Elsevier

Epilepsy & Behavior

Volume 25, Issue 1, September 2012, Pages 92-97
Epilepsy & Behavior

Finasteride inhibits the disease-modifying activity of progesterone in the hippocampus kindling model of epileptogenesis

https://doi.org/10.1016/j.yebeh.2012.05.024Get rights and content

Abstract

Progesterone (P) plays an important role in seizure susceptibility in women with epilepsy. Preclinical and experimental studies suggest that P appears to interrupt epileptogenesis, which is a process whereby a normal brain becomes progressively susceptible to recurrent, unprovoked seizures due to precipitating risk factors. Progesterone has not been investigated widely for its potential disease-modifying activity in epileptogenic models. Recently, P has been shown to exert disease-modifying effects in the kindling model of epileptogenesis. However, the mechanisms underlying the protective effects of P against epileptogenesis remain unclear. In this study, we investigated the role of P-derived neurosteroids in the disease-modifying activity of P. It is hypothesized that 5α-reductase converts P to allopregnanolone and related neurosteroids that retard epileptogenesis in the brain. To test this hypothesis, we utilized the mouse hippocampus kindling model of epileptogenesis and investigated the effect of finasteride, a 5α-reductase and neurosteroid synthesis inhibitor. Progesterone markedly retarded the development of epileptogenesis and inhibited the rate of kindling acquisition to elicit stage 5 seizures. Pretreatment with finasteride led to complete inhibition of the P-induced retardation of the limbic epileptogenesis in mice. Finasteride did not significantly influence the acute seizure expression in fully kindled mice expressing stage 5 seizures. Thus, neurosteroids that potentiate phasic and tonic inhibition in the hippocampus, such as allopregnanolone, may mediate the disease-modifying effect of P, indicating a new role of neurosteroids in acquired limbic epileptogenesis and temporal lobe epilepsy.

Highlights

► In this study, we investigated a potential mechanism underlying progesterone’s (P) disease-modifying activity. ► P has been shown to exert disease-modifying effects in rodent model of epileptogenesis. ► Finasteride pretreatment significantly inhibited the P-induced retardation of limbic epileptogenesis in mice. ► These studies support a role for neurosteroids in limbic epileptogenesis.

Introduction

Acquired limbic epilepsy or temporal lobe epilepsy (TLE) is caused by diverse precipitating factors, such as brain injury, stroke, infections, or prolonged seizures. The mechanisms underlying the development of acquired TLE are not very well understood. The term ‘epileptogenesis’ is used to describe the complex plastic changes in the brain that, following a precipitating injury, convert a normal brain into a brain debilitated by recurrent seizures [1], [2]. There is an unmet need for drugs that truly prevent the development of epilepsy (antiepileptogenic agents) or alter its natural course, delaying the appearance or severity of epileptic seizures (disease-modifying agents) in people at risk. Progesterone (P) plays an important role in women with epilepsy [3], [4]. Progesterone has long been known to have antiseizure activity in animal models [5], [6], [7] and in clinical studies [8], [9], [10], [11]. Therefore, it has been suggested that women with epilepsy are prone to seizures in response to decreased levels of P during perimenstrual periods [12], [13]. The antiseizure properties of P are due to its conversion in the brain to allopregnanolone (3α-hydroxy-5α-pregnane-20-one) and other structurally related neurosteroids [6], [7], [14], [15]. Recent studies show that neurosteroids are mainly present in principal neurons in many brain regions that are relevant to acquired TLE including the hippocampus [16]. Neurosteroids rapidly alter neuronal excitability through direct interaction with synaptic and extrasynaptic GABA-A receptors in the hippocampus [17]. Although they act on all GABA-A-receptor subtypes, they have pronounced effects on the extrasynaptic δ-subunit containing GABA-A receptors that mediate tonic inhibition [18]. Therefore, allopregnanolone and related 5α-reduced neurosteroids are powerful anticonvulsants in animal seizure models, and their synthetic analogues have been proposed for epilepsy therapy [19].

Neurosteroids are broad-spectrum anticonvulsants that confer seizure protection in various experimental models of evoked seizures in normal or healthy rats and mice [7], [20], [21], [22]. They protect against seizures induced by GABA-A receptor antagonists, 6-Hz model, pilocarpine-induced limbic seizures, and seizures in kindled animals. Unlike benzodiazepines, tolerance to their actions does not occur during chronic administration in rats [19], [23]. In addition to anticonvulsant activity, there is emerging evidence that neurosteroids play a role in regulating acquired epileptogenesis. Although the precise mechanisms underlying epileptogenesis remain unclear, epileptogenesis may involve an interaction of acute and delayed anatomic, molecular, and physiological events that are both complex and multifaceted. The initial precipitating factor activates diverse signaling events, such as inflammation, oxidation, apoptosis, neurogenesis and synaptic plasticity, which eventually lead to structural and functional changes in neurons [1], [2]. These changes are eventually manifested as abnormal hyperexcitability and spontaneous seizures. In the rodent brain, the 5α-reductase type I and II isozymes convert P to dihydroprogesterone ultimately leading to the production of allopregnanolone and related neurosteroids which may retard epileptogenesis by the interruption of one or more of the above events. In addition to catalyzing the rate-limiting step in the reduction of P, both isoforms of the 5α-reductase enzyme are responsible for the conversion of testosterone and deoxycorticosterone to dihydroprogesterone and dihydrodeoxycorticosterone, respectively [13]. Finasteride, an irreversible inhibitor of 5α-reductase, blocks the synthesis of all 5α-reduced neurosteroids, including testosterone-derived androstanediol [24] and deoxycorticosterone-derived allotetrahydrodeoxycorticosterone (THDOC) [25] and thereby may affect epileptogenesis.

As a precursor of neurosteroids, P is an appealing agent for the intervention of epileptogenesis. Three observations support this contention. First, P's inhibitory effect on kindling was observed in rats [26] and mice [27]. Second, following pilocarpine-induced status epilepticus, the neurosteroidogenic enzyme cytochrome P450 side chain cleavage (P450scc) was increased for several weeks, suggesting that it may be associated with the promotion of neurosteroidogenesis [28]. Cytochrome P450 side chain cleavage is a critical enzyme involved in the conversion of cholesterol into pregnenolone, a precursor for several neurosteroids. Third, inhibiting neurosteroid synthesis with finasteride accelerated the onset of spontaneous recurrent seizures [29], suggesting that endogenous neurosteroids play a role in restraining epileptogenesis. However, P has not been investigated widely for its potential disease-modifying activity in epileptogenic models. Using the mouse kindling model, we previously demonstrated that P inhibits the development of limbic epileptogenesis at doses that do not affect seizure expression and motor performance [27], indicating the disease-modifying activity of P. Kindling is a model of functional epileptogenesis, since seizures develop after repeated subthreshold stimulations applied in the hippocampus, amygdala or other brain regions, without induction of evident brain lesions [30]. However, the mechanisms underlying the protective effect of P against limbic epileptogenesis remain unclear.

In the present study, we sought to investigate the role of neurosteroids in the disease-modifying effect of P by using finasteride, a 5α-reductase inhibitor that blocks the synthesis of P-derived and 5α-reduced neurosteroids in the brain. Our results show for the first time that finasteride prevents P's inhibition of hippocampal kindling epileptogenesis, indicating that P-derived neurosteroids such as allopregnanolone may inhibit epileptogenesis, thus confirming the new role of neurosteroids in limbic epileptogenesis and TLE.

Section snippets

Animals

Adult female mice (3–5 months old) of C57BL6 strain weighing 25–30 g were used in the study. The animals were housed separately, four to a cage, and were fed mouse chow and water ad libitum. The animals were not checked for ovarian cycle stages due to a chronic experimental (kindling) protocol that lasted longer than the 5-day cycle. All procedures were performed in compliance with the NIH Guide for the Care and Use of Laboratory Animals, under a protocol approved by the Institutional Animal Care

Disease-modifying activity of P in the kindling epileptogenesis model

The development of kindling epileptogenesis in mice treated with vehicle, P (25 mg/kg), and finasteride (50 mg/kg) is shown in Fig. 1; there was no significant group difference in the initial AD threshold (control: 78 ± 10 μA; P treatment: 75 ± 13 μA; P + F treatment: 68 ± 19 μA). In vehicle-treated control mice, there was a progressive increase in behavioral seizure intensity with all animals achieving stage 5 seizures after 13 stimulations (Fig. 1A). Progesterone produced a marked retardation of

Discussion

The key observation of this study is that the disease-modifying effect of P in the kindling model of epileptogenesis is blocked by a pretreatment of finasteride, which is consistent with a protective role of 5α-reduced neurosteroids in limbic epileptogenesis. Finasteride inhibition of P's kindling-retarding effect has not been reported previously. Finasteride is a 5α-reductase inhibitor that blocks the biosynthesis of endogenous neurosteroids such as allopregnanolone and related 5α-reduced

Acknowledgment

This research was supported by the National Institutes of Health, National Institute of Neurological Disorders and Stroke [grant NS051398].

References (51)

  • R.J. Racine

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

    Electroencephalogr Clin Neurophysiol

    (1972)
  • B. Azzolina et al.

    Inhibition of rat 5α-reductases by finasteride: evidence for isozyme differences in mechanism of inhibition

    J Steroid Biochem Mol Biol

    (1997)
  • R.C. Melcangi et al.

    The 5α-reductase in the central nervous system: expression and modes of control

    J Steroid Biochem Mol Biol

    (1998)
  • O. Gangisetty et al.

    Neurosteroid withdrawal regulates GABA-A receptor α4-subunit expression and seizure susceptibility by activation of progesterone receptor-independent early growth response factor-3 pathway

    Neuroscience

    (2010)
  • C. Brandt et al.

    Prophylactic treatment with levetiracetam after status epilepticus: lack of effect on epileptogenesis, neuronal damage, and behavioral alterations in rats

    Neuropharmacology

    (2007)
  • U. Wahnschaffe et al.

    Lack of changes in seizure susceptibility during the estrous cycle in kindled rats

    Epilepsy Res

    (1992)
  • R.E. Gorin-Meyer et al.

    Sex differences in the effect of finasteride on acute ethanol withdrawal severity in C57BL/6J and DBA/2J mice

    Neuroscience

    (2007)
  • A. Pitkänen et al.

    From traumatic brain injury to posttraumatic epilepsy: what animal models tell us about the process and treatment options

    Epilepsia

    (2009)
  • D.S. Reddy et al.

    Development and persistence of limbic epileptogenesis are impaired in mice lacking progesterone receptors

    J Neurosci

    (2011)
  • A.G. Herzog et al.

    Sensitivity and specificity of the association between catamenial seizure patterns and ovulation

    Neurology

    (2008)
  • C.R. Craig

    Anticonvulsant activity of steroids: separability of anticonvulsant from hormonal effects

    J Pharmacol Exp Ther

    (1966)
  • T.G. Kokate et al.

    Finasteride, a 5α-reductase inhibitor, blocks the anticonvulsant activity of progesterone in mice

    J Pharmacol Exp Ther

    (1999)
  • D.S. Reddy et al.

    Antiseizure activity of progesterone and neurosteroids in progesterone receptor knockout mice

    J Pharmacol Exp Ther

    (2004)
  • T. Bäckström et al.

    Effect of intravenous progesterone infusions on the epileptic discharge frequency in women with partial epilepsy

    Acta Neurol Scand

    (1984)
  • A.G. Herzog

    Progesterone therapy in women with complex partial and secondary generalized seizures

    Neurology

    (1995)
  • Cited by (27)

    • Neurosteroids: A potential target for neuropsychiatric disorders

      2024, Journal of Steroid Biochemistry and Molecular Biology
    • Molecular mechanisms of sex differences in epilepsy

      2023, Principles of Gender-Specific Medicine: Sex and Gender-Specific Biology in the Postgenomic Era
    • Molecular mechanisms of sex differences in epilepsy and seizure susceptibility in chemical, genetic and acquired epileptogenesis

      2021, Neuroscience Letters
      Citation Excerpt :

      Finally, we adopted two separate pharmacological methods of inducing increased levels of neurosteroids: (a) chronic exogenous progesterone treatment, and (b) a gonadotropin administration to induce endogenous neurosteroid synthesis. The gonadotropin-induced neurosteroid synthesis and withdrawal paradigm appears more physiologically relevant than the exogenous progesterone treatment, as the withdrawal paradigm is induced by the neurosteroid synthesis inhibitor finasteride [132]. We have found that the mouse model of perimenstrual CE is useful for the investigation of disease mechanisms and exploring the efficacy of new therapeutic techniques.

    • Neurosteroid regulation of GABA<inf>A</inf> receptors: A role in catamenial epilepsy

      2019, Brain Research
      Citation Excerpt :

      Infusion of progesterone suppresses ictal activity in human EEG (Bäckström et al., 1984). Similarly, acute administration of progesterone also reduces the susceptibility to seizures evoked by chemical or electrical stimulation in experimental animals (Frye et al., 2002; Frye and Scalise, 2000; Kokate et al., 1999a; Reddy et al., 2004; Reddy and Ramanathan, 2012). Multiple lines of evidence show that the anticonvulsant effects of progesterone are mediated through its conversion to allopregnanolone.

    View all citing articles on Scopus
    View full text