Elsevier

Neuropharmacology

Volume 48, Issue 4, March 2005, Pages 576-583
Neuropharmacology

Opposite effects of lithium and valproic acid on trophic factor deprivation-induced glycogen synthase kinase-3 activation, c-Jun expression and neuronal cell death

https://doi.org/10.1016/j.neuropharm.2004.11.010Get rights and content

Abstract

Recent studies demonstrate that lithium and valproic acid (VPA), two commonly used mood-stabilizing drugs, have neuroprotective effects against a variety of insults. Inhibition of the proapoptotic enzyme, glycogen synthase kinase-3 (GSK-3), has been suggested to be the mechanism of action of neuroprotection for both drugs. In this study, we tested if lithium and VPA could protect cultured cerebellar granule neurons (CGNs) from GSK-3-mediated apoptosis induced by trophic factor withdrawal (serum/potassium deprivation). Both lithium and indirubin, a specific GSK-3 inhibitor, protected CGNs in a dose-dependent manner. In contrast, VPA did not provide any neuroprotection and even potentiated cell death. Immunoblot analysis revealed that lithium inhibited the trophic factor deprivation-induced activation of GSK-3 as well as the in vivo phosphorylation of the microtubule-associated protein Tau on Ser199, a specific target site for GSK-3. Under these same experimental conditions, however, VPA neither inhibited GSK-3 activation nor hindered GSK-3 mediated Tau phosphorylation. Furthermore, in accordance with their effects on neuronal survival, lithium prevented the induction of c-Jun expression in trophic factor-deprived CGNs, whereas VPA potentiated it. Collectively, these results show that VPA is not a universal inhibitor of neuronal GSK-3, and that instead of being neuroprotective, VPA can even exacerbate neuronal death under some conditions.

Introduction

Lithium and valproic acid (VPA) are commonly used mood-stabilizing drugs for the treatment of bipolar disorder (Harwood and Agam, 2003). The biochemical basis of their therapeutic effect, however, is poorly understood. Lithium inhibits two different signal transduction pathways: it suppresses inositol signaling through depletion of intracellular inositol, and directly inhibits glycogen synthase kinase-3 (GSK-3), a multifunctional protein kinase (Harwood and Agam, 2003). VPA has also been found to inhibit GSK-3 in a human neuroblastoma cell line (Chen et al., 1999a, De Sarno et al., 2002) and in immature cultures of mouse cerebellar granule cells (Hall et al., 2002). The GSK-3 inhibitory effect of VPA, however, was not confirmed in other studies using a mouse neuroblastoma cell line (Phiel et al., 2001) and cultured rat sensory neurons (Williams et al., 2002). These latter studies rather linked the action of VPA to inositol depletion (Williams et al., 2002) and to inhibition of the transcriptional repressor histone deacetylase (HDAC) (Phiel et al., 2001).

GSK-3, originally identified as the protein kinase that phosphorylates and inactivates glycogen synthase, is a multifunctional serine/threonine protein kinase (Frame and Cohen, 2001). There are two GSK-3 isoforms encoded by distinct genes: GSK-3α (51 kDa) and GSK-3β (47 kDa), and GSK-3 is a key participant in numerous signaling cascades, including the phosphatidylinositol 3-kinase (PI3-K) and Wingless (Wnt) pathways (Frame and Cohen, 2001). GSK-3 is also involved in a wide range of cellular processes, such as embryonic patterning, cell proliferation and axonal remodeling (Frame and Cohen, 2001).

GSK-3, and particularly the GSK-3β isoform, has recently been identified as an important key regulator of neuronal cell fate, having a proapoptotic effect in many settings (Hetman et al., 2000, Cross et al., 2001, Facci et al., 2003). Insulin and several other neurotrophic factors induce inhibitory phosphorylation of GSK-3, mediated by the survival-promoting kinase Akt (van Weeren et al., 1998), and thereby promote neuronal survival. Consistent with this, when neurons are deprived of trophic stimulation, GSK-3 becomes activated and apoptotic cell death is initiated (Hetman et al., 2000, Cross et al., 2001, Mora et al., 2001, Hongisto et al., 2003).

Recent studies have shown that lithium and VPA have neuroprotective effects against a variety of insults (Maggirwar et al., 1999, Mora et al., 2001, Tong et al., 2001, Dou et al., 2003, Hongisto et al., 2003), and it has been proposed that the inhibition of GSK-3 may represent a common molecular mechanism for the neuroprotective effect of both drugs (Li et al., 2000, Tong et al., 2001, Li et al., 2002).

In this study, we tested if lithium and VPA could inhibit GSK-3 and prevent GSK-3-mediated cell death in trophic factor deprived cultures of cerebellar granule neurons (CGNs). We found that lithium protected CGNs against trophic factor withdrawal-induced apoptosis, whereas VPA was not neuroprotective and even potentiated cell death. Furthermore, lithium, but not VPA, prevented GSK-3 activation and inhibited GSK-3β-mediated Tau phosphorylation in vivo.

Section snippets

Cell culture

Cerebellar brain tissue from 7 to 8-day-old Sprague–Dawley rats (Charles River) was harvested according to the guidelines of the Animal Welfare Act and NIH policies. Primary cultures of cerebellar granule neurons were prepared as previously described (Kovacs et al., 2002) with some modifications. Briefly, cerebella were dissected and meninges and blood vessels were removed. Then cerebella were minced using a single edged razor blade with two passes at 90° to each other at approximately 0.5 mm

Lithium, but not VPA, protects cultured cerebellar granule neurons from GSK-3-mediated death induced by trophic factor deprivation

Primary cultures of CGNs were maintained in medium containing serum (10%) and 25 mM KCl as survival factors. Serum and potassium deprivation triggers GSK-3-mediated apoptosis in these cultures (Cross et al., 2001, Mora et al., 2001, Hongisto et al., 2003). To investigate the effects of lithium and VPA on neuronal survival in this culture model, serum and potassium were withdrawn (K5-S) in the presence of increasing concentrations of either lithium chloride or valproic acid sodium salt (sodium

Discussion

Primary cultures of cerebellar granule neurons (CGNs) can be maintained in medium containing serum (10%) and 25 mM KCl as survival factors. The chronic depolarization induced by this high concentration of potassium is thought to imitate endogenous excitatory activity. Serum and potassium deprivation in these cultures results in inactivation of the survival-promoting PI3-K/Akt pathway, leading to dephosphorylation and hyperactivation of GSK-3 and to subsequent apoptosis (Cross et al., 2001, Mora

Acknowledgements

This study was supported by an NIH grant (PO1 MH64570).

References (36)

  • C.J. Phiel et al.

    Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen

    Journal of Biological Chemistry

    (2001)
  • P.C. van Weeren et al.

    Essential role for protein kinase B (PKB) in insulin-induced glycogen synthase kinase 3 inactivation. Characterization of dominant-negative mutant of PKB

    Journal of Biological Chemistry

    (1998)
  • G. Chen et al.

    The mood-stabilizing agent valproate inhibits the activity of glycogen synthase kinase-3

    Journal of Neurochemistry

    (1999)
  • R.W. Chen et al.

    Regulation of c-Jun N-terminal kinase, p38 kinase and AP-1 DNA binding in cultured brain neurons: roles in glutamate excitotoxicity and lithium neuroprotection

    Journal of Neurochemistry

    (2003)
  • R.M. Collins et al.

    Valproate increases glutaminase and decreases glutamine synthetase activities in primary cultures of rat brain astrocytes

    Journal of Neurochemistry

    (1994)
  • D.A. Cross et al.

    Selective small-molecule inhibitors of glycogen synthase kinase-3 activity protect primary neurones from death

    Journal of Neurochemistry

    (2001)
  • H. Dou et al.

    Neuroprotective activities of sodium valproate in a murine model of human immunodeficiency virus-1 encephalitis

    Journal of Neuroscience

    (2003)
  • L. Facci et al.

    Glycogen synthase kinase-3 inhibitors protect central neurons against excitotoxicity

    Neuroreport

    (2003)
  • Cited by (65)

    • Frontotemporal dementia

      2017, Disease-Modifying Targets in Neurodegenerative Disorders: Paving the Way for Disease-Modifying Therapies
    • Animal models of bipolar mania: The past, present and future

      2016, Neuroscience
      Citation Excerpt :

      Furthermore, polymorphisms in GSK-3β are associated with certain symptoms of BD, as well as treatment response to lithium (Jope and Roh, 2006; Lin et al., 2013; Tang et al., 2013). The antipsychotic VPA also inhibits GSK-3β activity via mechanisms less characterized than lithium action (Chen et al., 1999; De Sarno et al., 2002; Jin et al., 2005; Sintoni et al., 2013). GSK-3β has numerous cellular and molecular functions, including cell development, gene transcription, metabolic homeostasis, neurogenesis, and apoptosis (Doble and Woodgett, 2003).

    • Valproic acid in normal therapeutic concentration has no neuroprotective or differentiation influencing effects on long term expanded murine neural stem cells

      2014, Epilepsy Research
      Citation Excerpt :

      Using VPA concentrations of 0, 100, 200 and 300 μg/ml, we found a marked NFkB inhibition only after treatment with 300 μg/ml VPA after 1 h (p = 0.04) and 6 h (p = 0.05) in comparison to the untreated control (Fig. 5) in addition to the previously described treatment independent NFkB down regulation during the first hours of cell differentiation in our cell system (Sabolek et al., 2009). As previous publications had also pointed to a possible induction of the MAP kinase pathway (Jin et al., 2005; Yamauchi et al., 2007), we tested the effects of VPA on JUN kinase and p38 MAP kinase activity, respectively. Using the same concentrations of 0, 100, 200 and 300 μg/ml VPA, we found no significant influence on JUN kinase and p38 MAP kinase activity at 1, 3 and 6 h after starting the differentiation process (data not shown).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text