4001306 a

Upload: iwan-miswar

Post on 02-Jun-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/11/2019 4001306 A

    1/10

  • 8/11/2019 4001306 A

    2/10

    implicated in mood stabilization for the treatment of bipolar disorder.

    Lithium and phosphoinositide/protein kinase Csignaling pathway

    Lithium has been shown to be an inhibitor of anumber of structurally similar magnesium-dependentphosphomonoesterases at K i values within thetherapeutically relevant range of concentrations(0.81.2 mM). 16,17 It was recognized over two decadesago that lithium is a potent inhibitor of the intracel-lular enzyme, inositol monophosphatase (IMPase)(K i 0.8 mM), which converts inositol monopho-sphate to inositol. 1820 Biochemical and Geneticstudies subsequently identified the upstream inositolpolyphosphatase as an additional target for

    lithium. 21,22 Thus, receptor G-protein-coupled PIhydrolysis has been extensively investigated as a sitefor the action of lithium as a mood stabilizer (seereview Manji and Lenox; 23 see Figure 1). Furthermore,since there is evidence that the mode of enzymeinhibition of IMPase is un competitive, the preferen-tial site of action for lithium was proposed to be themost overactive receptor-mediated neuronal path-ways undergoing the highest rate of phosphatidyl(4,5) bisphosphate (PIP 2 ) hydrolysis. 24,25 While it wasposited that lithium produces its therapeutic effectsvia a consequent reduction in relative concentrationsof myo- inositol and PIP 2 concentrations, data-drivensupport for this hypothesis has been highly depen-dent upon the cell and animal models underinvestigation. 2631 Much of this inconsistency may be related to the relatively small size of the signal-

    Figure 1 Lithium-responsive signal transduction pathways focused on PI/PKC and GSK-3 signaling. Lithium directlyinhibits the enzyme inositol monophosphate phosphatase (IMPase; K i E 0.8mM). Binding of ligands to G-protein-coupledreceptors (GPCRs) activates PLC and causes hydrolysis of PIP 2 to inositol-1,4,5 trisphosphate (IP 3) and DAG. IP 3 stimulatesCa2 release from cellular store, and DAG activates DAG-dependent protein kinase C (PKC). In the presence of receptorligands, long-term inhibition of IMPase results in a depletion of myo -inositol ( myo -I) and an accumulation of DAG followed by a downregulation of PKC isozymes a and e. On the other hand, lithium is a potent inhibitor of glycogen synthase kinase-3 b(GSK-3b ; K i E 2 mM) leading to the stabilization of b-catenin, which enters the nucleus to activate LEF/TCF-dependent genes.Lithium-responsive gene network is hypothetically proposed to be activated through PKC and GSK-3 signaling pathways andtheir crosstalk at therapeutically relevant concentrations. Abbreviations: A, receptor agoinst; Akt/PKB, a serine/threoninekinase; APC, adenomatous polyposis protein; axin, a homolog of Drosophila product of the fused locus; CMP-PA, cytidinemonophosphate-phosphatidate; DAG, diacylglycerol; Dsh, dishevelled; G, G protein; GBP, GSK-3 binding protein; GSK-3,glycogen synthase kinase-3; IP3, inositol-1,4,5-trisphosphate; LEF/TCF, LEF(lymphoid enhancer element)/TCF(T cell factor-1) family transcription factors; LRE, lithium-responsive promoter element; MUNC, a synaptic protein that contains DAG binding domain; PI, phosphatidylinositol; PIP 2 , phosphatidylinositol 4,5 bisphosphate; PP2A, protein phosphatase 2A; PLC,phospholipase C; Wnt, a secreted glycoprotein ligand for Wnt/ b-catenin signaling pathway.

    Molecular basis of lithium actionRH Lenox and Le Wang

    136

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    3/10

    dependent pools of myo- inositol and PIP 2 , as well asthe fact that the lithium-induced reduction of myo-inositol appears to be dependent upon multiplefactors including cell-type and tonic activity of thereceptor-coupled PI signaling pathway. 3133 It is of interest in this regard that a significant lithium-induced reduction in myo -inositol levels in the rightfrontal lobe has been observed in bipolar patientsprior to clinical response using proton magneticresonance spectroscopy. 34 However, these and otherdata suggest that while inhibition of IMPase mayrepresent an initial effect of lithium, reducing myo-inositol levels may be more critical to the specificityof the cellular site of action for lithium than its long-term therapeutic efficacy (see review, Lenox andManji). 2

    Such data in turn supported the hypothesis thatshort-term lithium exposure in selective neuronalpopulations in brain undergoing heightened activa-tion would result in diacylglycerol (DAG)-mediated

    activation of protein kinase C (PKC), subsequentdownregulation of PKC isozymes, and long-termdownstream changes in brain. 5,35 PKC isozymes have been implicated in the regulation of neurotransmitterrelease, neuronal excitability, and long-term changesin PKC-regulated protein function, neuroplasticity,and gene expression. PKC activity is tightly regulated by virtue of not only the distinct distribution of itsfamily of isozymes, but its required translocation tothe membrane associated with phosphorylation and binding to a receptor for activated C-kinase (RACK)proteins, and ultimate autocatalysis upon prolongedactivation. Several laboratories including our ownhave demonstrated that short-term lithium treatmentactivates PKC, and long-term lithium treatment down-regulates PKC isozymes in a PI-signaling-dependentmanner in brain. 5,35 Studies of chronic lithium (1 mM)exposure in both in vivo and in vitro models from ourlaboratory and others have demonstrated a reductionin PKC isozymes a and e in rat subiculum and in CA1regions of the hippocampus, which has been repli-cated in immortalized hippocampal cell modelsystem. 19,36,37 Furthermore, administration of myo-inositol to rats has been reported to reverse thedownregulation of PKC e in brain following chroniclithium. These studies have led to investigationsexamining PKC regulation in man (see reviews, Manji

    and Lenox,19,35

    Jope and Song,38

    Wang et al ,39

    Soareset al 40 ). Recent studies using human post-mortem brain homogenates revealed an increased associationof the receptor for activated C kinase-1 (RACK1) withPKC isozymes in frontal cortex of subjects with BPD,suggesting that interactions between these proteinsmay be also altered in bipolar disease. 37 In addition,there is intriguing evidence that tamoxifen, a drugknown to inhibit PKC in vitro , was effective in aninitial clinical study of acutely manic patients withBPD.41 Thus, it appears that chronic lithium-depen-dent modulation of receptor-coupled PIPKC signal-ing pathways may contribute to the long-term actionof lithium in the brain.

    Lithium and Wnt signaling pathway

    Glycogen synthase kinase 3 b (GSK-3b) is a serine/threonine kinase that is constitutively active in cellsand negatively regulates its substrates, one of which isb-catenin, a downstream effector of the Wnt signalingpathway controlling dorsalventral axis specificationand cell fate determination in various organismsincluding Dictyostelium , sea urchins, zebrafish, andXenopus. 17 GSK-3 activity is regulated through multi-ple proteins in a complex (Figure 1), where GSK-3 isactivated in the presence of axin, adenomatouspolyposis coli (APC), dishelveled (Dsh) and pro-motes phosphorylation of b-catenin for ubiquitinproteosome-mediated degradation. 43 On the contrary,GSK-3 binding protein (GBP), which is requiredfor axis formation in Xenopus , could join the comp-lex to inhibit GSK-3 activity, in part, by preventingaxin from binding GSK-3. 44 When b-cateninaccumulates in cytoplasm, it translocates into nu-

    cleus and activates T-cell factor (TCF) and lymphoidenhancer element (LEF)-dependent gene transcrip-tion, through which the Wnt pathway controlsnumerous developmental processes. The full reper-toire of the genes containing LEF/TCF responsiveelements in their promoters has not been defined.Patterning genes Ultrabithorax in Drosophila, sia-mois , and nodal-related gene-3 in Xenopus ,4547 hu-man genes c-myc and cyclin D1 ,4850 and connexin43and E-cadherin 51,52 represent major LEF/TCF-respon-sive genes discovered to date. Other possibletargets of GSK-3 may include AP-1, CREB, NF- kB,heat shock protein 1, and CCAAT/enhancer binding proteins. 53 LEF1 was recently found to becritical to the generation of dentate gyrus granulecells and the development of the hippocampus inmice. 4547,54 In addition, GSK-3 could phosphorylateNF-AT, facilitating its export from the nucleus,and thus antagonizing NF-AT-dependent gene tran-scription. 55

    Lithium mimics Wnt signals by inhibition of GSK-3 both in vitro and in vivo . This suggests that the often-observed developmental effects of lithium, whichcould not be explained by inhibition of IMPase, may be in fact a result of inhibition of GSK-3 b .56,57 Valproicacid also inhibits GSK-3, 58 but its control over theb-catenin stability differs from lithium. While lithium

    stabilizes b-catenin via inhibition of GSK-3, VPA-induced b -catenin accumulation is through increasedexpression of b-catenin rather than stabilization of the protein. 59 GSK-3 has also been implicated insynaptic function. For example, GSK-3 b inhibitioninduces the synapsin I clustering in the formation of synapses and neurotransmitter release. 60 In addition,GSK-mediated phosphorylation of MAP-B and tauproteins regulate microtubule assembly and stabiliza-tion at synapses. 57,6163 Clinical studies have demon-strated that specific GSK-3 inhibitors mimic thetherapeutic action of mood stabilizers and mighttherefore be plausible drugs in treating bipolarpatients. 64

    Molecular basis of lithium actionRH Lenox and Le Wang

    137

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    4/10

  • 8/11/2019 4001306 A

    5/10

    development. 8890 Within brain and neurons in cul-ture, MARCKS protein is expressed in neurites andsynaptosomes, and is colocalized with synapticvesicles. 9193 PKC-MARCKS signaling has been im-plicated in vesicular trafficking of neurotransmitter inliving neurons, 94 and in the regulation of variousprocesses including synaptic transmission at nerveterminals. 80,81 Electrophysiological and behavioralstudies in heterozygote mutant mice have shown that50% reduction in MARCKS significantly affectslong-term potentiation (LTP) and hippocampallydependent behavior. 9597 These data indicate thatMARCKS plays an important role in the mediationof neuroplastic processes in the developing andmature CNS.

    Chronic treatment with lithium at therapeuticallyrelevant concentrations significantly downregulatesMARCKS protein and mRNA (50%) in both rat brainand immortalized hippocampal cells. 27,98,99 Directactivation of PKC by phorbol esters in immortalized

    hippocampal cells also downregulates the MARCKSprotein, 100 suggesting a role for PKC in the regulationof MARCKS gene ( Macs ) expression in brain. Thelithium-induced downregulation of MARCKS proteinis dependent upon the myo- inositol concentrationand the level of activation of receptor-coupled PIsignaling. 31 Addition of inositol completely reversesthe lithium-induced downregulation of MARCKS,indicating that the downregulation of MARCKS wasmediated via the phosphoinositol pathway. 31 Further-more, lithium-induced down regulation of MARCKSis only apparent after chronic, but not acute , admin-istration and persists beyond abrupt withdrawal of the drug for an extended period of time; parallelingthe clinical time course for the therapeutic effects of lithium during initial treatment as well as itsdiscontinuation. Of interest, the structurally unre-lated mood-stabilizer, valproic acid (VPA), shares theproperty of lithium in reducing the MARCKS expres-sion in hippocampus, but carbamazepine (CBZ) andother psychotropic agents (antianxiety, analgesic,antipsychotic, antidepressant, calcium-channel blocker) as well as other monovalent cations such asrubidium do not alter MARCKS regulation. 101,102 Onthe other hand, VPA-induced downregulation of MARCKS is inositol-independent, and has an addi-tive effect on MARCKS regulation, consistent with

    clinical data supporting greater efficacy of thecombined treatment in refractory bipolar patients. 101These findings point to a common mechanism, inwhich MARCKS may be a shared target for bothlithium and VPA. Thus, we have proposed that thereduction of MARCKS protein following long-termlithium administration alters pre/postsynaptic mem- brane structure and stabilizes aberrant neuronalsignaling in key brain regions of patients withBPD.3,5 The regulation of MARCKS expression repre-sents a highly valued target for the long-term action of mood stabilizers, not only by virtue of its underlyingneurobiological properties but also in light of itspharmacological sensitivity and selectivity for struc-

    turally unrelated drugs with efficacy in the prophy-lactic treatment of BPD. While MARCKS remains anattractive target for illustrative purposes in defining alithium-responsive gene network, it is evident thatother targets, such as prolyl oligopeptidase (POase),which appears to mediate the common effect of lithium, CBZ and VPA on growth cone stability andhas been implicated in affective disorders, may atsome point represent a similar opportunity. 103

    Perspective: linking the lithium-responsive genesas a network

    The mechanisms underlying the prophylactic treat-ment of BPD by lithium are likely to be hardwired inthe genomic DNA. Despite all the reports of howindividual gene expression can be modulated inresponse to lithiums exposure, there is as yet nostrategy available for the identification of the lithium-responsive genomic regulatory network in brain.

    Many studies have focused on determining the effectof lithium on one or a few genes at a time, anapproach that is not adequate for the analysis of largeregulatory control system organized as networks.Gene-specific expression is controlled by specificcis -regulatory target sequence embedded in promotersand the cognate binding transcription factors encoded by a set of regulatory genes. The functional linkages of such a genomic regulatory network include elementsthat exhibit multiple interactions between the outputsof regulatory genes and corresponding cis-regulatorygenomic sequences. Thus, identifying networks of transcription factors and the genes they regulate inresponse to lithium exposure is important to under-stand the biological responsiveness of an organism tolithium and the prophylactic properties of its actionin brain.

    Studies in our laboratory have demonstrated thatthe lithium-induced reduction of MARCKS proteinwas accompanied by a downregulation of MARCKSmRNA with no evidence for a change in the half-lifeof the mRNA. 99 Furthermore, synthesis of nascentRNA for MARCKS and the Macs promoter activitywere also found to be significantly reduced inchronic, but not acute, lithium-treated immortalizedhippocampal cells. Both reductions were signifi-cantly enhanced in the presence of activation of

    receptor-coupled PI signaling.99

    We have identifiedfor the first time a lithium-responsive promotersequence located in the upstream region ( 993/

    713) of the Macs promoter. 99 The mutant promoterlacking the 993/ 713 fragment not only did notrespond to chronic lithium expose but also had asignificantly reduced promoter activity, suggestingthat chronic lithium represses the transcriptionalactivator(s) bound to this region. Until now, however,no lithium-responsive transcription factor directly bound to this region has been identified. Whilespecific transcription factors that bind to lithium-responsive promoter element (LRE) remain to beidentified (Figure 2), current data suggest that the

    Molecular basis of lithium actionRH Lenox and Le Wang

    139

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    6/10

    mechanism by which chronic lithium represses theMacs gene transcription are likely through: (a)lithium-induced downregulation of DNA bindingactivity and/or expression of transcription activator(s)interacting with LRE; (b) lithium-induced down-regulation of activator function of transcriptionactivator(s) acting at LRE; (c) lithium-induced disrup-tion of the coordinate interaction between the distalactivating sequence-bound activator(s) and the prox-imal core promoter sequence-bound basal transcrip-tional machinery. The lithium-responsive 993/ 713fragment contains significant enhancer/activator DNAelements that are necessary to sustain the optimalMacs gene transcription in cells. 104 While an atypicalSp1 site, characterized by the presence of a prominentGA-rich sequence, was identified within the 993/

    713, a classical Sp1 site was found in the middle of the GC-rich box close to a potential Z-DNA-formingsegment near the transcription initiation site. 104Presence of Z-DNA forming segment signifies both

    compositional and conformational changes influen-cing the genomic landscape, the accessibility of cis-acting elements, and the activation of gene transcrip-tion in the core promoter region. 105 As summarized inFigure 2, Macs promoter lacks typical TATA box,and in the absence of a TATA box multiple Sp1 sitesin the promoter may control the transcription of Macsgene.

    A recent microarray experiment further showedthat the expression of as many as 37 genes from 4132rat genes is altered during the chronic lithiumtreatment. 106 If these data were extrapolated to theentire genome, there would be as many as 750 genesthat could potentially be regulated by lithium.However, the majority of such studies using differ-ential display and microarray analysis do not distin-guish between direct activation of target genes by atranscription factor and indirect effects resulting fromone transcription factor inducing the expression of asecond. Furthermore, the functional value of the vastmajority of these potential targets remains unknown.Using our knowledge regarding a high-value pharma-cologically relevant target such as MARCKS for theaction of chronic lithium in brain to identify lithium-responsive elements that could serve to link lithium-responsive genes as a regulatory network is bothintriguing and challenging. To establish such a genenetwork, some studies have coupled the overexpres-

    sion of a given transcription factor with microarrayexperiments. However, the genes affected by theoverexpressed transcription factors may not representthe true targets of those factors under physiologicalconditions. Recently, using finite perturbations of gene expression in conjunction with microarrayexperiments that may mimic physiological conditionshas been advocated; thus differing from the drastic

    Figure 2 Lithium-responsive promoter of the Macs gene. The Promoter region upstream of Macs is pharmacologicallyresponsive to chronic, but not acute, lithium exposure. A hypothetical activator complex binds to the upstream region of Macs promoter and crosstalk with proteins in proximal promoter region including TBP, TAFs, Sp1, hypothetical tetheringproteins, etc. The gray rectangle box represents cis-regulatory enhancer elements (LRE) that may interact with specificlithium-responsive transcription factors. Long-term treatment with lithium is postulated to repress the Macs transcriptionthrough: (a) lithium-induced reduction in the DNA binding of the transcription activator(s) acting at the LRE; (b) lithium-induced alteration in the expression and/or the transactivation function of lithium-responsive transcription activator(s)acting at the LRE; (c) lithium-induced disruption of the interaction between enhancer complex formed at LRE and theproximal transcription initiator. In the figure, # refers to potential intermediary factors mediating the indirect action of chronic lithium on the Macs promoter.

    Molecular basis of lithium actionRH Lenox and Le Wang

    140

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    7/10

    changes incurred during transgenic overexpression orknockouts. 107 In such studies, one can modulatesingle gene expression using techniques such asRNA interference (RNAi) one at a time and in asystematic fashion among all the genes of interest.Once the perturbed systems are in a new steady state,the levels of gene expression is measured against thatof reference by microarray or Taqman RCR. Theresulting data are arranged as a regulatory strengthmatrix, from which a regulatory network exhibitinggenegene interactions could be inferred using ap-proaches adapted from metabolic control analysis. 108The method, however, lacks genome-wide efficiencyand does not provide direct evidence of in vivointeraction of given transcription factors with theirrespective cis-regulatory elements across the genome.To address these questions, a new technology knownas chIpchip was developed, in which chromatinimmunoprecipitation (chIp) is coupled with micro-array experiment (chip) to accelerate the genome-

    wide detection of DNAprotein interactions in realtime and in real space. Using such a combinedapproach as chIpchip, genome-wide mapping of

    DNA binding sites for transcription factors (STE12,GAL4, RAP1, SCB, MCB, MCM1, SFF, and SW15) has been achieved in yeast. 109,110 This experimentalapproach has also been successfully extended tohuman genome, where DNA binding sites for E2Fand GATA-1 transcription factors are mapped genomewide. 111113

    A lithium-responsive gene network will offer anopportunity to define a pathway associated with thelong-term prophylactic properties of lithium, distinctfrom its side-effect profile, which will drive thediscovery of novel agents for stabilization of moodin patients with BPD (Figure 3). Recently, a regulatorygene network that directs specific developmentalevents has been identified in developing sea urchinembryo. 114,115 This provides a heuristic model forconstructing a lithium-responsive gene network asa means to identify signature genes that directthe therapeutic or nontherapeutic action of lithium.While chromosome immunoprecipitation could start

    with antibodies against several known lithium-responsive transcription factors such as AP-1, CREB,NF-kB, LEF/TCF, these are general transcription

    Figure 3 Model for a gene network for lithium-responsive genes: defining the therapeutic effect of chronic lithium. A genenetwork can be reconstructed by perturbation microarray and chlpchip methods (see test for details). A lithium-responsivegene network will consist of a distinctive subset of genes that share a lithium-responsive element (LRE) as defined within theMacs gene which is regulated by specific transcription factors referred to as TFs that are yet to be identified. Such a lithium-responsive gene network (TF1) may subserve critical processes in the brain underlying cytoskeletal remodeling andregulation of synaptic signaling, while a different subset of genes defined by lithium-responsive elements regulated bytranscription factors (TF2, TF3) may underlie processes such as cell proliferation. Since these different network of genesunderlie different physiological sequela of chronic lithium, modulation of gene expression involved in the cytoskeletalrestructuring and neuroplasticity by lithium may prove to be fundamental to the mood-stabilizing properties of lithium in the brain, whereas regulation of genes involved in cell proliferation and immune response may be linked to lithiumsleukocytotic and antiviral effects.

    Molecular basis of lithium actionRH Lenox and Le Wang

    141

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    8/10

    factors that elicit far more gene expression thannecessary for the therapeutic action of lithium. Thus,identifying lithium-responsive transcription factorsor promoter elements from highly valued targets suchas the Macs gene becomes extremely desirable. Wedemonstrated that Macs promoter is pharmacologi-cally responsive to chronic, but not acute, lithiumtreatment at therapeutically relevant concentrations,and possesses pharmacological properties consistentwith its expressed protein and clinical properties of lithium. 99 We further showed that the sites for lithiumresponsiveness lie within an enhancer element of thepromoter that has the potential to interact witha unique lithium-responsive activator complex(Figure 3). 104 This raises the possibility that uniquetranscription factors sensitive to lithium treatmentmay be identifiable through this promoter. Whilegenes like Macs involved in the cytoskeletal restruc-turing, synaptic transmission, and neuroplasticitymay relate preferentially to the long-term mood-

    stabilizing properties of chronic lithium in the brain,genes underlying cell proliferation and immuneresponse may serve to identify pathways associatedwith lithiums leukocytotic and antiviral effects(Figure 3). Thus, defining a lithium-responsive genenetwork will provide the data for pathway mapping of novel targets with better-defined mood-stabilizingproperties for the long-term treatment of BPD.

    References

    1 Goodwin FK, Ghaemi SN. The impact of the discovery of lithiumon psychiatric thought and practice in the USA and Europe. Aust NZ J Psychiatry 1999; 33(Suppl): S54S64.

    2 Lenox RH, Manji HK. Lithium. In: Nemeroff C, Schatzberg A(eds). APA Textbook of Psychopharmacology . Washington, DC:APA Press, Inc: 1998, pp. 303314.

    3 Lenox RH, Hahn CG. Overview of the mechanism of action of lithium in the brain: fifty-year update. J Clin Psychiatry 2000; 61:515.

    4 Hudson CJ, Young LT, Li PP, Warsh JJ. CNS signal transduction inthe pathophysiology and pharmacotherapy of affective disordersand schizophrenia. Synapse 1993; 13: 278293.

    5 Lenox RH. Role of receptor coupling to phosphoinositidemetabolism in the therapeutic action of lithium. In: Ehrlich Y(ed). Molecular Mechanisms of Neuronal Responsiveness . NewYork: Plenum. Adv Exp Med 1987; 221: 515530.

    6 Post RM. Transduction of psychosocial stress into the neuro- biology of recurrent affective disorder. Am J Psychiatry 1992; 149:9991010.

    7 Suppes T, Baldessarini RJ, Faedda GL, Tohen M. Risk of recurrence following discontinuation of lithium treatment in bipolar disorder. Arch Gen Psychiatry 1991; 48: 10821088.

    8 Faedda GL, Tondo L, Baldessarini RJ, Suppes T, Tohen M.Outcome after rapid vs gradual discontinuation of lithiumtreatment in bipolar disorders. Arch Gen Psychiatry 1993; 50:448455.

    9 Tondo L, Baldessarini RJ, Hennen J, Floris G. Lithium main-tenance treatment of depression and mania in bipolar I and bipolar II disorders. Am J Psychiatry 1998; 155: 638645.

    10 Lenox RH, McNamara RK, Papke RL, Manji HK. Neurobiology of lithium: an update. J Clin Psychiatry 1998; 59: 3747.

    11 Manji HK, Lenox RH. Lithium: a molecular transducer of mood-stabilization in the treatment of bipolar disorder. Neuropsycho- pharmacology 1998; 19: 161166.

    12 Jope RS. Anti-bipolar therapy: mechanism of action of lithium.Mol Psychiatry 1999; 4: 117128.

    13 Williams RS, Harwood AJ. Lithium therapy and signal transduc-tion. Trends Pharmacol Sci 2000; 21: 6164.

    14 Manji HK, Lenox RH. Signaling: cellular insights into thepathophysiology of bipolar disorder [In Process Citation]. Biol Psychiatry 2000; 48: 518530.

    15 Hyman SE, Nestler EJ. Initiation and adaptation: a paradigm forunderstanding psychotropic drug action. Am J Psychiatry 1996;153 : 151162.

    16 York JD, Ponder JW, Majerus PW. Definition of a metal-dependent/Li(+)-inhibited phosphomonoesterase protein family based upon a conserved three-dimensional core structure. Proc Natl Acad Sci USA 1995; 92: 51495153.

    17 Phiel C, Klein P. Molecular targets of lithium action. Annu Rev Pharmacol Toxicol 2001; 41: 789813.

    18 Hallcher LM, Sherman WR. The effects of lithium ion and otheragents on the activity of myo-inositol-1-phosphatase from bovine brain. J Biol Chem 1980; 255: 1089610901.

    19 Sherman WR, Gish BG, Honchar MP, Munsell LY. Effects of lithium on phosphoinositide metabolism in vivo . Fed Proc 1986;45: 26392646.

    20 Irvine RF. Nuclear lipid signaling. Sci STKE 2002; 2002: RE13.21 Majerus PW. Inositol phosphate biochemistry. Annu Rev Bio-

    chem 1992; 61: 225250.22 Acharya JK, Labarca P, Delgado R, Jalink K, Zuker CS. Synaptic

    defects and compensatory regulation of inositol metabolism ininositol polyphosphate 1-phosphatase mutants. Neuron 1998; 20:12191229.

    23 Manji HK, Lenox RH. Ziskind-Somerfeld Research Award.Protein kinase C signaling in the brain: molecular transductionof mood stabilization in the treatment of manic-depressiveillness. Biol Psychiatry 1999; 46: 13281351.

    24 Nahorski SR, Ragan CI, Challiss RA. Lithium and the phosphoi-nositide cycle: an example of uncompetitive inhibition and itspharmacological consequences. Trends Pharmacol Sci 1991; 12:297303.

    25 Berridge MJ, Downes CP, Hanley MR. Lithium amplifies agonist-dependent phosphatidylinositol responses in brain and salivaryglands. Biochem J 1982; 206: 587595.

    26 Kendall DA, Nahorski SR. Acute and chronic lithium treatmentsinfluence agoinst and depolarization-stimulated inositol phos-

    pholipid hydrolysis in rat cerebral cortex. J Pharmacol Exp Ther 1987; 241: 10231027.27 Kennedy ED, Challiss RA, Nahorski SR. Lithium reduces the

    accumulation of inositol polyphosphate second messengersfollowing cholinergic stimulation of cerebral cortex slices. J Neurochem 1989; 53: 16521655.

    28 Kennedy ED, Challiss RA, Ragan CI, Nahorski SR. Reducedinositol polyphosphate accumulation and inositol supply in-duced by lithium in stimulated cerebral cortex slices. Biochem J 1990; 267: 781786.

    29 Kofman O, Belmaker RH, Grisaru, N, Alpert C, Fuchs I, Katz V,Rigler O. Myo-inositol attenuates two specific behavioral effectsof acute lithium in rats. Psychopharmacol Bull 1991; 27: 185190.

    30 Tricklebank MD, Singh L, Jackson A, Oles RJ. Evidence that aproconvulsant action of lithium is mediated by inhibition of myo-inositol phosphatase in mouse brain. Brain Res 1991; 558:145148.

    31 Watson DG, Lenox RH. Chronic lithium-induced down-regula-tion of MARCKS in immortalized hippocampal cells: potentia-tion by muscarinic receptor activation. J Neurochem 1996; 67:767777.

    32 Fisher SK, Novak JE, Agranoff BW. Inositol and higher inositolphosphates in neural tissues: homeostasis, metabolism andfunctional significance. J Neurochem 2002; 82: 736754.

    33 Gani D, Downes CP, Batty I, Bramham J. Lithium and myo-inositol homeostasis. Biochim Biophys Acta 1993; 1177:253269.

    34 Moore GJ, Bedchuk JM, Parrish JK, Faulk MW, Arjken CL, Strahl-Bevacqua J, Marji HK. Temporal dissociation between lithium-induced changes in frontal lobe myo-inositol and clinicalresponse in manic-depressive illness. Am J Psychiatry 1999;156: 19021908.

    Molecular basis of lithium actionRH Lenox and Le Wang

    142

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    9/10

    35 Manji HK, Lenox RH. Long-term action of lithium: a role fortranscriptional and posttranscriptional factors regulated byprotein kinase C. Synapse 1994; 16: 1128.

    36 Manji HK, Etcheberrigaray R, Chen G, Olds JL. Lithium decreasesmembrane-associated protein kinase C in hippocampus: selectiv-ity for the alpha isozyme. J Neurochem 1993; 61: 23032310.

    37 Manji HK, Bersudsky Y, Chen G, Belmaker RH, Potter WZ.Modulation of protein kinase C isozymes and substrates bylithium: the role of myo-inositol. Neuropsychopharmacology 1996; 15: 370381.

    38 Jope RS, Song L. AP-1 and NF-kappaB stimulated by carbachol inhuman neuroblastoma SH-SY5Y cells are differentially sensitiveto inhibition by lithium. Brain Res Mol Brain Res 1997; 50: 171180.

    39 Wang HY, Markowitz P, Levinson D, Undie AS, Freidman E.Increased membrane-associated protein kinase C activity andtranslocation in blood platelets from bipolar affective disorderpatients. J Psychiatr Res 1999; 33: 171179.

    40 Soares JC, Chen G, Dippold CS et al . Concurrent measures of protein kinase C and phophoinositides in lithium-treated bipolarpatients and healthy individuals: a preliminiary study. Psychia-try Res 2000; 95: 109118.

    41 Wang H, Freidman E. Increased association of brain proteinkinase C with the receptor for activated C kinase-1 (RACK1) in

    bipolar affective disorder. Biol Psychiatry 2001; 50: 364370.42 Bebchuk JM, Arfken CL, Dolan-Manji S, Murphy J, Hasanat K,Manji HK. A preliminary investigation of a protein kinase Cinhibitor in the treatment of acute mania. Arch Gen Psychiatry 2000; 57: 9597.

    43 Orford K, Crockett C, Jensen JP, Weissman AM, Byers SW. Serinephosphorylation-regulated ubiquitination and degradation of beta-catenin. J Biol Chem 1997; 272: 2473524738.

    44 Farr III GH, Ferkey DM, Yost C, Pierce SB, Weaver C, KimelmanD. Interaction among GSK-3, GBP, axin, and APC in Xenopus axisspecification. J Cell Biol 2000; 148: 691702.

    45 McKendry R, Hsu SC, Harland RM, Grosschedl R. LEF-1/TCFproteins mediate wnt-inducible transcription from the Xenopusnodal-related 3 promoter. Dev Biol 1997; 192: 420431.

    46 Brannon M, Gomperts M, Sumoy L, Moon RT, Kimelman D. A beta-catenin/XTcf-3 complex binds to the siamois promoter toregulate drosal axis specification in Xenopus. Genes Dev 1997;11: 23592370.

    47 Riese J, Yu X, Munnerlyn A et al . LEF-1, a nuclear factorcoordinating signaling inputs from wingless and decapentaple-gic. Cell 1997; 88: 777787.

    48 Shtutman M, Zhurinsky J, Simcha I et al . The cyclin D1 gene is atarget of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA 1999; 96: 55225527.

    49 Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 1999; 398: 422426.

    50 He TC, Sparks AB, Rago C et al . Identification of c-MYC as atarget of the APC pathway. Science 1998; 281: 15091512.

    51 van der Heyden MA, Rook MB, Hermans MM et al . Identificationof connexin43 as a functional target for Wnt signalling. J Cell Sci 1998; 111: 17411749.

    52 Bradley RS, Cowin P, Brown AM. Expression of Wnt-1 in PC12cells results in modulation of plakoglobin and E-cadherin and

    increased cellular adhesion. J Cell Biol 1993; 123: 18571865.53 Mai L, Jope RS, Li X. BDNF-mediated signal transduction ismodulated by GSK3beta and mood stabilizing agents. J Neuro-chem 2002; 82: 7583.

    54 Galceran J, Miyashita-Lin EM, Devaney E, Rubenstein JL,Grosschedl R. Hippocampus development and generation of dentate gyrus granule cells is regulated by LEF1. Development 2000; 127: 469482.

    55 Graef IA, Mermelstein PG, Stankunas K et al . L-type calciumchannels and GSK-3 regulate the activity of NF-ATc4 inhippocampal neurons. Nature 1999; 401: 703708.

    56 Hedgepeth CM, Conrad LJ, Zhang J, Huang HC, Lee VM, Klein PS.Activation of the Wnt signaling pathway: a molecular mechanismfor lithium action. Dev Biol 1997; 185: 8291.

    57 Klein PS, Melton DA. A molecular mechanism for the effect of lithium on development. Proc Natl Acad Sci USA 1996; 93:84558459.

    58 Yuan P-X, Huang L-D, Jiang Y-M, Gutkind JS, Manji HK, Chen G.The mood stabilizer valproic acid activates mitogen-activatedprotein kinases and promotes neurite growth. J Biol Chem 2001;276: 3167431683.

    59 Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS.Histone deacetylase is a direct target of valproic acid, a potentanticonvulsant, mood stabilizer, and teratogen. J Biol Chem 2001;276: 3673436741.

    60 Hall AC, Brennan A, Goold RG et al . Valproate regulatesGSK-3-mediated axonal remodeling and synapsin I cluster-ing in developing neurons. Mol Cell Neurosci 2002; 20:257270.

    61 Lovestone S, Davis DR, Webster MT et al . Lithium reduces tauphosphorylation: effects in living cells and in neurons attherapeutic concentrations. Biol Psychiatry 1999; 45: 9951003.

    62 Hong M, Chen DC, Klein PS, Lee VM. Lithium reduces tauphosphorylation by inhibition of glycogen synthase kinase-3. J Biol Chem 1997; 272: 2532625332.

    63 Munoz-Montano JR, Moreno FJ, Avila J, Diaz-Nido J. Lithiuminhibits Alzheimers disease-like tau protein phosphorylation inneurons. FEBS Lett 1997; 411: 183188.

    64 Eldar-Finkelman H. Glycogen synthase kinase 3: an emergingtherapeutic target. Trends Mol Med 2002; 8: 126132.

    65 Goode N, Hughes K, Woodgett JR, Parker PJ. Differential

    regulation of glycogen synthase kinase-3 beta by protein kinaseC isotypes. J Biol Chem 1992; 267: 1687816882.66 Chen RH, Ding WV, McCormick F. Wnt signaling to beta-catenin

    involves two interactive components. Glycogen synthase kinase-3beta inhibition and activation of protein kinase. C. J Biol Chem2000; 275: 1789417899.

    67 Cook D, Fry M, Hughes K, Sumathipala R, Woodgett J, Dale T.Wingless inactivates glycogen synthase kinase-3 via an intracel-lular signalling pathway which involves a protein kinase C.EMBO J 1996; 15: 45264536.

    68 Sheldahl LC, Park M, Malbon CC, Moon RT. Protein kinase C isdifferentially stimulated by Wnt and Frizzled homologs in a G-protein-dependent manner. Curr Biol 1999; 9: 695698.

    69 Malbon CC, Wang H, Moon RT. Wnt signaling and heterotrimericG-proteins: strange bedfellows or a classic romance? BiochemBiophys Res Commun 2001; 287: 589593.

    70 Liu T, DeCostanzo AJ, Liu X et al . G protein signaling fromactivated rat frizzled-1 to the beta-catenin-Lef-Tcf pathway.Science 2001; 292: 17181722.

    71 Chen G, Hasanat KA, Bebchuk JM, Moore GJ, Glitz D, Manji HK.Regulation of signal transduction pathways and gene expression by mood stabilizers and antidepressants. Psychosom Med 1999;61: 599617.

    72 Chen RW, Chuang DM. Long term lithium treatment suppressesp53 and Bax expression but increases Bcl-2 expression. Aprominent role in neuroprotection against excitotoxicity. J Biol Chem 1999; 274: 60396042.

    73 Chalecka-Franaszek E, Chuang DM. Lithium activates the serine/threonine kinase Akt-1 and suppresses glutamate-induced in-hibition of Akt-1 activity in neutrons. Proc Natl Acad Sci USA1999; 96: 87458750.

    74 Berglund K, Midorikawa M, Tachibana M. Increase in the poolsize of releasable synaptic vesicles by the activation of protein

    kinase C in goldfish retinal bipolar cells. J Neurosci 2002; 22:47764785.75 Hori T, Takai Y, Takahashi T. Presynaptic mechanism for phorbol

    ester-induced synaptic potentiation. J Neurosci 1999; 19: 72627267.

    76 Voets T, Toonen RF, Brain EC et al . Munc18-1 promotes largedense-core vesicle docking. Neuron 2001; 31: 581591.

    77 Cordeiro ML, Umbach JA, Gundersen CB. Lithium ions enhancecysteine string protein gene expression in vivo and in vitro . J Neurochem 2000; 74: 23652372.

    78 Buchner E, Gundersen CB. The DnaJ-like cysteine string proteinand exocytotic neurotransmitter release. Trends Neurosci 1997;20: 223227.

    79 Heckmann M, Adelsberger H, Dudel J. Evoked transmitter releaseat neuromuscular junctions in wild type and cysteine stringprotein null mutant larvae of Drosophila. Neurosci Lett 1997; 228:167170.

    Molecular basis of lithium actionRH Lenox and Le Wang

    143

    Molecular Psychiatry

  • 8/11/2019 4001306 A

    10/10

    80 Aderem A. The MARCKS brothers: a family of protein kinase Csubstrates. Cell 1992; 71: 713716.

    81 Blackshear PJ. The MARCKS family of cellular protein kinase Csubstrates. J Biol Chem 1993; 268: 15011504.

    82 Laux T, Fukami K, Thelen M, Golub T, Frey D, Caroni P. GAP43,MARCKS, and CAP23 modulate PI(4,5)P(2) at plasmalemmalrafts, and regulate cell cortex actin dynamics through a commonmechanism. J Cell Biol 2000; 149: 14551472.

    83 Glaser M, Wanaski S, Buser CA et al . Myristoylated alanine-richC kinase substrate (MARCKS) produces reversible inhibition of phospholipase C by sequestering phosphatidylinositol 4,5-bi-sphosphate in lateral domains. J Biol Chem 1996; 271: 2618726193.

    84 Morash SC, Rose SD, Byers DM, Ridgway ND, Cook HW.Overexpression of myristoylated alanine-rich C-kinase substrateenhances activation of phospholipase D by protein kinase C inSK-N-MC human neuroblastoma cells. Biochem J 1998; 332:321327.

    85 Goodall AR, Turner NA, Walker JH, Ball SG, Vaughan PF.Activation of protein kinase C-alpha and translocation of themyristoylated alanine-rich C-kinase substrate correlate withphorbol ester-enhanced noradrenaline release from SH-SY5Yhuman neuroblastoma cells. J Neurochem 1997; 68: 392401.

    86 Li Y, Martin LD, Spizz G, Adler KB. MARCKS protein is a keymolecule regulating MUCIN secretion by human airway epithe-lial cells in vitro . J Biol Chem 2001; 276: 4098240990.

    87 Allen LH, Aderem A. A role for MARCKS, the alpha isozyme of protein kinase C and myosin I in zymosan phagocytosis bymacrophages. J Exp Med 1995; 182: 829840.

    88 McNamara RK, Lenox RH. Distribution of the protein kinase Csubstrates MARCKS and MRP in the postnatal developing rat brain. J Comp Neurol 1998; 397: 337356.

    89 Stumpo DJ, Bock CB, Tuttle JS, Blackshear PJ. MARCKSdeficiency in mice leads to abnormal brain development andperinatal death. Proc Natl Acad Sci USA 1995; 92: 944948.

    90 Swierczynski SL, Siddhanti SR, Tuttle JS, Blackshear PJ.Nonmyristoylated MARCKS complements some but not all of the developmental defects associated with MARCKS deficiencyin mice. Dev Biol 1996; 179: 135147.

    91 Wang JK, Walaas SI, Sihra TS, Aderem A, Greengard P.Phosphorylation and associated translocation of the 87-kDa

    protein, a major protein kinase C substrate, in isolatednerve terminals. Proc Natl Acad Sci USA 1989; 86:22532256.

    92 Lu D, Yang H, Lenox RH, Raizada MK. Regulation of angiotensinII-induced neuromodulation by MARCKS in brain neurons. J Cell Biol 1998; 142: 217227.

    93 Ouimet CC, Wang JK, Walaas SI, Albert KA, Greengard P.Localization of the MARCKS (87 kDa) protein, a major specificsubstrate for protein kinase C, in rat brain. J Neurosci 1990; 10:16831698.

    94 Yang H, Wang X, Sumners C, Raizada MK. Obligatory role of protein kinase Cbeta and MARCKS in vesicular trafficking inliving neurons. Hypertension 2002; 39: 567572.

    95 McNamara RK, Stumpo DJ, Morel LM et al . Effect of reducedmyristoylated alanine-rich C kinase substrate expressionon hippocampal mossy fiber development and spatiallearning in mutant mice: transgenic rescue and interactionswith gene background. Proc Natl Acad Sci USA 1998; 95: 1451714522.

    96 Ramakers GM, McNamara RK, Lenox RH, De Graan PN.Differential changes in the phosphorylation of the protein kinaseC substrates myristoylated alanine-rich C kinase substrate andgrowth-associated protein-43/B-50 following Schaffer collateral

    long-term potentiation and long-term depression. J Neurochem1999; 73: 21752183.

    97 Hussain R, McNamara RK, Stumpo DJ et al . Effect of MARCKSoverexpression on hippocampal long-term potentiation andhippocampal-dependent learning in transgenic mice. Soc Neu-rosci Abstracts 2000; 26: 1511.

    98 Lenox RH, Watson DG, Patel J, Ellis J. Chronic lithiumadministration alters a prominent PKC substrate in rat hippo-campus. Brain Res 1992; 570: 333340.

    99 Wang L, Liu X, Lenox RH. Transcriptional down-regulation of MARCKS gene expression in immortalized hippocampal cells bylithium. J Neurochem 2001; 79: 816825.

    100 Watson DG, Wainer BH, Lenox RH. Phorbol ester- and retinoicacid-induced regulation of the protein kinase C substrateMARCKS in immortalized hippocampal cells. J Neurochem1994; 63: 16661674.

    101 Watson DG, Watterson JM, Lenox RH. Sodium valproate down-regulates the myristoylated alanine-rich C kinase substrate(MARCKS) in immortalized hippocampal cells: a property of protein kinase C-mediated mood statilizers. J Pharmacol ExpTher 1998; 285: 307316.

    102 Lenox RH, McNamara RK, Watterson JM, Watson DG. Myristoy-lated alanine-rich C kinase substrate (MARKCS): a moleculartarget for the therapeutic action of mood stabilizers in the brain?

    J Clin Psychiatry 1996; 57: 2331; discussion 3233.103 Williams RS, Cheng L, Mudge AW, Harwood AJ. A common

    mechanism of action for three mood-stabilizing drugs. Nature2002; 417: 292295.

    104 Wang L, Liu X, Lenox RH. Transcriptional regulation of mouseMARCKS promoter in immortalized hippocampal cells. BiochemBiophys Res Commun 2002; 292: 969979.

    105 Herbert A, Rich A. The biology of left-handed Z-DNA. J Biol Chem 1996; 271: 1159511598.

    106 Bosetti F, Seemann R, Bell JM et al . Analysis of gene expressionwith cDNA microarrays in rat brain after 7 and 42 days of orallithium administration. Brain Res Bull 2002; 57: 205209.

    107 de la Fuente A, Brazhnik P, Mendes P. Linking the genes:inferring quantitative gene networks from microarray data.Trends Genet 2002; 18: 395398.

    108 Bowden AC. Metabolic control analysis in biotechnology and

    medicine. Nat Biotechnol 1999; 17: 641643.109 Iyer VR, Horak CE, Scafe CS, Botstein D, Snyder M, Brown PO.Genomic binding sites of the yeast cell-cycle transcription factorsSBF and MBF. Nature 2001; 409: 533538.

    110 Ren B, Robert F, Wyrick JJ et al . Genome-wide location andfunction of DNA binding proteins. Science 2000; 290: 23062309.

    111 Weinmann AS, Yan PS, Oberley MJ, Huang TH, Famham PJ.Isolating human transcription factor targets by coupling chroma-tin immunoprecipitation and CpG island microarray analysis.Genes Dev 2002; 16: 235244.

    112 Horak CE, Mahajan MC, Luscombe NM, Gerstein M, WeissmanSM, Snyder M. GATA-1 binding sites mapped in the beta-globinlocus by using mammalian chlpchip analysis. Proc Natl Acad Sci USA 2002; 99: 29242929.

    113 Weinmann AS, Farnham PJ. Identification of unknown targetgenes of human transcription factors using chromatin immuno-precipitation. Methods 2002; 26: 3747.

    114 Davidson EH, Rast JP, Oliveri P et al . A provisional regulatorygene network for specification of endomesoderm in the seaurchin embryo. Dev Biol 2002; 246: 162190.

    115 Oliveri P, Carrick DM, Davidson EH. A regulatory gene networkthat directs micromere specification in the sea urchin embryo.Dev Biol 2002; 246: 209228.

    Molecular basis of lithium actionRH Lenox and Le Wang

    144

    Molecular Psychiatry