469 0.001
Docosahexaenoic (22:6 n-3) 12.8 ± 5.7 53.9 ± 44.0 14.6 ± 7.5 64.8 ± 30.0 0.657 0.532 < 0.001

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

Regional brain AA incorporation coefficients, k*

Effects of NMDA in vehicle-treated rats

Mean AA incorporation coefficients, k*, in each of 82 brain regions (Eq. 1), were subjected to a two-way ANOVA (Table 3, Fig. 1). Interactions between CBZ and NMDA were statistically significant in 68 of the 82 regions. In these 68 regions, t-tests showed that NMDA compared with saline significantly increased mean k* by 29–122% in the chronic vehicle-treated rats. Affected were prefrontal, frontal, anterior cingulate, motor, somatosensory, auditory and visual cortical areas (33–122%), preoptic area (44%), globus pallidus (60%), olfactory tubercle (57%), diagonal band (52–72%), amygdala (41%), hippocampus (29–87%), nucleus accumbens (83%), caudate-putamen (58–88%), septal nuclei (62–64%), habenular nuclei (67–68%), lateral geniculate nucleus dorsal (56%), medial geniculate nucleus (67%), regions of the thalamus (46–104%), areas in the hypothalamus (46–86%), interpeduncular nucleus (35%), substantia nigra (77%), superior and inferior colliculi (46–79%), and cerebellar gray matter (32%). Where CBZ x NMDA interactions were statistically insignificant, NMDA increased k* significantly in the pyriform cortex (33%). Thus, NMDA increased k* significantly in 69 (68 + 1) of the 82 brain regions.

Table 3.

Effect of chronic carbamazepine on NMDA-induced regional AA incorporation coefficients k* in rat brain

Vehicle Carbamazepine (CBZ) CBZ x NMDA Interaction CBZ effect NMDA effect
k*, ml/sec/gram brain × 10−4
Brain region saline (n = 8) NMDA (n = 8) saline (n = 7) NMDA (n = 7) p-value p-value p-value
Prefrontal cortex layer I 3.30 ± 0.33 5.46 ± 0.37*** 3.60 ± 0.38 3.55 ± 0.14 < 0.001
Prefrontal cortex layer IV 3.76 ± 0.49 6.08 ± 0.33*** 3.75 ± 0.34 3.93 ± 0.60 < 0.001
Primary olfactory cortex 3.28 ± 0.25 5.47 ± 0.59*** 3.33 ± 0.61 3.68 ± 0.33 < 0.001
Frontal cortex (10)
 Layer I 3.26 ± 0.28 5.82 ± 0.47*** 3.51 ± 0.56 3.64 ± 0.20 < 0.001
 Layer IV 3.66 ± 0.34 6.49 ± 0.17*** 3.98 ± 0.44 3.90 ± 0.38 < 0.001
Frontal cortex (8)
 Layer I 3.59 ± 0.34 6.28 ± 0.34*** 3.72 ± 0.52 3.97 ± 0.46 < 0.001
 Layer IV 4.13 ± 0.39 6.96 ± 0.50*** 4.27 ± 0.48 4.32 ± 0.45 < 0.001
Pyriform cortex 3.13 ± 0.30 4.15 ± 0.74 3.04 ± 0.80 3.39 ± 0.34 0.189 NS 0.002
Anterior cingulate cortex 5.33 ± 1.02 7.77 ± 0.41*** 4.60 ± 0.52 4.46 ± 0.73 < 0.001
Motor cortex
 Layer I 3.50 ± 0.18 6.24 ± 0.63*** 3.73 ± 0.38 3.62 ± 0.40 < 0.001
 Layer II –III 3.52 ± 0.18 6.53 ± 0.72*** 3.98 ± 0.24 3.42 ± 0.52 < 0.001
 Layer IV 3.97 ± 0.30 7.09 ± 0.93*** 4.39 ± 0.33 4.18 ± 0.53 < 0.001
 Layer V 3.00 ± 0.15 5.35 ± 0.45*** 3.22 ± 0.33 3.52 ± 0.68 < 0.001
 Layer VI 2.91 ± 0.19 5.13 ± 0.34*** 3.12 ± 0.43 3.10 ± 0.24 < 0.001
Somatosensory cortex
 Layer I 3.66 ± 0.44 6.55 ± 1.27*** 3.81 ± 0.33 3.95 ± 0.48 < 0.001
 Layer II – III 3.48 ± 0.21 6.45 ± 0.92*** 3.98 ± 0.25 3.90 ± 0.45 < 0.001
 Layer IV 4.34 ± 0.57 7.32 ± 0.93*** 4.41 ± 0.27 4.52 ± 0.68 < 0.001
 Layer V 3.55 ± 0.23 6.31 ± 0.98*** 3.98 ± 0.40 4.04 ± 0.53 < 0.001
 Layer VI 3.46 ± 0.15 6.14 ± 0.91*** 3.82 ± 0.31 4.11 ± 0.61 < 0.001
Auditory cortex
 Layer I 3.86 ± 0.19 7.86 ± 0.97*** 4.29 ± 0.77 4.27 ± 0.94 < 0.001
 Layer IV 3.72 ± 0.32 8.26 ± 0.88*** 4.46 ± 0.45 4.59 ± 1.10 < 0.001
 Layer VI 4.09 ± 0.15 6.86 ± 0.70*** 4.18 ± 0.56 4.01 ± 0.74 < 0.001
Visual cortex
 Layer I 3.92 ± 0.14 6.57 ± 0.49*** 4.06 ± 0.50 3.98 ± 0.82 < 0.001
 Layer IV 4.09 ± 0.16 6.99 ± 0.43*** 4.53 ± 0.40 4.58 ± 1.07 < 0.001
 Layer VI 3.82 ± 0.32 6.58 ± 0.68*** 4.03 ± 0.48 4.18 ± 0.78 < 0.001
Preoptic area (LPO/MPO) 3.28 ± 0.24 4.74 ± 0.79*** 3.07 ± 0.56 3.23 ± 0.15 0.002
Suprachiasmatic nu 3.06 ± 0.33 3.47 ± 0.25 2.60 ± 0.55 3.39 ± 0.33 NS NS NS
Globus pallidus 3.04 ± 0.42 4.86 ± 0.68*** 2.56 ± 0.62 3.11 ± 0.29 0.003
Bed nu stria terminalis 3.06 ± 0.20 5.17 ± 0.64*** 2.87 ± 0.57 3.20 ± 0.30 < 0.001
Olfactory tubercle 3.84 ± 0.14 6.02 ± 0.33*** 3.71 ± 0.46 3.75 ± 0.55 < 0.001
Diagonal band Dorsal 3.62 ± 0.11 5.49 ± 0.90*** 3.67 ± 0.76 3.68 ± 0.23 < 0.001
 Ventral 3.45 ± 0.14 5.94 ± 1.16*** 3.41 ± 0.70 3.66 ± 0.21 < 0.001
Amygdala basolateral/med ial 3.60 ± 0.29 5.07 ± 0.41 3.04 ± 0.99 3.65 ± 0.44 0.050
Hippocampus
 CA1 3.58 ± 0.38 4.65 ± 0.40*** 3.03 ± 0.41 2.94 ± 0.19 < 0.001
 CA2 3.59 ± 0.38 4.63 ± 0.43*** 3.27 ± 0.56 3.11 ± 0.26 0.001
 CA3 3.55 ± 0.35 4.96 ± 0.59*** 3.28 ± 0.56 3.29 ± 0.28 < 0.001
 Dentate gyrus 3.12 ± 0.24 5.84 ± 0.54*** 3.88 ± 0.95 3.58 ± 0.20 < 0.001
 SLM 3.48 ± 0.16 6.03 ± 1.34*** 4.71 ± 0.67 4.34 ± 0.88 < 0.001
Accumbens nucleus 2.99 ± 0.13 5.48 ± 1.23*** 3.45 ± 0.38 3.70 ± 0.43 < 0.001
Caudate putamen
 Dorsal 3.47 ± 0.10 5.49 ± 0.41*** 3.67 ± 0.51 3.75 ± 0.38 < 0.001
 Ventral 3.33 ± 0.19 5.68 ± 0.66*** 3.65 ± 0.46 3.85 ± 0.31 < 0.001
 Lateral 3.30 ± 0.17 5.68 ± 0.82*** 3.63 ± 0.50 3.84 ± 0.47 < 0.001
 Medial 3.35 ± 0.22 6.30 ± 0.34*** 3.57 ± 0.53 3.72 ± 0.31 < 0.001
Septal nu lateral 3.11 ± 0.29 5.05 ± 0.83*** 2.76 ± 0.67 3.27 ± 0.16 0.002
Septal nu medial 3.37 ± 0.17 5.51 ± 0.62*** 3.46 ± 0.68 3.85 ± 0.44 < 0.001
Habenular nu lateral 5.20 ± 0.21 8.75 ± 1.26*** 4.97 ± 0.48 5.43 ± 0.83 < 0.001
Habenular nu medial 5.11 ± 0.20 8.52 ± 1.00*** 5.31 ± 0.99 5.26 ± 0.88 < 0.001
Lateral geniculate nu dorsal 3.89 ± 0.19 6.07 ± 0.47*** 4.21 ± 0.48 4.65 ± 0.32 < 0.001
Medial geniculate nu 4.27 ± 0.16 7.15 ± 1.08*** 4.78 ± 0.54 5.02 ± 0.62 < 0.001
Thalamus
Ventroposterior lateral nu 3.75 ± 0.15 6.20 ± 1.44*** 4.09 ± 0.80 4.11 ± 0.52 0.001
Ventroposterior medial nu 3.76 ± 0.14 6.69 ± 0.89*** 4.03 ± 0.47 4.32 ± 0.34 < 0.001
Paratenial nu 3.52 ± 0.29 5.50 ± 0.52*** 3.89 ± 1.00 4.01 ± 0.23 < 0.001
Anteroventral nu 5.33 ± 0.70 7.87 ± 0.89*** 4.69 ± 0.37 5.02 ± 1.12 0.007
Anteromedial nu 3.50 ± 0.21 7.15 ± 1.21*** 4.22 ± 1.19 4.15 ± 0.50 < 0.001
Reticular nu 3.53 ± 0.37 6.13 ± 0.35*** 3.72 ± 0.56 4.41 ± 0.61 < 0.001
Paraventricular nu 3.41 ± 0.22 6.07 ± 0.50*** 3.55 ± 0.61 3.90 ± 0.50 < 0.001
Parafascicular nu 3.26 ± 0.16 6.31 ± 0.42*** 3.51 ± 0.45 3.25 ± 0.49 < 0.001
Subthalamic nu 5.14 ± 0.49 7.52 ± 1.61** 4.21 ± 0.65 4.32 ± 0.50 < 0.001
Hypothalamus
Supraoptic nu 3.14 ± 0.13 3.92 ± 0.67 3.46 ± 0.89 3.08 ± 0.60 NS NS NS
Lateral 2.68 ± 0.23 4.99 ± 0.93*** 2.64 ± 0.62 3.12 ± 0.39 < 0.001
Anterior 3.16 ± 0.30 4.98 ± 0.89*** 2.77 ± 0.69 3.36 ± 0.26 0.010
Periventricular 2.56 ± 0.27 4.42 ± 0.41*** 2.35 ± 0.43 2.90 ± 0.26 < 0.001
Arcuate 3.19 ± 0.29 4.66 ± 0.51*** 3.66 ± 0.56 3.93 ± 0.82 < 0.001
Ventromedial 3.09 ± 0.31 4.96 ± 0.73*** 2.94 ± 0.56 3.51 ± 0.38 0.002
Posterior 5.38 ± 0.21 6.20 ± 1.37 5.24 ± 0.54 5.65 ± 0.68 NS NS NS
Mammillary nu 3.04 ± 0.14 4.05 ± 1.82 3.90 ± 0.55 4.05 ± 0.94 NS NS NS
Interpeduncular nu 6.05 ± 0.72 8.15 ± 0.80*** 5.57 ± 0.81 5.56 ± 0.60 0.028
Substantia ni gra 3.59 ± 0.31 6.34 ± 1.24*** 3.77 ± 0.49 3.56 ± 0.49 < 0.001
Pretectal area 3.77 ± 0.62 5.44 ± 1.09** 4.38 ± 0.45 4.36 ± 0.26 0.003
Grey layer superior colliculus 4.00 ± 0.15 7.17 ± 1.50*** 4.49 ± 0.52 4.23 ± 0.59 < 0.001
Superior colliculus 3.89 ± 0.45 5.99 ± 0.51*** 4.03 ± 0.56 4.20 ± 0.84 0.009
Inferior colliculus 6.18 ± 0.51 9.04 ± 0.41*** 5.82 ± 1.24 5.79 ± 0.77 0.006
Flocculus 4.56 ± 0.13 5.30 ± 1.11 4.67 ± 1.04 4.81 ± 0.95 NS NS NS
Cerebellar gray matter 4.20 ± 0.26 5.55 ± 0.48*** 3.98 ± 0.31 3.82 ± 0.29 < 0.001
Molecular layer cerebellar gray 7.14 ± 0.65 7.13 ± 1.23 7.01 ± 0.77 7.09 ± 0.70 NS NS NS
White matter
Corpus callosum 3.18 ± 0.50 3.57 ± 0.29 2.90 ± 0.41 3.12 ± 0.35 NS NS NS
Zone incerta 3.08 ± 0.31 3.83 ± 1.15 3.40 ± 0.61 3.45 ± 0.53 NS NS NS
Internal capsule 2.69 ± 0.31 3.09 ± 0.35 2.43 ± 0.41 2.66 ± 0.56 NS NS NS
Cerebellar white matter 3.00 ± 0.09 3.31 ± 0.98 3.12 ± 0.78 3.24 ± 0.55 NS NS NS
Non-blood-brain barrier regions
Subfornical organ 5.45 ± 0.20 5.67 ± 1.26 5.39 ± 0.75 3.75 ± 0.67 NS NS NS
Median eminence 3.30 ± 0.12 3.83 ± 0.40 3.52 ± 0.67 3.81 ± 0.50 NS NS NS
Choroid plexus 22.6 ± 2.49 22.8 ± 2.69 24.0 ± 5.46 22.8 ± 4.0 NS NS NS
*

Abbreviations: nu, nucleus; Values of k are means ± S.D.

Main effects are not reported if statistically significant CBZ x NMDA interaction. In cases of statistically significant CBZ x NMDA interaction, unpaired t-tests were determined:

**

p < 0.01;

***

p < 0.001; vehicle plus NMDA versus vehicle plus saline, CBZ plus saline versus vehicle plus saline, and CBZ plus NMDA versus CBZ plus sali

Figure 1. Coronal autoradiographs of brain showing effects of NMDA and carbamazepine on regional AA incorporation coefficients k* in rats.

Figure 1

Values of k* (ml/sec/g brain × 10−4) are given on a color scale from 3 (blue) to 8 (yellow-orange). Abbreviations: Acg, anterior cingulate cortex; Aud, auditory cortex; AV, anteroventral thalamus nucleus; CPu, caudate-putamen; Fr, frontal cortex; Hipp, hippocampus; IPC, interpeduncular nucleus; MG, medial geniculate nucleus; Mot, motor cortex; Pyr, pyriform cortex; Vis, visual cortex.

Effects of chronic CBZ

In the 68 regions in which CBZ x NMDA interactions were statistically significant, t-tests showed that chronic CBZ vs. vehicle did not significantly change mean baseline (response to acute saline) k* in any region. In the 14 regions with statistically insignificant CBZ x NMDA interactions, CBZ did not have any main effect (Table 3). NMDA compared with saline did not change k* significantly in any of the 68 regions in which CBZ x NMDA interactions were statistically significant, nor did it affect k* in the pyriform cortex, where the CBZ x NMDA interaction was insignificant. Thus, chronic CBZ blocked each of the 69 NMDA-induced k* increments that were seen in the chronic vehicle rats.

Regional rates of incorporation of unlabeled unesterified plasma AA

Rates of incorporation of unlabeled unesterified AA from plasma into brain, Jin, for AA (Table were calculated by Eq. 2 using regional values of k* (Table 3) and cplasma did not differ significantly 2) (results not shown). Because baseline values of cplasmabetween chronic CBZ- and vehicle-treated rats (Table 2), the baseline relations of regional Jin corresponded to respective relations in regional values of k* between the two groups. In vehicle-treated rats, baseline Jin ranged from 3.3 fmol/sec/g in the periventricular area of the hypothalamus to 28.9 fmol/sec/g in the choroid plexus. by 2.1–2.4 fold (Table 2), a steady-However, because NMDA acutely increased cplasma state plasma (and likely brain) AA concentration was unavailable to calculate Jin in response to NMDA, and thus we did not calculate or compare Jin responses to acute NMDA and saline.

Brain PGE2 and TXB2 concentrations

A two-way ANOVA demonstrated significant interactions between CBZ and NMDA for brain PGE2 (p = 0.031) and TXB2 (p = 0.014) concentrations. Consequent t-tests showed that chronic CBZ decreased both basal concentrations, PGE2 by 45% (p = 0.023) and TXB2 by 51% (p = 0.0014) (Table 4). NMDA administration to chronic saline-treated rats significantly increased concentrations of PGE2 (p = 0.009) and TXB2 (p = 0.0009) by 1.8 and 2.2 fold, respectively, whereas NMDA had no significant effect in the chronic CBZ-treated rats.

Table 4.

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

Vehicle CBZ CBZ x NMDA interaction
Saline NMDA Saline NMDA p-value
PGE2 (ng/g brain) 7.8 ± 1.6 13.8 ± 4.7** 4.3 ± 1.7 ** 3.2 ± 1.4 0.031
TXB2 (pg/g brain) 43.4 ± 2.6 97.8 ± 17.6*** 21.2 ± 7.6** 38.2 ± 17.2 0.014

Each value is a mean ± S.D (n = 4).

**

p < 0.01;

***

p < 0.001; Vehicle plus NMDA versus vehicle plus saline, CBZ plus saline versus vehicle plus saline, and CBZ plus NMDA versus CBZ plus saline

Discussion

In this study, acute NMDA in chronic vehicle-treated unanesthetized adult rats increased k* for AA, a marker of cPLA2 activation and AA release from phospholipids, in widespread brain regions, and also increased brain concentrations of the AA metabolites, PGE2 and TXB2. Chronic CBZ blocked the NMDA-induced increments of k* as well as of PGE2 and TXB2. As chronic lithium also blocks NMDA-induced increments in k* for AA in unanesthetized rats (38), taken together the results suggest that effective antibipolar agents generally block NMDA-mediated signaling via AA. Supporting this interpretation, valproic acid, another effective agent, has been reported to interfere with NMDA neurotransmission (56,57), but it remains to be seen whether chronic valproate also blocks the NMDA-initiated AA signal in unanesthetized rats and, furthermore, whether abnormal NMDAR-mediated signaling, suggested to exist in BD (see Introduction), might be modified in humans by the agents (58).

Mechanisms underlying attenuation of NMDA-induced increments in k* by chronic CBZ or lithium are uncertain. Inhibition of AA cascade enzymes (e.g., cPLA2 and COX), as well as direct interference with the NMDAR, may be involved. Both chronic lithium and CBZ reduce rat brain cPLA2 activity, protein and mRNA levels, without changing activities of calcium-independent iPLA2 or secretory sPLA2 (33,34,59). cPLA2 downregulation by both drugs is accompanied by reduced expression and DNA-binding of activator protein 2 (AP-2), a cPLA2 transcription factor (47,60). Both drugs also reduce the brain basal (this paper and (34)) PGE2 concentration, and in this paper we show that CBZ also reduces brain basal TXB2 concentration and increments in PGE2 and TXB2 concentrations in response to acute NMDA. PGE2 and TXB2 are AA metabolites produced preferentially via COX-2 and COX-1, respectively, and their reductions agree with evidence of a 40% reduction in net brain COX activity caused by chronic CBZ (34,6165).

Reduced k* responses to NMDA also might arise from chronic CBZ’s effect on the NMDAR receptor itself, causing reduced entry of Ca2+ into the cell and reduced activation of cPLA2 and other Ca2+-dependent intracellular enzymes. Indeed, CBZ has been reported to interfere with multiple NMDAR signaling processes in in vitro studies (see Introduction) (3943,66). Additionally, chronic CBZ is reported to inhibit brain cyclic AMP-dependent protein kinase A, which phosphorylates the NMDAR (47,67). NMDAR activation can increase acetylation of histones H3 and H2A.X via the PKC/ERK signaling cascade, and CBZ can inhibit histone deacetylases. Thus, CBZ may affect histones as well as NMDAR transcription factors that are regulated by acetylation, such as specificity protein-1 (SP-1) (6871). CBZ’s blocking of NMDAR-mediated intracellular events is consistent with evidence that it has neuroprotective properties (44,7274). Neuroprotection may involve pro-apoptotic proteins p53, cytoprotective protein Bcl-2, and cell survival protein kinase Akt, and would be interesting to know the effects of CBZ on these proteins.

The ability of chronic CBZ (this paper) and of chronic lithium (38) to attenuate k* responses to AA in unanesthetized rats is consistent with proposals that inhibition of glutamatergic neurotransmission would have an antimanic effect in BD (1,2,75). In this regard, chronic lithium and valproic acid administration to rats reduced expression of the glutamatergic alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit, GluR1, an effect that been associated with reduced AMPA receptor function (76).

We did not determine if any of the CBZ effects arose from short-term brain exposure to drug. This is unlikely, as chronic CBZ exposure is accompanied by significant changes in cPLA2 mRNA and in the AP-2 transcription that modulates cPLA2 transcription, as well as by other neuroplastic changes that likely require long term exposure (34,47,7779). Acute CBZ effects include altering serotonergic transmission in vivo and in vitro and reducing high frequency firing in cultured neurons (8082). The brains in the present study were sampled within 4 hours after the last of the daily CBZ doses, and pharmacokinetic data show that, following an i.p. CBZ in rats, the brain concentration peaks at 100 min and then declines with a half-life of 205-min (83). Thus, the brain would have been exposed to a therapeutic level of CBZ for only 1–2 hours.

Our baseline values in Table 3 of k* (after acute saline in chronic vehicle-treated rats) ranged from 2.69 to 7.14 × 10−4 ml/sec/g brain, agreeing with published values (38,84,85). NMDA 25 mg/k.g. i.p. significantly increased k* in 69/82 brain structures that have high NMDAR densities, including the cerebral cortex, caudate-putamen, globus pallidus, hippocampus, thalamus, hypothalamus, superior and inferior colliculi, and substantia nigra (86, 87). In a prior study, 49/83 regions were significantly activated by the same NMDA dose (38). The discrepancy may be due to the lower variance of k* in the present study, a DMSO (vehicle) effect, or just to experimental variation.

Chronic CBZ did not significantly affect baseline values of k*, consistent with its lack of effect on baseline values of k* in individual phospholipid classes in microwaved rat brain (31). Regional baseline values for Jin in vehicle-treated rats, 33–289 × 10−4 nmol/sec/g, agree with a published global value of 65.7 × 10−4 nmol/sec/g (31). Given that Jin and k* represent regional rates of AA metabolic loss from brain (8890), our data indicate comparable baseline rates of AA loss, but reduced incremental rates following NMDA, in chronic CBZ- compared with vehicle-treated rats. The reduced incremental rates likely reflect the reduced formation of PGE2 and TXB2. In this regard, k* responses in rats to the cholinergic agonist arecoline, which activates PLA2  via muscarinic cholinergic receptors, were absent in COX-2 knockout compared with wild-type mice (89), and in rats treated with a mixed COX-1/2 inhibitor (Basselin et al., unpublished data).

The behavioral effects of NMDA noted in Table 1 were not altered by chronic CBZ, and also are not altered by chronic lithium (38). Consistent with the lack of effect of both agents, neither modifies the seizure threshold to NMDA in rodents (9193). Thus, the behavioral and seizure responses to NMDA do not appear to directly involve the AA cascade.

Acute NMDA significantly increased many unesterified fatty acid concentrations in plasma in both the chronic vehicle and CBZ groups (Table 2), likely through by stimulating corticosterone secretion (94,95). The inability of CBZ treatment to block these increments agrees with our previous report (31), and is consistent with evidence in normal human volunteers that CBZ increases cortisol blood levels (96).

If disturbed NMDAR-initiated signaling via AA contributes to the mania of BD, as has been suggested (see above), then our finding in this paper that CBZ suppresses the NMDA signal, and elsewhere that chronic LiCl feeding does so as well, may mean that the efficacy of both agents in BD involves such an effect. Experiments with valproic acid help to test this possibility, as valproic acid, like lithium and CBZ, downregulates the AA cascade in rat brain and has been shown to interfere with NMDAR function (29,36,56,57). Additionally, chronic lithium has been reported to reduce k* for AA in response to quinpirole, an agonist of dopaminergic D2-like receptors coupled to cPLA2 by a G-protein (84,97). It would be worthwhile to see if CBZ and VPA do so as well, which would suggest a more general receptor action of these agents on cPLA2-mediated AA signaling.

Acknowledgments

We thank the editorial assistance of the NIH Fellows’ Editorial Board.

Financial Disclosures

This work was supported entirely by the Intramural Research Program of the National Institute on Aging, National Institutes of Health. All authors have no conflicts of interest or financial interest.

Abbreviations

AA

arachidonic acid (20:4n-6)

AP-2

activator protein-2

BD

bipolar disorder

CBZ

carbamazepine

COX

cyclooxygenase

NMDA

N-methyl-D-aspartic acid

NMDAR

NMDA receptor

cPLA2

cytosolic phospholipase

sPLA2

secretory PLA2

iPLA2

calcium independent PLA2

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.

References

  • 1.Krystal JH, Sanacora G, Blumberg H, Anand A, Charney DS, Marek G, et al. Glutamate and GABA systems as targets for novel antidepressant and mood-stabilizing treatments. Mol Psychiatry. 2002;7(Suppl 1):S71–80. doi: 10.1038/sj.mp.4001021. [DOI] [PubMed] [Google Scholar]
  • 2.Zarate CA, Jr, Du J, Quiroz J, Gray NA, Denicoff KD, Singh J, et al. Regulation of cellular plasticity cascades in the pathophysiology and treatment of mood disorders: role of the glutamatergic system. Ann N Y Acad Sci. 2003;1003:273–291. doi: 10.1196/annals.1300.017. [DOI] [PubMed] [Google Scholar]
  • 3.Michael N, Erfurth A, Ohrmann P, Gossling M, Arolt V, Heindel W, 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]
  • 4.Castillo M, Kwock L, Courvoisie H, Hooper SR. Proton MR spectroscopy in children with bipolar affective disorder: preliminary observations. AJNR Am J Neuroradiol. 2000;21:832–838. [PMC free article] [PubMed] [Google Scholar]
  • 5.Itokawa M, Yamada K, Iwayama-Shigeno Y, Ishitsuka Y, Detera-Wadleigh S, Yoshikawa T. Genetic analysis of a functional GRIN2A promoter (GT)n repeat in bipolar disorder pedigrees in humans. Neurosci Lett. 2003;345:53–56. doi: 10.1016/s0304-3940(03)00501-9. [DOI] [PubMed] [Google Scholar]
  • 6.Martucci L, Wong AH, De Luca V, Likhodi O, Wong GW, King N, et al. N-methyl-D-aspartate receptor NR2B subunit gene GRIN2B in schizophrenia and bipolar disorder: Polymorphisms and mRNA levels. Schizophr Res. 2006;84:214–221. doi: 10.1016/j.schres.2006.02.001. [DOI] [PubMed] [Google Scholar]
  • 7.Mundo E, Tharmalingham S, Neves-Pereira M, Dalton EJ, Macciardi F, Parikh SV, et al. Evidence that the N-methyl-D-aspartate subunit 1 receptor gene (GRIN1) confers susceptibility to bipolar disorder. Mol Psychiatry. 2003;8:241–245. doi: 10.1038/sj.mp.4001218. [DOI] [PubMed] [Google Scholar]
  • 8.Scarr E, Pavey G, Sundram S, MacKinnon A, Dean B. Decreased hippocampal NMDA, but not kainate or AMPA receptors in bipolar disorder. Bipolar Disord. 2003;5:257–264. doi: 10.1034/j.1399-5618.2003.00024.x. [DOI] [PubMed] [Google Scholar]
  • 9.McCullumsmith RE, Kristiansen LV, Beneyto M, Scarr E, Dean B, Meador-Woodruff JH. Decreased NR1, NR2A, and SAP102 transcript expression in the hippocampus in bipolar disorder. Brain Res. 2007;1127:108–118. doi: 10.1016/j.brainres.2006.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gascon S, Deogracias R, Sobrado M, Roda JM, Renart J, Rodriguez-Pena A, et al. Transcription of the NR1 subunit of the N-methyl-D-aspartate receptor is down-regulated by excitotoxic stimulation and cerebral ischemia. J Biol Chem. 2005;280:35018–35027. doi: 10.1074/jbc.M504108200. [DOI] [PubMed] [Google Scholar]
  • 11.Ciabarra AM, Sullivan JM, Gahn LG, Pecht G, Heinemann S, Sevarino KA. Cloning and characterization of chi-1: a developmentally regulated member of a novel class of the ionotropic glutamate receptor family. J Neurosci. 1995;15:6498–6508. doi: 10.1523/JNEUROSCI.15-10-06498.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sucher NJ, Akbarian S, Chi CL, Leclerc CL, Awobuluyi M, Deitcher DL, et al. Developmental and regional expression pattern of a novel NMDA receptor-like subunit (NMDAR-L) in the rodent brain. J Neurosci. 1995;15:6509–6520. doi: 10.1523/JNEUROSCI.15-10-06509.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Das S, Sasaki YF, Rothe T, Premkumar LS, Takasu M, Crandall JE, 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]
  • 14.Dumuis A, Sebben M, Haynes L, Pin JP, Bockaert J. NMDA receptors activate the arachidonic acid cascade system in striatal neurons. Nature. 1988;336:68–70. doi: 10.1038/336068a0. [DOI] [PubMed] [Google Scholar]
  • 15.Kim DK, Rordorf G, Nemenoff RA, Koroshetz WJ, Bonventre JV. Glutamate stably enhances the activity of two cytosolic forms of phospholipase A2 in brain cortical cultures. Biochem J. 1995;310(Pt 1):83–90. doi: 10.1042/bj3100083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tapia-Arancibia L, Rage F, Recasens M, Pin JP. NMDA receptor activation stimulates phospholipase A2 and somatostatin release from rat cortical neurons in primary cultures. Eur J Pharmacol. 1992;225:253–262. doi: 10.1016/0922-4106(92)90027-s. [DOI] [PubMed] [Google Scholar]
  • 17.Weichel O, Hilgert M, Chatterjee SS, Lehr M, Klein J. Bilobalide, a constituent of Ginkgo biloba, inhibits NMDA-induced phospholipase A2 activation and phospholipid breakdown in rat hippocampus. Naunyn Schmiedebergs Arch Pharmacol. 1999;360:609–615. doi: 10.1007/s002109900131. [DOI] [PubMed] [Google Scholar]
  • 18.Shimizu T, Wolfe LS. Arachidonic acid cascade and signal transduction. J Neurochem. 1990;55:1–15. doi: 10.1111/j.1471-4159.1990.tb08813.x. [DOI] [PubMed] [Google Scholar]
  • 19.Rapoport SI. In vivo fatty acid incorporation into brain phospholipids in relation to plasma availability, signal transduction and membrane remodeling. J Mol Neurosci. 2001;16:243–261. doi: 10.1385/JMN:16:2-3:243. [DOI] [PubMed] [Google Scholar]
  • 20.Rapoport SI. In vivo approaches to quantifying and imaging brain arachidonic and docosahexaenoic acid metabolism. J Pediatr. 2003;143:S26–34. doi: 10.1067/s0022-3476(03)00399-8. [DOI] [PubMed] [Google Scholar]
  • 21.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]
  • 22.Dawson E, Gill M, Curtis D, Castle D, Hunt N, Murray R, et al. Genetic association between alleles of pancreatic phospholipase A2 gene and bipolar affective disorder. Psychiatr Genet. 1995;5:177–180. doi: 10.1097/00041444-199524000-00005. [DOI] [PubMed] [Google Scholar]
  • 23.Maida ME, Hurley SD, Daeschner JA, Moore AH, O'Banion MK. Cytosolic prostaglandin E2 synthase (cPGES) expression is decreased in discrete cortical regions in psychiatric disease. Brain Res. 2006;1103:164–172. doi: 10.1016/j.brainres.2006.05.048. [DOI] [PubMed] [Google Scholar]
  • 24.Chiu CC, Huang SY, Su KP, Lu ML, Huang MC, Chen CC, et al. Polyunsaturated fatty acid deficit in patients with bipolar mania. Eur Neuropsychopharmacol. 2003;13:99–103. doi: 10.1016/s0924-977x(02)00130-x. [DOI] [PubMed] [Google Scholar]
  • 25.Calabrese JR, Bowden C, Woyshville MJ. Lithium and the anticonvulsants in the treatment of bipolar disorder. In: Bloom FE, Kupfer DJ, editors. Psychopharmacology: The Fourth Generation of Progress. New York: Raven; 1995. pp. 1099–1111. [Google Scholar]
  • 26.Bowden CL, Karren NU. Anticonvulsants in bipolar disorder. Aust N Z J Psychiatry. 2006;40:386–393. doi: 10.1080/j.1440-1614.2006.01815.x. [DOI] [PubMed] [Google Scholar]
  • 27.Goodnick PJ. Anticonvulsants in the treatment of bipolar mania. Expert Opin Pharmacother. 2006;7:401–410. doi: 10.1517/14656566.7.4.401. [DOI] [PubMed] [Google Scholar]
  • 28.Post RM, Ketter TA, Uhde T, Ballenger JC. Thirty years of clinical experience with carbamazepine in the treatment of bipolar illness: principles and practice. CNS Drugs. 2007;21:47–71. doi: 10.2165/00023210-200721010-00005. [DOI] [PubMed] [Google Scholar]
  • 29.Chang MC, 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 MC, 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 1222:1141. [DOI] [PubMed] [Google Scholar]
  • 31.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. 2006;59:401–407. doi: 10.1016/j.biopsych.2005.07.024. [DOI] [PubMed] [Google Scholar]
  • 32.Bazinet RP, Weis MT, Rapoport SI, Rosenberger TA. Valproic acid selectively inhibits conversion of arachidonic acid to arachidonoyl-CoA by brain microsomal long-chain fatty acyl-CoA synthetases: relevance to bipolar disorder. Psychopharmacology (Berl) 2006;184:122–129. doi: 10.1007/s00213-005-0272-4. [DOI] [PubMed] [Google Scholar]
  • 33.Rintala J, Seemann R, Chandrasekaran K, Rosenberger TA, Chang L, Contreras MA, 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]
  • 34.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]
  • 35.Bosetti F, Rintala J, Seemann R, Rosenberger TA, Contreras MA, Rapoport SI, et al. 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]
  • 36.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]
  • 37.Rapoport SI, Bosetti F. Do lithium and anticonvulsants target the brain arachidonic acid cascade in bipolar disorder? Arch Gen Psychiatry. 2002;59:592–506. doi: 10.1001/archpsyc.59.7.592. [DOI] [PubMed] [Google Scholar]
  • 38.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. 2006;31:1659–1674. doi: 10.1038/sj.npp.1300920. [DOI] [PubMed] [Google Scholar]
  • 39.Lampe H, Bigalke H. Carbamazepine blocks NMDA-activated currents in cultured spinal cord neurons. Neuroreport. 1990;1:26–28. doi: 10.1097/00001756-199009000-00008. [DOI] [PubMed] [Google Scholar]
  • 40.Lancaster JM, Davies JA. Carbamazepine inhibits NMDA-induced depolarizations in cortical wedges prepared from DBA/2 mice. Experientia. 1992;48:751–753. doi: 10.1007/BF02124294. [DOI] [PubMed] [Google Scholar]
  • 41.Sofia RD, Gordon R, Gels M, Diamantis W. Comparative effects of felbamate and other compounds on N-methyl-D-aspartic acid-induced convulsions and lethality in mice. Pharmacol Res. 1994;29:139–144. doi: 10.1016/1043-6618(94)80037-5. [DOI] [PubMed] [Google Scholar]
  • 42.Hough CJ, Irwin RP, Gao XM, Rogawski MA, Chuang DM. Carbamazepine inhibition of N-methyl-D-aspartate-evoked calcium influx in rat cerebellar granule cells. J Pharmacol Exp Ther. 1996;276:143–149. [PubMed] [Google Scholar]
  • 43.Ambrosio AF, Silva AP, Malva JO, Soares-da-Silva P, Carvalho AP, Carvalho CM. Carbamazepine inhibits L-type Ca2+ channels in cultured rat hippocampal neurons stimulated with glutamate receptor agonists. Neuropharmacology. 1999;38:1349–1359. doi: 10.1016/s0028-3908(99)00058-1. [DOI] [PubMed] [Google Scholar]
  • 44.Bown CD, Wang JF, Young LT. Attenuation of N-methyl-D-aspartate-mediated cytoplasmic vacuolization in primary rat hippocampal neurons by mood stabilizers. Neuroscience. 2003;117:949–955. doi: 10.1016/s0306-4522(02)00743-1. [DOI] [PubMed] [Google Scholar]
  • 45.Farber NB, Jiang XP, Heinkel C, Nemmers B. Antiepileptic drugs and agents that inhibit voltage-gated sodium channels prevent NMDA antagonist neurotoxicity. Mol Psychiatry. 2002;7:726–733. doi: 10.1038/sj.mp.4001087. [DOI] [PubMed] [Google Scholar]
  • 46.Bialer M, Levy RH, Perucca E. Does carbamazepine have a narrow therapeutic plasma concentration range? Ther Drug Monit. 1998;20:56–59. doi: 10.1097/00007691-199802000-00010. [DOI] [PubMed] [Google Scholar]
  • 47.Rao JS, Bazinet RP, Rapoport SI, Lee HJ. Chronic administration of carbamazepine downregulates AP-2 DNA binding activity and AP-2α protein expression in rat frontal cortex. Biol Psychiatry. 2006 doi: 10.1016/j.biopsych.2006.03.029. [DOI] [PubMed] [Google Scholar]
  • 48.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]
  • 49.DeGeorge JJ, Noronha JG, Bell J, 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]
  • 50.Makrides M, Neumann MA, Byard RW, Simmer K, Gibson RA. Fatty acid composition of brain, retina, and erythrocytes in breast- and formula-fed infants. Am J Clin Nutr. 1994;60:189–194. doi: 10.1093/ajcn/60.2.189. [DOI] [PubMed] [Google Scholar]
  • 51.Paxinos G, Watson C. The rat brain in stereotaxic coordinates. 3. New York: Academic Press; 1987. [DOI] [PubMed] [Google Scholar]
  • 52.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]
  • 53.Radin NS. Extraction of tissue lipids with a solvent of low toxicity. Methods Enzymol. 1981;72:5–7. doi: 10.1016/s0076-6879(81)72003-2. [DOI] [PubMed] [Google Scholar]
  • 54.Tabachnick BG, Fidell LS. Computer-assisted research design and analysis. Boston: Allyn and Bacon ed; 2001. [Google Scholar]
  • 55.Motulsky HJ. Prism 4 Statistics Guide- Statistical analyses for laboratory and clinical researchers. San Diego CA: GraphPad Software Inc; 2003. www.graphpad.com. [Google Scholar]
  • 56.Zeise ML, Kasparow S, Zieglgansberger W. Valproate suppresses N-methyl-D-aspartate-evoked, transient depolarizations in the rat neocortex in vitro. Brain Res. 1991;544:345–348. doi: 10.1016/0006-8993(91)90078-a. [DOI] [PubMed] [Google Scholar]
  • 57.Gean PW, Huang CC, Hung CR, Tsai JJ. Valproic acid suppresses the synaptic response mediated by the NMDA receptors in rat amygdalar slices. Brain Res Bull. 1994;33:333–336. doi: 10.1016/0361-9230(94)90202-x. [DOI] [PubMed] [Google Scholar]
  • 58.Esposito G, Giovacchini G, Der M, Liow JS, Bhattacharjee AK, Ma K, et al. Imaging signal transduction via arachidonic acid in the human brain during visual stimulation, by means of positron emission tomography. Neuroimage. 2007;34:1342–1351. doi: 10.1016/j.neuroimage.2006.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Weerasinghe GR, Rapoport SI, Bosetti F. The effect of chronic lithium on arachidonic acid release and metabolism in rat brain does not involve secretory phospholipase A2 or lipoxygenase/cytochrome P450 pathways. Brain Res Bull. 2004;63:485–489. doi: 10.1016/j.brainresbull.2004.04.005. [DOI] [PubMed] [Google Scholar]
  • 60.Morri H, Ozaki M, Watanabe Y. 5′-flanking region surrounding a human cytosolic phospholipase A2 gene. Biochem Biophys Res Commun. 1994;205:6–11. doi: 10.1006/bbrc.1994.2621. [DOI] [PubMed] [Google Scholar]
  • 61.Bosetti F, Langenbach R, Weerasinghe GR. Prostaglandin E2 and microsomal prostaglandin E synthase-2 expression are decreased in the cyclooxygenase-2-deficient mouse brain despite compensatory induction of cyclooxygenase-1 and Ca2+-dependent phospholipase A2. J Neurochem. 2004;91:1389–1397. doi: 10.1111/j.1471-4159.2004.02829.x. [DOI] [PubMed] [Google Scholar]
  • 62.Choi SH, Langenbach R, Bosetti F. Cyclooxygenase-1 and -2 enzymes differentially regulate the brain upstream NF-kappaB pathway and downstream enzymes involved in prostaglandin biosynthesis. J Neurochem. 2006;98:801–811. doi: 10.1111/j.1471-4159.2006.03926.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pepicelli O, Fedele E, Bonanno G, Raiteri M, Ajmone-Cat MA, Greco A, et al. In vivo activation of N-methyl-D-aspartate receptors in the rat hippocampus increases prostaglandin E2 extracellular levels and triggers lipid peroxidation through cyclooxygenase-mediated mechanisms. J Neurochem. 2002;81:1028–1034. doi: 10.1046/j.1471-4159.2002.00897.x. [DOI] [PubMed] [Google Scholar]
  • 64.Iadecola C, Niwa K, Nogawa S, Zhao X, Nagayama M, Araki E, et al. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci U S A. 2001;98:1294–1299. doi: 10.1073/pnas.98.3.1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Okada S, Murakami Y, Nishihara M, Yokotani K, Osumi Y. 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]
  • 66.Cai Z, McCaslin PP. Amitriptyline, desipramine, cyproheptadine and carbamazepine, in concentrations used therapeutically, reduce kainate- and N-methyl-D-aspartate-induced intracellular Ca2+ levels in neuronal culture. Eur J Pharmacol. 1992;219:53–57. doi: 10.1016/0014-2999(92)90579-s. [DOI] [PubMed] [Google Scholar]
  • 67.Leonard AS, Hell JW. Cyclic AMP-dependent protein kinase and protein kinase C phosphorylate N-methyl-D-aspartate receptors at different sites. J Biol Chem. 1997;272:12107–12115. doi: 10.1074/jbc.272.18.12107. [DOI] [PubMed] [Google Scholar]
  • 68.Beutler AS, Li S, Nicol R, Walsh MJ. Carbamazepine is an inhibitor of histone deacetylases. Life Sci. 2005;76:3107–3115. doi: 10.1016/j.lfs.2005.01.003. [DOI] [PubMed] [Google Scholar]
  • 69.Crowe SL, Movsesyan VA, Jorgensen TJ, Kondratyev A. Rapid phosphorylation of histone H2A.X following ionotropic glutamate receptor activation. Eur J Neurosci. 2006;23:2351–2361. doi: 10.1111/j.1460-9568.2006.04768.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bai G, Kusiak JW. Functional analysis of the proximal 5′-flanking region of the N-methyl-D-aspartate receptor subunit gene, NMDAR1. J Biol Chem. 1995;270:7737–7744. doi: 10.1074/jbc.270.13.7737. [DOI] [PubMed] [Google Scholar]
  • 71.Ryu H, Lee J, Olofsson BA, Mwidau A, Dedeoglu A, Escudero M, et al. Histone deacetylase inhibitors prevent oxidative neuronal death independent of expanded polyglutamine repeats via an Sp1-dependent pathway. Proc Natl Acad Sci U S A. 2003;100:4281–4286. doi: 10.1073/pnas.0737363100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Costa C, Martella G, Picconi B, Prosperetti C, Pisani A, Di Filippo M, et al. Multiple mechanisms underlying the neuroprotective effects of antiepileptic drugs against in vitro ischemia. Stroke. 2006;37:1319–1326. doi: 10.1161/01.STR.0000217303.22856.38. [DOI] [PubMed] [Google Scholar]
  • 73.Mark RJ, Ashford JW, Goodman Y, Mattson MP. Anticonvulsants attenuate amyloid beta-peptide neurotoxicity, Ca2+ deregulation, and cytoskeletal pathology. Neurobiol Aging. 1995;16:187–198. doi: 10.1016/0197-4580(94)00150-2. [DOI] [PubMed] [Google Scholar]
  • 74.McDonald JW, Johnston MV. Pharmacology of N-methyl-D-aspartate-induced brain injury in an in vivo perinatal rat model. Synapse. 1990;6:179–188. doi: 10.1002/syn.890060210. [DOI] [PubMed] [Google Scholar]
  • 75.Abelli M, Martini C, Pini S, Gesi C, Cassano GB. The role of newer antiglutamatergic agents as mood-stabilizers in bipolar disorder. Curr Drug Targets. 2005;6:607–614. doi: 10.2174/1389450054546015. [DOI] [PubMed] [Google Scholar]
  • 76.Du J, Gray NA, Falke CA, Chen W, Yuan P, Szabo ST, et al. Modulation of synaptic plasticity by antimanic agents: the role of AMPA glutamate receptor subunit 1 synaptic expression. J Neurosci. 2004;24:6578–6589. doi: 10.1523/JNEUROSCI.1258-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Jakobsen SN, Wiborg O. Selective effects of long-term lithium and carbamazepine administration on G-protein subunit expression in rat brain. Brain Res. 1998;780:46–55. doi: 10.1016/s0006-8993(97)01181-5. [DOI] [PubMed] [Google Scholar]
  • 78.Lesch KP, Aulakh CS, Tolliver TJ, Hill JL, Wolozin BL, Murphy DL. Differential effects of long-term lithium and carbamazepine administration on Gs alpha and Gi alpha protein in rat brain. Eur J Pharmacol. 1991;207:355–359. doi: 10.1016/0922-4106(91)90011-6. [DOI] [PubMed] [Google Scholar]
  • 79.Bachmann RF, Schloesser RJ, Gould TD, Manji HK. Mood stabilizers target cellular plasticity and resilience cascades: implications for the development of novel therapeutics. Mol Neurobiol. 2005;32:173–202. doi: 10.1385/MN:32:2:173. [DOI] [PubMed] [Google Scholar]
  • 80.Macdonald RL, McLean MJ. Mechanisms of anticonvulsant drug action. Electroencephalogr Clin Neurophysiol Suppl. 1987;39:200–208. [PubMed] [Google Scholar]
  • 81.Pranzatelli MR. Effect of antiepileptic and antimyoclonic drugs on serotonin receptors in vitro. Epilepsia. 1988;29:412–419. doi: 10.1111/j.1528-1157.1988.tb03740.x. [DOI] [PubMed] [Google Scholar]
  • 82.Okada M, Hirano T, Mizuno K, Kawata Y, Wada K, Murakami T, et al. Effects of carbamazepine on hippocampal serotonergic system. Epilepsy Res. 1998;31:187–198. doi: 10.1016/s0920-1211(98)00025-4. [DOI] [PubMed] [Google Scholar]
  • 83.Graumlich JF, McLaughlin RG, Birkhahn D, Shah N, Burk A, Jobe PC, et al. Subcutaneous microdialysis in rats correlates with carbamazepine concentrations in plasma and brain. Epilepsy Res. 2000;40:25–32. doi: 10.1016/s0920-1211(00)00110-8. [DOI] [PubMed] [Google Scholar]
  • 84.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]
  • 85.Basselin M, Chang L, Seemann R, Bell JM, Rapoport SI. Chronic lithium administration to rats selectively modifies 5-HT2A/2C receptor-mediated brain signaling via arachidonic acid. Neuropsychopharmacology. 2005;30:461–472. doi: 10.1038/sj.npp.1300611. [DOI] [PubMed] [Google Scholar]
  • 86.Cotman CW, Monaghan DT. Anatomical organization of excitatory amino acid receptors and their properties. Adv Exp Med Biol. 1986;203:237–252. doi: 10.1007/978-1-4684-7971-3_18. [DOI] [PubMed] [Google Scholar]
  • 87.Jarvis MF, Murphy DE, Williams M. Quantitative autoradiographic localization of NMDA receptors in rat brain using [3H]CPP: comparison with [3H]TCP binding sites. Eur J Pharmacol. 1987;141:149–152. doi: 10.1016/0014-2999(87)90423-7. [DOI] [PubMed] [Google Scholar]
  • 88.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]
  • 89.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 mouse. J Neurochem. 2006;96:669–679. doi: 10.1111/j.1471-4159.2005.03612.x. [DOI] [PubMed] [Google Scholar]
  • 90.DeMar JC, Jr, Lee HJ, Ma K, Chang L, Bell JM, Rapoport SI, et al. 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]
  • 91.Steppuhn KG, Turski L. Modulation of the seizure threshold for excitatory amino acids in mice by antiepileptic drugs and chemoconvulsants. J Pharmacol Exp Ther. 1993;265:1063–1070. [PubMed] [Google Scholar]
  • 92.Turski L, Niemann W, Stephens DN. Differential effects of antiepileptic drugs and beta-carbolines on seizures induced by excitatory amino acids. Neuroscience. 1990;39:799–807. doi: 10.1016/0306-4522(90)90262-3. [DOI] [PubMed] [Google Scholar]
  • 93.Ormandy GC, Song L, Jope RS. Analysis of the convulsant-potentiating effects of lithium in rats. Exp Neurol. 1991;111:356–361. doi: 10.1016/0014-4886(91)90103-j. [DOI] [PubMed] [Google Scholar]
  • 94.Kent S, Kernahan SD, Levine S. Effects of excitatory amino acids on the hypothalamic-pituitary-adrenal axis of the neonatal rat. Brain Res Dev Brain Res. 1996;94:1–13. doi: 10.1016/0165-3806(96)00023-5. [DOI] [PubMed] [Google Scholar]
  • 95.Boonstra R, Tinnikov AA. Increased corticosteroid binding capacity of plasma albumin but not of corticosteroid-binding globulin caused by ACTH-induced changes in free fatty acid concentrations in snowshoe hares and rabbits. J Endocrinol. 1998;156:205–212. doi: 10.1677/joe.0.1560205. [DOI] [PubMed] [Google Scholar]
  • 96.Perini GI, Devinsky O, Hauser P, Gallucci WT, Theodore WH, Chrousos GP, et al. Effects of carbamazepine on pituitary-adrenal function in healthy volunteers. J Clin Endocrinol Metab. 1992;74:406–412. doi: 10.1210/jcem.74.2.1309836. [DOI] [PubMed] [Google Scholar]
  • 97.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]

RESOURCES