Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Molecular Psychiatry (1999) 4, 117–128

 1999 Stockton Press All rights reserved 1359–4184/99 $12.00

MECHANISMS OF DRUG ACTION

Anti-bipolar therapy: mechanism of action of lithium


RS Jope
Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294,
USA

This review introduces the concepts that multiple actions of lithium are critical for its thera-
peutic effect, and that these complex effects stabilize neuronal activities, support neural plas-
ticity, and provide neuroprotection. Three interacting systems appear most critical. (i) Modu-
lation of neurotransmitters by lithium likely readjusts balances between excitatory and
inhibitory activities, and decreased glutamatergic activity may contribute to neuroprotection.
(ii) Lithium modulates signals impacting on the cytoskeleton, a dynamic system contributing
to neural plasticity, at multiple levels, including glycogen synthase kinase-3␤, cyclic AMP-
dependent kinase, and protein kinase C, which may be critical for the neural plasticity involved
in mood recovery and stabilization. (iii) Lithium adjusts signaling activities regulating second
messengers, transcription factors, and gene expression. The outcome of these effects
appears likely to result in limiting the magnitudes of fluctuations in activities, contributing to
a stabilizing influence induced by lithium, and neuroprotective effects may be derived from
its modulation of gene expression.
Keywords: lithium; bipolar disorder; inositol; glutamate; cyclic AMP; AP-1

Recent research suggests that understanding how lith- and depression, as a relatively weak antidepressant,
ium works in the treatment of bipolar disorder requires and as an augmenter of the effects of many antidepress-
a different way of thinking about drug action. The vast ants. These multiple therapeutic effects of lithium
majority of drugs are designed, by evolution or syn- along with the complexities of mood disorders and the
thesis, to interact with single proteins, such as a spe- multiplicity of affected systems inherent in these ill-
cific receptor or enzyme. However, it now seems likely nesses suggest that it is unlikely that all these effects
that multiple sites affected by lithium contribute to its are due to a single biochemical site of action.
mood-stabilizing action. Thus, the therapeutic action This review focuses on advances made in recent
of lithium in bipolar disorder appears not to result years and is organized to follow the steps of neuro-
from an effect at a single target site, but rather as the transmission, beginning with presynaptic actions lead-
culmination of an integrated re-orchestration of a com- ing to receptor-coupled signaling systems which
plex concert of events which effectively adjusts neu- eventually culminate in modulating gene expression.
ronal activity at multiple levels. Numerous effects of Many comprehensive reviews1–15 discuss other aspects
lithium have been identified, and some of these may of bipolar disorder and the use of lithium, and provide
each provide a necessary, but individually insufficient, detailed coverage of important earlier progress in this
component of the therapeutic response. One of the cur- field that set the stage for the recent developments that
rent challenges is to distinguish those critical effects are covered here. Because the therapeutic concen-
from the many known biochemical actions of lithium,
tration of lithium is near 1 mM, coverage focuses on
many of which may have little discernible effect, con-
studies where significant effects were obtained using
tribute to side effects, or lead to toxicity. Another chal-
lithium near this concentration. Two directly associa-
lenge is to integrate the multiple effects of lithium into
ted targets of lithium, signaling systems and gene
a comprehensive picture of how neuronal function is
modulated by lithium. In retrospect, considering the expression, have attracted the greatest amount of
complexities of mood, it perhaps should not be surpris- investigative attention recently, and so are emphas-
ing that mood stabilization by lithium is also a com- ized. However, this emphasis should not detract from
plex process involving multiple actions. It is also other intriguing findings that remain to be investigated
important to keep in mind that lithium has efficacy as more completely. In accordance with the many actions
an antimanic agent, as a prophylactic agent for mania of lithium that are discussed in this review, the author
suggests that it is the integration of multiple effects of
lithium which is necessary to achieve the neurochem-
Correspondence: Dr RS Jope, Department of Psychiatry and ical balance facilitating the complex processes of mood
Behavioral Neurobiology, Sparks Center 1057, University of recovery and stabilization.
Alabama at Birmingham, Birmingham, AL 35294-0017, USA.
E-mail: jope얀uab.edu
Received 17 September 1998; accepted 14 October 1998
Mechanism of action of lithium
RS Jope

118
Neurotransmitter effects lithium was found to effectively inhibit glutamate-
induced neuronal calcium uptake and cell death28
Much of the early research on the actions of lithium
(Figure 1). These discoveries raise the intriguing possi-
focused on modulation of neurotransmitter synthesis,
bility that modulation of excitatory glutamatergic neur-
release, and uptake. Lithium was found to influence to
otransmission constitutes an important therapeutic
some extent virtually every neurotransmitter that was
action of lithium. Furthermore, it was recently shown
investigated, although most research focused on the
that glutamate receptors expressed in oocytes are
catecholamines and acetylcholine.2 This intensive
highly permeable to lithium, raising the possibility that
study of lithium’s presynaptic effects has largely, but
not entirely, been supplanted by other avenues of this may be an important mechanism and site for
research in recent years. This has occurred despite the localized accumulation of lithium.32 Taken together,
fact that many significant effects of lithium were ident- these reports indicate that some types of apoptosis and
ified without consensus conclusions being drawn. excitatory neurodegeneration can be attenuated by lith-
One of the most provocative recent developments in ium. This neuroprotective action may be mediated by
the study of neurotransmitter effects of lithium con- increased glutamate uptake, attenuated increases in
cerns its modulation of glutamatergic neurotransmis- intracellular calcium levels associated with excitatory
sion. Hokin and colleagues found that lithium acutely neurotransmission, and the recent finding that lithium
increased synaptic concentrations of glutamate which, up-regulated the anti-apoptotic protein bcl-2.33,34
upon chronic lithium administration led to an increase Investigations of other neurotransmitters have also
and stabilization in glutamate uptake transporter continued, although not with the intensity of previous
capacity16–20 (Figure 1). They speculated that this could years. In accordance with earlier studies, recent reports
mediate a reduction and stabilization in excitatory neu- continue to find that lithium reduces dopaminergic
rotransmission after lithium treatment. This action activity,35–38 and enhances cholinergic activity39–41 and
may contribute to a relatively large and diverse group the activity of inhibitory GABAergic neurotransmis-
of findings in the literature indicating that lithium has sion.23,42,43 Recent studies also report that lithium
neuroprotective effects, an action of lithium that has alters serotonergic neurotransmission,44–48 but as
been observed most often, but not exclusively, in cer- reviewed previously49 the varied observed effects of
ebellar cells.21–30 For example, chronic lithium admin- lithium on this system remain to be incorporated into
istration was found to block excitotoxicity induced by a cohesive scheme.
glutamate agonists,26–28 although little protection was Thus a clear outcome has not been reached concern-
afforded by lithium from kainate-induced damage.31 ing which, if any, neurotransmitter synthesis, uptake,
Perhaps most remarkable in these studies was the and release processes contribute to the therapeutic
recent finding that lithium administration to rats action of lithium, but the reported effects on cat-
reduced by 56% the size of the ischemic infarct meas- echolamines, acetylcholine, and GABA, as well as the
ured 24 h after occlusion of the left middle cerebral recent findings with glutamate, suggest that these may
artery.29 In a study of its neuroprotective mechanisms, make important, if as yet not completely defined, con-

Figure 1 Lithium modulates glutamatergic neurotransmission. The figure depicts a glutamatergic synaptic area, with the post-
synaptic sites containing N-methyl-d-aspartate (NMDA) receptors enlarged in the lower diagrams. Chronic lithium treatment
can increase the uptake of glutamate, and also can reduce the increase in intracellular calcium subsequent to activation of N-
methyl-d-aspartate receptors. These actions may provide mechanisms for lithium to reduce excitatory neurotransmission and
contribute to lithium’s neuroprotective effects against certain excitatory insults.
Mechanism of action of lithium
RS Jope

119
tributions. The overall consequences of these actions There is little doubt that signals emanating from
may be that lithium readjusts the balance between receptors coupled to the phosphoinositide signal trans-
excitatory and inhibitory activities, as well as the earl- duction system are affected by lithium, but exactly how
ier postulated balance between catecholamines and this occurs remains under intense scrutiny. Sherman
acetylcholine, so that it is the adjusted balances and colleagues1 first found lithium to be a potent
(increased GABA/glutamate and acetylcholine/ inhibitor of inositol monophosphatase, the enzyme
catecholamine activity ratios, in a generalized manner), that converts inositol monophosphates to free inositol.
rather than actions on a single neurotransmitter, that This led to the ingenious proposal,50 known as the
facilitate mood recovery and stabilization. inositol depletion hypothesis, that this action of lith-
ium might result in sequestration of inositol in the form
of inositol monophosphate. It was proposed that this
Signal transduction systems/protein
action may result in the depletion of inositol-contain-
phosphorylation
ing lipids and thereby attenuate receptor-coupled
A concerted effort has been directed towards under- phosphoinositide signaling. However, only marginal
standing how lithium influences signal transduction effects of lithium on phosphoinositide concentrations
systems, especially the phosphoinositide and cyclic have been found.11 Having explored this hypothesis in
AMP signaling systems, which followed earlier investi- exquisite detail, several groups suggested that lithium
gations of lithium on receptors using binding methods may cause not a global depletion of phosphoinositides,
which failed to provide conclusive evidence about but rather highly selective localized depletions.51–56
whether or not lithium altered receptor-binding This modification limits the hypothetical conse-
properties.2,49 Instead of having a single target site quences of lithium exposure to those cells with the
within each of these signaling systems, it appears that lowest endogenous sources of inositol, which may
multiple sites are affected by lithium and that the over- make them most susceptible to inositol depletion,
all outcome has to do with the balances achieved and/or to cells experiencing the greatest stimulation of
between negative and positive modulatory effects of phosphoinositide signaling activity, which would
lithium rather than resulting from an individual site of make them most dependent on inositol recycling to
action15 (Figure 2). maintain signaling activity. An important strength of

Figure 2 Lithium can bimodally modulate signaling systems dependent on the endogenous level of basal and stimulated
activities. (a) Depiction of signaling with normal basal and stimulated activities. (b) Signaling system with depressed basal
activity can be corrected by lithium-induced increases in basal activity. (c) Signaling system with excessive response to a
stimulus can be corrected by lithium-induced inhibition of the maximal response. (d) Signaling system with depressed basal
activity and excessive response to a stimulus can be corrected by lithium-induced increases in basal activity and inhibition
of the maximal response. Evidence suggests that this bimodal mechanism of action of lithium may be applicable to its effects
on cyclic AMP production and AP-1 activation.
Mechanism of action of lithium
RS Jope

120
this hypothesis is that it pinpoints a specific locus of plexity in studies of lithium by indicating that only
lithium action, the inhibition of inositol monophos- abnormal signaling activity may be affected by lithium.
phatase. Support for this modified inositol depletion Although providing direct support in human brain for
hypothesis has been obtained in a number of creative the thesis that phosphoinositide signaling is abnormal
experiments using model systems,51–56 but demonstrat- in bipolar brain, these methods lack the capability of
ing such effects in more intact systems, especially in identifying more precisely the mechanism of action of
human brain,57,58 has been less successful and remains lithium, the sample number was small, it is difficult to
a formidable challenge. Nonetheless, an intriguing out- precisely correlate in vitro measures with in vivo sig-
growth of the inositol depletion hypothesis has been naling activity, and the use of postmortem tissue
the novel application of inositol as a pharmacological encompasses many uncontrollable factors concerning
agent which has shown promising utility in animal disease and the drug history of each subject.
models and therapeutic applications.13,59 Additionally, In part because of the ambiguities of if and how lith-
the concentration and uptake of inositol differs among ium actually affects phosphoinositide signaling,
regions of rat brain and reduced inositol levels in rat especially in human brain and in association with
brain following lithium administration were recently bipolar disorder, many investigators have turned to
reported to be limited to the hypothalamus,60–62 and studying lithium’s effects on signaling processes down-
lithium was also found to reduce inositol in astro- stream from phosphoinositide hydrolysis. Two pri-
cytes.63 Thus, each of these factors, brain region, cell- mary second messengers produced by the phospho-
type, and endogenous inositol cycles, introduces com- inositide signal transduction system are inositol
plexities in the relationship between lithium and inosi- trisphosphate, which raises intracellular calcium levels
tol which have made it difficult to define precisely how by releasing it from sequestered sites, and diacylgly-
lithium’s inhibition of inositol monophosphatase cerol, which activates protein kinase C. Surprisingly
affects phosphoinositide signaling. little has been published about lithium’s effects on cal-
Besides possibly causing inositol depletion in some cium, with a limited number of reports, mostly involv-
cells, there is also substantial evidence that lithium can ing non-neuronal cells, indicating reductions in stimu-
attenuate phosphoinositide signaling by inhibiting the lated increases in intracellular calcium concentrations
activation of G-proteins that couple receptors to phos- in tissue exposed to lithium.73–78 In contrast, modu-
phoinositide-selective phospholipase C.5,64,65 The lation of protein kinase C by lithium has been studied
majority of studies indicate that lithium reduces the extensively. Early studies found decreased protein kin-
functional activity, not the level, of G-proteins ase C activity in lithium-treated tissues using a variety
mediating phosphoinositide hydrolysis (but see66), of methods79–82 and evidence of activated protein kin-
with evidence that lithium interferes with magnesium- ase C associated with mania.83 Subsequently, specific
binding sites.67 Thus, lithium-induced impaired G-pro- isotypes of protein kinase C were reported to be
tein activity may contribute to some cases in which reduced by lithium.84 Since lithium was found to
lithium was reported to decrease phosphoinositide sig- reduce protein kinase C activity, a recent clinical
naling activity. At this time it may be most parsimoni- evaluation of tamoxifen, which is known to inhibit
ous to conclude from these studies that lithium has the protein kinase C, was undertaken to determine its effi-
potential to reduce the activation of phosphoinositide cacy in bipolar disorder. Initial results indicate that
signaling by more than one mechanism. tamoxifen, at doses sufficient to inhibit protein kinase
However, not all evidence supports the conclusion C, had a striking, and in many cases, rapid antimanic
that lithium directly impedes phosphoinositide sig- effect in 苲80% of the patients.85 Clearly the possible
naling. In rat brain, in vivo levels of inositol tris- contribution of estrogen receptor blockade or other
phosphate derived from phosphoinositide hydrolysis actions of tamoxifen cannot be ruled out, but these
are not reduced by lithium treatment.68–70 Hokin and intriguing results clearly warrant further studies
colleagues16–18 have emphasized species differences in related to protein kinase C activity. One of the major
the inositol-depleting effects of lithium and showed substrates of protein kinase C, myristoylated alanine-
that in primate brain slices lithium increases phospho- rich C kinase substrate (MARCKS), has been studied
inositide signaling through activation of glutamatergic extensively and lithium was found to reduce the levels
N-methyl-d-aspartate receptors. Therapeutic concen- of MARCKS, an effect that is also caused by direct acti-
trations of lithium also increased muscarinic receptor- vation of protein kinase C, suggesting a modulatory
stimulated phosphoinositide signaling in cultured cer- effect of lithium on protein kinase C activity which
ebellar granule cells.71 In studies using postmortem modulates MARCKS.81,86 MARCKS is involved in cyto-
human brain,72 a regionally-selective deficit in phos- skeletal architecture, indicating that this function may
phoinositide signaling was evident in bipolar com- be affected by lithium. MARCKS also inhibits phos-
pared with matched control tissue. Additionally, in the phoinositide signaling,87 suggestive that lithium may
affected brain region there was a direct relationship reduce this inhibitory influence on phosphoinositide
between bipolar brain lithium levels and phosphoinos- signaling as a consequence of down-regulating
itide signaling activity so that the deficit was greatest MARCKS. The extent and consequences of lithium’s
in tissues with the lowest lithium, suggestive of an effects on protein kinase C and its substrates remains
amelioration by lithium of deficient phosphoinositide a topic of great interest.
signaling activity. This also raises an additional com- Numerous studies have demonstrated that the cyclic
Mechanism of action of lithium
RS Jope

121
AMP second messenger system is modulated by lith- in cyclic AMP production, such as by norepinephrine,
ium.3,5,11 In general, these show that lithium increases may be caused by an inhibitory effect of lithium on
basal levels of cyclic AMP but impairs receptor- the activation of Gs,3,5,92,93 but the specific mechanism
coupled stimulation of cyclic AMP production (Figure mediating this action is not entirely clear. Direct
3). One hypothesis is that these dual effects of lithium inhibitory effects of lithium on adenylyl cyclase also
are due to inhibition by lithium of the G-proteins that may contribute to its modulatory effects on the pro-
mediate cyclic AMP production. Receptor-mediated duction of cyclic AMP.92 Overall, it appears that these
production of cyclic AMP is controlled by a stimu- actions of lithium reduce the magnitude of fluctuations
latory G-protein, Gs, and a counter-balancing inhibi- in cyclic AMP levels by increasing the lowest basal lev-
tory G-protein, Gi. Under basal conditions, cyclic AMP els and decreasing maximal stimulated increases, thus
production is tonically inhibited by the prevailing Gi stabilizing the activity of this signaling system (Figures
influence. Increased basal cyclic AMP levels caused by 2 and 3).
lithium may occur at least in part because lithium One intriguing mechanism by which lithium may
reduces the activity of Gi, apparently by shifting its influence G-protein function is by modulating the post-
equilibrium between a free active conformation and an translational modification of ADP-ribosylation of G-
inactive heterotrimeric conformation towards the inac- proteins catalyzed by endogenous enzymes (ie, not by
tive form,5,66,88–90 but not all studies find such inhibi- added ADP-ribosylating toxins such as pertussis toxin).
tory effects.91 Therefore, inhibitory effects of lithium Administration of lithium was found to increase the
on Gi can elevate the basal level of cyclic AMP. On the endogenous ADP-ribosylation of G-proteins.94,95
other hand, attenuation of stimulus-induced increases Although the ramifications of this modification on sig-
naling activities remain to be established, these find-
ings raise the possibility of a new mechanism for mod-
ulating G-protein function.
A primary action of cyclic AMP is to stimulate the
activity of cyclic AMP-dependent protein kinase
(protein kinase A). Several studies found that lithium
can modulate stimulated protein kinase A-mediated
protein phosphorylation.80,96–98 Especially interesting
are a series of reports that lithium inhibits protein kin-
ase A-induced phosphorylation of cytoskeletal pro-
teins,99,100 an action that may contribute to long-term
modulation of neuronal structure and function by lith-
ium.
An exciting new story is rapidly developing involv-
ing the regulation by lithium of cytoskeletal protein
phosphorylation and function, and the consequential
effects on neuronal architecture. Protein phosphoryl-
ation of cytoskeletal proteins was recently found to be
affected by a newly discovered inhibitory effect of lith-
ium on glycogen synthase kinase-3␤101 (Figure 4). In
addition to its originally described function of phos-
phorylating glycogen synthase, this kinase also phos-
phorylates several other proteins, including microtu-
bule-associated proteins (MAPs) such as tau and MAP-
1B, which contribute to regulating neuronal cytoskele-
tal networks. A long-overlooked report had shown that
lithium inhibits an unidentified kinase that phos-
phorylates glycogen synthase,102 which was later ident-
ified as glycogen synthase kinase-3␤ and recently
shown to be the kinase directly inhibited by lithium.101
Figure 3 The production of cyclic AMP is bimodally modu- Several recent studies found that inhibition of glycogen
lated by lithium. (a) Basal cyclic AMP production is predomi- synthase kinase-3␤ by lithium reduces tau phosphoryl-
nantly controlled by the action of the ␣-subunit of the inhibi- ation.101,103–107 The majority of these studies have been
tory G-protein, Gi. Lithium appears to facilitate the formation conducted by investigators interested in developmen-
of the inactive heterotrimeric conformation of Gi, resulting in tal effects of glycogen synthase kinase-3␤ or microtu-
increased cyclic AMP production under basal conditions. (b)
bule dynamics, rather than lithium’s therapeutic
Receptor activation can stimulate the heterotrimeric G-pro-
tein Gs to dissociate, allowing the active ␣-subunit to activate effects, so acute treatments and supra-therapeutic con-
adenylyl cyclase (AC) to increase cyclic AMP production. centrations of lithium, generally 5–20 mM, were util-
Lithium appears to reduce receptor-activated stimulation of ized. Nonetheless, the dramatic effects of lithium that
Gs, resulting in decreased cyclic AMP production under were observed suggest that lower concentrations of
stimulated conditions. ␣␤␥ = heterotrimeric G-protein. lithium will have significant, though more subtle,
Mechanism of action of lithium
RS Jope

122
ings indicate that lithium can inhibit phospholipase
A2, an enzyme that mediates the production of arachi-
donate.115,116 Since arachidonate can contribute to the
activation of protein kinase C, it was suggested that its
reduced production after lithium may contribute to
reduced activation of protein kinase C.116 Further
investigations of these signaling systems are needed to
clarify how their regulation is integrated into the over-
all metabolic and signaling activity modulatory effects
of lithium.

Transcription factors, gene expression, and


protein levels

Figure 4 Inhibition of glycogen synthase kinase-3␤ (GSK-3␤)


Transcription factors fulfill the critical functions of
by lithium modulates the phosphorylation and function of transmitting and regulating signals to the nucleus,
the microtubule-associated proteins tau and MAP-1B. GSK- allowing cells to respond to a changing environment
3␤ phosphorylates tau (␶), which decreases its binding to through alterations in gene expression. Thus transcrip-
microtubules, and phosphorylates MAP-1B, which increases tion factors may be selectively influenced by lithium
its binding to microtubules. By inhibiting GSK-3␤, lithium both as a result of lithium’s modulation of the activities
decreases the phosphorylation of tau and MAP-1B, increasing of specific signaling systems and as targets for lithium
tau microtubule binding and decreasing MAP-1B microtubule to modulate the expression of genes selectively.
binding. Although each binds microtubules, tau and MAP-1B Studies with lithium have focused on transcription
are not functionally interchangeable and each of their actions
factors coded by immediate early genes, those
may predominate in different neuronal compartments.
responding rapidly and transiently to cell signals, such
as the AP-1 transcription factor which is composed of
modulatory effects on neuronal cytoskeletal modifi- dimers of the Fos and Jun protein families. Following
cations mediated by glycogen synthase kinase-3␤, and the initial discovery that lithium increased c-fos mRNA
evidence of such effects with 1 mM lithium were levels,117 several reports have appeared with a confus-
reported briefly in some studies.101,103–105 Dephos- ing array of results after lithium treatment, including
phorylation of tau at the glycogen synthase kinase-3␤ increases, decreases, and no changes in c-fos mRNA
sites, which is caused by lithium, enhances the binding levels in a variety of cultured cells and in regions of
of tau to microtubules, which promotes microtubule rat brain,118–128 but only limited information is avail-
assembly.108 Therefore, this action of lithium suggests able about the effects of lithium on the expression of
that it may stabilize microtubule-based neuronal struc- Jun family members.121,125,128 These inconsistent
ture. However, inhibition of glycogen synthase kinase- results with c-fos may be related to cellular and brain
3␤ by lithium also dramatically decreased MAP-1B regional differences in the response to lithium com-
phosphorylation.109,110 Dephosphorylation of MAP-1B, pounded by the complexity of factors regulating mRNA
in contrast to tau dephosphorylation, decreases its levels and the activation of immediate early genes.
ability to bind and stabilize microtubules and therefore More functional information may be gained from
results in more dynamic microtubules, and this examining transcription factor DNA binding activity.
response to lithium treatment also was reported to This was first employed only in 1995, but is now
cause increased axonal spreading and increased receiving much attention. Stimulation of AP-1 DNA
growth cone area.109 These recent findings suggest that binding activity in rat brain was found not only to be
neuronal cytoskeletal rearrangements may be a signifi- attenuated by lithium, but also to be influenced by cir-
cant consequence of lithium administration. The cadian rhythm.129 This attenuation of stimulated AP-1
opposing effects of dephosphorylation of tau and of by lithium was confirmed in cultured cells, but elev-
MAP-1B, promoting microtubule disassembly and ated basal AP-1 DNA binding activity was noted as the
assembly, respectively, caused by lithium’s inhibition concentration of lithium was increased.125,130 Lithium
of glycogen synthase kinase-3␤, support the theme of also increased basal AP-1 in several cultured cell lines
this review that lithium acts at multiple sites to adjust and in rat brain, although with evidence of differential
the balance between opposing influences rather than lithium concentration sensitivities among cell
having unidirectional effects (Figure 4). Undoubtedly types.126,128,131 The opposite actions of lithium on
ongoing studies will shed more light on the precise stimulated (decrease) and basal (increase) AP-1 DNA
effects of lithium on these critical components of neu- binding activity at first appeared to be contradictory.
ronal structure. However, these have been proposed to reflect actions
Other signaling systems also have been shown to be of lithium at different sites which are cellularly inte-
modulated by lithium. In accordance with earlier grated to reduce the magnitude of fluctuations in AP-
reports, more recent evidence was obtained that lith- 1, and consequently of AP-1-responsive gene
ium may modulate the production of cyclic GMP111,112 expression, so after lithium treatment minimal basal
as well as nitric oxide.41,113,114 In addition, new find- levels are elevated and maximal stimulated levels are
Mechanism of action of lithium
RS Jope

123
dampened15 (Figure 2). The decrease in receptor- The mRNA levels for neuropeptide Y and somatostatin
coupled stimulation of AP-1 could result from inhi- were increased, and cholecystokinin decreased, by
bition by lithium of phosphoinositide hydrolysis, lithium administration in some rat brain regions.141,142
reduced second messenger activities (protein kinase C Especially intriguing is the recent discovery that lith-
or calcium), or other effects that regulate stimulation ium increases levels of the anti-apoptotic protein bcl-
of AP-1. On the other hand, the increased basal level 2 which may have important consequences for the neu-
of AP-1 activity is likely due to inhibition by lithium roprotective actions of lithium.33,34 Additionally, the
of glycogen synthase kinase-3␤, an enzyme that is con- method of differential display holds great promise as a
stituitively inhibitory towards AP-1, so blockade by method capable of distinguishing drug-induced alter-
lithium can elevate basal AP-1. These combined effects ations in the expression of specific genes, and the first
of lithium on basal and stimulated AP-1 may model study with lithium using this method identified 2⬘,3⬘-
how multiple actions of lithium are integrated to stabil- cyclic nucleotide 3⬘-phosphodiesterase, an enzyme
ize fluctuations in this, and other, signaling systems potentially involved in neuronal growth and repair, as
which impact to also stabilize gene expression activi- being up-regulated after lithium treatment.143
ties within a lithium-modified range of maximal and Overall, although a variety of effects of lithium on
minimal levels. Investigators are now in the process of mRNA and protein levels have been reported, there is
identifying other transcription factors and genes that not yet a consensus about which of these may be of
are regulated by lithium. Recently lithium was also primary importance in the therapeutic response, and
shown to modulate two other transcription factors, the the mechanisms accounting for these changes caused
cyclic AMP responsive element and NF-␬B, confirming by lithium have not been identified. At present these
that lithium’s effects doubtlessly extend to additional findings are most useful as leads for further investi-
transcription factors besides AP-1.126,130,132 Current gations aimed at identifying the critical affected pro-
investigations should provide greater insight into the ducts, as well as for confirming that lithium clearly
mechanisms and outcomes of these modulatory effects does have selective modulatory influences on neuronal
of lithium on transcription factors. gene expression and protein concentrations.
It seems likely that alterations in neuronal gene
expression and consequently protein levels induced by
Physiological effects
lithium contribute to its therapeutic effect. Not surpris-
ingly considering the foregoing discussion, lithium has Potentially important additional actions of lithium
been reported to alter the expression of a number of appear to warrant further investigation. Modulation by
genes and/or the levels of proteins, processes that may lithium of the Na+-K+-ATPase remains a viable con-
underlie the necessity for long-term treatment with tributory factor in lithium’s actions.144 There has been
lithium to achieve optimal therapeutic responses. This surprisingly little research in recent years examining
time-delay may be required for neuronal proteins to hormonal effects of lithium, although hormonal fluc-
reach new steady state levels in the presence of lithium tuations likely contribute to susceptibility and episodic
that mediate structural and/or functional neuronal onset. However, several reports provide confirmation
alterations. However, it remains to be determined of earlier studies that thyroid hormone activity is affec-
which of these are therapeutically critical products. ted by lithium.145–147 Circadian rhythm disturbances
As previously reviewed,10,11 lithium has been may be a factor in bipolar disorder, and recent findings
reported to alter the expression of several proteins continue to support earlier reports that the circadian
involved in signaling systems, and recent reports con- period can be altered by lithium.148 This limited num-
tinue to find effects on G-proteins.133–135 Lithium also ber of recent studies clearly indicates the need for
increased mRNA levels for nitric oxide synthase type- further investigations of the hormonal and circadian
2.114 It is difficult to know whether this preponderance influences of lithium.
of evidence that the expression of proteins involved
in signaling systems is affected by lithium reflects a
Summary
predominant action of lithium on these proteins or
reflects the current interests of investigators examining Although many clues have been obtained in recent
the effects of lithium. Nonetheless, it appears likely years, there is still not a clear understanding of the
that altered levels of signaling proteins contribute to mechanism of action of lithium. One of the primary
the signal and mood stabilizing actions of lithium. difficulties is that there is still no means to test which
Several additional mRNA or protein levels were of the multitude of described actions of lithium are
modulated by lithium. Glial fibrillary acidic protein critical for its therapeutic effect, and there is a dearth
was increased by lithium136 and lithium modulated of schemes which consolidate this information. Thus,
stress-induced alterations in polyamine enzymes.137 In from the many biochemical actions of lithium that have
PC12 cells, treatment with a high concentration of lith- been discovered, investigators are faced with the formi-
ium (20 mM) in combination with other agents dable tasks of identifying which of these are critical for
increased neurotensin/neuromedin N expression,138 lithium’s therapeutic actions and integrating these into
increased melanin-concentrating hormone mRNA and a comprehensive picture of how neuronal function is
decreased tyrosine hydroxylase mRNA.139 Lower levels modulated by lithium. There is clearly a need for ani-
of lithium increased tyrosine hydroxylase levels.140 mal models that would help to distinguish the actions
Mechanism of action of lithium
RS Jope

124
of lithium that are critical for a therapeutic on the cytoskeleton at multiple levels, including sig-
response.4,149,150 There is also a need for information nals from glycogen synthase kinase 3␤, cyclic AMP-
about the intracellular concentrations of lithium that dependent kinase, and protein kinase C, and these
are required for therapeutic responses. The intracellu- actions may be critical for the neural plasticity
lar concentration of lithium has traditionally been esti- involved in mood recovery and stabilization. (3) Lith-
mated from gross measurements of extracts of brain ium adjusts signaling activities regulating second mess-
regions. Such estimates are clearly inaccurate, since in engers, transcription factors, and gene expression. The
recent years it has become evident that spatial, as well outcome of these effects appears likely to result in lim-
as temporal, gradients of molecules are achieved in iting the magnitudes of fluctuations in activities, con-
neurons. Identifying the spatial and temporal gradients tributing to a stabilizing influence induced by lithium,
of lithium’s distribution is a critical, and challenging, and neuroprotective effects of lithium may be derived
issue. This limitation is emphasized by a recent study from its modulation of the expression of genes
addressing the intraneuronal concentration of lithium involved in regulating degenerative and regenerative
which concluded that localized concentrations of lith- mechanisms.
ium near transport sites are likely to be much higher In summary, this review introduces the concepts that
than 1 mM.32 multiple actions of lithium must be considered critical
Even with these limitations, however, there is begin- in the therapeutic response, and that these complex
ning to emerge an understanding that rather than look- effects support neural plasticity and stabilize neuro-
ing for a single site of action, many actions of lithium transmitter balances, signaling activities, and gene
must, and can, be integrated to obtain a cohesive pic- expression, each of which may make a necessary, but
ture of how neuronal function is modulated by long- individually insufficient, contribution to lithium’s
term exposure to lithium (Figure 5). Three systems therapeutic effects (Figure 5). Multiple actions of lith-
appear especially enticing as critical sites of actions of ium, rather than a single site of action, may be neces-
lithium which appear most likely to contribute to the sary because lithium has antimanic, antidepressant,
multiple therapeutic actions of lithium, and each of and prophylactic stabilizing actions, and because
which can influence the others. (1) Lithium seems to bipolar disorder is a complex illness involving mul-
alter the balance among neurotransmitter activities and tiple systems. Thus, it may be too much to expect that
to have neuroprotective effects. Modulation of neuro- these multiple effects of lithium could be accounted for
transmitter balances, in conjunction with decreasing by either an action that is unidirectional, rather than a
glutamatergic activity and toxicity, is likely to modu- bimodal balancing action, or a single site of action,
late excitatory and inhibitory synapses and networks, rather than the culmination of multiple actions. The
as well as contributing to neuroprotection. (2) The neu- critical property of lithium appears to be that it is able
ronal cytoskeleton is much more than a structural to act at multiple sites to adjust the balance, and
framework, but is a dynamic system contributing to dampen the magnitudes of fluctuations, of positive and
neural plasticity. Lithium modulates signals impacting negative inputs to neuronal function, allowing it to

Figure 5 Summary of the major sites of action of lithium emphasized in this review. Evidence reviewed in this article indicates
that more than a single site of action of lithium is likely to contribute to its therapeutic effects. Furthermore, this review notes
that lithium has a widespread tendency to alter balances between opposing pathways, rather than having unidirectional actions,
to modulate the magnitudes of fluctuations in systems. The figure depicts sites of signaling systems that appear to be the most
critical sites of action of lithium. NT = neurotransmitter. Rec = receptor.
Mechanism of action of lithium
RS Jope

accumulation in brain cortex slices but not accumulation of inosi-


125
normalize the complex components of mood disorders
tol monophosphates and bisphosphates. Proc Natl Acad Sci USA
and to have multiple therapeutic effects.
1997; 94: 4757–4760.
20 Dixon JF, Hokin LE. Lithium acutely inhibits and chronically up-
regulates and stabilizes glutamate uptake by presynaptic nerve end-
Acknowledgements ings in mouse cerebral cortex. Proc Natl Acad Sci USA 1998; 95:
8363–8368.
I am very grateful to Drs John Challiss, De-Maw Chu- 21 Volonte C, Rukenstein A. Lithium chloride promotes short-term
ang, Lowell Hokin, and Trevor Young for their valuable survival of PC12 cells after serum and NGF deprivation. Lithium
comments on an earlier version of this review, and 1993; 4: 211–219.
especially to Dr Husseini Manji for many stimulating 22 D’Mello SR, Anelli R, Calissano P. Lithium induces apoptosis in
immature cerebellar granule cells but promotes survival of mature
discussions regarding the contents of this review. neurons. Exp Cell Res 1994; 211: 332–338.
Research in the author’s laboratory was supported by 23 Volonte C, Ciotti MT, Merlo D. LiCl promotes survival of GABA-
grants from the Theodore and Vada Stanley Foun- ergic neurons from cerebellum and cerebral cortex: LiCl induces
dation and the National Institutes of Health survival of GABAergic neurons. Neurosci Lett 1994; 172: 6–10.
24 Li R, Shen Y, El-Mallakh RS. Lithium protects against ouabain-
(MH38752). induced cell death. Lithium 1994; 5: 211–216.
25 Inouye M, Yamamura H, Nakano A. Lithium delays the radiation-
induced apoptotic process in external granule cells of mouse cere-
References bellum. J Radiat Res 1995; 36: 203–208.
26 Pascual T, Gonzalez, JL. A protective effect of lithium on rat
1 Sherman WR, Gish BG, Honchar MP, Munsell LY. Effects of lith-
behavior altered by ibotenic acid lesions of the basal forebrain chol-
ium on phosphoinositide metabolism in vivo. Fed Proc 1986; 45:
inergic system. Brain Res 1995; 695: 289–292.
2639–2646.
27 Grignon S, Levy N, Couraud F, Bruguerolle B. Tyrosine kinase
2 Wood AJ, Goodwin GM. A review of the biochemical and neuro-
inhibitors and cycloheximide inhibit Li+ protection of cerebellar
pharmacological actions of lithium. Psychol Med 1987; 17: 579–
granule neurons switched to non-depolarizing medium. Eur J Phar-
600.
macol 1996; 315: 111–114.
3 Mork A, Geisler A, Hollund P. Effects of lithium on second messen-
28 Nonaka S, Hough CJ, Chuang D-M. Chronic lithium treatment
ger systems in the brain. Pharmacol Toxicol 1992; 71: 4–17.
robustly protects neurons in the central nervous system against
4 Post RM. Transduction of psychosocial stress into the neurobiology
excitotoxicity by inhibiting N-methyl-d-aspartate receptor-
of recurrent affective disorder. Am J Psychiatry 1992; 149: 999–
mediated calcium influx. Proc Natl Acad Sci USA 1998; 95:
1010.
2642–2647.
5 Avissar S, Schreiber G. The involvement of guanine nucleotide
29 Nonaka S, Chuang D-M. Neuroprotective effects of lithium on focal
binding proteins in the pathogenesis and treatment of affective dis-
cerebral ischemia in rats. Neuroreport 1998; 9: 2081–2084.
orders. Biol Psychiatry 1992; 31: 435–459.
30 Nonaka S, Katsube N, Chuang D-M. Lithium protects rat cerebellar
6 Gani D, Downes CP, Batty I, Bramham J. Lithium and myo-inositol
granule cells against apoptosis induced by anticonvulsants, pheny-
homeostasis. Biochim Biophys Acta 1993; 1177: 253–269.
7 Duman RS, Heninger GR, Nestler EJ. Adaptations of receptor- toin and carbamazepine. J Pharmacol Exp Ther 1998; 286: 539–547.
coupled signal transduction pathways underlying stress- and drug- 31 Sparapani M, Virgili M, Ortali F, Contestabile A. Effects of lithium
induced neural plasticity. J Nerv Ment Dis 1994; 182: 692–700. treatment on ornithine decarboxylase induction and excitotoxic
8 Price LH, Heninger GR. Lithium in the treatment of mood dis- neuropathology in the rat. Brain Res 1997; 765: 164–168.
orders. New Engl J Med 1994; 331: 591–598. 32 Kabakov AY, Karkanias NB, Lenox RH, Papke RL. Synapse-specific
9 Lachman HM, Papolos DF. A molecular model for bipolar affective accumulation of lithium in intracellular microdomains: a model for
disorder. Med Hypoth 1995; 45: 255–264. uncoupling coincidence detection in the brain. Synapse 1998; 28:
10 Manji HK, Lenox RH. Long-term action of lithium: a role for tran- 271–279.
scriptional and posttranscriptional factors regulated by protein kin- 33 Manji HK, Zeng WZ, Huang D, Ikonomov OC, Chen G. A new com-
ase C. Synapse 1994; 16: 11–28. mon transcriptional target for the mood-stabilizing agents lithium
11 Jope RS, Williams MB. Lithium and brain signal transduction sys- and valproic acid. Soc Neurosci Abstr 1997; 23: 2045.
tems. Biochem Pharmacol 1994; 47: 429–441. 34 Chuang D-M, Chen R-W. Chronic lithium induces p53 downregul-
12 Atack JR, Broughton HB, Pollack SJ. Inositol monophosphatase—a ation and bcl-2 upregulation in cerebellar neurons: a potential role
putative target for Li+ in the treatment of bipolar disorder. Trends in neuroprotection against excitotoxicity. Soc Neurosci Abstr 1998;
Neurosci 1995; 18: 344–349. 24: 997.
13 Belmaker RH, Bersudsky Y, Agam G, Levine J, Kofman O. How 35 Dunigan CD, Shamoo AE. Li+ stimulates ATP-regulated dopamine
does lithium work on manic depression? Clinical and psychologi- uptake in PC12 cells. Neuroscience 1995; 65: 1–4.
cal correlates of the inositol theory. Annu Rev Med 1996; 47: 47–56. 36 Dziedzicka-Wasylewska M, Mackowiak M, Figat K, Wedzony K.
14 Soares JC, Gershon S. The lithium ion: a foundation for psycho- Adaptive changes in the rat dopaminergic transmission following
pharmacological specificity. Neuropsychopharmacology 1998; 19: repeated lithium administration. J Neural Trans 1996; 103: 765–
167–182. 776.
15 Jope RS. A bimodal model of the mechanism of action of lithium. 37 Acquas E, Fibiger, HC. Chronic lithium attenuates dopamine D1-
Mol Psychiatry 1999; 4: 21–25. receptor mediated increases in acetylcholine release in rat frontal
16 Dixon JF, Lee CH, Los GV, Hokin LE. Lithium enhances accumu- cortex. Psychopharmacology 1996; 125: 162–167.
lation of [3H]inositol radioactivity and mass of second messenger 38 Carli M, Morissette M, Hebert C, Di Paolo T, Reader TA. Effects of
inositol 1,4,5-trisphosphate in monkey cerebral cortex slices. J Neu- chronic lithium treatment on central dopamine transporters.
rochem 1992; 59: 2332–2335. Biochem Pharmacol 1997; 54: 391–397.
17 Dixon JF, Los GV, Hokin LE. Lithium stimulates glutamate ‘release’ 39 Ormandy GC, Song L, Jope RS. Analysis of the convulsant-
and inositol 1,4,5-trisphosphate accumulation via activation of the potentiating effects of lithium. Exp Neurol 1991; 111: 356–361.
N-methyl-d-aspartate receptor in monkey and mouse cerebral cor- 40 Jope RS, Gu X. Seizures increase acetylcholine and choline concen-
tex slices. Proc Natl Acad Sci USA 1994; 91: 8358–8362. trations in rat brain regions. Neurochem Res 1991; 16: 1219–1226.
18 Los GV, Artemenko IP, Hokin LE. Time-dependent effects of lith- 41 Bagetta G, Rodino P, Paoletti AM, Arabia A, Massoud R, Nistico G.
ium on the agonist-stimulated accumulation of second messenger Systemic administration of lithium chloride and tacrine but not
inositol 1,4,5-trisphosphate in SH-SY5Y human neuroblastoma kainic acid augments citrulline content of rat brain. Eur J Pharma-
cells. Biochem J 1995; 311: 225–232. col 1995; 294: 341–433.
19 Dixon JF, Hokin LE. The antibipolar drug valproate mimics lithium 42 Motohashi N. GABA receptor alterations after chronic lithium
in stimulating glutamate release and inositol 1,4,5-trisphosphate administration. Comparison with carbamazepine and sodium valp-
Mechanism of action of lithium
RS Jope

126 roate. Prog Neuropsychopharmacol Biol Psychiatry 1992; 16: and inositol levels may be abnormal in postmortem brain from
571–579. bipolar patients. Neuropsychopharmacology 1998; 19: 220–232.
43 Vargas C, Tannhauser M, Barros HM. Dissimilar effects of lithium 63 Wolfson M, Hertz E, Belmaker RH, Hertz L. Chronic treatment with
and valproic acid on GABA and glutamine concentrations in rat lithium and pretreatment with excess inositol reduce inositol pool
cerebrospinal fluid. Gen Pharmacol 1998; 30: 601–604. size in astrocytes by different mechanisms. Brain Res 1998; 787:
44 Williams NB, Jope RS. Lithium potentiates phosphoinositide- 34–40.
linked 5-HT receptor stimulation in vivo. NeuroReport 1994; 5: 64 Avissar S, Schreiber G, Danon A, Belmaker RH. Lithium inhibits
1118–1120. adrenergic and cholinergic increases in GTP binding in rat cortex.
45 Bourin M, Hascoet M, Colombel M-C, Redrobe JP, Baker GB. Differ- Nature 1988; 331: 440–442.
ential effects of clonidine, lithium and quinine in the forced swim- 65 Song L, Jope RS. Chronic lithium treatment impairs phosphatidyli-
ming test in mice for antidepressants; possible roles of serotoni- nositol hydrolysis in membranes from rat brain regions. J Neuro-
nergic systems. Eur Neuropsychopharmacol 1996; 6: 231–236. chem 1992; 58: 2200–2206.
46 Carli M, Reader TA. Regulation of central serotonin transporters 66 Manji HK, Chen G, Shimon H, Hsiao JK, Potter WZ, Belmaker RH.
by chronic lithium; an autoradiographic study. Synapse 1997; 27: Guanine nucleotide-binding proteins in bipolar affective disorder.
83–89. Arch Gen Psychiatry 1995; 52: 135–144.
47 Carli M, Afkhami-Dastjerdian S, Reader TA. Effects of a chronic 67 Avissar S, Murphy DL, Schreiber G. Magnesium reversal of lithium
lithium treatment on cortical serotonin uptake sites and 5-HT1A inhibition of beta-adrenergic and muscarinic receptor coupling to
receptors. Neurochem Res 1997; 22: 427–435. G-proteins. Biochem Pharmacol 1991; 41: 171–175.
48 Moorman JM, Leslie RA. Paradoxical effects of lithium on sero- 68 Whitworth P, Heal DJ, Kendall DA. The effect of acute and chronic
tonergic receptor function: an immunocytochemical, behavioral lithium treatment on pilocarpine-stimulated phosphoinositide
and autoradiographic study. Neuropharmacology 1998; 37: 357– hydrolysis in mouse brain in vivo. Br J Pharmacol 1990; 101: 39–44.
69 Jope RS, Song L, Kolasa K. Inositol trisphosphate, cyclic AMP and
374.
cyclic GMP in rat brain regions after lithium and seizures. Biol Psy-
49 Odagaki Y, Koyama T, Yamashita I. Lithium and serotonergic neu-
chiatry 1992; 31: 505–514.
ral transmission: a review of pharmacological and biochemical
70 Gur E, Lerer B, Newman ME. Acute or chronic lithium does not
aspects in animal studies. Lithium 1992; 3: 95–107.
affect agonist-stimulated inositol trisphosphate formation in rat
50 Berridge MJ, Downes CP, Hanley MR. Lithium amplifies agonist-
brain in vivo. Neuroreport 1996; 7: 393–396.
dependent phosphatidylinositol responses in brain and salivary
71 Gao X-M, Fukamauchi F, Chuang D-M. Long-term biphasic effects
glands. Biochem J 1982; 206: 587–595.
of lithium treatment on phospholipase C-coupled M3-muscarinic
51 Jenkinson S, Patel N, Nahorski SR, Challiss RAJ. Comparative effect
acetylcholine receptors in cultured cerebellar granule cells. Neuro-
of lithium on the phosphoinositide cycle in rat cerebral cortex, hip-
chem Int 1993; 22: 395–403.
pocampus, and striatum. J Neurochem 1993; 61: 1082–1090.
72 Jope RS, Song L, Li PP, Young LT, Kish SJ, Pacheco MA et al. The
52 Gray DW, Challiss RAJ, Nahorski SR. Differential effects of lithium
phosphoinositide signal transduction system is impaired in bipolar
on muscarinic cholinoceptor-stimulated CMP-phosphatidate
affective disorder brain. J Neurochem 1996; 66: 2402–2409.
accumulation in cerebellar granule cells, CHO-M3 cells, and SH-
73 van Calker D, Forstner U, Bohus M, Gebicke-Harter P, Hecht H,
SY5Y neuroblastoma cells. J Neurochem 1994; 63: 1354–1360.
Wark H-J et al. Increased sensitivity to agonist stimulation of Ca2+
53 Jenkinson S, Nahorski SR, Challiss RAJ. Disruption by lithium of response in neutrophils of manic-depressive patients: effect of lith-
phosphatidylinositol-4,5-bisphosphate supply and inositol-1,4,5- ium therapy. Neuropsychobiology 1993; 27: 180–183.
trisphosphate generation in Chinese hamster ovary cells expressing 74 Forstner U, Bohus PJ, Gebicke-Harter PJ, Baumer B, Berger M, van
human recombinant m1 muscarinic receptors. Mol Pharmacol Calker D. Decreased agonist-stimulated Ca2+ response in neutro-
1994; 46: 1138–1148. phils from patients under chronic lithium therapy. Psychiatry Clin
54 del Rio E, Shinomura T, van der Kaay J, Nicholls DG, Downes CP. Neurosci 1994; 243: 240–243.
Disruption by lithium of phosphoinositide signalling in cerebellar 75 Varney MA, Galione A, Watson SP. Lithium-induced decrease in
granule cells in primary culture. J Neurochem 1998; 70: 1662–1669. spontaneous Ca2+ oscillations in single GH3 rat pituitary cells. Br J
55 Batty IH, Currie RA, Downes CP. Evidence for a model of integrated Pharmacol 1994; 112: 390–395.
inositol phospholipid pools implies an essential role for lipid 76 Okamoto Y, Kagaya A, Motohashi N, Yamawaki S. Inhibitory
transport in the maintenance of receptor-mediated phospholipase effects of lithium ion on intracellular Ca2+ mobilization in rat hip-
C activity in 1321N1 cells. Biochem J 1998; 330: 1069–1077. pocampal slices. Neurochem Int 1995; 26: 233–238.
56 Willars GB, Nahorski SR, Challiss RA. Differential regulation of 77 Berk M, Kirchmann NH, Butkow N. Lithium blocks 45 Ca2+ uptake
muscarinic acetylcholine receptor-sensitive polyphosphoinositide into platelets in bipolar affective disorder and controls. Clin Neuro-
pools and consequences for signaling in human neuroblastoma pharmacol 1996; 19: 48–51.
cells. J Biol Chem 1998; 273: 5037–5046. 78 Chen Y, Hertz L. Inhibition of noradrenaline stimulated increase
57 Silverstone PH, Hanstock CC, Fabian J, Staab R, Allen PS. Chronic in [Ca2+]i in cultured astrocytes by chronic treatment with a thera-
lithium does not alter human myo-inositol or phosphomonoester peutically relevant lithium concentration. Brain Res 1996; 711:
concentrations as measured by 1H and 31P MRS. Biol Psychiatry 245–248.
1996; 40: 235–246. 79 Wang H-Y, Friedman E. Lithium inhibition of protein kinase C acti-
58 Shimon H, Agam G, Belmaker RH, Hyde TM, Kleinman JE. vation-induced serotonin release. Psychopharmacology 1989; 99:
Reduced frontal cortex inositol levels in postmortem brain of suic- 213–218.
ide victims and patients with bipolar disorder. Am J Psychiatry 80 Casebolt TL, Jope RS. Effects of chronic lithium treatment on pro-
1997; 154: 1148–1150. tein kinase C and cyclic AMP-dependent protein phosphorylation.
59 Cohen H, Kotler M, Kaplan Z, Matar MA, Kofman O, Belmaker RH. Biol Psychiatry 1991; 29: 233–243.
Inositol has behavioral effects with adaptation after chronic admin- 81 Lenox RH, Watson DG, Patel J, Ellis J. Chronic lithium adminis-
istration. J Neural Trans 1997; 104: 299–305. tration alters a prominent PKC substrate in rat hippocampus. Brain
60 Patishi Y, Lubrich B, Berger M, Kofman O, van Calker D, Belmaker Res 1992; 570: 333–340.
RH. Differential uptake of myo-inositol in vivo into rat brain areas. 82 Li PP, Sibony D, Green MA, Warsh JJ. Lithium modulation of phos-
Eur Neuropsychopharmacol 1996; 6: 73–75. phoinositide signaling system in rat cortex: selective effect on phor-
61 Lubrich B, Patishi Y, Kofman O, Agam G, Berger M Belmaker RH bol ester binding. J Neurochem 1993; 61: 1722–1730.
et al. Lithium-induced inositol depletion in rat brain after chronic 83 Friedman E, Wang H-Y, Levinson D, Connell TA, Singh H. Altered
treatment is restricted to the hypothalamus. Mol Psychiatry 1997; protein kinase C activity in bipolar affective disorder, manic epi-
2: 407–412. sode. Biol Psychiatry 1993; 33: 520–525.
62 Belmaker RH, Agam G, van Calker D, Richards MH, Kofman O. 84 Manji HK, Etcheberrigaray R, Chen G, Olds JL. Lithium decreases
Behavioral reversal of lithium effects by four inositol isomers corre- membrane-associated protein kinase C in hippocampus: selectivity
lates perfectly with biochemical effects on the PI cycle: depletion for the ␣ isozyme. J Neurochem 1993; 61: 2303–2310.
by chronic lithium of brain inositol is specific to hypothalamus, 85 Bebchuk JM, Arfken CL, Dolan-Manji S, Murphy J, Manji HK. A
Mechanism of action of lithium
RS Jope

preliminary investigation of a protein kinase C inhibitor 107 Xie H, Litersky JM, Hartigan JA, Jope RS, Johnson GVW. The inter-
127
(tamoxifen) in the treatment of acute mania. Arch Gen Psychiatry relationship between selective tau phosphorylation and microtu-
1999; (in press). bule association. Brain Res 1998; 798: 173–183.
86 Watson DG, Lenox RH. Chronic lithium-induced down-regulation 108 Lovestone S, Hartley CL, Pearce J, Anderton BH. Phosphorylation
of MARCKS in immortalized hippocampal cells: potentiation by of tau by glycogen synthase kinase-3␤ in intact mammalian cells:
muscarinic receptor activation. J Neurochem 1996; 67: 767–777. the effects on the organization and stability of microtubules. Neu-
87 Glaser M, Wanaski S, Buser CA, Boguslavsky V, Rashidzada W, roscience 1996; 73: 1145–1157.
Morris A et al. Myristoylated alanine-rich C kinase substrate 109 Lucas FR, Goold RG, Gordon-Weeks PR, Salinas PC. Inhibition of
(MARCKS) produces reversible inhibition of phospholipase C by GSK-3␤ leading to the loss of phosphorylated MAP-1B is an early
sequestering phosphatidylinositol 4,5-bisphosphate in lateral event in axonal remodeling induced by WNT-7a or lithium. J Cell
domains. J Biol Chem 1996; 271: 26187–26193. Sci 1998; 111: 1351–1361.
88 Hsiao JK, Manji HK, Chen G, Bitran JA, Risby ED, Potter WZ. Lith- 110 Garcia-Perez J, Avila J, Diaz-Nido J. Implication of cyclin-depen-
ium administration modulates platelet Gi in humans. Life Sci 1991; dent kinases and glycogen synthase kinase 3 in the phosphoryl-
50: 227–233. ation of microtubule-associated protein 1B in developing neu-
89 Ormandy GC, Jope RS. Pertussis toxin potentiates seizures induced ronal cells. J Neurosci Res 1998; 52: 445–452.
by pilocarpine, kainic acid and N-methyl-d-aspartate. Brain Res 111 Harvey BH, Carstens ME, Taljaard JJF. Evidence that lithium
1991; 553: 51–57. induces a glutamatergic, nitric oxide-mediated response in rat
90 Masana MI, Bitran JA, Hsiao JK, Potter WZ. In vivo evidence that brain. Neurochem Res 1994; 19: 469–474.
lithium inactivates Gi modulation of adenylate cyclase in brain. J 112 Shintani F, Kanba S, Nakaki T, Nakamura R, Sato K, Yagi G et al.
Neurochem 1992; 59: 200–205. Inhibition by lithium of cyclic GMP formation without inhibition
91 Odagaki Y, Nishi N, Koyama T. Lack of interfering effects of lith- of nitric oxide generation in the mouse neuroblastoma cell (N1E-
ium on receptor/G protein coupling in human platelet and rat brain 115). Neuropsychopharmacology 1994; 11: 119–124.
membranes. Biol Psychiatry 1997; 42: 697–703. 113 Bagetta G, Corasaniti MT, Melino G, Paoletti AM, Finazzi-Agro,
92 Mork A, Geisler A. Effects of chronic lithium treatment on agonist- Nistico G. Lithium and tacrine increase the expression of nitric
enhanced extracellular concentrations of cyclic AMP in the dorsal oxide synthase mRNA in the hippocampus of rat. Biochem
hippocampus of freely moving rats. J Neurochem 1995; 65: 134– Biophys Res Commun 1993; 197: 1132–1139.
139. 114 Feinstein DL. Potentiation of astroglial nitric oxide synthase type-
93 Carli M, Anand-Srivastava MB, Molina-Holgado E, Dewar KM, 2 expression by lithium chloride. J Neurochem 1998; 71: 883–886.
Reader TA. Effects of chronic lithium treatment on central dopami- 115 Chang MCJ, Grange E, Rabin O, Bell JM, Allen DD, Rapoport SI.
nergic receptor systems: G proteins as possible targets. Neurochem Lithium decreases turnover of arachidonate in several brain phos-
Int 1994; 24: 13–22. pholipids. Neurosci Lett 1996; 220: 171–174.
94 Nestler EJ, Terwilliger RZ, Duman RS. Regulation of endogenous 116 Chang MCJ, Jones CR. Chronic lithium treatment decreases brain
ADP-ribosylation by acute and chronic lithium in rat brain. J phospholipase A2 activity. Neurochem Res 1998; 23: 887–892.
Neurochem 1995; 64: 2319–2324. 117 Arenander AT, de Vellis J, Herschman HR. Induction of c-fos and
95 Young LT, Woods CM. Mood stabilizers have differential effects on TIS genes in cultured rat astrocytes by neurotransmitters. J Neuro-
endogenous ADP ribosylation in C6 glioma cells. Eur J Pharmacol sci Res 1989; 24: 107–114.
1996; 309: 215–218. 118 Kalasapudi VD, Sheftel G, Divish MM, Papolos DF, Lachman HM.
96 Guitart X, Nestler EJ. Chronic administration of lithium or other Lithium augments fos protooncogene expression in PC12 pheoch-
antidepressants increases levels of DARPP-32 in rat frontal cortex. romocytoma cells: implications for therapeutic action. Brain Res
J Neurochem 1992; 59: 1164–1167. 1990; 521: 47–54.
97 Jensen JB, Mork A. Altered protein phosphorylation in the rat brain 119 Divish MM, Sheftel G, Boyle A, Kalasapudi VD, Papolos DF, Lach-
following chronic lithium and carbamazepine treatments. Eur Neu- man HM. Differential effect of lithium on fos protooncogene
ropsychopharmacol 1997; 7: 173–179. expression mediated by receptor and postreceptor activators of
98 Zanardi R, Racagni G, Smeraldi E, Perez E. Differential effects of protein kinase C and cyclic adenosine monophosphate: model for
lithium on platelet protein phosphorylation in bipolar patients and its antimanic action. J Neurosci Res 1991; 28: 40–48.
healthy subjects. Psychopharmacology 1997; 129: 44–47. 120 Weiner ED, Kalasapudi VD, Papolos DF, Lachman HM. Lithium
99 Mori S, Zanardi R, Popoli M, Smeraldi E, Racagni G, Perez J. Inhibi- augments pilocarpine-induced fos gene expression in rat brain.
tory effect of lithium on cAMP dependent phosphorylation system. Brain Res 1991; 553: 117–122.
Life Sci 1996; 59: 99–104. 121 Williams MB, Jope RS. Distinctive rat brain immediate early gene
100 Mori S, Tardito D, Dorigo A, Zanardi R, Smeraldi E, Racagni G et responses to seizures induced by lithium plus pilocarpine. Mol
al. Effects of lithium on cAMP-dependent protein kinase in rat Brain Res 1994; 25: 80–89.
brain. Neuropsychopharmacology 1998; 19: 233–240. 122 Mathe AA, Miller JC, Stenfors C. Chronic dietary lithium inhibits
101 Klein PS, Melton DA. A molecular mechanism for the effect of basal c-fos mRNA expression in rat brain. Prog Neuropsychophar-
lithium on development. Proc Natl Acad Sci USA 1996; 93: macol Biol Psychiatry 1995; 19: 1177–1187.
8455–8459. 123 Harada H, Sugiyama T, Suketa Y. Characterization of inhibition
102 Horn RS, Walaas O, Walaas E. The influence of sodium, potassium by chronic lithium treatment with lithium ion on nerve growth
and lithium on the response of glycogen synthetase I to insulin factor-induced neuronal differentiation of rat PC12 pheochromo-
and epinephrine in the isolated rat diaphragm. Biochim Biophys cytoma cells. J Toxicol Environ Health 1996; 49: 197–206.
Acta 1973; 313: 296–309. 124 Miller JC, Mathe AA. Basal and stimulated c-fos mRNA
103 Stambolic V, Ruel L, Woodgett JR. Lithium inhibits glycogen syn- expression in the rat brain: effect of chronic dietary lithium. Neur-
thase kinase-3 activity and mimics Wingless signaling in intact opsychopharmacology 1997; 16: 408–418.
cells. Current Biol 1996; 6: 1664–1668. 125 Unlap MT, Jope RS. Lithium attenuates nerve growth factor-
104 Hong M, Chen DCR, Klein PS, Lee VMY. Lithium reduces tau induced activation of AP-1 DNA binding activity in PC12 cells.
phosphorylation by inhibition of glycogen synthase kinase-3. J Neuropsychopharmacology 1997; 17: 12–17.
Biol Chem 1997; 272: 25326–25332. 126 Ozaki N, Chuang D-M. Lithium increases transcription factor
105 Munoz-Montano JR, Moreno FJ, Avila J, Diaz-Nido J. Lithium binding to AP-1 and cyclic AMP-responsive element in cultured
inhibits Alzheimer’s disease-like tau protein phosphorylation in neurons and rat brain. J Neurochem 1997; 69: 2336–2344.
neurons. FEBS Lett 1997; 411: 183–188. 127 Namima M, Sugihara K, Okamoto K. Lithium inhibits the reverse
106 Zheng-Fischhofer Q, Biernet J, Mandelkow E-M, Illenberger S, tolerance and the c-Fos expression induced by methamphetamine
Godemann R, Mandelkow E. Sequential phosphorylation of tau in mice. Brain Res 1998; 782: 83–90.
by glycogen synthase kinase-3␤ and protein kinase A at thr212 128 Asghari V, Wang JF, Reiach JS, Young LT. Differential effects of
and ser214 generates the Alzheimer-specific epitope of antibody mood stabilizers on Fos/Jun proteins and AP-1 DNA binding
AT100 and requires a paired-helical-filament-like conformation. activity in human neuroblastoma SH-SY5Y cells. Mol Brain Res
Eur J Biochem 1998; 252: 542–552. 1998; 58: 95–102.
Mechanism of action of lithium
RS Jope

128 129 Williams MB, Jope RS. Circadian variation in rat brain AP-1 DNA 140 Chen G, Yuan P-X, Jiang Y-M, Huang L-D, Manji HK. Lithium
binding activity after cholinergic stimulation: modulation by lith- increases tyrosine hydroxylase levels both in vivo and in vitro. J
ium. Psychopharmacology 1995; 122: 363–368. Neurochem 1998; 70: 1768–1771.
130 Jope RS, Song L. AP-1 and NF-␬B stimulated by carbachol in 141 Zachrisson O, Mathe AA, Stenfors C, Lindefors N. Region-specific
human neuroblastoma SH-SY5Y cells are differentially sensitive effects of chronic lithium administration on neuropeptide Y and
to inhibition by lithium. Mol Brain Res 1997; 50: 171–180. somatostatin mRNA expression in the rat brain. Neurosci Lett
131 Yuan P-X, Chen G, Huang L-D, Manji HK. Lithium stimulates gene 1995; 194: 89–92.
expression through the AP-1 transcription factor pathway. Mol 142 Zachrisson O, Mathe AA, Lindefors N. Effects of chronic lithium
Brain Res 1998; 58: 225–230. and electroconvulsive stimuli on cholecystokinin mRNA
132 Wang JF, Asghari V, Rockel C, Young LT. Cyclic AMP responsive expression in the rat brain. Mol Brain Res 1996; 43: 347–350.
element binding protein phosphorylation and DNA binding is 143 Wang JF, Young LT. Differential display PCR reveals increased
decreased by chronic lithium but not valproate treatment of SH- expression of 2⬘,3⬘ cyclic nucleotide 3⬘-phosphodiesterase by lith-
SY5Y neuroblastoma cells. Neuroscience 1999; (in press). ium. FEBS Lett 1996; 386: 225–229.
133 McGowan S, Eastwood SL, Mead A, Burnet PWJ, Smith C, Flani- 144 El-Mallakh RS, Wyatt RJ. The Na, K-ATPase hypothesis for bipolar
gan TP et al. Hippocampal and cortical G protein Gs␣, Go␣, Gi2␣ disorder. Biol Psychiatry 1995; 37: 235–244.
mRNA expression after electroconvulsive shock or lithium treat- 145 Baumgartner A, von Stuckrad M, Muller-Oerlinghausen B, Graf
ment. Eur J Pharmacol 1996; 306: 249–255. K-J, Kurten I. The hypothalamic-pituitary-thyroid axis in patients
134 Dwivedi Y, Pandey GN. Effects of subchronic administration of maintained on lithium prophylaxis for years: high triiodothyron-
ine serum concentrations are correlated to the prophylactic effi-
antidepressants and anxiolytics on levels of the ␣ subunits of G
cacy. J Affect Dis 1995; 34: 211–218.
proteins in the rat brain. J Neural Trans 1997; 104: 747–760.
146 Bolaris S, Margarity M, Valcana T. Effects of LiCl on triiodothy-
135 Jakobsen SN, Wiborg O. Selective effects of long-term lithium and
ronine (T3) binding to nuclei from rat cerebral hemispheres. Biol
carbamazepine administration on G-protein subunit expression in
Psychiatry 1995; 37: 106–111.
rat brain. Brain Res 1998; 780: 46–55.
147 Kallner G, Petterson KG. Renal, thyroid and parathyroid function
136 Rocha E, Rodnight R. Chronic administration of lithium chloride during lithium treatment: laboratory tests in 207 people treated
increases immunodetectable glial fibrillary acidic protein in the for 1–30 years. Acta Psychiatr Scand 1995; 91: 48–51.
rat hippocampus. J Neurochem 1994; 63: 1582–1584. 148 Hafen T, Wollnik F. Effect of lithium carbonate on activity level
137 Gilad GM, Gilad VH. Brain polyamine stress response: recurrence and circadian period in different strains of rats. Pharmacol
after repetitive stressor and inhibition by lithium. J Neurochem Biochem Behavior 1994; 49: 975–983.
1996; 67: 1992–1996. 149 Antelman SM, Caggiula AR, Kucinski BJ, Fowler H, Gershon S,
138 Bullock BP, McNeil GP, Dobner PR. Synergistic induction of Edwards DJ et al. The effects of lithium on a potential cycling
neurotensin gene transcription in PC12 cells parallels changes in model of bipolar disorder. Prog Neuropsychopharmacol Biol Psy-
AP-1 activity. Mol Brain Res 1994; 27: 232–242. chiatry 1998; 22: 495–510.
139 Presse F, Cardona B, Borsu L, Nahon J-L. Lithium increases mel- 150 Kucinski BJ, Antelman SM, Caggiula AR, Fowler H, Gershon S,
anin-concentrating hormone mRNA stability and inhibits tyrosine Edwards DJ et al. Oscillatory effects of repeated morphine on
hydroxylase gene expression in PC12 cells. Mol Brain Res 1997; shock-induced hypoalgesia and beta-endorphin. Synapse 1998;
52: 270–283. 30: 30–37.

You might also like