Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Print ISSN: 2319-2003 | Online ISSN: 2279-0780

IJBCP International Journal of Basic & Clinical Pharmacology


doi: 10.5455/2319-2003.ijbcp004212
Review Article

Neuroactive steroids and their role in epilepsy


Chetan Y. Patil*, Shamsundar A. Jadhav, Sudhakar M. Doifode, Mirza Shiraz Baig

ABSTRACT

Neuroactive steroids are the certain steroids that alter neuronal excitability via
the cell surface through interaction with certain neurotransmitter receptors.
Neuroactive steroids regulate physiological functions of the central nervous
Department of Pharmacology, system and have possible therapeutic potential in neurological diseases. They
Government Medical College have been shown to affect neuronal excitability via their interaction with the
and Hospital, Aurangabad- ligand-gated ion channel family, such as the GABAA receptor by acting
431001, Maharashtra, India genomically as well as nongenomically. Positive modulators of GABA A
receptor have anticonvulsant action as they enhance GABAergic transmission
Received: 18 October 2012 thereby increasing the seizure threshold. By virtue of these properties,
Accepted: 31 October 2012 neurosteroids appear to be relevant to pathophysiology and pharmacological
treatment of many neurological diseases including catamenial epilepsy, stress
*Correspondence to: induced epilepsy, temporal lobe epilepsy, alcohol withdrawal seizures,
Dr. Chetan Y. Patil, infantile spasm and status epilepticus. So far, only synthetic neurosteroid,
Email: ganaxolone has been tried in treatment of epilepsy and has shown good
drchetanpatil.gmc@gmail.com efficacy and tolerability. But, human data of trials are limited and hence, large
double-blinded, placebo-controlled, randomized clinical trials are required
before their use. The paper reviews the biosynthesis and GABAA receptor
modulation of neurosteroids and their potential role in epilepsy.

Keywords: Neuroactive steroids, Neurosteroids, Allopregnanolone,


Tetrahydrodeoxycorticosterone (THDOC), GABAA receptor, Catamenial
epilepsy, Seizure, Epilepsy

INTRODUCTION high voltage-activated Ca+2 channel, Na+ channel, Ca+2-


activated K channel, and anion channels.2
Steroid hormones act as transcription factors and regulate
gene expression which is exerted by binding to their In general, neuroactive steroids are the steroids which
respective intracellular receptors.1 However, certain regulate the neuronal activities and physiological
steroids may alter neuronal excitability via the cell functions of the CNS.2,3 They are classified as
surface through interaction with certain neurotransmitter endogenous steroids and exogenous (synthetic) steroids
receptors. The term neuroactive steroid has been coined based on the source or production site. Endogenous
for steroids with these particular properties. Molecular steroids are subdivided into hormonal steroids (produced
basis for the action of neurosteroids is explained by their by the endocrine glands) and neurosteroids (produced by
genomic effects (limited by the rate of protein the nervous tissue) [Figure 1].2
biosynthesis and requiring a time period from minutes to
hours) and nongenomic effects (the modulatory effects Neuroactive steroids have shown numerous important
requiring only milliseconds to seconds). This provides a modulatory effects on brain functions and brain diseases.
broad spectrum action of neuroactive steroids on These effects are exerted by modulating almost all kinds
neuronal function and plasticity.1 Neuroactive steroids of classical synaptic transmission including
also act on various neurotransmitter receptors and glutamatergic, GABAergic, cholinergic, noradrenergic,
voltage-dependent ion channels, especially - dopaminergic and serotonergic synaptic transmission,
aminobutyric acid (GABA), N-methyl-D-aspartate either by altering the responsiveness of postsynaptic
(NMDA), -amino-3-hydroxy-5-methyl-4- receptors or by the presynaptic release of
isoxazolepropionic acid (AMPA), kainate, glycine, neurotransmitters.2 As they modulate GABAergic
serotonin, sigma-1, nicotinic and muscarinic transmission, endogenous neurosteroids though to play a
acetylcholine receptors, as well as T-type Ca+2 channel, role in the physiological regulation of seizure

www.ijbcp.com International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 150
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

susceptibility in individuals with epilepsy. Many studies tetrahydroprogesterone), deoxycorticosterone (3,5- and
have proven that they play important roles in the 3,5 tetrahydrodeoxycorticosterone),
pathology and treatment of neurological disorders dehydroepiandrosterone (DHEA) (3,5- and 3,5-
including, epilepsy such as catamenial epilepsy, androsterone) and testosterone (3,5- and 3,5-
childhood epilepsy, stress induced seizures, temporal lobe androstanol) enhance GABAergic neurotransmission and
epilepsy, alcohol withdrawal seizure, infantile spasm and produce inhibitory neurobehavioral effects such as
status epilepticus. However, there is no evidence that anticonvulsant, anxiolytic and sedative actions. The
alterations in neurosteroid levels can cause excitatory neuroactive steroids include the sulfated
epileptogenesis in the absence of pre-existing epilepsy.4 derivatives of pregnenolone and DHEA as well as the 3,
5- and 3, 5-reduced metabolites of cortisol.1,5,6 The
BIOSYNTHESIS OF NEUROSTEROIDS biosynthetic pathway for these steroids is shown in
Figure 2.5
Endogenous neuroactive steroids can be synthesized de
novo in the brain as well as in adrenal glands, ovaries and NOMENCLATURE
testes. Among these compounds, the 3,5- and 3,5-
reduced metabolites of progesterone (3,5- and 3,5- The nomenclature of the steroids is provided in Table 1.5

Figure 1: Classification of neuroactive steroids.2

Table 1: IUPAC and conventional nomenclature.5

IUPAC name Conventional (trivial) names Abbreviation


5-pregnane-3-ol-20-one,
3-hydroxy-5-pregnan-20-one 3,5-tetrahydroprogesterone, 3,5-THP
allopregnanolone
5-pregnane-3-ol-20-one,
3-hydroxy-5-pregnan-20-one 3,5-tetrahydroprogesterone, 3,5-THP
pregnanolone
5-pregnan-3, 21-diol-20-one,
3,21-dihydroxy-5-pregnan-20-one 3,5-THDOC
3,5-tetrahydrodeoxycorticosterone
5-pregnan-3, 21-diol-20-one,
3,21-dihydroxy-5-pregnan-20-one 3,5-THDOC
3,5-tetrahydrodeoxycorticosterone

IUPAC: International Union of Pure and Applied Chemistry; THP: Tetrahydroprogesterone;


THDOC: Tetrahydrodeoxycorticosterone.

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 151
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

Figure 2: The biosynthetic pathway for neuroactive steroids.5


The inhibitory neuroactive steroids with potent GABAA receptor-positive modulatory effects are highlighted in light blue
box; whereas the excitatory neuroactive steroids with weak GABA A receptor antagonist effects are highlighted in dark
blue box.

Figure 3 (A and B): Schematic and binding sites of GABAA receptor7, (A) A single subunit of the GABAA receptor,
highlighting topology. M1-M4 represent transmembrane domains. The M2 transmembrane domain forms an important
part of the chloride channel pore. (B) Pentameric structure of a typical GABA A receptor. Several putative sites of
modulatory drugs, including neurosteroids, are shown. It also shows steroids act on the GABAA receptor from within the
transmembrane domains which is supported by pharmacological studies and by recent site-directed mutagenesis studies.

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 152
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

MODULATION OF GABAA RECEPTORS BY Recent studies have shown evidence of two sites of
NEUROACTIVE STEROID neurosteroid action on GABAA receptors. One site spans
the M1 and M4 transmembrane domains of the -subunit
GABAA receptors and accounts for the potentiating actions of some
GABA receptors are pentameric-heteromer and belong to steroids. Another site, between the M1 domain of the -
the Cys-loop family of ligand-gated ion channels.1,7 subunit and the M3 domain of the -subunit, is
Though the GABA-gated conductance has inhibitory responsible for direct gating of the channel by steroids.
influence on the cell, postsynaptic GABA response can Schematic representation of these sites is shown in Figure
be excitatory or inhibitory. Variations in GABAA 4.7 Various studies conducted have shown direct gating
receptors depend upon structure of the subunits. by steroids is inefficient, with maximum responses less
Functional channels of GABAA receptor are formed by than the responses generated by saturating GABA. In
the assembly of two subunits (from 1, 2, 3, 4, 5, spite of this, small currents resulting from direct gating
6), two subunits (from 1, 2, 3) plus one additional can have a significant impact on cellular excitability.
subunit, often a subunit (from 1, 2, 3) but sometimes Furthermore, studies done recently have reported that the
from a , , , or subunit.7 The 122 subunit onset and offset kinetics of direct gating are particularly
combination is the most common combination in the slow.7 This may be the reason which has led to an under-
mammalian brain.7 Moreover, the 2 subunit is appreciation of the potency of neurosteroids directly
particularly important for synaptic localization/clustering activating the channel.
of GABAA receptors as it contains sequence motifs,
which are responsible for synaptic targeting.

A schematic of a single GABAA receptor subunit and the


pentameric receptor with sites of action for important
modulatory drugs are shown in Figure 3A and 3B
respectively.7

Interaction with GABAA receptors


The 3-reduced metabolites of progesterone and
deoxycorticosterone ( 3,5-tetrahydroprogesterone
/3,5-THP and 3,5 tetrahydrodeoxycorticosterone
/3,5-THDOC respectively) were the first steroids that
have shown to have modulatory effect on neuronal
excitability via their interaction with GABA type A Figure 4: Top-down view of the pentameric GABAA
receptors.1,6 The most prominent interaction of receptor showing proposed sites of potentiation and
neurosteroids with GABAA receptors is potentiating direct gating for neurosteroids.7
action rather antagonistic actions and is seen with the
endogenous 3,5-THP (allopregnanolone), 3,5-THP Neuroactive steroids are among the most potent and
(pregnanolone), 35-THDOC and 35-THDOC.1,5,6,7 efficacious modulators of GABAA receptors. When
This positive allosteric modulatory action is because they compared with benzodiazepines their potency is less
enhance the GABA-evoked chloride current either by (half-maximum effects in the nanomolar concentration
increasing frequency and/or by increasing duration of range), but generate higher maximum potentiation of
openings of the GABA-gated chloride channel.5,7,8 As GABA responses and therefore they are more efficacious.
neurosteroids are synthesized from cholesterol via On the other hand, neuroactive steroids are less
progesterone or deoxycorticosterone; therefore, efficacious and more potent than barbiturate. 7 Many
fluctuations in their levels may influence the modulatory review articles in the literature show that the action of
effects that neurosteroids have over the function of the neurosteroids is highly specific, being both brain region
GABAA receptors.5,7 and neuron dependent. This selectivity is due to the
composition of the GABAA receptor subunit, the
Non-competitive antagonist action is seen with steroids differential expression of steroid-synthesizing and
sulfated at C3, typified by pregnenolone sulfate (PS) and metabolizing enzymes, and local steroid metabolism and
sulfated pregnane steroids, with either or phosphorylation mechanisms.5,9 Neuroactive steroids act
stereochemistry at C3 and at C5.5,7 The IC50 (half on both synaptic and extrasynaptic GABAA receptors.
maximal inhibitory concentration) values for the action of Moreover, extrasynaptic GABAA receptors are highly
this class of neuroactive steroids is typically in the high sensitive to neurosteroids9 and appear to play a vital role
nanomolar to micromolar range. Finally, 3-OH steroids in the neuronal plasticity changes that accompany stress,
can also antagonize GABAA receptors but the potency puberty and the ovarian cycle. 5,9
and perhaps efficacy of 3-OH steroids is weaker than
that of sulfated steroids.7 Expression of the GABAA receptors
Previously, it was assumed that the neuroactive steroids
do not regulate gene expression because they do not bind

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 153
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

to a known intracellular steroid hormone receptor. Ganaxolone, the synthetic 3b-methyl analogue of
Research done using a cotransfection system with allopregnanolone (3- hydroxy-3-methyl-5-pregnan-
recombinant progesterone receptors and the mouse 20-one) is the only neurosteroid that has been evaluated
mammary tumour virus (MTV) has shown that the in human clinical trials for the treatment of epilepsy.23
neuroactive steroids like 3,5-THP and 3,5-THDOC Various safety and pharmacokinetic studies conducted on
activate gene expression effectively and enhance the human subjects including children have shown that
nuclear translocation.1 ganaxolone is well tolerated. However, it is associated
with dose limiting adverse effects such as sedation and
Recently, it has been shown that both chronic exposure somnolence which are reversible. Ganaxolone is
and withdrawal from exogenous neurosteroids increase metabolized by CYP3A to 16-hydroxyganaxolone, which
expression of the GABAA receptor 4-subunit to produce is inactive and then to various multihydroxylated
CNS hyperexcitability.3 This increase in the expression of metabolites. Despite the metabolism by microsomal
the 4-subunit is responsible for benzodiazepine enzyme, the information available to date indicates that
insensitivity, one common factor in experimental rodent ganaxolone will have low drug-drug interactions.4,24
models and the clinical presentation of premenstrual Therefore, it could be administered easily with
dysphoric disorder. concomitant antiepileptic drugs. However, clinical trials
having large sample size and with randomized, controlled
CLINICAL IMPORTANCE OF NEUROSTEROIDS and double blind study design are needed before its use in
IN EPILEPSY clinical practice.

Neurosteroids including, allopregnanolone and Catamenial Epilepsy


tetrahydrodeoxycorticosterone (THDOC) that act as Catamenial epilepsy is a form of pharmacoresistant
positive GABAA receptor modulators have shown to epilepsy in which seizure exacerbations occur in cyclical
exhibit broad-spectrum anticonvulsant effects in diverse manner during particular phases of the menstrual cycle in
rodent seizure models. They protect against seizures women with pre-existing epilepsy. It is estimated that up
induced by GABAA receptor antagonists including to 70% of women of child-bearing age with epilepsy
pentylenetetrazol (PTZ) and bicuculline and they are experience catamenial seizure exacerbations.25-27
effective against pilocarpine-induced limbic seizures and Catamenial epilepsy is classified into three forms: (1)
seizures in kindled animals.10-13 Neurosteroids are also perimenstrual (C1: days -3 to 3) and (2) periovulatory
highly effective in suppressing seizures due to (C2: days 10 to -13) in normal cycles, and (3) luteal (C3:
withdrawal of GABAA receptor modulator drugs days 10 to 3) in inadequate luteal phase cycles, wherein
including neurosteroids and benzodiazepines. However, it day 1 is the first day of menstrual flow and ovulation is
is seen in animal studies that neurosteroids may presumed to occur 14 days before the subsequent onset of
exacerbate generalized absence seizures.4,14,15 menses (day -14).24 Neurosteroids have been implicated
Neurosteroids differ from benzodiazepine that they do only in the seizure exacerbations which occur during
not act on the benzodiazepine site of GABAA receptors perimenstrual period which is also the most common
and also they are able to modulate all isoforms of form of catamenial epilepsy. It is hypothesized that
GABAA receptors, including benzodiazepine-insensitive withdrawal of progesterone derived neurosteroids
one. As a result anticonvulsant tolerance is not seen with predispose to seizure because of enhanced brain
neurosteroids in contrast to benzodiazepines.16,17 Thus, excitability.
neurosteroids have the potential to be used in the chronic
treatment of epilepsy and laid the basis for clinical trials. During follicular phase of the menstrual cycle, circulating
progesterone along with its metabolite allopregnanolone
The sulfated neurosteroids pregnenolone sulfate and levels are low but their level rise in the mid-luteal phase
dehydroepiandosterone sulfate, which act as GABAA for about 10 to 11 days. However during late luteal phase
receptor antagonists are proconvulsant when circulating progesterone levels decline causing
administered at high doses into the brain, producing withdrawal of the anticonvulsant effects of
seizures and status epilepticus.18 It is unlikely that they neurosteroids.28 Circulating THDOC levels though lower
exist endogenously even if they do, their concentrations than those of allopregnanolone fluctuate during the
is not sufficiently high to exert proconvulsant effects. menstrual cycle, with higher levels in the luteal phase.
Therefore, their physiological relevance is unclear. Study done recently suggested that serum THDOC levels
However, it is known that the proconvulsant effects of could contribute to catamenial epilepsy, but it appears to
these steroids can be blocked by co-administration of be less relevant.4,24 An important question, whether the
allopregnanolone or other neurosteroids that positively local brain synthesis of neurosteroids also fluctuates is
modulate GABAA receptors.19 There is some limited still unanswered.
evidence that endogenous neurosteroids play a role in
regulating epileptogenesis making them potentially useful Pliability in GABAA receptors during perimenstrual
for disease modifying therapeutic activity.20-22 period is also thought to play a role in the enhanced brain
excitability, which leads to increase in seizure
susceptibility in perimenstrual catamenial epilepsy.24

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 154
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

However, there is a lack of conclusive evidence that catamenial epilepsy, there is only limited circumstantial
changes in expression of brain GABAA receptor subunit data available to support their use.36
occurs during the human menstrual cycle. Studies in rats
have shown that prolonged exposure to allopregnanolone A neurosteroid could be administered in a pulse prior
causes increased expression of the 4 GABAA receptor to menstruation but continuous administration throughout
subunit in hippocampus. This results in decreased the month is preferred. Continuous administration would
benzodiazepine sensitivity of GABAA receptor as avoid withdrawal of the therapeutic agent, which itself
benzodiazepines only act on GABAA receptors containing could predispose to seizures, also there is difficulty in
2 subunits and not containing 4 or 6 subunits. 29 predicting the time of menstrual periods in many women.
Treatment of rats with allopregnanolone also results in They are administered at low doses to avoid sedative side
transient increased expression of the subunit in effects. Though, it contributes little to anticonvulsant
hippocampus. It can coassemble with 4 to form activity during most of the menstrual cycle, at the time of
nonsynaptic (perisynaptic/extrasynaptic) GABAA menstruation increased potency of the neurosteroid would
receptors and increased benzodiazepine-insensitive tonic confer protection against perimenstrual seizure
current.30,31 However, it is unclear that increased exacerbations. It is noteworthy that there is no
subunit expression has relevance in catamenial epilepsy corresponding increases in side effects (mainly
as increase in subunit may be transitory. sedation).24 Therefore, enhanced side effects which would
negate the potential of the therapeutic approach, would
Therefore, an important result of increased 4 subunit not be expected to occur.
into synaptic GABAA receptors causes accelerated decay
of synaptic currents generated by these receptors, which No neurosteroid is currently approved. The data from
results in reduced inhibition.32 Neurosteroids cause a human clinical trials suggests that adjunctive cyclic
prolongation of the decay of GABA-mediated synaptic progesterone supplementation may be useful in women
currents. As a result during the luteal phase, presence of with catamenial epilepsy.37,38 However, there are chances
high levels of allopregnanolone and the acceleration due that Progesterone therapy in women may cause hormonal
to 4 substitution are balanced.24 However, at the time of effects such as breakthrough vaginal bleeding, breast
menstruation when neurosteroids are withdrawn; synaptic tenderness, weight gain, sedation and emotional
inhibition is diminished resulting in enhanced excitability depression. In the treatment regimen used in the clinical
predisposing to seizures. trials, progesterone is administered only after cycle day
14 and is tapered and discontinued during days 26 to 28,
The neurosteroid withdrawal rodent model of catamenial as it is believed that starting earlier than mid-cycle would
epilepsy was used to investigate therapies for interfere with normal cycling causing irregular bleeding
perimenstrual catamenial epilepsy.33,34 A major outcome and abrupt discontinuation can result in rebound seizure
was that conventional antiepileptic drugs , like exacerbation. An advantage of hormonally-inactive
benzodiazepines and valproate, have reduced potency in neurosteroids is that they have no hormonal adverse
protecting against seizures during the period of enhanced effects and can be administered throughout the cycle,
seizure susceptibility following neurosteroid withdrawal. simplifying the treatment regimen.24,36
This situation was similar to the catamenial epilepsy in
women where breakthrough seizures occur despite Preliminary uncontrolled open label study was conducted
treatment with antiepileptic drugs. Moreover, to evaluated safety and efficacy of ganaxolone in women
neurosteroids including allopregnanolone, THDOC and with catamenial epilepsy.22,24,36 These women received
their 5-isomers, were found to have enhanced activity in oral ganaxolone (300 mg/day, twice a day) starting on
the perimenstrual catamenial epilepsy model.33 This day 21 of the menstrual cycle and continued up to third
suggested a novel approach of neurosteroid day of next menstrual cycle. It showed that ganaxolone
replacement for the treatment of catamenial seizure appeared to reduce their catamenial seizures. Study also
exacerbations.24 showed that it was well tolerated and was associated with
no hormonal adverse effects. However, further clinical
While neurosteroid reduction by itself does not lead to trial with a randomized, controlled study design is
epilepsy, it is well established that endogenous required before this agent can be recommended for
neurosteroids do modulate seizure susceptibility in clinical use.
epileptic animals. Moreover, in the catamenial epilepsy
model neurosteroid withdrawal causes markedly greater Stress-Induced Seizure
seizure provocation, consistent with earlier studies Body respond to physiological stress by increasing
demonstrating enhanced seizure susceptibility in acute circulating levels of neurosteroids. Stress results in
seizure models. Importantly, it supports the concept that stimulation of hypothalamo-pituitary-adrenal (HPA) axis
neurosteroid replacement may be useful in the treatment leading to enhanced synthesis of adrenal cortisol and
of seizures associated with neurosteroid fluctuations such neurosteroid, deoxycorticosterone (DOC).39,40 THDOC is
as catamenial epilepsy.35Although neurosteroids seems to derived nearly exclusively from adrenal sources, while
be the most direct approach to the treatment of allopregnanolone is derived either from brain, adrenal
gland and/or gonads proved in animal studies.41 Acute

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 155
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

stress such as swimming, foot shock or carbon dioxide properties.4,49,50 Studies have shown that serum levels of
exposure elicit rapid rise in plasma and brain levels of these neurosteroids are reduced in men with epilepsy
THDOC and allopregnanolone.41-43 Normal plasma levels compared with control subjects.4 Therefore, neurosteroid
of THDOC fluctuate between 1 and 5 nM, but following replacement might be a useful therapeutic approach.
the acute stress it increase to 15-30 nM and might reach
40-60 nM during pregnancy. On the other hand, However in temporal lobe epilepsy there are some
allopregnanolone levels typically reach 70-160 nM biological factors which may influence the sensitivity of
during the third trimester of pregnancy and have been endogenous neurosteroids and therefore could have an
measured as high as 220 nM.44 influence on the efficacy of exogenous neurosteroids
used in epilepsy therapy. Studies in a status epilepticus
Various animal studies have shown that stress-induced model of TLE have shown that in the dentate gyrus there
neurosteroids elevate seizure threshold and have is a reduction in -subunit containing GABAA receptors,51
anticonvulsant effects against seizures induced by PTZ also the neurosteroids modulation of synaptic currents is
and other GABAA receptor antagonists.41This diminished along with presence of 4 subunit containing
anticonvulsant action of stress induced neurosteroids receptors at synapses.52 All of these changes may reduce
could be due to circulating neurosteroids synthesized in the efficacy of endogenous neurosteroids and may
peripheral tissues or to those produced locally in the facilitate seizures in epileptic animals. Therefore, before
brain. Though, stress-induced enhanced neurosteroid considering the neurosteroid replacement therapy in TLE
production and their protective effect against seizure is these biological factures should be considered.
seen in animals,41 it is unlikely to see reduction in seizure
frequency associated with stress in epileptic patients. Alcohol Withdrawal Seizures
Indeed, stress has been reported to trigger seizure activity Ethanol, when administered systemically at moderate
in persons with epilepsy.45 During the stressful episodes, doses of (1-2.5 g/kg) is believed to activate HPA axis.
it is expected to have fluctuation in adrenal hormone This causes increase in plasma and brain neurosteroids
levels and may be the withdrawal of THDOC during such that may contribute to many of the its behavioural
fluctuations is associated with seizure provocation. effects in rodents.53 Chronic ethanol administration
Alternatively, other unidentified hormonal factors with causes elevation of neurosteroids like, allopregnanolone
proconvulsant activity may be responsible for stress- and THDOC and their enhanced anticonvulsant actions
induced increases in seizures. However, the type of stress These effects are associated with increases in the
experienced by patients with epilepsy is a chronic type, sensitivity of GABAA receptors to neurosteroids.53
and it is likely that it has different endocrinological Though endogenous neurosteroids may protect against
consequences than acute stress. The effects of ethanol withdrawal seizures but, ethanol induction of
fluctuations in neurosteroids on seizures in chronic stress allopregnanolone is diminished in tolerant and dependent
remain to be studied. animals. Reduced availability of allopregnanolone under
such circumstances may be a factor that predisposes to
Temporal Lobe Epilepsy alcohol withdrawal seizures. Therefore, neurosteroid
Epilepsy is commonly associated with sexual and replacement could possibly be useful in the treatment of
reproductive dysfunction.46 Patients of temporal lobe alcohol withdrawal seizures.54
epilepsy (TLE) particularly men often have diminished
libido and sexual potency that is associated with low Status Epilepticus
testosterone levels.47 There are two factors which can be Status epilepticus is an emergency neurological
attributed to this hypogonadal state, firstly, effects of condition. It is characterized by persistent seizures lasting
certain hepatic enzyme-inducing antiepileptic drugs and more than 30 minutes, resistance to benzodiazepine
secondly, the connections between temporal lobe treatment and progressive internalization of synaptic
structures such as the amygdala and hypothalamic nuclei GABAA receptors. Therefore, new therapies with novel
that governs the production and secretion of gonadotropin mechanism of actions are needed for refractory status
releasing hormone, which is responsible for suppression epilepticus.4 During the attack of status epilepticus, the
of the hypothalamic-pituitary-gonadal axis by limbic extra synaptic -subunit containing GABAA receptors
seizures.46,47 that generate tonic inhibition do not internalize.
Considering this factor, neurosteroids having positive
A study conducted on men have shown that after modulatory action on GABAA receptors with strong
temporal lobe epilepsy surgery, serum androgens level anticonvulsant activity, could be more effective for
becomes normal which results in successful seizure treatments of status epilepticus.4,55 Preclinical studies
control. However, normalization of serum androgens is involving animal model have shown that neurosteroids
not seen in those that continue to have seizures, like THDOC can effectively terminate chemically
supporting the observation that seizures are responsible electrographic and behavioural status epilepticus.55
for the hypoandrongenic state.48 As noted earlier, Treatment with THDOC successfully aborted seizures
testosterone serves as a precursor for neurosteroids like with sustained suppression of status epilepticus, with
androstanediol which are a positive modulator of GABAA efficacy superior to the benzodiazepine. In addition,
receptor that is responsible for their anticonvulsant THDOC therapy may confer significant neuroprotection

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 156
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

by diminishing the neuronal cell death associated with mechanism and possible significance. Prog
status epilepticus. Further studies are needed to clarify Neurobiol 2009;89:134-52.
whether neurosteroids might be valuable in the treatment 3. Melcangi RC, Panzica G, Garcia-Segura LM.
of status epilepticus. Neuroactive steroids: Focus on human brain.
Neuroscience 2011;191:1-5.
Infantile Spasm 4. Reddy DS. Role of anticonvulsant and
Infantile spasm is catastrophic form of epileptic antiepileptogenic neurosteroids in the
encephalopathy in infants and young children. ACTH has pathophysiology and treatment of epilepsy. Front
been used for many years as a treatment for infantile Endocrinol (Lausanne) 2011;2:38.
spasms. Despite the numerous studies in literature 5. Morrow AL. Recent developments in the
reporting effectiveness of ACTH and oral steroids in significance and therapeutic relevance of
treatment of infantile spasm, the exact mechanism is neuroactive steroids--Introduction to the special
unknown.56 Clinical benefits of ACTH in infantile issue. Pharmacol Ther 2007;116:1-6.
spasms thought to be due to release of 6. Rupprecht R, di Michele F, Hermann B, Strhle A,
adrenocorticosteroids and neurosteroids. Recent studies Lancel M, Romeo E, et al. Neuroactive steroids:
show that several newer anticonvulsant agents (such as Molecular mechanisms of action and implications
vigabatrin and benzodiazepines), which are positive for neuropsychopharmacology. Brain Res Brain Res
allosteric modulators of GABAA receptors, are as Rev 2001;37:59-67.
effective as ACTH in acutely controlling infantile 7. Akk G, Covey DF, Evers AS, Steinbach JH,
spasms. Ganaxolone, a novel neuroactive steroid, Zorumski CF, Mennerick S. Mechanisms of
however has demonstrated better efficacy and tolerability neurosteroid interactions with GABA(A) receptors.
in children with intractable infantile spasms. Results of Pharmacol Ther 2007;116:35-57.
an open-label, add-on trial of ganaxolone showed that it 8. Mellon SH, Griffin LD. Neurosteroids:
is safe and effective in treating refractory infantile spasms Biochemistry and clinical significance. Trends
patients.57 However further investigation is required Endocrinol Metab 2002;13:35-43.
before starting its clinical use. 9. Herd MB, Belelli D, Lambert JJ. Neurosteroid
modulation of synaptic and extrasynaptic GABA(A)
CONCLUSIONS receptors. Pharmacol Ther 2007;116:20-34.
10. Kokate TG, Svensson BE, Rogawski MA.
Awareness of the prominent role of neurosteroids in Anticonvulsant activity of neuroactive steroids:
neurological disorders is rapidly increasing. Numerous correlation with gamma-aminobutyric acid-evoked
basic and clinical investigations carried out over the past chloride current potentiation. J Pharmacol Exp Ther
few year support their central role in normal 1994;270:1223-9.
physiological functioning as well as in illness. 11. Belelli D, Bolger MB, Gee KW. Anticonvulsant
Neurosteroids that are positive modulator of GABAA profile of the progesterone metabolite 5-pregnan-
receptor have potent anticonvulsants action and may 3-ol-20-one. Eur J Pharmacol 1989;166:325-9.
regulate various neuronal excitability networks. 12. Frye CA. The neuroactive steroid 3,5-THP has
Neurosteroids are believed to play a role in the regulation anti-seizure and possible neuroprotective effects in
of seizure susceptibility in the setting of pre-existing an animal model of epilepsy. Brain Res
epilepsy. Although, preliminary investigation into the use 1995;696:113-20.
of neurosteroid analogues or manipulation of the 13. Wieland S, Belluzzi JD, Stein L, Lan NC.
neurosteroid system in treatment remain promising, such Comparative behavioral characterization of the
as the use of ganaxolone in epilepsy. Further studies are neuroactive steroids 3-OH,5-pregnan-20-one and
required to determine whether neurosteroid 3-OH,5-pregnan-20-one in rodents.
replacement is a useful approach for epileptic seizures Psychopharmacology 1995;118:65-71.
related to endogenous neurosteroid uctuations, such as 14. Citraro R, Russo E, Di Paola ED, Ibbadu GF,
in catamenial epilepsy. Gratteri S, Marra R, De Sarro G. Effects of some
neurosteroids injected into some brain areas of
Funding: No funding sources WAG/Rij rats, an animal model of generalized
Competing interests: There are no competing interests to absence epilepsy. Neuropharmacology
declare 2006;50:1059-71.
15. Snead OC 3rd. Ganaxolone, a selective, high-
REFERENCES affinity steroid modulator of the -aminobutyric
acid-A receptor, exacerbates seizures in animal
1. Rupprecht R. Neuroactive steroids: Mechanisms of models of absence. Ann Neurol 1998;44:688-91.
action and neuropsychopharmacological properties. 16. Reddy DS, Rogawski MA. Chronic treatment with
Psychoneuroendocrinology 2003;28:139-68. the neuroactive steroid ganaxolone in the rat induces
2. Zheng P. Neuroactive steroid regulation of anticonvulsant tolerance to diazepam but not to
neurotransmitter release in the CNS: Action, itself. J Pharmacol Exp Ther 2000;295:1241-8.

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 157
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

17. Kokate TG, Yamaguchi S, Pannell LK, Rajamani U, pharmacological and behavioral effects.
Carroll DM, Grossman AB, Rogawski MA. Lack of Neuropharmacology 2005;49:573-86.
anticonvulsant tolerance to the neuroactive steroid 31. Maguire JL, Stell BM, Rafizadeh M, Mody I.
pregnanolone in mice. J Pharmacol Exp Ther Ovarian cycle-linked changes in GABAA receptors
1998;287:553-8. mediating tonic inhibition alter seizure susceptibility
18. Williamson J, Mtchedlishvili Z, Kapur J. and anxiety. Nat Neurosci 2005;8:797-804.
Characterization of the convulsant action of 32. Smith SS, Gong QH. Neurosteroid administration
pregnenolone sulfate. Neuropharmacology and withdrawal alter GABAA receptor kinetics in
2004;46:856-64. CA1 hippocampus of female rats. J Physiol
19. Reddy DS, Kulkarni SK. Proconvulsant effects of 2005;564:421-36.
neurosteroid pregnenolone sulfate and 33. Reddy DS, Rogawski MA. Enhanced anticonvulsant
dehydroepiandrosterone sulfate in mice. Eur J activity of neuroactive steroids in a rat model of
Pharmacol 1998;345:55-9. catamenial epilepsy. Epilepsia 2001;42:303-10.
20. Biagini G, Panuccio G, Avoli M. Neurosteroids and 34. Reddy DS, Rogawski MA. Enhanced anticonvulsant
epilepsy. Curr Opin Neurol 2010;23:170-6. activity of ganaxolone after neurosteroid withdrawal
21. Biagini G, Longo D, Baldelli E, Zoli M, Rogawski in a rat model of catamenial epilepsy. J Pharmacol
MA, Bertazzoni G, Avoli M. Neurosteroids and Exp Ther 2000;294:909-15.
epileptogenesis in the pilocarpine model: Evidence 35. Lawrence C, Martin BS, Sun C, Williamson J,
for a relationship between P450scc induction and Kapur J. Endogenous neurosteroid synthesis
length of the latent period. Epilepsia 2009;50 Suppl modulates seizure frequency. Ann Neurol
1:53-8. 2010;67:689-93.
22. Biagini G, Baldelli E, Longo D, Pradelli L, Zini I, 36. McAuley JW, Moore JL, Reeves AL, Flyak J,
Rogawski MA, Avoli M. Endogenous neurosteroids Monaghan EP, Data J. A pilot study of the
modulate epileptogenesis in a model of temporal neurosteroid ganaxolone in catamenial epilepsy:
lobe epilepsy. Exp Neurol 2006;201:519-24. clinical experience in two patients. Epilepsia
23. Monaghan EP, McAuley JW, Data JL. Ganaxolone: 2001;42:85.
a novel positive allosteric modulator of the GABAA 37. Herzog AG, Fowler KM, Smithson SD, Kalayjian
receptor complex for the treatment of epilepsy. LA, Heck CN, Sperling MR, Liporace JD, Harden
Expert Opin Investig Drugs 1999;8:1663-71. CL, Dworetzky BA, Pennell PB, Massaro JM.
24. Reddy DS, Rogawski MA. Neurosteroid Progesterone vs placebo therapy for women with
replacement therapy for catamenial epilepsy. epilepsy: A randomized clinical trial. Neurology
Neurotherapeutics 2009;6:392-401. 2012;78:1959-66.
25. Reddy DS. The role of neurosteroids in the 38. Herzog AG. Hormonal therapies: progesterone.
pathophysiology and treatment of catamenial Neurotherapeutics 2009;6:383-91.
epilepsy. Epilepsy Res 2009;85:1-30. 39. Biggio G, Concas A, Follesa P, Sanna E, Serra M.
26. Bazan AC, Montenegro MA, Cendes F, Min LL, Stress, ethanol, and neuroactive steroids. Pharmacol
Guerreiro CA. Menstrual cycle worsening of Ther 2007;116:140-71.
epileptic seizures in women with symptomatic focal 40. Reddy DS. Physiological role of adrenal
epilepsy. Arg Neuropsiguiatr 2005;63(3B):751-6. deoxycorticosterone-derived neuroactive steroids in
27. Herzog AG, Harden CL, Liporace J, Pennell P, stress-sensitive conditions. Neuroscience
Schomer DL, Sperling M, Fowler K, Nikolov B, 2006;138:911-20.
Shuman S, Newman M. Frequency of catamenial 41. Reddy DS, Rogawski MA. Stress-induced
seizure exacerbation in women with localization- deoxycorticosterone-derived neuroactive steroids
related epilepsy. Ann Neurol 2004;56:431-4. modulates GABAA receptor function and seizure
28. Tuveri A, Paoletti AM, Orr M, Melis GB, Marotto susceptibility. J Neurosci 2002;42:3795-805.
MF, Zedda P, Marrosu F, Sogliano C, Marra C, 42. Barbaccia ML, Roscetti G, Trabucchi M, Mostallino
Biggio G, Concas A. Reduced serum level of MC, Concas A, Purdy RH, Biggio G.
THDOC, an anticonvulsant steroid, in women with Timedependent changes in rat brain neuroactive
perimenstrual catamenial epilepsy. Epilepsia steroid concentrations and GABAA receptor
2008;49:1221-9. function after acute stress. Neuroendocrinology
29. Gangisetty O, Reddy DS. Neurosteroid withdrawal 1996;63:166-72.
regulates GABAA receptor alpha4-subunit 43. Barbaccia ML, Roscetti G, Trabucchi M, Purdy RH,
expression and seizure susceptibility by activation Mostallino MC, Concas A, Biggio G. The effects of
of progesterone receptor-independent early growth inhibitors of GABAergic transmission and stress on
response factor-3 pathway. Neuroscience brain and plasma allopregnanolone concentrations.
2010;170:865-80. Br J Pharmacol 1997;120:1582-8.
30. Shen H, Gong QH, Yuan M, Smith SS. Short-term 44. Parzek A, Hill M, Kancheva R, Havlkov H,
steroid treatment increases GABAA receptor Kancheva L, Cindr J, Paskov A, Pouzar V, Cerny I,
subunit expression in rat CA1 hippocampus: Drbohlav P, Hjek Z, Strka L. Neuroactive

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 158
Patil CY et al. Int J Basic Clin Pharmacol. 2012 Dec;1(3):150-159

pregnanolone isomers during pregnancy. J Clin granule cell dendrites influences tonic and phasic
Endocrinol Metab 2005;90:395-403. inhibition in a mouse model of epilepsy. J Neurosci
45. Frucht MM, Quigg M, Schwaner C, Fountain NB. 2007;27:7520-31.
Distribution of seizure precipitants among epilepsy 52. Sun C, Mtchedlishvili Z, Erisir A, Kapur J.
syndromes. Epilepsia 2000;41:1534-9. Diminished neurosteroid sensitivity of synaptic
46. Edwards HE, MacLusky NJ, Burnham WM. inhibition and altered location of the 4 subunit of
Epileptic seizures: Do they cause reproductive GABAA receptors in an animal model of epilepsy. J
dysfunction? Univ Toronto Med J 2000;77:104-11. Neurosci 2007;27:12641-50.
47. Herzog AG. Altered reproductive endocrine 53. Morrow AL, Porcu P, Boyd KN, Grant KA.
regulation in men with epilepsy: implications for Hypothalamic-pituitary-adrenal axis modulation of
reproductive function and seizures. Ann Neurol GABAergic neuroactive steroids influences ethanol
2002;51:539-42. sensitivity and drinking behavior. Dialogues Clin
48. Bauer J, Stoffel-Wagner B, Flgel D, Kluge M, Neurosci 2006;8:463-77.
Schramm J, Bidlingmaier F, Elger CE. Serum 54. NGouemo P, Rogawski MA. Alcohol withdrawal
androgens return to normal after temporal lobe seizures. In: Pitknen A, Schwartzkroin PA, Mosh
epilepsy surgery in men. Neurology 2000;55:820- SL, editors. Models of Seizures and Epilepsy.
24. Amsterdam: Elsevier Academic Press; 2006:161-77.
49. Reddy DS. Anticonvulsant activity of the 55. Kuruba R, Reddy DS. Neuroprotective effects of
testosterone-derived neurosteroid 3-androstanediol. GABAergic agents in the rat model of refractory
Neuroreport 2004;15:515-8. status epilepticus. Soc Neurosci Abstr 2011;PN
50. Reddy DS. Testosterone modulation of seizure 338.08.
susceptibility is mediated by neurosteroids 3- 56. Shields WD. Infantile Spasms: Little Seizures, BIG
androstanediol and 17-estradiol. Neuroscience Consequences. Epilepsy Curr 2006;6:63-9.
2004;129:195-207. 57. Anovadiya AP, Sanmukhani JJ, Tripathi CB.
51. Zhang N, Wei W, Mody I, Houser CR. Altered Epilepsy: Novel therapeutic targets. J Pharmacol
localization of GABAA receptor subunits on dentate Pharmacother 2012;3:112-7.

doi: 10.5455/2319-2003.ijbcp004212
Cite this article as: Patil CY, Jadhav SA, Doifode
SM, Baig MS. Neuroactive steroids and their role in
epilepsy. Int J Basic Clin Pharmacol 2012;1:150-9.

International Journal of Basic & Clinical Pharmacology | November-December 2012 | Vol 1 | Issue 3 Page 159

You might also like