Associate editor: B.L. Roth
Lithium and valproic acid: parallels and contrasts in diverse signaling contexts

https://doi.org/10.1016/S0163-7258(02)00299-1Get rights and content

Abstract

The introduction of lithium salts to treat bipolar disorder (BPD) revolutionized the therapy of psychiatric illnesses, but the pathogenesis of the disease and the mechanism of lithium action remain unknown. While several direct molecular targets of lithium have been identified, it is unknown which, if any, of these targets plays a role in the therapeutic response to lithium. Exposure to lithium evokes a wide spectrum of behavioral, physiological, and developmental responses in diverse organisms, and these effects have been exploited to explore the mechanisms of lithium action. Valproic acid (VPA), a widely used anticonvulsant, is also an effective therapy for BPD, and again its mechanism of action is not known, although new in vitro targets have been identified recently. In this review, the clinical, physiological, and developmental effects of lithium and VPA are summarized and recent work on direct targets for lithium and VPA is discussed in the context of these effects. We then describe some of the physiological effects common to the two drugs, in addition to treatment of BPD, and address signaling pathways that could be regulated by both lithium and VPA.

Introduction

The introduction of lithium salts for the treatment of bipolar disorder (BPD) by John Cade in 1949 revolutionized the therapy of this and other severe psychiatric illnesses. This landmark discovery not only brought an effective pharmacotherapy to this disorder for the first time, it also changed the public and scientific perception of psychiatric illnesses and triggered still ongoing attempts to define a pathophysiological mechanism for the origin and treatment of this common disorder (Manji et al., 1999a).

BPD is a prevalent and debilitating illness, affecting ∼1% of the population worldwide (Weissman et al., 1996). BPD is associated with significant morbidity and mortality, with suicide as a cause of death in as many as 10% of patients. Little is known about the pathogenesis of this or other affective disorders, but it responds remarkably well to mood-stabilizing drugs, such as lithium and the anticonvulsant valproic acid (VPA). More recently, other anticonvulsants, including carbamazepine (CBZ) and lamotrigine, have also been used in bipolar patients, and the effectiveness of these alternative regimens is reviewed in depth elsewhere (De Leon, 2001). In this review, we will focus on the physiological effects and potential molecular targets of lithium and VPA.

Despite their wide use in the pharmacotherapy of BPD, the mechanism of action of these drugs remains unknown. Both lithium and VPA have multiple actions in humans, model organisms, and cell culture systems, and these actions only partially overlap. Furthermore, a large number of indirect targets, as well as a more limited number of direct in vitro targets, for these two drugs has been described. The direct targets described in vitro offer plausible, but unproven, candidates to explain the in vivo effects of these drugs in various settings. In some cases, a strong argument can be made for inhibition of a given target molecule to explain in vivo drug effects. For example, inhibition of glycogen synthase kinase-3 (GSK-3) likely explains the effects of lithium on the developmental program of Xenopus embryos and Dictyostelium discoideum, inhibition of inositol polyphosphate 1-phosphatase (IPPase) provides a compelling explanation for the effect of lithium on the neuromuscular junction in Drosophila, and inhibition of phosphoglucomutase (PGM) is a likely explanation for the toxicity of lithium in yeast grown on galactose. However, most of the known, direct targets of lithium and VPA are ubiquitously expressed and are implicated as regulatory factors in diverse settings. Therefore, it is important to keep in mind that perturbation of a single molecular target is unlikely to explain all of the effects of either drug, and also that modulation of more than one direct target may be responsible for their efficacy in a complex disease process such as BPD Jope, 1999, Manji et al., 1996.

Several recent reviews have addressed the possible mechanisms of action for either lithium or VPA Baraban, 1994, Johannessen, 2000, Jope & Williams, 1994, Lenox et al., 1998, Lenox et al., 2002, Manji & Lenox, 1999, Stoll & Severus, 1996, Phiel & Klein, 2001, Tunnicliff, 1999, Williams & Harwood, 2000 and, in addition, numerous reviews and an excellent monograph are available concerning the indirect, as well as clinical, effects of these drugs (Goodwin & Jamison, 1990). Thus, we will only briefly discuss the clinical, physiological, and developmental effects of lithium and VPA to provide a context for the molecular discussion to follow. We will also summarize some of the recent work on direct, in vitro targets for lithium and VPA. Finally, physiological effects and potential signaling pathways that may be regulated by both drugs will be discussed, as these may reflect a common molecular pathway for the action of lithium and VPA in the treatment of BPD, and this would provide insight into the pathogenesis and pharmacotherapy of BPD.

Section snippets

Clinical effects

Lithium salts were used therapeutically in the 19th century as soporifics and gout remedies El-Mallakh & Jefferson, 1999, Felber, 1987, Johnson & Amdisen, 1983, Rogers, 1989 and have been used for BPD since the late 1940s (Cade, 1949). Cade's work was followed up and validated by a series of European studies (Schou, 2001), and despite some setbacks, especially in the United States, it has been firmly established as the first line of therapy for BPD Goodwin & Jamison, 1990, Lenox et al., 1998,

Clinical effects

VPA is a short-chain, branched fatty acid originally used as a solvent (Fig. 4). The effectiveness of VPA as an anticonvulsant was discovered serendipitously when other compounds were dissolved in VPA for administration to animals used in experimental models of epilepsy Meunier et al., 1963, Tunnicliff, 1999. Since then, VPA has been used to control a variety of seizures, including generalized, partial, and absence seizures Johannessen, 2000, Tunnicliff, 1999. VPA has also been shown to provide

Common molecular targets for lithium and valproic acid

Lithium and VPA are highly effective in the treatment of BPD, and yet, in other aspects, such as anticonvulsant activity, teratogenicity, and clinically significant side effects, they appear to differ considerably. Thus, identification of a common target or signaling pathway that is regulated by both lithium and VPA may provide insights specifically into the mechanism of these drugs in BPD. So far, no molecule has been identified as a common direct target for lithium and VPA, but a number of

Conclusion

While the literature on lithium and VPA effects is vast, surprisingly few direct targets of either drug have been identified. This is especially surprising because these two drugs are simple in structure, and, at least in the case of lithium, inhibition occurs through a simple mechanism (competition for divalent cations). The few molecular targets of lithium and VPA identified so far share the characteristic that each is involved in basic regulatory pathways and thus, their inhibition is likely

Acknowledgements

We wish to thank Patricia Salinas and Armin Manoukian for permission to discuss their unpublished data. We also thank Jonathon Raper and Christopher Phiel for helpful discussions. In addition, we greatly appreciate the thoughtful comments of the referees. Work on lithium and VPA in this laboratory is supported by grants from the NIH. P.S.K. is an Assistant Investigator in the Howard Hughes Medical Institute.

References (293)

  • W.B. Busa et al.

    Lithium-induced teratogenesis in frog embryos prevented by a polyphosphoinositide cycle intermediate or a diacylglycerol analog

    Dev Biol

    (1989)
  • M.C. Chang et al.

    Lithium decreases turnover of arachidonate in several brain phospholipids

    Neurosci Lett

    (1996)
  • G. Chen et al.

    Increase in AP-1 transcription factor DNA binding activity by valproic acid

    Neuropsychopharmacology

    (1997)
  • R.H. Chen et al.

    Wnt signaling to β-catenin involves two interactive components. Glycogen synthase kinase-3β inhibition and activation of protein kinase C

    J Biol Chem

    (2000)
  • R.W. Chen et al.

    Long term lithium treatment suppresses p53 and Bax expression but increases Bcl-2 expression. A prominent role in neuroprotection against excitotoxicity

    J Biol Chem

    (1999)
  • W.S. Choi et al.

    Effects of lithium and insulin on glycogen synthesis in L6 myocytes: additive effects on inactivation of glycogen synthase kinase-3

    Biochim Biophys Acta

    (2000)
  • M.P. Coghlan et al.

    Selective small molecule inhibitors of glycogen synthase kinase-3 modulate glycogen metabolism and gene transcription

    Chem Biol

    (2000)
  • A.F. Collins et al.

    Increased fetal hemoglobin production in patients receiving valproic acid for epilepsy

    Blood

    (1994)
  • R.J. Crowder et al.

    Glycogen synthase kinase-3β activity is critical for neuronal death caused by inhibiting phosphatidylinositol 3-kinase or Akt but not for death caused by nerve growth factor withdrawal

    J Biol Chem

    (2000)
  • A.A. Culbert et al.

    GSK-3 inhibition by adenoviral FRAT1 overexpression is neuroprotective and induces Tau dephosphorylation and β-catenin stabilisation without elevation of glycogen synthase activity

    FEBS Lett

    (2001)
  • M.P. Czech

    Signal transmission by the insulin-like growth factors

    Cell

    (1989)
  • J.A. Davies et al.

    Induction of early stages of kidney differentiation by lithium ions

    Dev Biol

    (1995)
  • J.R. Fredieu et al.

    Xwnt-8 and lithium can act upon either dorsal mesodermal or neurectodermal cells to cause a loss of forebrain in Xenopus embryos

    Dev Biol

    (1997)
  • E. Friedman et al.

    Altered platelet protein kinase C activity in bipolar affective disorder, manic episode

    Biol Psychiatry

    (1993)
  • C.A. Grimes et al.

    The multifaceted roles of glycogen synthase kinase 3β in cellular signaling

    Prog Neurobiol

    (2001)
  • V. Grimm et al.

    The level of GABA in the brain and locomotor behavior

    Pharmacol Biochem Behav

    (1975)
  • A.C. Hall et al.

    Axonal remodeling and synaptic differentiation in the cerebellum is regulated by WNT-7a signaling

    Cell

    (2000)
  • L.M. Hallcher et al.

    The effects of lithium ion and other agents on the activity of myo-inositol-1-phosphatase from bovine brain

    J Biol Chem

    (1980)
  • A.J. Harwood et al.

    Glycogen synthase kinase 3 regulates cell fate in Dictyostelium

    Cell

    (1995)
  • R. Hashimoto et al.

    Lithium protection against glutamate excitotoxicity in rat cerebral cortical neurons: involvement of NMDA receptor inhibition possibly by decreasing NR2B tyrosine phosphorylation

    J Neurochem

    (2002)
  • H. Aberle et al.

    β-Catenin is a target for the ubiquitin-proteasome pathway

    EMBO J

    (1997)
  • S. Acharya et al.

    Hematologic toxicity of sodium valproate

    J Pediatr Hematol Oncol

    (2000)
  • H. Albus et al.

    Electrophysiologic analysis of the actions of valproate on pyramidal neurons in the rat hippocampal slice

    Epilepsia

    (1998)
  • L. Alland et al.

    Role for N-CoR and histone deacetylase in Sin3-mediated transcriptional repression

    Nature

    (1997)
  • J.H. Allison et al.

    The effects of lithium on myo-inositol levels in layers of frontal cerebral cortex, in cerebellum, and in corpus callosum of the rat

    J Neurochem

    (1980)
  • C.L. Antos et al.

    Activated glycogen synthase-3β suppresses cardiac hypertrophy in vivo

    Proc Natl Acad Sci USA

    (2002)
  • J.R. Atack et al.

    In vitro and in vivo inhibition of inositol monophosphatase by the bisphosphonate L-690,330

    J Neurochem

    (1993)
  • K.T. Ault et al.

    Modulation of Xenopus embryo mesoderm-specific gene expression and dorsoanterior patterning by receptors that activate the phosphatidylinositol cycle signal transduction pathway

    Development

    (1996)
  • R. Baldessarini et al.

    Release of 3H-dopamine and analogous monoamines from rat striatal tissue

    Cell Mol Neurobiol

    (1988)
  • J. Baraban

    Towards a crystal-clear view of lithium's site of action

    Proc Natl Acad Sci USA

    (1994)
  • G.L. Barnes et al.

    Valproic acid-induced somite teratogenesis in the chick embryo: relationship with Pax-1 gene expression

    Teratology

    (1996)
  • C.R. Beals et al.

    Nuclear export of NF-ATc enhanced by glycogen synthase kinase-3

    Science

    (1997)
  • G.S. Bennett et al.

    Lithium chloride inhibits the phosphorylation of newly synthesized neurofilament protein, NF-M, in cultured chick sensory neurons

    J Neurochem

    (1991)
  • M.J. Berridge et al.

    Inositol phosphates and cell signalling

    Nature

    (1989)
  • A.N. Billin et al.

    β-Catenin-histone deacetylase interactions regulate the transition of LEF1 from a transcriptional repressor to an activator

    Mol Cell Biol

    (2000)
  • D.R. Boggs et al.

    The hematopoietic effects of lithium

    Semin Hematol

    (1983)
  • R. Bone et al.

    Structural studies of metal binding by inositol monophosphatase: evidence for two-metal ion catalysis

    Biochemistry

    (1994)
  • C.L. Bowden et al.

    History of the development of valproate for treatment of bipolar disorder

    J Clin Psychiatry

    (1995)
  • C.L. Bowden et al.

    Efficacy of divalproex vs lithium and placebo in the treatment of mania

    JAMA

    (1994)
  • H. Brantjes et al.

    All Tcf HMG box transcription factors interact with Groucho-related co-repressors

    Nucleic Acids Res

    (2001)
  • Cited by (154)

    View all citing articles on Scopus
    View full text