Skip to main content
ACS Chemical Neuroscience logoLink to ACS Chemical Neuroscience
. 2014 May 1;5(6):459–467. doi: 10.1021/cn500058v

Lithium and the Other Mood Stabilizers Effective in Bipolar Disorder Target the Rat Brain Arachidonic Acid Cascade

Stanley I Rapoport 1,*
PMCID: PMC4063504  PMID: 24786695

Abstract

graphic file with name cn-2014-00058v_0002.jpg

This Review evaluates the arachidonic acid (AA, 20:4n-6) cascade hypothesis for the actions of lithium and other FDA-approved mood stabilizers in bipolar disorder (BD). The hypothesis is based on evidence in unanesthetized rats that chronically administered lithium, carbamazepine, valproate, or lamotrigine each downregulated brain AA metabolism, and it is consistent with reported upregulated AA cascade markers in post-mortem BD brain. In the rats, each mood stabilizer reduced AA turnover in brain phospholipids, cyclooxygenase-2 expression, and prostaglandin E2 concentration. Lithium and carbamazepine also reduced expression of cytosolic phospholipase A2 (cPLA2) IVA, which releases AA from membrane phospholipids, whereas valproate uncompetitively inhibited in vitro acyl-CoA synthetase-4, which recycles AA into phospholipid. Topiramate and gabapentin, proven ineffective in BD, changed rat brain AA metabolism minimally. On the other hand, the atypical antipsychotics olanzapine and clozapine, which show efficacy in BD, decreased rat brain AA metabolism by reducing plasma AA availability. Each of the four approved mood stabilizers also dampened brain AA signaling during glutamatergic NMDA and dopaminergic D2 receptor activation, while lithium enhanced the signal during cholinergic muscarinic receptor activation. In BD patients, such signaling effects might normalize the neurotransmission imbalance proposed to cause disease symptoms. Additionally, the antidepressants fluoxetine and imipramine, which tend to switch BD depression to mania, each increased AA turnover and cPLA2 IVA expression in rat brain, suggesting that brain AA metabolism is higher in BD mania than depression. The AA hypothesis for mood stabilizer action is consistent with reports that low-dose aspirin reduced morbidity in patients taking lithium, and that high n-3 and/or low n-6 polyunsaturated fatty acid diets, which in rats reduce brain AA metabolism, were effective in BD and migraine patients.

Keywords: Lithium, bipolar disorder, arachidonic acid, carbamazepine, mood stabilizers, valproic acid, rat, brain, antidepressant, antipsychotics, biotype

1. Introduction

Bipolar disorder (BD) is characterized by recurring cycles of depressive and manic symptoms (Bipolar I) or hypomanic symptoms (Bipolar II). The depressive phase is three times more common than the manic or hypomanic phase, and the lifetime suicide risk is 10–20%. BD is a life-long malady that is not diagnosed on average until 10 years after symptoms appear, and treatment may be delayed for another 10 years.1 Two serial BD “biotypes” or “biostages” are recognized. An initial one, perhaps explaining presenting symptoms, involves an imbalanced neurotransmission consisting of excessive dopaminergic and glutamatergic transmission, reduced cholinergic muscarinic transmission, with disturbed serotonergic transmission.1b,2 The later appearing biotype additionally includes cognitive decline, brain atrophy, and symptom worsening, and overlaps with the biotypes of schizophrenia, schizoaffective disorder, and major depressive disorder.3

Even with intensive treatment in academic centers, BD therapy is inadequate and produces frequent side effects; on average, patients remain symptomatic for half the year. Thus, major challenges in the field are to identify more effective, less toxic drugs for treatment, and to deal with poor compliance. But drug development has not progressed markedly in the last decades, in part because BD pathology is not sufficiently understood and there is no accepted behavioral animal model for the disease.1a Identifying a drug target also is difficult because genomic studies implicate 90 or more risk alleles, each with only a small statistically significant effect.4 One possible approach, however, is to try to understand mechanisms of action of the already FDA-approved mood stabilizers, using cell or animal biochemical models. These agents, the univalent ion lithium, valproate (2-propylpentanoate), carbamazepine (5H-dibenz[b,f ]azepine-5-carboxamide), and lamotrigine (3,5-diamino-6-(2,3-dichlorophenyl)-as-triazine), have no common structure that would suggest a specific common target.1

Since the discovery of lithium’s efficacy against BD some 65 years ago,5 multiple hypotheses have been suggested to explain its action,1 some of which are presented in this volume. In this Review, I present evidence for the arachidonic acid (AA) cascade hypothesis, while other actively investigated hypotheses include the following: (1) Myo-inositol depletion (inhibition of inositol monophosphatase (IMPase) in the phosphatidylinositide cycle).6 (2) Inhibition of glycogen synthase kinase-3β (GSK-3β).7 (3) Inhibition of protein kinase C. This hypothesis has been proposed to explain the action of Tamoxifen against bipolar mania.8 (4) Inhibition of NMDA/AMPA receptors. This hypothesis is consistent with evidence that both the N-methyl-d-aspartate (NMDA) receptor antagonist ketamine, and the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonist riluzole, showed efficacy in bipolar depression.9 It overlaps to some extent the AA cascade hypothesis, since each of the mood stabilizers blocks an AA signal caused by injected NMDA in rats (see below).

2. The Arachidonic Cascade Hypothesis

2.1. The AA Cascade

Unlike many of the other hypotheses, the AA cascade hypothesis for lithium’s action does not identify a single enzyme, protein, or receptor target of lithium. Rather, this hypothesis encompasses a system of ordered metabolic reactions involving AA and its metabolites, which can be modulated by receptor or biochemical events.10 Within this “system,” there can be several points of therapeutic intervention. The hypothesis also applies to each of the four mood stabilizers approved by the FDA for treating BD (but not to potential mood stabilizers proven ineffective), and further suggests related mechanisms of action of certain antidepressants and atypical antipsychotics, of aspirin, and of dietary intervention, with regard to BD symptoms (see below).1a,11

AA (20:4n-6) is a long-chain n-6 polyunsaturated fatty acid (PUFA) that, like the n-3 PUFA, docosahexaenoic acid (DHA, 22:6n-3), is esterified in millimolar concentrations in brain phospholipids, triacylglycerols, and cholesteryl esters, or is found in lower μmolar concentrations in its unesterified (free) form within cells, often bound to fatty acid binding proteins. Neither AA nor DHA can be synthesized de novo in vertebrates. Each must be absorbed through the diet or synthesized mainly within the liver by elongation of its shorter-chain nutritionally essential precursor, linoleic acid (LA, 18:2n-6) and α-linolenic acid (α-LNA, 18:3n-3), respectively.10 AA and DHA are metabolized by separate but interacting metabolic systems or cascades within brain, which are regulated by enzymes often showing specificity for one or the other PUFA and its metabolites.12 These enzymes often are functionally and transcriptionally coupled within the separate cascades, during brain development and aging.13 Both AA and DHA and their metabolites have important second messenger actions in brain, affecting gene transcription, membrane fluidity, neurotransmission, electrical excitability, neuroinflammation, excitotoxicity, energy consumption, and other functions.10

Figure 1 illustrates some relevant pathways within the AA cascade, superimposed on an outline of synaptic and cell structure.11a In this illustration, the cascade is initiated at the outer plasma membrane surface when an agonist binds to a neuroreceptor that is coupled to Ca2+-dependent AA-selective cytosolic phospholipase A2 (cPLA2).12a cPLA2 hydrolyzes esterified AA from the stereospecifically numbered (sn)-2 position of synaptic membrane phospholipid, and can be activated via G-protein coupled cholinergic muscarinic M1,3,5,14 dopaminergic D2-like,15 or serotonergic 5-HT2A/2C receptors,16 or following entry of extracellular Ca2+ into the cell due to glutamate binding to ionotropic NMDA or AMPA receptors.17 Of the unesterified AA released into the cytoplasm, the largest fraction (about 95%) is recycled by conversion to AA-CoA by an acyl-CoA synthetase (Acsl) (selectively Acsl-4) with the consumption of 2 ATP,18 and then is re-esterified by an acyltransferase (selectively lysophospholipid acyltransferase (LPCAT)-313,19) into an available sn-2 position of membrane lysophospholipid. The smaller fraction is metabolized through enzymatic oxidation by cyclooxygenase (COX)-2, COX-1, cytochrome P450 epoxygenase (CYP450), or lipoxygenase (LOX), to produce multiple bioactive eicosanoid metabolites, including pro-inflammatory prostaglandin E2 (PGE2) and thromboxane B2 (TXB2). AA also can be β-oxidized within mitochondria, nonenzymatically converted to reactive oxygen species (ROS), or follow other degradative pathways.

Figure 1.

Figure 1

Model of brain arachidonic acid cascade initiated at synapse, with sites of action of mood stabilizers and atypical antipsychotics, based on studies in unanesthetized rats and in vitro. Arachidonic acid (AA), esterified within synaptic membrane phospholipid, is liberated following ligand binding to a neuroreceptor on the outer surface of the plasma membrane, which is coupled cPLA2 activation by a G protein or Ca2+. A fraction of liberated unesterified AA is converted to bioactive eicosanoids (e.g., PGE2) by COX-2, lipoxygenase (LOX), COX-1, or cytochrome P450 epoxygenase (CYP450), which together with AA produce cellular actions. The larger remaining fraction is converted to AA-CoA by AA-selective acyl-CoA synthetase (Acsl)-4, then is re-esterified into membrane by lysophospholipid choline acyltransferase (LPCAT)-3. When administered chronically to rats, each of the four mood stabilizers interferes with neuroreceptor-mediated activation of cPLA2, reduces COX-2 activity and PGE2 concentration in the brain. Valproate, lamotrigine, and the antipsychotics olanzapine and clozapine also each reduce COX-2 gene transcription within the nucleus via NF-κB. Lithium and carbamazepine each reduce cPLA2 IVA expression by reducing its gene transcription by AP-2, whereas valproate uncompetitively inhibits AA-selective Acsl-4. Both lithium and carbamazepine increase GRK-3, which may reduce G-protein neuroreceptor coupled activation of cPLA2. The figure also illustrates diffusion of circulating unlabeled unesterified AA and radiolabeled AA* into the cellular unesterified AA pool that is available for reacylation.18 See text for details. Prepared by Dr. Chuck T. Chen as adapted from Rao et al.11a

In the pathological condition of neuroinflammation, which is associated with microglial activation, the AA cascade is chronically upregulated by a number of mechanisms. These include secretion of cytokines (e.g., interleukin (IL)-1β or tumor necrosis factor (TNF)-α) that stimulate astrocytic receptors that are coupled to activation of cPLA2 and secretory sPLA2,20 and excess glutamatergic levels that stimulate neuronal NMDA and AMPA receptors and cause excitotoxicity.21 Synaptic loss and apoptosis often accompany these changes.22

2. Effects of Drugs Used in Bipolar Disorder on Arachidonic Acid Cascade

2.1. Lithium and Other Mood Stabilizers Downregulate Rat Brain AA Cascade

Figure 1 also identifies suggested sites of action of the four FDA-approved mood stabilizers within the AA cascade, as well as of olanzapine and clozapine (see below), based on experiments in unanesthetized rats and in vitro. At the apex of the cascade, each of the mood stabilizers can modulate AA hydrolysis by cPLA2, initiated by agonist activation of certain neuroreceptors. The experimental pattern of modulation is consistent with their ability to rectify the proposed neurotransmission imbalance in early biostage BD (see above).1a This was shown by neuroimaging studies in partially restrained unanesthetized rats that had been treated chronically with vehicle or a mood stabilizer, having indwelling femoral vein and artery catheters.18 Just before acute saline or drug was injected, radiolabeled [1-14C]AA (*AA in Figure 1) was infused intravenously for 5 min, the animal was killed, and its brain was removed, frozen, and sectioned coronally for quantitative autoradiography. Regional incorporation coefficients k* were quantified as the ratio of brain radioactivity to integrated arterial plasma radioactivity (input function), from which regional rates of incorporation, Jin, the product of k* and unesterified unlabeled plasma AA, were estimated. Jin equals the rate of replacement by circulating unesterified AA of the AA that has been metabolically lost within brain.18,23 Both k* and Jin are unaffected by changes in cerebral blood flow, and thus, the imaging method can be used under pathological conditions and with changing functional activity.

As shown in Table 1, chronically administered lithium, valproate, carbamazepine, or lamotrigine, at a therapeutically relevant plasma level, each blocked the AA signal in response to 25 or 50 (lithium) mg/kg i.p. NMDA in unanesthetized rats.24 Blockage by lithium and carbamazepine is consistent with their in vitro inhibition of NMDA-induced Ca2+ influxes,25 and with therapeutic effects of NMDA or AMPA antagonists in bipolar depression9 (see above). Lithium, carbamazepine, and valproate each also dampened the AA signal in rats injected with the D2-like receptor agonist quinpirole,26 while lithium decreased the signal in brain auditory and visual areas in response to DOI ((±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane hydrochloride), a serotonergic 5-HT2A/2C receptor agonist.27 Consistent with the proposed hypocholinergic neurotransmission in BD (see above), and lithium’s proconvulsant action with physostigmine, chronic lithium increased the rat brain AA signal to the muscarinic M1,3,5 agonist arecoline,28 while also increasing brain glucose metabolism.29 In separate experiments, each of the acute agonist-induced signals could be blocked by pretreatment with the specific receptor antagonist, confirming its specific receptor origin (Table 2).

Table 1. Effects of Each of Four Chronically Administered Mood Stabilizers in Unanesthetized Rats on Arachidonic Acid Signaling Provoked by Acute Administration of NMDA, Dopaminergic D2, Cholinergic muscarinic M1,3,5 or Serotonergic 5-HT2A/2C agonist, NMDA, Quinpirole, Arecoline, and 2,5-Dimethoxy-4-iodoamphetamine (DOI)a.

  drug effects on arachidonic acid signal
receptor subtype glutamatergic NMDA signal dopaminergic D2 signal cholinergic muscarinic M1,3,5 signal serotonergic 5-HT2A/2C signal
coupling to cPLA2 Ca2+ coupled G-protein coupled
agonistb NMDA quinpirole arecoline DOI
antagonistc MK-801 raclopride or butaclamol atropine mianserin
mood stabilizers
lithium d
carbamazepine e e
valproate e e
lamotrigine e e e
a

Specificity of a receptor effect was confirmed by blocking the AA signal in independent experiments with pretreatment with MK-801, raclopride or butaclamol, atropine, or mianserin prior to agonist injection. See text for references.

b

Agonist used to provoke signal in brain.

c

Antagonist that blocked signal in independent experiments.

d

Selective to auditory and visual brain areas.

e

Not tested. See Text for references.

Table 2. Effects of Chronic Administration of Each of Four FDA Approved Mood Stabilizers, and of Topiramate and Gabapentin, on Different Aspects of the Rat Brain Arachidonic Cascadea.

drug AA turnover DHA turnover cPLA2 activity, protein, mRNA iPLA2 activity, protein, mRNA Acsl-4 activity COX-1 protein COX-2 protein COX activity PGE2 concen- tration TXB2 concen- tration AP-2 NF-κB
lithium NCb NC NC NC NC NCf
carbamazepine NC NC   NC NC
valproate NCb NC NC d NC
lamotrigine c   NC NC   NC d NC  
topiramate NC NC NC NC   NC NC NC        
gabapentin     e NC   NC NC NC NC    
a

See text for references. NC, no significant change.

b

Also no effect on palmitate turnover.

c

AA incorporation coefficient decreased.

d

mRNA also reduced.

e

Only mRNA reduced.

f

Chronic lithium did not reduce NF-κB in intact rat, but does so in neuroblastoma SH-SY5Y cells in vitro.

Downstream in the cascade at the inner plasma membrane (Figure 1), G-protein receptor kinases (GRKs) modulate homologous desensitization of agonist activated G-protein coupled receptors, like those identified in Table 1. In rat brain, chronic lithium and carbamazepine each significantly increased GRK-3 expression in the membrane but not cytosolic fraction, which might desensitize the AA signal initiated at G-protein coupled D2-like and other receptors.30

It is possible to quantify turnover of long chain fatty acids in brain phospholipids of partially restrained unanesthetized rats by infusing the radiolabeled fatty acid intravenously for 5 min, determining integrated plasma specific activity by repeated arterial sampling, then killing the rat and subjecting its brain to high energy microwaving to prevent post-mortem metabolic changes.18 Fatty acid specific activity (radioactive/cold concentration) is measured in brain acyl-CoA (Figure 1), the precursor pool for fatty acid incorporation into phospholipid, and in plasma to calculate, as a ratio, a dilution factor λ. A mathematical model then is applied to determine fatty acid turnover in individual phospholipids and other kinetic parameters.18 Using this approach, we showed that chronic lithium, carbamazepine or valproate each reduced AA turnover (deacylation–reacylation31 (Figure 1)) in brain phospholipids of unanesthetized rats, while lamotrigine reduced AA incorporation into brain from plasma32 (Table 2). The reductions were selective for AA, since lithium, valproate, or carbamazepine did not reduce DHA turnover, and lithium or valproate did not reduce palmitate turnover.1a

Related to their selective reduction of AA turnover, chronic lithium and carbamazepine each reduced transcription (mRNA level) and activity of AA-selective Ca2+-dependent cPLA2 IVA12a in rat brain, and expression of activator protein-2 (AP-2), a cPLA2 IVA transcription factor, without changing expression of DHA-selective Ca2+-independent iPLA2 VIA or of sPLA2 IIA (Table 2).1a Valproate did not modify expression of any of the three PLA2 enzymes, but uncompetitively inhibited AA activation to AA-CoA by recombinant AA-selective Acsl-4 and by a microsomal rat brain preparation.33 Lithium did not produce such inhibition. On this basis, we are testing in our turnover rat model proven nonteratogenic inhibitors of recombinant Acsl-4, such as the valproate amide derivative valnoctamide,34 as potential new mood stabilizers for treating BD via their effect on the AA cascade.35

Further downstream in the cascade (Figure 1), COX-2 colocalizes and is functionally coupled with cPLA2 IVA at postsynaptic sites in brain.36 Each of the four mood stabilizers reduced brain COX-2 activity and PGE2 concentration when given chronically to rats (Table 2). Chronic lithium did not reduce NF-κB in the intact rat,37 but does so in neuroblastoma SH-SY5Y cells in vitro.38 Valproate and lamotrigine additionally reduced COX-2 protein and mRNA and the COX-2 transcription factor, NF-κB.1a Chronic valproate also reduced brain mRNA levels for 87 genes and increased levels for 34 genes by at least 40%, indicating that its AA cascade effects are embedded in multiple other brain changes.37a Lithium’s selectivity for the COX-2 pathway was illustrated by showing that it did not change expression of COX-1, 5-LOX, CYP450, or membrane PGE synthase-2 (mPGES-2) in rat brain.39 At the resting state, it is thought that PGE2 is produced from AA primarily via COX-2, TXB2 via COX-1.40 Lithium, lamotrigine or gabapentin did not change the rat brain TXB2 concentration, while carbamazepine and VPA each reduced TXB2.24c,24d,26c,41 Additionally, neither valproate nor carbamazepine altered 5-LOX or its product leukotriene B4 (LTB4), and carbamazepine did not change CYP450.42

An unexpected and as yet incompletely understood observation was that chronic lithium elevated rat brain concentration of antiinflammatory 17-hydroxy-DHA and other as yet unidentified DHA metabolites.43 This elevation may contribute to the reported synergy between aspirin and lithium in BD patients, since 17-hydroxy-DHA is formed from DHA by acetylated COX-2 following exposure to aspirin (see below).44

In contrast to the overlapping actions of the four FDA approved mood stabilizers, the antiepileptic topiramate (2,3:4,5-bis-O-(1-methylethylidene)-β-d-fructopyranose 1-sulfamate), which failed in phase III trials in BD I patients, did not change any measured rat brain AA cascade marker,45 nor did gabapentin (1-(aminomethyl)cyclohexaneacetic acid), which also lacks efficacy.41 Thus, the AA cascade model for lithium’s action has a strong clinical-experimental correlation based on the rat studies with the six drugs.

2.2. Antidepressants That Switch Bipolar Depression to Mania Increase Rat Brain AA Metabolism

The AA cascade hypothesis has relevance for the effects of certain antidepressants and atypical antipsychotics in BD.1b,11b For example, the tricyclic antidepressant imipramine and the selective serotonin reuptake inhibitor (SSRI) fluoxetine are reported to increase “switching” of bipolar depression to mania when used as monotherapy or with mood stabilizers.46 Bupropion, also an antidepressant but a norepinephrine and dopamine reuptake inhibitor and nicotinic antagonist, does not increase switching.46 These clinical distinctions correlated with the different effects of the three drugs on rat brain AA metabolism. Thus, chronic fluoxetine and imipramine at therapeutically relevant doses increased AA turnover in rat brain phospholipids, as well as expression (activity, protein, mRNA, and phosphorylation) of cPLA2 IVA and of its transcription factor subunit, AP-2α,47 whereas chronic bupropion had no comparable effect. These results imply that the manic or hypomanic phase of BD has a higher brain AA metabolic rate than does the depression phase, a hypothesis that can be tested directly by PET imaging of brain AA metabolism using [1-11C]AA or [18F]AA.48

2.3. Atypical Antipsychotics Used in Bipolar Disorder Indirectly Decrease Rat Brain AA Metabolism

Atypical antipsychotics may act in part in BD by reducing the brain AA cascade, albeit indirectly and secondary to their effect on hepatic PUFA metabolism.49 Olanzapine is an atypical antipsychotic that is FDA-approved for maintenance therapy in Bipolar I, as well as for bipolar mania, and it can rapidly dampen hyperactive motoric symptoms before mood stabilizers start to act.1b Clozapine shows efficacy in acute BD mania, rapid cycling BD, and as maintenance therapy in patients with refractory BD.1b As a class, atypical antipsychotics have a high affinity as antagonists for dopaminergic D2 and serotonergic 5-HT2 receptors. They may produce fewer motor side effects than typical antipsychotics such as haloperidol because of their ability to rapidly dissociate from D2 receptors.50

Using our in vivo kinetic method, we found that both olanzapine and clozapine when chronically administered to rats reduced brain COX activity and PGE2 concentration, plasma unesterified AA concentration, and AA incorporation into brain from plasma (Figure 1).51 Olanzapine alone also reduced AA turnover within brain phospholipids, while clozapine alone also increased expression of DHA-selective iPLA2 VIA and COX-1.

3. Additional Therapeutic Approaches to BD Involving the Brain Arachidonic Acid Cascade

3.1. Low Dose Aspirin

In a pharmacoepidemiological study of patients taking lithium for an average duration of 847 days, patients receiving low-dose (30 or 80 mg/day) acetylsalicylic acid (aspirin) were significantly less likely to have a “medication event” (evidence of disease worsening) than patients on lithium alone, independently of use duration.44 High dose aspirin given for short periods of time, nonselective COX inhibitors, selective COX-2 inhibitors, or glucocorticoids were not beneficial. As low dose aspirin does not increase serum lithium,52 aspirin’s synergistic effect with lithium likely was centrally mediated, particularly because it can enter the brain and inhibit AA metabolism.53 Clinical trials with aspirin in BD currently are underway.54

A central positive effect of aspirin in BD is consistent with a report that aspirin given to men undergoing coronary angiography reduced depression and anxiety.55 Of relevance, the COX-2 inhibitor celecoxib, although having low brain penetrability,56 showed significant positive effects as adjunctive therapy in BD patients experiencing depressive or mixed episodes, and in depressed patients.57

The clinical data are consistent with the AA cascade hypothesis. Acetylation of COX-2 by aspirin reduces the ability of the enzyme to convert AA to pro-inflammatory PGE2. Additionally, acylated COX-2 can convert AA to anti-inflammatory mediators such as lipoxin A4 and 15-epi-lipoxin A4, as well as DHA to anti-inflammatory 17-(R)-OH-DHA.43a Lithium similarly reduces rat brain COX-2 activity and PGE2 concentration (Table 2), while increasing brain concentrations of 17-hydroxy-DHA and other potential DHA-derived anti-inflammatory metabolites.43b

3.2. Changing Dietary PUFA Composition Can Suppress Brain Arachidonic Acid Cascade

Brain concentrations of AA and DHA can be altered reciprocally by changing dietary PUFA concentrations, since brain AA and DHA concentrations depend on dietary intake and hepatic elongation from nutritionally essential LA and α-LNA, respectively.49 Furthermore, decreases in dietary LA and increases in dietary α-LNA have been reported to be neuroprotective in animal models. In rats, reducing dietary α-LNA below a level considered to be PUFA “adequate” reduces brain DHA concentration and uptake, expression of DHA-selective iPLA2 VIA, and of brain derived growth factor (BDNF) critical for neuronal integrity,58 while it increases AA-metabolizing cPLA2 IVA, sPLA2 IIA and COX-2 activities. In contrast, reducing dietary LA below the “adequate” level reduces brain AA concentration, kinetics and enzyme expression, while reciprocally increasing corresponding DHA parameters.59

While data are controversial with regard to dietary intervention in the clinic, a cross-national study did identify a significant relation between greater DHA-containing seafood consumption and lower prevalence rates of BD.60 Also, a review of clinical trials reported that increased dietary n-3 PUFA in combination with standard treatment improved bipolar depression, even taking into account sample bias.61 In the future, one might maximize effects of dietary intervention by combining dietary n-3 PUFA supplementation with reduced dietary n-6 PUFA, which when compared to a standard diet was effective in a phase III trial in patients with migraine.62 Migraine occurs in 30% of BD patients.63

4. Arachidonic Acid Cascade and Bipolar Disorder Brain

BD genetics provide minimal evidence if any for the AA cascade hypothesis. A significant association with a calcium-independent iPLA2β (VIA) rs3788533 SNP (PLA2G6) in BD I has been reported, and the activity of this enzyme was elevated in plasma of patients with a history of psychoses.64 However, iPLA2 VIA is largely selective for DHA hydrolysis.12a Association with the gene for sPLA2 on chromosome 12q23-q24.1 also has been noted.65

On the other hand, there is abundant evidence for general and central inflammation as major contributing factors to BD, and inflammation is classically associated with upregulated AA metabolism and treated with nonsteroidal anti-inflammatory drugs like aspirin and Celebrex (see above).66 Disturbed circulating concentrations of proinflammatory interleukins, TNF-α, and other cytokines have been reported in BD patients, related to the manic or depressive phase of the disease as well as to response to therapy.66

Studies on the post-mortem BD brain do show upregulated AA cascade markers, thus a potential target of the mood stabilizers, accompanied by evidence of neuroinflammation, excitotoxicity, apoptosis, and synaptic loss. The changes are not entirely specific to BD, as similar changes were demonstrated in schizophrenic and Alzheimer’s disease brain tissue by the same investigators.67 The changes may represent common late-stage neurodegeneration in each disease, consistent with evidence of biotype overlap (see above).3

In post-mortem BD compared with control prefrontal cortex, mean protein and mRNA levels of AA-selective cPLA2 IVA, sPLA2 IIA, COX-2, and membrane prostaglandin E synthase (mPGES) were significantly elevated, while levels of COX-1 and cytosolic cPGES were reduced. mRNA and protein levels of DHA-selective iPLA2 VIA, of 5-, 12-, and 15-LOX, of thromboxane synthase (TXS) and of CYP450 did not differ from control values.67a In relation to the changes, BD compared with control cortex demonstrated decreased expression of antiapoptotic factors B-cell lymphoma (Bcl)-2 and BDNF, but increased expression of pro-apoptotic Bcl-2-associated X protein (BAX), Bcl-2 associated death promoter (BAD), and active caspase-3 and -9.68 Higher levels also were noted of interleukin (IL)-1β, the IL-1 receptor (IL-1R), and of astrocyte and microglia activation markers, glial fibrillary acidic protein (GFAP), inducible nitric oxide synthase (iNOS), c-fos, and CD11b.69 Significant synaptic loss, shown as reduced levels of presynaptic synaptophysin and postsynaptic dendritic spine drebrin, might explain the reported cognitive decline in BD.68 Indeed, synaptic loss often is evident in conditions of neuroinflammation and excitotoxicity, associated with an upregulated AA cascade.67b,70

Despite the many similar changes, some differences between changes in post-mortem BD and schizophrenia brain are noteworthy. The dopamine reuptake transporter (DAT) is downregulated in post-mortem schizophrenia and BD frontal cortex,71 consistent with responsiveness of both diseases to atypical antipsychotics. However, therapeutic responsiveness of BD but not schizophrenia to lithium and the other mood stabilizers that block the AA signal to NMDA in rat brain may relate to an increased glutamate signaling in BD but not schizophrenia brain. More than 90% of released glutamate is cleared from the synaptic cleft by the excitatory amino acid (reuptake) transporter (EAAT)2.72 EAAT2 is downregulated in the BD cortex, as are NMDA receptor (NR) subunits NR1 and NR2, consistent with increased glutamatergic activity (see above).9,69 EAAT3 and EAAT4 are unchanged while EAAT1 is elevated in BD.71 In schizophrenic cortex, EAAT1, EAAT3 and EAAT4 are each upregulated while EAAT2 expression is unchanged,71 consistent with decreased NMDA function in schizophrenia.73 Decreased binding of cholinergic muscarinic receptors in BD74 is consistent with lithium having an effect by upregulating the cholinergic muscarinic AA signal (Table 1).

5. Discussion

BD represents a complex set of symptoms that evolve over time, incompletely characterized pathophysiology, with multiple contributing genetic factors of low effects, and without an agreed-on behavioral animal model. There appear to be two biostages, an initial one involving imbalance in neurotransmission–hyperglutamatergic and hyperdopaminergic transmission, reduced cholinergic and altered serotonergic transmission, and a later appearing stage with superimposed neurodegenerative components associated with cognitive decline, symptom worsening and brain atrophy, which overlaps with biotypes of other neuropsychiatric disorders.

In this section, I review the AA hypothesis for the action of lithium, and show that the hypothesis extrapolates to the actions of the other FDA-approved mood stabilizers carbamazepine, valproate and lamotrigine, but not to topiramate or gabapentin, each of which failed phase III trials in BD patients.

The AA cascade hypothesis proposes that lithium and the other mood stabilizers downregulate brain AA metabolism at different entry points (Figure 1). This suggested target of mood stabilizers is consistent with studies showing upregulated cascade markers in post-mortem BD prefrontal cortex, and can be tested further in patients using PET to image brain AA metabolism. The AA cascade hypothesis also may explain high switching rates of BD depression to mania caused by the antidepressants fluoxetine and imipramine, and some treatment effects of olanzapine and clozapine. Further, it is supported by a pharmacoepidemiological study showing that chronic low dose aspirin reduced morbidity of patients taking lithium, and by evidence that high n-3 and/or low n-6 PUFA diets are helpful in BD and migraine patients.

Lithium and the other mood stabilizers have proven effective in BD, but they do not always work, and they may work in some individuals based on genetic specificity.75 Each also has unwanted side effects, leading to incomplete compliance and polypharmacy.1 Based on the AA cascade hypothesis and the suggested targets of the current drugs at different sites within the cascade, future research should aim to develop less toxic and more effective mood stabilizers. This Review suggests that this aim might be promoted by prescreening potential drug candidates for their ability to reduce brain AA turnover and metabolic markers in unanesthetized rodents, using our established kinetic methods and model.1a,18 Our data also suggest that the animal model for drug screening need not be a behavioral model, but rather might be the intact unanesthetized rodent on which brain lipid metabolic measurements can be performed. In the future, combining mood stabilizers with low dose aspirin or dietary intervention (high n-3/low n-6 PUFAs) may provide synergistic amelioration of BD symptoms and reduce disease progression.

Acknowledgments

I am indebted to Drs. Chuck T. Chen and Erika F. H. Saunders as well as Mr. Christopher T. Primiani and Ms. Veronica H. Ryan for their very helpful comments and suggestions.

Glossary

Abbreviations

AA

arachidonic acid

AMPA

alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

AP

activator protein

BAD

Bcl-2 associated death promoter

BAX

Bcl-2-associated X protein

BD

bipolar disorder

BDNF

brain derived neurotrophic factor

Bcl-2

B-cell lymphoma-2

COX

cyclooxygenase

CSF

cerebrospinal fluid

CYP450

cytochrome P450 epoxygenase

DAT

dopamine reuptake transporter

DHA

docosahexaenoic acid

DOI

(±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane hydrochloride

EAAT

excitatory amino acid (reuptake) transporter

GFAP

glial fibrillary acidic protein

GRK

G-protein receptor kinase

GSK

glycogen synthase kinase

5-HT

5-hydroxytryptamine (serotonin)

IL-1

interleukin-1

IL-1R

interleukin receptor

IMPase

inositol monophosphatase

iNOS

inducible nitric oxide synthase

LPCAT

lysophospholipid acyltransferase

LOX

lipoxygenase

LTB4

leukotriene B4

m-PGES-2

membrane PGE synthase-2

NMDA

N-methyl-d-aspartic acid

NF-κB

nuclear factor kappa B

NR

NMDA receptor

PET

positron emission tomography

PGE2

prostaglandin E2

PLA2

phospholipase A2

cPLA2

cytosolic PLA2

iPLA2

calcium independent PLA2

sPLA2

secretory PLA2

PUFA

polyunsaturated fatty acid

sn

stereospecifically numbered

SSRI

selective serotonin reuptake inhibitor

TNFα

tumor necrosis factor alpha

TXS

thromboxane synthase

This work was entirely supported by the Intramural Program of the National Institute on Aging.

The authors declare no competing financial interest.

Funding Statement

National Institutes of Health, United States

References

  1. a Rapoport S. I.; Basselin M.; Kim H. W.; Rao J. S. (2009) Bipolar disorder and mechanisms of action of mood stabilizers. Brain Res. Rev. 61(2), 185–209. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Goodwin F. K., and Jamison K. R. (2007) Manic-Depressive Illness: Bipolar Disorders and Recurrent Depression, 2nd ed., p 1262, Oxford University Press, New York. [Google Scholar]
  2. a Post R. M.; Jimerson D. C.; Bunney W. E. Jr.; Goodwin F. K. (1980) Dopamine and mania: behavioral and biochemical effects of the dopamine receptor blocker pimozide. Psychopharmacology (Berlin, Ger.) 67(3), 297–305. [DOI] [PubMed] [Google Scholar]; b Janowsky D. S., and Overstreet D. H. (1995) The role of acetylcholine mechanisms in mood disorders. In Psychopharmacology. The Fourth Generation of Progress (Bloom F. E., and Kupfer D. J., Eds.), pp 945–956, Raven, New York. [Google Scholar]
  3. Tamminga C. A. (2013) Validating psychosis biotypes. ACNP J. 38(S2), 22.4. [Google Scholar]
  4. Baum A. E.; Akula N.; Cabanero M.; Cardona I.; Corona W.; Klemens B.; Schulze T. G.; Cichon S.; Rietschel M.; Nothen M. M.; Georgi A.; Schumacher J.; Schwarz M.; Abou Jamra R.; Hofels S.; Propping P.; Satagopan J.; Detera-Wadleigh S. D.; Hardy J.; McMahon F. J. (2008) A genome-wide association study implicates diacylglycerol kinase eta (DGKH) and several other genes in the etiology of bipolar disorder. Mol. Psychiatry 13(2), 197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Cade J. F. J. (1949) Lithium salts in the treatment of psychotic excitement. Med. J. Aust. 36, 349–352. [DOI] [PubMed] [Google Scholar]
  6. Berridge M. J. (1989) The Albert Lasker Medical Awards. Inositol trisphosphate, calcium, lithium, and cell signaling. JAMA, J. Am. Med. Assoc. 262(13), 1834–1841. [PubMed] [Google Scholar]
  7. Gould T. D.; Chen G.; Manji H. K. (2004) In vivo evidence in the brain for lithium inhibition of glycogen synthase kinase-3. Neuropsychopharmacology 29(1), 32–38. [DOI] [PubMed] [Google Scholar]
  8. Yildiz A.; Guleryuz S.; Ankerst D. P.; Ongur D.; Renshaw P. F. (2008) Protein kinase C inhibition in the treatment of mania: a double-blind, placebo-controlled trial of tamoxifen. Arch. Gen. Psychiatry 65(3), 255–263. [DOI] [PubMed] [Google Scholar]
  9. Zarate C. A. Jr.; Brutsche N. E.; Ibrahim L.; Franco-Chaves J.; Diazgranados N.; Cravchik A.; Selter J.; Marquardt C. A.; Liberty V.; Luckenbaugh D. A. (2012) Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol. Psychiatry 71(11), 939–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Rapoport S. I. (2008) Brain arachidonic and docosahexaenoic acid cascades are selectively altered by drugs, diet and disease. Prostaglandins, Leukotrienes, Essent. Fatty Acids 79(3–5), 153–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. a Rao J. S.; Lee H. J.; Rapoport S. I.; Bazinet R. P. (2008) Mode of action of mood stabilizers: is the arachidonic acid cascade a common target?. Mol. Psychiatry 13(6), 585–596. [DOI] [PubMed] [Google Scholar]; b Rapoport S. I.; Bosetti F. (2002) Do lithium and anticonvulsants target the brain arachidonic acid cascade in bipolar disorder?. Arch. Gen. Psychiatry 59(7), 592–596. [DOI] [PubMed] [Google Scholar]
  12. a Six D. A.; Dennis E. A. (2000) The expanding superfamily of phospholipase A(2) enzymes: classification and characterization. Biochim. Biophys. Acta 1488(1–2), 1–19. [DOI] [PubMed] [Google Scholar]; b Soupene E.; Kuypers F. A. (2008) Mammalian long-chain acyl-CoA synthetases. Experimental biology and medicine 233(5), 507–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Ryan V. H.; Primiani C. T.; Rao J. S.; Ahn K.; Rapoport S. I.; Blanchard H.. Coordination of gene expression of arachidonic and docosahexaenoic acid cascade enzymes during human brain development and aging. PloS One 2014, in press [DOI] [PMC free article] [PubMed]
  14. Felder C. C. (1995) Muscarinic acetylcholine receptors: signal transduction through multiple effectors. FASEB J. 9, 619–625. [PubMed] [Google Scholar]
  15. Vial D.; Piomelli D. (1995) Dopamine D2 receptors potentiate arachidonate release via activation of cytosolic, arachidonate-specific phospholipase A2. J. Neurochem 64(6), 2765–2772. [DOI] [PubMed] [Google Scholar]
  16. Berg K. A.; Maayani S.; Goldfarb J.; Scaramellini C.; Leff P.; Clarke W. P. (1998) Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus. Mol. Pharmacol. 54(1), 94–104. [PubMed] [Google Scholar]
  17. Ramadan E.; Rosa A. O.; Chang L.; Chen M.; Rapoport S. I.; Basselin M. (2010) Extracellular-derived calcium does not initiate in vivo neurotransmission involving docosahexaenoic acid. J. Lipid Res. 51(8), 2334–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Robinson P. J.; Noronha J.; DeGeorge J. J.; Freed L. M.; Nariai T.; Rapoport S. I. (1992) 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. 17(3), 187–214. [DOI] [PubMed] [Google Scholar]
  19. Yamashita A.; Sugiura T.; Waku K. (1997) Acyltransferases and transacylases involved in fatty acid remodeling of phospholipids and metabolism of bioactive lipids in mammalian cells. J. Biochem. 122(1), 1–16. [DOI] [PubMed] [Google Scholar]
  20. Dinarello C. A. (2002) The IL-1 family and inflammatory diseases. Clin. Exp. Rheumatol. 20(5 Suppl 27), S1–13. [PubMed] [Google Scholar]
  21. Chang Y. C.; Kim H. W.; Rapoport S. I.; Rao J. S. (2008) Chronic NMDA administration increases neuroinflammatory markers in rat frontal cortex: cross-talk between excitotoxicity and neuroinflammation. Neurochem. Res. 33(11), 2318–2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Kim H. W.; Chang Y. C.; Chen M.; Rapoport S. I.; Rao J. S. (2009) Chronic NMDA administration to rats increases brain pro-apoptotic factors while decreasing anti-Apoptotic factors and causes cell death. BMC Neurosci. 10, 123. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  23. DeMar J. C. Jr.; Lee H. J.; Ma K.; Chang L.; Bell J. M.; Rapoport S. I.; Bazinet R. P. (2006) Brain elongation of linoleic acid is a negligible source of the arachidonate in brain phospholipids of adult rats. Biochim. Biophys. Acta 1761(9), 1050–1059. [DOI] [PubMed] [Google Scholar]
  24. a Basselin M.; Chang L.; Bell J. M.; Rapoport S. I. (2006) Chronic lithium chloride administration attenuates brain NMDA receptor-initiated signaling via arachidonic acid in unanesthetized rats. Neuropsychopharmacology 31(8), 1659–1674. [DOI] [PubMed] [Google Scholar]; b Basselin M.; Chang L.; Chen M.; Bell J. M.; Rapoport S. I. (2008) Chronic administration of valproic acid reduces brain NMDA signaling via arachidonic acid in unanesthetized rats. Neurochem. Res. 33(11), 2229–2240. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Ramadan E.; Basselin M.; Rao J. S.; Chang L.; Chen M.; Ma K.; Rapoport S. I. (2012) Lamotrigine blocks NMDA receptor-initiated arachidonic acid signalling in rat brain: implications for its efficacy in bipolar disorder. Int. J. Neuropsychopharmacol. 15(7), 931–943. [DOI] [PMC free article] [PubMed] [Google Scholar]; d Basselin M.; Villacreses N. E.; Chen M.; Bell J. M.; Rapoport S. I. (2007) Chronic carbamazepine administration reduces N-methyl-d-aspartate receptor-initiated signaling via arachidonic acid in rat brain. Biol. Psychiatry 62(8), 934–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hough C. J.; Irwin R. P.; Gao X. M.; Rogawski M. A.; Chuang D. M. (1996) Carbamazepine inhibition of N-methyl-d-aspartate-evoked calcium influx in rat cerebellar granule cells. J. Pharmacol. Exp. Ther. 276(1), 143–149. [PubMed] [Google Scholar]
  26. a Basselin M.; Chang L.; Bell J. M.; Rapoport S. I. (2005) Chronic lithium chloride administration to unanesthetized rats attenuates brain dopamine D2-like receptor-initiated signaling via arachidonic acid. Neuropsychopharmacology 30(6), 1064–1075. [DOI] [PubMed] [Google Scholar]; b Basselin M.; Chang L.; Chen M.; Bell J. M.; Rapoport S. I. (2008) Chronic carbamazepine administration attenuates dopamine D2-like receptor-initiated signaling via arachidonic acid in rat brain. Neurochem. Res. 33(7), 1373–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Ramadan E.; Basselin M.; Taha A. Y.; Cheon Y.; Chang L.; Chen M.; Rapoport S. I. (2011) Chronic valproate treatment blocks D2-like receptor-mediated brain signaling via arachidonic acid in rats. Neuropharmacology 61(8), 1256–1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Basselin M.; Chang L.; Seemann R.; Bell J. M.; Rapoport S. I. (2005) Chronic lithium administration to rats selectively modifies 5-HT2A/2C receptor-mediated brain signaling via arachidonic acid. Neuropsychopharmacology 30(3), 461–472. [DOI] [PubMed] [Google Scholar]
  28. Basselin M.; Chang L.; Seemann R.; Bell J. M.; Rapoport S. I. (2003) Chronic lithium administration potentiates brain arachidonic acid signaling at rest and during cholinergic activation in awake rats. J. Neurochem 85(6), 1553–1562. [DOI] [PubMed] [Google Scholar]
  29. Basselin M.; Chang L.; Rapoport S. I. (2006) Chronic lithium chloride administration to rats elevates glucose metabolism in wide areas of brain, while potentiating negative effects on metabolism of dopamine D(2)-like receptor stimulation. Psychopharmacology (Berlin, Ger.) 187(3), 303–311. [DOI] [PubMed] [Google Scholar]
  30. Ertley R. N.; Bazinet R. P.; Lee H. J.; Rapoport S. I.; Rao J. S. (2007) Chronic treatment with mood stabilizers increases membrane GRK3 in rat frontal cortex. Biol. Psychiatry 61(2), 246–249. [DOI] [PubMed] [Google Scholar]
  31. Sun G. Y.; MacQuarrie R. A. (1989) Deacylation-reacylation of arachidonoyl groups in cerebral phospholipids. Ann. N.Y. Acad. Sci. 559, 37–55. [DOI] [PubMed] [Google Scholar]
  32. a Bazinet R. P.; Rao J. S.; Chang L.; Rapoport S. I.; Lee H. J. (2006) 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 59(5), 401–407. [DOI] [PubMed] [Google Scholar]; b Lee H. J.; Rao J. S.; Chang L.; Rapoport S. I.; Bazinet R. P. (2007) 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 193(4), 467–474. [DOI] [PubMed] [Google Scholar]; c Chang M. C.; Contreras M. A.; Rosenberger T. A.; Rintala J. J.; Bell J. M.; Rapoport S. I. (2001) Chronic valproate treatment decreases the in vivo turnover of arachidonic acid in brain phospholipids: a possible common effect of mood stabilizers. J. Neurochem. 77(3), 796–803. [DOI] [PubMed] [Google Scholar]; d Chang M. C.; Grange E.; Rabin O.; Bell J. M.; Allen D. D.; Rapoport S. I. (1996) Lithium decreases turnover of arachidonate in several brain phospholipids. Neurosci. Lett. 220(3), 171–174. [DOI] [PubMed] [Google Scholar]
  33. a Shimshoni J. A.; Basselin M.; Li L. O.; Coleman R. A.; Rapoport S. I.; Modi H. R. (2011) Valproate uncompetitively inhibits arachidonic acid acylation by rat acyl-CoA synthetase 4: relevance to valproate’s efficacy against bipolar disorder. Biochim. Biophys. Acta 1811(3), 163–169. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Bazinet R. P.; Weis M. T.; Rapoport S. I.; Rosenberger T. A. (2006) 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 (Berlin, Ger.) 184(1), 122–129. [DOI] [PubMed] [Google Scholar]
  34. Bersudsky Y.; Applebaum J.; Gaiduk Y.; Sharony L.; Mishory A.; Podberezsky A.; Agam G.; Belmaker R. H. (2010) Valnoctamide as a valproate substitute with low teratogenic potential in mania: a double-blind, controlled, add-on clinical trial. Bipolar Disord. 12(4), 376–382. [DOI] [PubMed] [Google Scholar]
  35. a Modi H. R.; Basselin M.; Rapoport S. I.. Valnoctamide, a non-teratogenic amide derivative of valproic acid, inhibits arachidonic acid activation in vitro by recombinant acyl-CoA synthetase-4. Bipolar Disord. 2014, in press [DOI] [PMC free article] [PubMed]; b Modi H. R.; Basselin M.; Taha A. Y.; Li L. O.; Coleman R. A.; Bialer M.; Rapoport S. I. (2013) Propylisopropylacetic acid (PIA), a constitutional isomer of valproic acid, uncompetitively inhibits arachidonic acid acylation by rat acyl-CoA synthetase 4: a potential drug for bipolar disorder. Biochim. Biophys. Acta 1831(4), 880–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Kaufmann W. E.; Worley P. F.; Pegg J.; Bremer M.; Isakson P. (1996) COX-2, a synaptically induced enzyme, is expressed by excitatory neurons at postsynaptic sites in rat cerebral cortex. Proc. Natl. Acad. Sci. U.S.A. 93(6), 2317–2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. a Bosetti F.; Bell J. M.; Manickam P. (2005) Microarray analysis of rat brain gene expression after chronic administration of sodium valproate. Brain Res. Bull. 65(4), 331–338. [DOI] [PubMed] [Google Scholar]; b Rao J. S.; Rapoport S. I.; Bosetti F. (2005) 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 30(11), 2006–2013. [DOI] [PubMed] [Google Scholar]
  38. Jope R. S.; Song L. (1997) AP-1 and NF-KappaB stimulated by carbachol in human neuroblastoma SH-SY5Y cells are differentially sensitive to inhibition by lithium. Brain Res. Mol. Brain Res. 50, 171–180. [DOI] [PubMed] [Google Scholar]
  39. Weerasinghe G. R.; Rapoport S. I.; Bosetti F. (2004) 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. 63(6), 485–489. [DOI] [PubMed] [Google Scholar]
  40. Choi S. H.; Langenbach R.; Bosetti F. (2006) Cyclooxygenase-1 and -2 enzymes differentially regulate the brain upstream NF-kappaB pathway and downstream enzymes involved in prostaglandin biosynthesis. J. Neurochem. 98(3), 801–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Reese E. A.; Cheon Y.; Ramadan E.; Kim H. W.; Chang L.; Rao J. S.; Rapoport S. I.; Taha A. Y. (2012) Gabapentin’s minimal action on markers of rat brain arachidonic acid metabolism agrees with its inefficacy against bipolar disorder. Prostaglandins, Leukotrienes, Essent. Fatty Acids 87(2–3), 71–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. a Ghelardoni S.; Tomita Y. A.; Bell J. M.; Rapoport S. I.; Bosetti F. (2004) Chronic carbamazepine selectively downregulates cytosolic phospholipase A2 expression and cyclooxygenase activity in rat brain. Biol. Psychiatry 56(4), 248–254. [DOI] [PubMed] [Google Scholar]; b Bosetti F.; Weerasinghe G. R.; Rosenberger T. A.; Rapoport S. I. (2003) Valproic acid down-regulates the conversion of arachidonic acid to eicosanoids via cyclooxygenase-1 and −2 in rat brain. J. Neurochem. 85(3), 690–696. [DOI] [PubMed] [Google Scholar]
  43. a Serhan C. N.; Gotlinger K.; Hong S.; Arita M. (2004) Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their aspirin-triggered endogenous epimers: an overview of their protective roles in catabasis. Prostaglandins Other Lipid Mediators 73(3–4), 155–172. [DOI] [PubMed] [Google Scholar]; b Basselin M.; Kim H. W.; Chen M.; Ma K.; Rapoport S. I.; Murphy R. C.; Farias S. E. (2010) Lithium modifies brain arachidonic and docosahexaenoic metabolism in rat lipopolysaccharide model of neuroinflammation. J. Lipid Res. 51(5), 1049–1056. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  44. Stolk P.; Souverein P. C.; Wilting I.; Leufkens H. G.; Klein D. F.; Rapoport S. I.; Heerdink E. R. (2010) Is aspirin useful in patients on lithium? A pharmacoepidemiological study related to bipolar disorder. Prostaglandins, Leukotrienes, Essent. Fatty Acids 82(1), 9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Lee H. J.; Ghelardoni S.; Chang L.; Bosetti F.; Rapoport S. I.; Bazinet R. P. (2005) Topiramate does not alter the kinetics of arachidonic or docosahexaenoic acid in brain phospholipids of the unanesthetized rat. Neurochem. Res. 30(5), 677–683. [DOI] [PubMed] [Google Scholar]
  46. Post R. M.; Altshuler L. L.; Leverich G. S.; Frye M. A.; Nolen W. A.; Kupka R. W.; Suppes T.; McElroy S.; Keck P. E.; Denicoff K. D.; Grunze H.; Walden J.; Kitchen C. M.; Mintz J. (2006) Mood switch in bipolar depression: comparison of adjunctive venlafaxine, bupropion and sertraline. Br. J. Psychiatry 189, 124–131. [DOI] [PubMed] [Google Scholar]
  47. a Lee H. J.; Rao J. S.; Ertley R. N.; Chang L.; Rapoport S. I.; Bazinet R. P. (2007) Chronic fluoxetine increases cytosolic phospholipase A(2) activity and arachidonic acid turnover in brain phospholipids of the unanesthetized rat. Psychopharmacology (Berlin, Ger.) 190(1), 103–115. [DOI] [PubMed] [Google Scholar]; b Lee H. J.; Rao J. S.; Chang L.; Rapoport S. I.; Kim H. W. (2010) Chronic imipramine but not bupropion increases arachidonic acid signaling in rat brain: is this related to ’switching’ in bipolar disorder?. Mol. Psychiatry 15(6), 602–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. a Esposito G.; Giovacchini G.; Liow J. S.; Bhattacharjee A. K.; Greenstein D.; Schapiro M.; Hallett M.; Herscovitch P.; Eckelman W. C.; Carson R. E.; Rapoport S. I. (2008) Imaging neuroinflammation in Alzheimer’s disease with radiolabeled arachidonic acid and PET. J. Nucl. Med. 49(9), 1414–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Pichika R.; Taha A. Y.; Gao F.; Kotta K.; Cheon Y.; Chang L.; Kiesewetter D.; Rapoport S. I.; Eckelman W. C. (2012) The synthesis and in vivo pharmacokinetics of fluorinated arachidonic acid: implications for imaging neuroinflammation. J. Nucl. Med. 53(9), 1383–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Rapoport S. I.; Rao J. S.; Igarashi M. (2007) Brain metabolism of nutritionally essential polyunsaturated fatty acids depends on both the diet and the liver. Prostaglandins, Leukotrienes, Essent. Fatty Acids 77(5–6), 251–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Kapur S.; Seeman P. (2001) Does fast dissociation from the dopamine d(2) receptor explain the action of atypical antipsychotics?: A new hypothesis. Am. J. Psychiatry 158(3), 360–369. [DOI] [PubMed] [Google Scholar]
  51. a Cheon Y.; Park J. Y.; Modi H. R.; Kim H. W.; Lee H. J.; Chang L.; Rao J. S.; Rapoport S. I. (2011) Chronic olanzapine treatment decreases arachidonic acid turnover and prostaglandin E(2) concentration in rat brain. J. Neurochem. 119(2), 364–376. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Modi H. R.; Taha A. Y.; Kim H. W.; Chang L.; Rapoport S. I.; Cheon Y. (2013) Chronic clozapine reduces rat brain arachidonic acid metabolism by reducing plasma arachidonic acid availability. J. Neurochem. 124(3), 376–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Ragheb M. A. (1987) Aspirin does not significantly affect patients’ serum lithium levels. J. Clin. Psychiatry 48(10), 425. [PubMed] [Google Scholar]
  53. Blanchard H., Taha A. Y., Rapoport S. I., and Yuan Z. X.. Low-dose aspirin prevents increments in brain PGE2, 15-epi-lipoxin A4 and 8-isoprostane concentrations in 9 month-old HIV-1 transgenic rats. Submitted for publication. [DOI] [PMC free article] [PubMed]
  54. a Savitz J.; Preskorn S.; Teague T. K.; Drevets D.; Yates W.; Drevets W. (2012) Minocycline and aspirin in the treatment of bipolar depression: a protocol for a proof-of-concept, randomised, double-blind, placebo-controlled, 2 × 2 clinical trial. BMJ Open 2(1), e000643. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Soares J. Clinical Trial: N-Acetyl Cysteine and Aspirin as an Adjunctive Treatment for Bipolar Disorder (SMRI-Bipolar): Clinical Trial.gov identifier NCT01797575, http://www.clinicaltrials.gov/ct2/show/NCT01797575.
  55. Ketterer M. W.; Brymer J.; Rhoads K.; Kraft P.; Lovallo W. R. (1996) Is aspirin, as used for antithrombosis, an emotion-modulating agent?. J. Psychosom. Res. 40(1), 53–58. [DOI] [PubMed] [Google Scholar]
  56. Dembo G.; Park S. B.; Kharasch E. D. (2005) Central nervous system concentrations of cyclooxygenase-2 inhibitors in humans. Anesthesiology 102(2), 409–415. [DOI] [PubMed] [Google Scholar]
  57. a Nery F. G.; Monkul E. S.; Hatch J. P.; Fonseca M.; Zunta-Soares G. B.; Frey B. N.; Bowden C. L.; Soares J. C. (2008) Celecoxib as an adjunct in the treatment of depressive or mixed episodes of bipolar disorder: a double-blind, randomized, placebo-controlled study. Hum. Psychopharmacol. 23(2), 87–94. [DOI] [PubMed] [Google Scholar]; b Müller N.; Schwarz M. J.; Dehning S.; Douhe A.; Cerovecki A.; Goldstein-Muller B.; Spellmann I.; Hetzel G.; Maino K.; Kleindienst N.; Möller H. J.; Arolt V.; Riedel M. (2006) The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Mol. Psychiatry 11, 680–684. [DOI] [PubMed] [Google Scholar]
  58. Kim H. W.; Rao J. S.; Rapoport S. I.; Igarashi M. (2011) Regulation of rat brain polyunsaturated fatty acid (PUFA) metabolism during graded dietary n-3 PUFA deprivation. Prostaglandins, Leukotrienes, Essent. Fatty Acids 85(6), 361–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. a Igarashi M.; Gao F.; Kim H. W.; Ma K.; Bell J. M.; Rapoport S. I. (2009) Dietary n-6 PUFA deprivation for 15 weeks reduces arachidonic acid concentrations while increasing n-3 PUFA concentrations in organs of post-weaning male rats. Biochim. Biophys. Acta 1791(2), 132–139. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Kim H. W.; Rao J. S.; Rapoport S. I.; Igarashi M. (2011) Dietary n-6 PUFA deprivation downregulates arachidonate but upregulates docosahexaenoate metabolizing enzymes in rat brain. Biochim. Biophys. Acta 1811(2), 111–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Noaghiul S.; Hibbeln J. R. (2003) Cross-national comparisons of seafood consumption and rates of bipolar disorders. Am. J. Psychiatry 160(12), 2222–2227. [DOI] [PubMed] [Google Scholar]
  61. Sarris J.; Mischoulon D.; Schweitzer I. (2012) Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J. Clin. Psychiatry 73(1), 81–86. [DOI] [PubMed] [Google Scholar]
  62. Ramsden C. E.; Faurot K. R.; Zamora D.; Suchindran C. M.; Macintosh B. A.; Gaylord S.; Ringel A.; Hibbeln J. R.; Feldstein A. E.; Mori T. A.; Barden A.; Lynch C.; Coble R.; Mas E.; Palsson O.; Barrow D. A.; Mann J. D. (2013) Targeted alteration of dietary n-3 and n-6 fatty acids for the treatment of chronic headaches: a randomized trial. Pain 154(11), 2441–2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Saunders E. F. H.; Kamali M.; Gelenberg A. J.; McInnis M. (2012) Migraine Comorbidity Worsens Course of Illness in a Longitudinal Study of Bipolar Disorder. Neuropsychopharmacology 38, W135. [Google Scholar]
  64. Xu C.; Warsh J. J.; Wang K. S.; Mao C. X.; Kennedy J. L. (2013) Association of the iPLA2beta gene with bipolar disorder and assessment of its interaction with TRPM2 gene polymorphisms. Psychiatr. Genet. 23(2), 86–89. [DOI] [PubMed] [Google Scholar]
  65. Meira-Lima I.; Jardim D.; Junqueira R.; Ikenaga E.; Vallada H. (2003) Allelic association study between phospholipase A2 genes and bipolar affective disorder. Bipolar Disord. 5(4), 295–299. [DOI] [PubMed] [Google Scholar]
  66. Goldstein B. I.; Kemp D. E.; Soczynska J. K.; McIntyre R. S. (2009) Inflammation and the phenomenology, pathophysiology, comorbidity, and treatment of bipolar disorder: a systematic review of the literature. J. Clin. Psychiatry 70(8), 1078–1090. [DOI] [PubMed] [Google Scholar]
  67. a Kim H. W.; Rapoport S. I.; Rao J. S. (2011) Altered arachidonic acid cascade enzymes in postmortem brain from bipolar disorder patients. Mol. Psychiatry 16(4), 419–428. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Rao J. S.; Kellom M.; Kim H. W.; Rapoport S. I.; Reese E. A. (2012) Neuroinflammation and synaptic loss. Neurochem. Res. 37(5), 903–910. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Rao J. S.; Kim H. W.; Harry G. J.; Rapoport S. I.; Reese E. A. (2013) Increased neuroinflammatory and arachidonic acid cascade markers, and reduced synaptic proteins, in the postmortem frontal cortex from schizophrenia patients. Schizophr. Res. 147(1), 24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]; d Rao J. S.; Rapoport S. I.; Kim H. W. (2011) Altered neuroinflammatory, arachidonic acid cascade and synaptic markers in postmortem Alzheimer’s disease brain. Transl. Psychiatry 1, e31. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  68. Kim H. W.; Rapoport S. I.; Rao J. S. (2010) Altered expression of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiol. Dis. 37(3), 596–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Rao J. S.; Harry G. J.; Rapoport S. I.; Kim H. W. (2010) Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol. Psychiatry 15(4), 384–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hatanpaa K.; Isaacs K. R.; Shirao T.; Brady D. R.; Rapoport S. I. (1999) Loss of proteins regulating synaptic plasticity in normal aging of the human brain and in Alzheimer disease. J. Neuropathol. Exp. Neurol. 58(6), 637–643. [DOI] [PubMed] [Google Scholar]
  71. Rao J. S.; Kellom M.; Reese E. A.; Rapoport S. I.; Kim H. W. (2012) Dysregulated glutamate and dopamine transporters in postmortem frontal cortex from bipolar and schizophrenic patients. J. Affective Disord. 136(1–2), 63–71. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  72. Bunch L.; Erichsen M. N.; Jensen A. A. (2009) Excitatory amino acid transporters as potential drug targets. Expert Opin. Ther. Targets 13(6), 719–731. [DOI] [PubMed] [Google Scholar]
  73. Olney J. W.; Farber N. B. (1995) Glutamate receptor dysfunction and schizophrenia. Arch. of Gen. Psychiatry 52(12), 998–1007. [DOI] [PubMed] [Google Scholar]
  74. Gibbons A. S.; Scarr E.; McLean C.; Sundram S.; Dean B. (2009) Decreased muscarinic receptor binding in the frontal cortex of bipolar disorder and major depressive disorder subjects. J. Affective Disord. 116(3), 184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. McCarthy M. J.; Leckband S. G.; Kelsoe J. R. (2010) Pharmacogenetics of lithium response in bipolar disorder. Pharmacogenomics 11(10), 1439–1465. [DOI] [PubMed] [Google Scholar]

Articles from ACS Chemical Neuroscience are provided here courtesy of American Chemical Society

RESOURCES