Elsevier

Neurobiology of Disease

Volume 63, March 2014, Pages 194-200
Neurobiology of Disease

Chronic antidepressant treatment accelerates kindling epileptogenesis in rats

https://doi.org/10.1016/j.nbd.2013.11.020Get rights and content

Highlights

  • We assessed the effects of antidepressant treatment on kindling epileptogenesis.

  • Citalopram and fluoxetine both accelerated disease progression in this model.

  • This may have implications for epilepsy patients being treated with SSRIs.

Abstract

Objectives

Due to the high comorbidity of epilepsy and depression, antidepressant treatment is commonly indicated for patients with epilepsy. Studies in humans and animal models suggest that selective serotonin reuptake inhibitors (SSRIs) may reduce seizure frequency and severity, and these drugs are generally considered safe for use in epilepsy. No studies have investigated the effects of SSRIs on epileptogenesis, the neurobiological process underlying the development of the epileptic state.

Methods

The effect of continuous infusion of the SSRI, fluoxetine (10 mg/kg/day sc), versus vehicle control on amygdala kindling was examined in adult male Wistar rats. Seizure threshold and kindling rates were compared between SSRI-treated rats and controls. The study was then repeated examining the effect of a different SSRI, citalopram (10 mg/kg/day sc), versus vehicle control. Hippocampal mRNA expression of the serotonin transporter (SERT) and the 5-HT1A receptor was examined in the brains of the rats post-mortem.

Results

Treatment with either fluoxetine or citalopram significantly accelerated kindling epileptogenesis, as evidenced by fewer stimulations to reach Class V seizures compared to their respective vehicle-treated group (p < 0.01 for both drugs). Seizure duration was also increased in fluoxetine-treated rats. No differences in seizure threshold were observed between treatments (p > 0.05). mRNA analysis did not reveal any molecular changes which were common to both treatments.

Conclusions

The rate of epileptogenesis in rats is enhanced by chronic treatment with SSRIs. This could potentially have implications regarding the effect of SSRIs on the development or progression of human epilepsy.

Introduction

Mood and anxiety disorders are common co-morbidities in patients with epilepsy, in particular those with drug-resistant epilepsy (Gaitatzis et al., 2004). Mesial temporal lobe epilepsy (MTLE), the most common form of drug-resistant epilepsy in adults, has a particularly strong association with depression (Kanner, 2006a). These mood disorders have a major adverse impact on the quality of life of patients, and are associated with poorer seizure control, increased drug resistance, and a higher likelihood of suicide (Hesdorffer et al., 2006). The presence of psychopathology in patients with epilepsy has been consistently found to be one of the most important determinants of quality of life (Boylan et al., 2004). Active management of depression and anxiety is therefore an essential part of comprehensive treatment of people with epilepsy.

The mainstay of the pharmacological treatment of mood disorders is antidepressant medication. There has been a commonly held concern amongst clinicians about the safety of the use of these medications in people with epilepsy, stemming from the propensity of ‘first generation’ antidepressants, particularly tricyclic antidepressants (TCAs), to induce seizures in a few patients (Salzberg and Vajda, 2001). However, a growing body of evidence suggests that this should be less of a concern for the newer, or ‘second generation’, antidepressants. These newer classes, notably the selective serotonin reuptake inhibitors (SSRIs), may even decrease seizure frequency (Alper et al., 2007, Bagdy et al., 2007, Kanner, 2009). These clinical reports largely agree with animal literature on the influence of SSRIs on experimentally-induced seizures (reviewed in Cardamone et al., 2013).

While previous clinical and experimental studies have examined effects of antidepressants on seizure frequency, to our knowledge, none have investigated the effects of these drugs on epileptogenesis, despite strong reasons to suspect such effects (Cardamone et al., 2013). Epileptogenesis refers to the neurobiological processes which transform normal brain networks into an epileptic state, with an increased propensity to generate seizures (Goldberg and Coulter, 2013). These processes continue even after the epileptic state is reached, and are likely compounded by the effects of the spontaneous recurrent seizures themselves.

There are several good reasons for considering effects of SSRIs on epileptogenesis, especially in MTLE. First, the brain structures where most epileptogenic changes occur in MTLE show considerable overlap with those involved with the neurobiology of mood disorders, notably the hippocampus and amygdala (Nestler et al., 2002). Secondly, serotonergic neurotransmission is a key aspect of the function of both these structures (Bagdy et al., 2007). Thirdly, there is increasing epidemiological evidence that depression and other psychopathologies pre-date, and are a risk factor for the emergence of epilepsy (Hesdorffer et al., 2012), and so the possibility should be considered that treatment with antidepressants of some depressed patients may influence the onset of epilepsy (Hesdorffer et al., 2000). Based on these and other considerations, an emerging theory proposes that the pathogenesis of depression and epilepsy share common mechanisms (Kanner, 2006b, Kanner, 2012), leading to the proposal that antidepressants may have antiepileptic efficacy (Kanner, 2012). The influence of antidepressants on epileptogenesis is thus a question of clear clinical importance: depression in epilepsy is often under-recognised and undertreated; authoritative bodies advocate screening and treatment to address this; however, a serious gap exists regarding knowledge of potential effects of antidepressants on epileptogenesis and disease progression.

With these issues in mind, this study was designed to investigate the effect of chronic SSRI treatment on the progression of epileptogenesis using the amygdala kindling rat model of temporal lobe epilepsy (Morimoto et al., 2004). We also investigated potential molecular mediators underlying any effects by studying key players in the mechanism of action of the antidepressants: the serotonin transporter (SERT) as the primary target of SSRIs, and 5-HT1A receptors which may be relevant to depression in epilepsy (Hasler et al., 2007, Theodore et al., 2007). We tested the effect of two commonly prescribed SSRI antidepressants, fluoxetine and then citalopram, first-line medications for the treatment of depression in epilepsy.

Section snippets

Surgical implantation of osmotic pumps and stimulating/recording electrodes

Male Wistar rats aged 9–11 weeks underwent electrode implantation as previously described (Jones et al., 2009). Briefly, rats were anaesthetised with isofluorane (5% induction, 1.5–2.5% maintenance) and placed into a stereotactic frame. Holes were drilled into the skull to allow implantation of extradural recording electrodes, and a bipolar stimulating electrode (Plastics One, USA) into the left basolateral amygdala complex (AP: − 3.0; ML: − 5.25 relative to bregma; DV: − 6.5 relative to the dura (

Fluoxetine serum drug and metabolite levels

Serum was taken from animals at the completion of the fluoxetine study, and analysed for drug levels. All drug-treated animals had detectable levels of fluoxetine (mean: 0.38 ± 0.03 μmol/l) and norfluoxetine (3.02 ± 0.16 μmol/l), confirming effectiveness of the pumps to deliver drug over the treatment period (Fig. 1). There were no detectable drug levels in vehicle-treated rats. In addition, despite using a very high concentration of DMSO in the pumps, albeit at a very low release rate (60 μl/day), we

Discussion

The key finding of this study was that chronic SSRI antidepressant treatment in rats, using both citalopram and fluoxetine, increases susceptibility to limbic epileptogenesis. This did not appear to be due to effects on the innate excitability of the brain, since neither drug influenced seizure threshold. This finding suggests that the use of antidepressants may adversely impact the onset and progression of limbic epilepsy in patients.

The majority of studies that have examined antidepressant

References (49)

  • E. Le Poul

    Differential adaptation of brain 5-HT1A and 5-HT1B receptors and 5-HT transporter in rats treated chronically with fluoxetine

    Neuropharmacology

    (2000)
  • K. Morimoto

    Kindling and status epilepticus models of epilepsy: rewiring the brain

    Prog. Neurobiol.

    (2004)
  • E.J. Nestler

    Neurobiology of depression

    Neuron

    (2002)
  • R.J. Racine

    Modification of seizure activity by electrical stimulation. I. After-discharge threshold

    Electroencephalogr. Clin. Neurophysiol.

    (1972)
  • R.J. Racine

    Modification of seizure activity by electrical stimulation. II. Motor seizure

    Electroencephalogr. Clin. Neurophysiol.

    (1972)
  • S.S. Raju

    Effect of fluoxetine on maximal electroshock seizures in mice: acute vs chronic administration

    Pharmacol. Res.

    (1999)
  • M.R. Salzberg et al.

    Epilepsy, depression and antidepressant drugs

    J. Clin. Neurosci.

    (2001)
  • S.M. Stahl

    Mechanism of action of serotonin selective reuptake inhibitors. Serotonin receptors and pathways mediate therapeutic effects and side effects

    J. Affect. Disord.

    (1998)
  • Y. Wada

    Effects of the 5-HT3 receptor agonist 1-(m-chlorophenyl)-biguanide in the rat kindling model of epilepsy

    Brain Res.

    (1997)
  • Y. Wada

    Role of serotonin receptor subtypes in the development of amygdaloid kindling in rats

    Brain Res.

    (1997)
  • K. Watanabe

    The effect of the acute administration of various selective 5-HT receptor antagonists on focal hippocampal seizures in freely-moving rats

    Eur. J. Pharmacol.

    (2000)
  • G. Bagdy

    Serotonin and epilepsy

    J. Neurochem.

    (2007)
  • L.S. Boylan

    Depression but not seizure frequency predicts quality of life in treatment-resistant epilepsy

    Neurol.

    (2004)
  • L. Cardamone

    Antidepressant therapy in epilepsy: can treating the comorbidities affect the underlying disorder?

    Br. J. Pharmacol.

    (2013)
  • Cited by (0)

    Financial disclosure: This study was supported by an NHMRC project grant to TJO, MS and NJ (#566843) and an NHMRC Career Development Award to NJ (#628466). The authors report no disclosures, financial or otherwise, associated with this work.

    View full text