Sei sulla pagina 1di 9

Progress in Neurobiology 113 (2014) 79–87

Contents lists available at ScienceDirect

Progress in Neurobiology
journal homepage: www.elsevier.com/locate/pneurobio

The role of allopregnanolone in depression and anxiety


Cornelius Schüle a,*, Caroline Nothdurfter b, Rainer Rupprecht b
a
Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University, Nussbaumstr. 7, 80336 Munich, Germany
b
Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany

A R T I C L E I N F O A B S T R A C T

Article history: Neuroactive steroids such as allopregnanolone do not only act as transcriptional factors in the regulation
Received 4 June 2013 of gene expression after intracellular back-oxidation into the 5-a pregnane steroids but may also alter
Received in revised form 21 September 2013 neuronal excitability through interactions with specific neurotransmitter receptors. In particular, certain
Accepted 21 September 2013
3a-reduced metabolites of progesterone such as 3a,5a-tetrahydroprogesterone (allopregnanolone) and
Available online 8 November 2013
3a,5b-tetrahydroprogesterone (pregnanolone) are potent positive allosteric modulators of the GABAA
receptor complex. During the last years, the downregulation of neurosteroid biosynthesis has been
Keywords:
intensively discussed to be a possible contributor to the development of anxiety and depressive disorder.
Allopregnanolone
Reduced levels of allopregnanolone in the peripheral blood or cerebrospinal fluid were found to be
Neuroactive steroids
Depression associated with major depression, anxiety disorders, premenstrual dysphoric disorder, negative
Anxiety symptoms in schizophrenia, or impulsive aggression. The importance of allopregnanolone for the
Affective disorders regulation of emotion and its therapeutical use in depression and anxiety may not only involve
GABAergic mechanisms, but probably also includes enhancement of neurogenesis, myelination,
neuroprotection, and regulatory effects on HPA axis function. Certain pharmacokinetic obstacles limit
the therapeutic use of natural neurosteroids (low bioavailability, oxidation to the ketone). Until now
synthetic neuroactive steroids could not be established in the treatment of anxiety disorders or
depression. However, the translocator protein (18 kDa) (TSPO) which is important for neuroster-
oidogenesis has been identified as a potential novel target. TSPO ligands such as XBD 173 increase
neurosteroidogenesis and have anxiolytic effects with a favorable side effect profile.
ß 2013 Published by Elsevier Ltd.

Contents

1. Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
2. Allopregnanolone and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
2.1. Allopregnanolone in animal models of depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
2.2. Allopregnanolone and clinical studies in depressed patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
3. Allopregnanolone and anxiety. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
3.1.1. Allopregnanolone in animal models of anxiety . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
3.2. Allopregnanolone and clinical studies in patients suffering from anxiety disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
4. Conclusions and future directions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

1. Background one receptor active 5 alpha-pregnane steroids (Rupprecht et al.,


1993). However, neuroactive steroids may also alter neuronal
Neuroactive steroids act as transcriptional factors in the excitability through interactions with specific neurotransmitter
regulation of gene expression. The induction of DNA binding and receptors (Rupprecht and Holsboer, 1999; Rupprecht, 2003).
transcriptional activation of the progesterone receptor requires These steroids are synthesized from cholesterol or steroidal
intracellular oxidation of the neuroactive steroids into progester- precursors (Fig. 1). The formation of pregnenolone from choles-
terol is regulated by the translocator protein TSPO (18 kD),
formerly called peripheral or mitochondrial benzodiazepine
* Corresponding author. Tel.: +49 89 5160 5335. receptor, which is mainly located in the outer mitochondrial
E-mail address: Cornelius.Schuele@med.uni-muenchen.de (C. Schüle). membrane and favors the transport of cholesterol to the inner

0301-0082/$ – see front matter ß 2013 Published by Elsevier Ltd.


http://dx.doi.org/10.1016/j.pneurobio.2013.09.003
80 C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87

Fig. 1. Synthesis of neurosteroids (Nothdurfter et al., 2012a). The cholesterol side-chain-cleaving cytochrome-P-450 enzyme (P-450scc, CYP11A1) is located at the inner
mitochondrial membrane and converts cholesterol to pregnenolone. In the cytoplasm (diffusion marked by black arrow), pregnenolone is metabolized to progesterone by the
microsomal 3b-dehydrogenase/D5–D4 isomerase. Progesterone is further converted to deoxycorticosterone by the 21-hydroxylase (CYP21B). The reduction of progesterone
and deoxycorticosterone by the 5a-reductase leads to 5a-dihydroprogesterone and 5a-dihydrocorticosterone (5a-DHDOC). The neurosteroids allopregnanolone and
allotetrahydrodeoxycorticosterone (3a, 5a-tetrahydrodeoxycorticosterone, 3a, 5a-THDOC) are formed by further reduction through 3a-hydroxysteroid dehydrogenase
(3a-HSD).

mitochondrial membrane, thereby promoting neurosteroido- study hypothesized that these results may explain the rapid
genesis (Papadopoulos et al., 2006; Rupprecht et al., 2010). amelioration of dysphoria and anxiety symptoms associated with
Pregnenolone is further converted into an array of different major depression and late luteal phase dysphoria by SSRIs.
steroids (Fig. 1). Progesterone may be formed by the 3b- However, another study, which did not find any change in the
hydroxysteroid dehydrogenase and serves as the main precursor activity of this enzyme after administration of various SSRIs, came
molecule for 3a-reduced neuroactive steroids. Progesterone and to the conclusion that the interference with the 3a-HSD type 3
deoxycorticosterone (DOC) are irreversibly reduced by the 5a- might not be the crucial mechanism by which SSRIs increase
reductase into 5a-dihydroprogesterone (5a-DHP) and 5a-dihy- allopregnanolone concentrations (Trauger et al., 2002).
drodeoxycorticosterone (5a-DHDOC). These pregnane steroids In our research group we examined the effects of the
may be further reduced to 3a,5a-tetrahydroprogesterone antidepressant drug mirtazapine (a2-, 5-HT2-, 5-HT3- and H1-
(3a,5a-THP; 3a-hydroxy-5a-pregnan-20-one; allopregnano- antagonist) on the reduction of 5a-DHP (into 3a,5a-THP = allo-
lone), 3a,5b-tetrahydroprogesterone (3a,5b-THP; 5b-pregnan- pregnanolone) catalyzed by the cytosolic 3a-HSD type 3 and on
3a-ol-20-one; pregnanolone) and 3a,5a-tetrahydrodeoxycorti- the oxidation of 3a,5a-THP (into 5a-DHP) catalyzed by the
costerone (3a,5a-THDOC; 3a,21-dihydroxy-5a-pregnan-20- microsomal 3a-HSD (Schule et al., 2006b). Pure recombinant
one; allotetrahydrodeoxycorticosterone) by the 3a-hydroxyster- human cytosolic 3a-HSD type 3 protein was expressed in
oid dehydrogenase (3a-HSD). Escherichia coli, whereas the cDNA for human microsomal 3a-
In particular, certain 3a-reduced metabolites of progesterone HSD was stably expressed in HEK-293 cells. Our in vitro
such as 3a,5a-THP (allopregnanolone) and 3a,5b-THP (pregna- investigations demonstrated a dose-dependent inhibitory effect
nolone) are potent positive allosteric modulators of the GABAA of mirtazapine on the activity of the microsomal 3a-HSD in the
receptor complex, whereas 3b,5a-THP or DHEA exert an inhibitory oxidative direction (conversion of 3a,5a-THP into 5a-DHP);
modulation of the GABAA receptor (Paul and Purdy, 1992; however, in contrast to SSRIs no effect of mirtazapine on the
Rupprecht and Holsboer, 1999). In preclinical studies at the cytosolic 3a-HSD catalyzing the reductive pathway (conversion of
molecular level, selective serotonin reuptake inhibitors (SSRIs) 5a-DHP into 3a,5a-THP) was observed. Nevertheless, these in
were able to enhance the formation of 3a-reduced neuroactive vitro investigations support the view that both SSRIs and
steroids (e.g. allopregnanolone) via shifting the activity of the mirtazapine may enhance the formation of 3a-reduced neuroac-
cytosolic 3a-HSD type 3 toward the reductive direction; only tive steroids such as allopregnanolone thereby potentiating the
sertraline also inhibited the reverse oxidative reaction. The GABAergic tone in the brain. The effects of psychotropic drugs on
tricyclic antidepressant imipramine had no effect on the formation neurosteroidogenesis in humans, in particular depressed patients,
of allopregnanolone (Griffin and Mellon, 1999). The authors of this are summarized in Table 1.
C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87 81

Table 1
Effects of psychotropic drugs on neurosteroidogenesis in humans (in particular depressed patients) (for details see Schule et al., 2011).

Drugs Possible mechanism of action Effects of neurosteroidogenesis

SSRIs Shifting of the activity of 3a-HSD toward No change in 5a-DHP; increase of 3a-THP (allopregnanolone),
the reductive direction but decrease of of 3a-THDOC; decrease of 3b-THP; in some studies
decrease of DHEA-S
TCAs Unclear (no effect on 3a-HSD) No change in 5a-DHP; increase of 3a-THP (allopregnanolone), no effect
on 3a-THDOC; decrease of 3b-THP; in some studies decrease of DHEA-S
Mirtazapine Inhibition of the oxidative pathway catalyzed Weak but significant increase in 5a-DHP; increase of 3a-THP
by the microsomal 3a-HSD (allopregnanolone), no effect on 3a-THDOC; decrease of 3b-THP;
reduction of both cortisol and DHEA-S (no impact on cortisol/DHEA-S-ratio)
Lithium Unclear Given as an additional treatment reversal of increase in 5a-reduced
neuroactive steroids as compared to mirtazapine monotherapy
Carbama-mazepine Unclear (may be enzyme induction via Given as an additional treatment reversal of increase in 5a-reduced
Cytochrom P450 system) neuroactive steroids as compared to mirtazapine monotherapy
SD – No effect on neurosteroidogenesis
rTMS – No effect on neurosteroidogenesis
ECT – No effect on neurosteroidogenesis
TSPO-ligands (e.g. XBD 173) TSPO facilitates the transport of cholesterol o General stimulation of neurosteroidogenesis
the inner mitochondrial membrane

Abbreviations: SSRIs, selective serotonin reuptake inhibitors; TCAs, tricyclic antidepressants; 3a-HSD, 3a-hydroxysteroid-dehydrogenase; 5a-DHP, 5a-dihydroprogester-
one; 5a-THP, 5a-tetrahydroprogesterone; 3a-THDOC, 3a-tetrahydroprodeoxycorticosterone; DHEA, dehydroandrostendione; SD, sleep deprivation; rTMS, repeated
transcranial magnetic stimulation; ECT, electroconvulsive therapy; TSPO, translocator protein (18 kDa).

During the last 15 years, the downregulation of neurosteroid 1991). However, repeated and chronic stress leads to a significant
biosynthesis has been implicated as a possible contributor to the reduction in serum concentrations of allopregnanolone (Dong et al.,
development of anxiety and depressive disorder (Nothdurfter 2001; Matsumoto et al., 2005; Serra et al., 2000, 2006). 5a-
et al., 2012a; Rupprecht et al., 2010; Schule et al., 2011). It could be reductase, the rate-limiting enzyme in the synthesis of allopreg-
shown in quite a number of clinical trials that reduction in the nanolone has been demonstrated to be down-regulated in an
serum/plasma and cerebrospinal fluid content of neuroactive animal model of chronic stress (Agis-Balboa et al., 2007), suggesting
steroids, in particular the GABAA receptor-agonistic progesterone that chronic stress may alter allopregnanolone synthesis leading to
metabolite allopregnanolone, may be associated with several a dysregulation of the HPA axis (Reddy, 2006). Moreover,
affective disorders, including major depression (Romeo et al., allopregnanolone has been demonstrated to restore hippocampal
1998; Schule et al., 2006b; Uzunova et al., 1998), anxiety disorders neurogenesis in a transgenic mouse model of Alzheimer’ disease
(Rupprecht et al., 2009; Strohle et al., 2002), premenstrual (Singh et al., 2012; Wang et al., 2010; Brinton, 2013). In a recent
dysphoric disorder (Amin et al., 2006; Backstrom et al., 2003; study in rodents, chronic stress was induced using the social
Pearlstein, 2010; Rapkin et al., 1997), postpartum depression isolation model (Evans et al., 2012). In this model, chronic stress
(Bloch et al., 2000; Nappi et al., 2001), posttraumatic stress was associated with a significant reduction in endogenous
disorder (Rasmusson et al., 2006), negative symptoms in schizo- allopregnanolone levels and an enhancement of depressive/
phrenia (Marx et al., 2009, 2011), or impulsive aggression (Nelson anxiety-like behavior in the novelty-suppressed feeding test and
and Pinna, 2011). These findings gave reason to the hypothesis that forced-swim test. Exogenous administration of allopregnanolone
downregulation of allopregnanolone and other GABAergic neuro- either from the onset of isolation-induced chronic stress or
active steroids in the brain could be a risk factor for the following a period of chronic stress was able to impede the
development of the mentioned psychiatric disorders, but may occurrence of depression/anxiety-like behavior, to prevent or
be also involved in the pathophysiology of psychiatric and normalize HPA axis dysfunction and to avoid impairment of
neurological disorders such as epilepsy (Herzog, 1995; Reddy, hippocampal neurogenesis, respectively (Evans et al., 2012).
2011), learning and memory deficit disorders such as Alzheimer’ Interestingly, the behavioral effects of allopregnanolone and its
disease (Luchetti et al., 2011), or alcohol withdrawal (Follesa et al., regulation by antidepressant drugs such as selective serotonin
2006; Kumar et al., 2009) (Table 2). The hypothesis of dysregulated reuptake inhibitors (SSRIs) are brain region specific (Nelson and
neuroactive steroids in emotional disorders opened a variety of Pinna, 2011). In this animal study, male mice underwent four
new targets for treatment. Thus, several researchers worldwide weeks of social isolation, thereby inducing dysfunctional behaviors
were stimulated to develop new molecules enhancing neuroster- including fear or aggression in association with a reduction in
oidogenesis and allopregnanolone biosynthesis, in particular to allopregnanolone biosynthesis in selected corticolimbic brain
treat anxiety disorders and depression (Schule et al., 2011). structures. A decrease of allopregnanolone synthesis was found
New hypotheses concerning putative anxiolytic and antidepres- in the basolateral amygdala, olfactory bulb, hippocampus, and
sant properties of allopregnanolone also involve the role of chronic medial prefrontal cortex but not in the striatum and in the
stress, hypothalamic-pituitary-adrenocortical (HPA) axis dysregu- cerebellum. Moreover, direct infusion of S-norfluoxetine (SSRI) or
lation and neurogenesis. The importance of allopregnanolone for pregnanolone (an analog of allopregnanolone) into the basolateral
the regulation of emotion and its therapeutical use in depression amygdala, but not into the striatum dose-dependently reduced
and anxiety may not only involve GABAergic mechanisms, but aggression in socially isolated mice to a same-sex intruder (Nelson
probably also includes enhancement of neurogenesis (Wang et al., and Pinna, 2011). These findings suggest an involvement of
2005), myelination (Ghoumari et al., 2003), neuroprotection allopregnanolone in the regulation of aggressiveness in socially
(Djebaili et al., 2005; Griffin et al., 2004; Mellon et al., 2008; isolated mice and point to the pivotal role of the basolateral
Sayeed et al., 2006), or regulation of HPA axis function (Reddy, amygdala and the hippocampus in this context.
2006). Acute stress produces an increase in allopregnanolone levels The aim of the review is to provide an overview of the animal
which negatively modulates the stress-induced HPA axis activation, and human studies investigating the 3a-reduced neuroactive
thereby facilitating the recovery of physiological homeostasis steroid allopregnanolone and to discuss its potential in the
following stressful stimuli (Girdler and Klatzkin, 2007; Purdy et al., treatment of depression and anxiety.
82 C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87

Table 2
The role of allopregnanolone and other neuroactive steroids in psychiatric and neurological disorders (for details see Reddy, 2010).

Disease Role of allopregnanolone and other NAS Therapeutic implications


Epilepsy Broad-spectrum anticonvulsant properties of 3a-reduced Positive controlled trials for the synthetic neurosteroid
NAS (e.g. allopregnanolone). Sulfated neurosteroids PS and ganaxolone (analog for allopregnanolone) in the treatment
DHEA-S are proconvulsants (blocking of GABAA receptors of epilepsy
and facilitating of NMDA receptor family
Anxiety Anxiolytic properties of progesterone, allopregnanolone, Replacement of neurosteroids by synthetic analogs of
and THDOC. Anxiogenic effects of the sulphated allopregnanolone. Stimulation of endogenous neurosteroid
neurosteorids PS (biphasic in higher dosages) and DHEA-S. synthesis by SSRIs (in this context also called ‘‘SBSSs’’).
In patients with CCK-4 induced panic attacks, there is a Anxiolytic actions of selective ligands for TSPO such as
pronounced decrease in allopregnanolone levels, and FGIN-1-27 or XBD 173
elevated neurosteroids may counteract the occurrence of
spontaneous panic attacks
Premenstrual snydrome (PMS), In normal women, allopregnanolone varies very similarly to Natural progesterone supplementation in women with PMS
pre-menstrual dysphoric progesterone throughout the menstrual cycle with greater has no clear beneficial effect
disorder (PMDD) levels in the luteal phase than in the follicular phase. During
the luteal phase, PMDD women have significantly greater
allopregnanolone levels, coupled with significantly lower
cortisol levels, during both baseline and mental stress
(enhanced ratio of allopregnanolone/cortisol). Marked
insensitivity to benzodiazepine therapy in patients with
PMS (possibly due to cross-tolerance between
benzodiazepines and neurosteroids)
Depression Dysequilibrium of NAS in depressed patients (decreased Increase of 3a-reduced NAS such as allopregnanolone
levels of 3a-reduced NAS Such as allopregnanolone, during treatment with SSRIs, TCAs, and mirtazapine as a
increased concentrations of 3b-THP). Correction of this putative therapeutic principle. Indications, that adjunct
dysequilibrium by antidepressant drugs (SSRIs, TCAs, and DHEA has beneficial effects in depressed patients
mirtazapine). Pregnancy is associated with a marked rise in
progesterone-derived neurosteroid levels, which decline
rapidly after delivery, suggesting a key role of NAS in the
pathophysiology of depression
Learning and memory Neurosteroids such as allopregnanolone regulate both There is preclinical evidence suggesting that
regeneration and repair systems in the brain. In the brain of allopregnanolone may achieve efficacy as a disease
mice with Alzheimer disease, allopregnanolone increases modifying therapeutic to promote regeneration while
expression of liver-X-receptor, pregnane-X-receptor, and 3- simultaneously decreasing the pathology associated with
hydroxy-3-methyl-glutaryl-CoA-reductase (HMG-CoA-R), Alzheimer’s disease
three proteins that regulate cholesterol homeostasis and
clearance from brain. Moreover, allopregnanolone reduces
amyloid-b and microglial activation, and increases markers
of myelin and white matter generation
Alcohol withdrawal Specific GABAA receptor subtypes are sensitive to ethanol at There are new opportunities to intervene in the effects of
doses attained during social drinking while other subtypes ethanol on different aspects of brain function (e.g.
respond to ethanol at doses attained by severe intoxication. inhibition of ethanol effects by blockade of a4-GABAA
Moreover, Furthermore, ethanol increases GABAergic receptors or by inhibition of neuroactive steroid synthesis).
neurotransmission through indirect effects, including the Likewise, it is also possible to enhance ethanol sensitivity
elevation of endogenous GABAergic neuroactive steroids, by targeting a4-GABAA receptors with selective agonists or
presynaptic release of GABA, and dephosphorylation of promoting neuroactive steroid synthesis. The clinical
GABAA receptors promoting increases in GABA sensitivity importance of these findings has to be tested

Abreviations: NAS, neuroactive steroids; PS, pregnenolone sulfate; DHEA-S, dehydroepiandrosterone sulfate; GABAA receptor, gamma-aminobutyric acid type A receptor;
NMDA receptor, N-methyl-D-aspartate receptor; CCK-4, cholecystokinin-tetrapeptide; SSRI, selective serotonin reuptake inhibitor; SBSS, selective brain steroidogenic
stimulant; TCA, tricyclic antidepressant; TSPO, translocator protein (18 kDa); PMS, premenstrual syndrome; PMDD, premenstrual dysphoric disorder; THP,
tetrahydroprogesterone.

2. Allopregnanolone and depression sant-like effects of fluoxetine but not imipramine in the FST were
mediated by modulation of GABAergic neurotransmission since
2.1. Allopregnanolone in animal models of depression the reduction of the immobility caused by fluoxetine was
potentiated by the GABAA agonist muscimol and blocked by the
First evidence for the assumption that 3a-reduced neuroactive GABAA antagonist bicuculline, whereas no impact of bicuculline
steroids may play a role in the pathophysiology of depression came was found on the antidepressant-like effect of imipramine (Khisti
from an animal study in sham-operated or adrenalectomized/ et al., 2000). In addition, serotonergic agents such as fluoxetine or
castrated male rats (Uzunov et al., 1996). In these rats acute fenfluramine have been shown to enhance the antidepressant-like
treatment with selective serotonin reuptake inhibitors such as action of allopregnanolone in the FST by potentiating the
fluoxetine and to a lesser extent paroxetine increased the brain GABAergic tone as a likely consequence of increased brain content
levels of allopregnanolone (3a,5a-THP) in a dose- and time- of this neurosteroid (Khisti and Chopde, 2000).
dependant manner whereas the tricyclic antidepressant imipra- Besides the Porsolt FST, olfactory bulbectomy is one of the most
mine failed to have an influence on the allopregnanolone content validated animal models of depression. Bilateral removal of the
(Uzunov et al., 1996). Interestingly, in this animal study fluoxetine olfactory bulbs of rats produces a strong region-specific dysregula-
increased the allopregnanolone biosynthesis without changing the tion of allopregnanolone homeostasis in the brain which may play
brain content of its precursor 5a-DHP. Moreover, the administra- a role in the induction of the bulbectomy syndrome by reducing
tion of allopregnanolone reduced the duration of immobility in the the physiological GABAergic tone in particular in the amygdala and
forced swim test (FST) in a dose-dependent manner (Khisti et al., the frontal cortex (Uzunova et al., 2003). Moreover, it has been
2000). The same authors could demonstrate that the antidepres- demonstrated that this decline of the allopregnanolone content in
C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87 83

selected brain areas can be reversed by sub-chronic (3-week) et al., 1998), but not after determination of neuroactive steroid
treatment with different classes of antidepressants (desipramine, levels in the plasma (Schule et al., 2006a,b).
fluoxetine, sertraline, and venlafaxine) supporting the view that In patients suffering from bipolar disorder (BD) or major
normalization of allopregnanolone cerebrocortical levels may depressive disorder (MDD), but in a condition of well-being
contribute to the antidepressant-like profile of these drugs in without relapse or recurrence during the 3 months before study
the olfactory-bulbectomized rat model of depression (Uzunova enrolment, the plasma concentrations of allopregnanolone and
et al., 2004). progesterone were significantly higher than in healthy controls,
Taken together, multiple antidepressant agents (including also being higher in BD patients than in MDD patients (Hardoy
fluoxetine, norfluoxetine, fluvoxamine, and paroxetine) (Marx et al., 2006). The elevated allopregnanolone concentrations did not
et al., 2006; Matsumoto et al., 2007; Uzunov et al., 1996), but also depend on concomitant psychopharmacological treatment or
antipsychotic drugs such as olanzapine (Marx et al., 2006) or mood comorbidity with other psychiatric disorder. In contrast to the
stabilizers such as carbamazepine (Biggio et al., 1998; Jaworska- described former studies (Romeo et al., 1998; Uzunova et al., 1998)
Feil et al., 2000; Serra et al., 2000) have been demonstrated to which were performed with patients during a manifest depressive
elevate allopregnanolone brain levels in animal studies, suggesting episode, the patients of the Hardoy-study were in a state of well-
that enhancement of allopregnanolone might be an important being which may be the reason for the different results. However, it
mechanism for the antidepressant effects of these drugs. has to be mentioned in this context that in another trial, women
with a history of depression, even during full remission, showed
2.2. Allopregnanolone and clinical studies in depressed patients lower concentrations of GABAergic neuroactive steroids such as
allopregnanolone (Girdler et al., 2012).
The finding in animal studies that SSRIs such as fluoxetine or In two clinical trials our research group investigated the
paroxetine increase the content of allopregnanolone in various rat influence of mood stabilizers such as lithium or carbamazepine on
brain areas (Uzunov et al., 1996) gave reason perform human trials neuroactive steroid levels in depressed patients (Schule et al.,
testing the hypothesis that clinical depression would be associated 2009). In both studies, mirtazapine monotherapy was compared
with reduced allopregnanolone levels in patients also and with combination therapy using mirtazapine and additional
administration of SSRIs would normalize the brain allopregnano- lithium or carbamazepine. In the first study, the mirtazapine-
lone levels in depressed patients thereby improving depressive induced rises in 3a-reduced neuroactive steroids (allopregnano-
symptomatology. Uzunova and coworkers performed a small, but lone, pregnanolone) were abolished by additional lithium admin-
landmark study in 15 patients suffering from unipolar depression, istration, as compared to mirtazapine monotherapy. In the second
measuring the cerebrospinal fluid (CSF) allopregnanolone levels study, the mirtazapine-evoked increase in allopregnanolone was
before and after 8–10 weeks of treatment with fluoxetine or reversed after additional administration of carbamazepine also,
fluvoxamine (Uzunova et al., 1998). Before initiation of antide- suggesting that the mood stabilizers lithium and carbamazepine
pressant pharmacotherapy, the allopregnanolone concentrations do not enhance but rather reverse the increase in plasma
were about 60% lower in depressed patients than that determined concentrations of 3a-reduced neuroactive steroids in depressed
in age- and sex-matched non-psychiatric subjects. At the end of the patients pretreated with antidepressants such as mirtazapine
treatment period, a normalization and re-increase of allopregna- (Schule et al., 2009).
nolone concentrations in CSF could be observed which was Investigating depressed patients treated with transcranial
significantly correlated with the improvement of depressive magnetic stimulation (Padberg et al., 2002), sleep deprivation
symptoms as measured by the Hamilton Rating Scale for (Schule et al., 2003), or electroconvulsive therapy (Baghai et al.,
Depression (HRDS). Interestingly, in a recent study in patients 2005), we found no impact of these non-pharmacological
suffering from posttraumatic stress disorder (PTSD), allopregna- treatment strategies on the levels of neuroactive steroids including
nolone levels in the CSF were decreased being the lowest in those allopregnanolone, indicating that changes in neuroactive steroid
patients with PTSD and comorbid depression (Rasmusson et al., concentrations are not a general therapeutic principle of all kind of
2006). Moreover, a downregulation of allopregnanolone concen- antidepressant treatment, but are rather related to specific
trations in the CSF could also been shown in this study which was pharmacological properties of antidepressant drugs.
associated with an increase of PTSD re-experiencing and comorbid In recent human studies, functional brain imaging was
depressive symptoms (Rasmusson et al., 2006). introduced to further investigate the impact of allopregnanolone
Consistent with the finding in the CSF in depressed patients, our on the regulation of emotion (Sripada et al., 2013; van Wingen
research group determined neuroactive steroids in the plasma of et al., 2008, 2007). In humans, progesterone administration
patients suffering from depression, demonstrating that during (increasing downstream allopregnanolone) modulated amygdala
depression, the concentrations of 3a-reduced neuroactive steroids responses to emotional faces (van Wingen et al., 2007) and
such as allopregnanolone were decreased, while the levels of enhanced functional connectivity between amygdala and dorsal
isopregnanolone (3b,5a-THP), a stereoisomer of 3a,5a-THP, medial prefrontal cortex (van Wingen et al., 2008), a regulatory
which may act as an antagonist for GABAergic steroids (Rupprecht region interconnected with limbic structures (Price and Drevets,
and Holsboer, 1999) were increased (Romeo et al., 1998). 2010) and crucial to emotional regulation (Phan et al., 2002). In the
Moreover, in contrast to preclinical data in these human trials trial of Sripada et al. (2013) participants received either 400 mg
not only SSRIs (Romeo et al., 1998; Uzunova et al., 1998), but also pregnenolone (n = 16) or placebo (n = 15). A 3T functional
other classes of antidepressant drugs, e.g. tricyclic antidepressants magnetic resonance imaging was done while performing the
(Romeo et al., 1998), or mirtazapine (Schule et al., 2006b) were able shifted-attention emotion appraisal task to probe emotional
to correct this disequilibrium of neuroactive steroids throughout regulation and processing. Compared with placebo, allopregna-
several weeks in depressed patients by enhancing 3a-reduced nolone elevations following pregnenolone administration were
neuroactive steroids and decreasing 3b-reduced isopregnanolone associated with reduced activity in the amygdala and insula (all
(Romeo et al., 1998; Schule et al., 2006b). However, an association conditions), but increased activity in the dorsal medial prefrontal
between amelioration of depressive symptoms (as measured by cortex and enhanced connectivity between amygdala and dorsal
the HRDS) and increase of 3a-reduced neuroactive steroids such as medial prefrontal cortex (appraisal condition). Moreover, these
allopregnanolone could only be demonstrated when measuring effects on emotion-regulatory neurocircuits were associated with
neuroactive steroids in the CSF of depressed patients (Uzunova reduced self-reported anxiety in the 31 healthy male volunteers
84 C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87

(Sripada et al., 2013). Notably, amygdala and medial prefrontal (Brambilla et al., 2003). Thus, it has been hypothesized that
cortex are rich in GABAA receptors (Pirker et al., 2000) and GABAergic 3a-reduced neuroactive steroids may serve as an
endogenous allopregnanolone (Marx et al., 2006), suggesting that endogenous counter-regulatory mechanisms against the occur-
allopregnanolone could feasibly have a direct influence on the rence of spontaneous panic attacks (Rupprecht, 2003).
activity in these brain regions. No data are available concerning alteration of allopregnanolone
or other neuroactive steroids levels during spontaneous panic
3. Allopregnanolone and anxiety attacks. However, when using infusion with sodium lactate or
cholecystokinin-tetrapeptide (CCK-4) to provoke panic attacks in
3.1.1. Allopregnanolone in animal models of anxiety an experimental challenge design in patients with panic disorder,
panic symptoms were accompanied by marked decreases in the
Consistent with their ability to facilitate GABAergic neuro- 3a-reduced allopregnanolone and pregnanolone concentrations,
transmission, 3a-reduced neuroactive steroids exhibit potent whereas 3b-reduced isopregnanolone was increased (Ströhle
anxiolytic-like effects in a variety of animal models (Gasior et al., 2003). In contrast, no such changes in neuroactive steroid
et al., 1999). Allopregnanolone and pregnanolone produce compositions were seen in healthy controls following experimen-
anxiolytic-like effects in different animal models of anxiety such tal panic induction with CCK-4 at dosages of 25 mg i.v. (Ströhle
as light/dark transition (Wieland et al., 1995), elevated plus-maze et al., 2003) or 50 mg i.v. (Zwanzger et al., 2004). Therefore,
(Bitran et al., 1991; Rodgers and Johnson, 1998), defensive burying alterations of allopregnanolone or other neuroactive steroid levels
(Picazo and Fernandez-Guasti, 1995), mirrored chamber (Reddy do not merely reflect a level of state anxiety but appear to be
and Kulkarni, 1997), and ultrasonic vocalization (Vivian et al., related to the pathophysiology of panic attacks in panic disorder
1997; Zimmerberg et al., 1994). Thereby, the amygdala was (Ströhle et al., 2003; Zwanzger et al., 2004). Studies investigating
identified as a key structure for mediating the anxiolytic effects of the impact of paroxetine treatment on allopregnanolone in panic
allopregnanolone, as direct infusion into the amygdala was disorder patients revealed conflicting results: In one study,
followed by a significant increase in the number of entries and paroxetine treatment did not affect allopregnanolone levels or
the time spent in the open arms of the elevated plus maze (Akwa the concentrations of other neuroactive steroids, which were
et al., 1999; Engin and Treit, 2007) or produced antidepressant highly stable over 24 weeks (Strohle et al., 2002). On the contrary,
effects on the learned helplessness in rats (Shirayama et al., 2011). in another trial the allopregnanolone levels were indistinguishable
Interestingly, allopregnanolone attenuates anxiety-related behav- between panic disorder patients and healthy controls before
ior, but does not affect spontaneous locomotor activity in contrast pharmacotherapy; however, 2-month paroxetine treatment in
to benzodiazepines (Reddy and Kulkarni, 1997; Rodgers and patients significantly increased the plasma concentrations of
Johnson, 1998). allopregnanolone which were higher than those measured in
In the meanwhile, allopregnanolone has been shown in several controls at the end of the treatment period (Brambilla et al., 2005).
animal studies to reduce stress and anxiety-like behavior in
rodents (Belelli et al., 2005; Darbra and Pallares, 2010; Frye and 4. Conclusions and future directions
Rhodes, 2006; Harrison and Simmonds, 1984; Lambert et al., 2003;
Majewska et al., 1986; Martin-Garcia and Pallares, 2005). The neuroactive steroid allopregnanolone possesses GABAergic
Moreover, blockade of metabolism from progesterone to allo- properties and may have importance for the regulation of emotion
pregnanolone impairs social behavior and induces anxiety-related playing an important role in the pathophysiology of affective
behavior in rats (Frye and Paris, 2011; Koonce et al., 2012) which disorders such as depression or anxiety. However, the mechanisms
can be reversed by allopregnanolone infusion (Frye et al., 2011). of action of allopregnanolone are not only related to the GABAA
Decreased corticolimbic allopregnanolone expression during receptor but also include effects on neurogenesis and the
social isolation may cause increased emotional hyperreactivity, regulation of the HPA system. Studies using animal models and
enhanced contextual fear, and impaired contextual fear extinction also trials in patients strongly suggest that allopregnanolone and
in mice and may serve as a model relevant for posttraumatic stress other neuroactive steroids are of therapeutic use in the treatment
disorder (Pibiri et al., 2008). Remarkably, circulating and hippo- of affective disorders including depression or anxiety spectrum
campal concentrations of allopregnanolone but not of corticoste- diseases. Antidepressant agents, in particular selective serotonin
rone, progesterone, or estrogen are significantly correlated with reuptake inhibitors (SSRIs) such as fluoxetine, paroxetine, sertra-
low-anxiety and exploratory behaviors in rats (Koonce et al., 2012). line have been shown to reverse the decrease of brain allopregna-
These results led to the assumption that allopregnanolone may be nolone in depression or other affective disorders which may at
one of the most promising candidates within neuroactive steroids least in part explain their antidepressant action. Moreover, SSRIs
as a new anxiolytic treatment option. have been demonstrated to increase allopregnanolone levels and
to correct behavioral deficits expressed by socially isolated mice
3.2. Allopregnanolone and clinical studies in patients suffering from even at dosages that are 50fold lower than those required to cause
anxiety disorders an effective 5-HT reuptake inhibition (Pinna et al., 2003, 2006).
Therefore, it has been proposed to change the term ‘‘SSRI’’ for these
In humans, no alterations of allopregnanolone concentrations drugs into the more appropriate term ‘‘SBSS’’ (selective brain
could be detected in patients suffering from generalized anxiety steroidogenic stimulant) (Pinna et al., 2006). However, it remains a
disorder (Semeniuk et al., 2001), generalized social phobia matter of debate if this proposition will become accepted in
(Heydari and Le Melledo, 2002), or mixed anxiety-depressive psychiatry.
disorder (Bicikova et al., 2000). However, opposite to the findings Besides mediation of therapeutic effects of conventional
in major depression, in patients with panic disorder 3a-reduced antidepressant drugs by endogenous metabolites of progesterone,
neuroactive steroid levels such as allopregnanolone were in- neuroactive steroids themselves have been used in the treatment of
creased while the concentrations of 3b-reduced isopregnanolone epilepsy (Herzog, 1995; Reddy, 2011). However, certain pharmaco-
were decreased (Strohle et al., 2002). In line with this finding, in kinetic obstacles limit the therapeutic use of natural neurosteroids.
women suffering from panic disorder elevated plasma concentra- First, natural neurosteroids such as 3a,5a-THP (allopregnanolone)
tions of allopregnanolone were found to be elevated both during have a low bioavailability due to rapid inactivation by glucuronida-
the follicular and premenstrual phase of the menstrual cycle tion and sulphate conjugation. Second, the 3a-hydroxyl group of
C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87 85

allopregnanolone may undergo oxidation to the ketone. Neverthe- neurosteroidogenic compounds still are promising avenues for
less, synthetic neurosteroids may represent a rational alternative the treatment of affective disorders such as depression and
approach to therapy (Reddy, 2010; Reddy and Kulkarni, 2000). Three anxiety.
synthetic neurosteroids have been investigated with respect to their
therapeutic potential (ganaxolone, alphaxalone, and minaxolone); References
however, until now synthetic neuroactive steroids could not be
Agis-Balboa, R.C., Pinna, G., Pibiri, F., Kadriu, B., Costa, E., Guidotti, A., 2007. Down-
established in the treatment of anxiety disorders or depression regulation of neurosteroid biosynthesis in corticolimbic circuits mediates social
(Nothdurfter et al., 2012b). isolation-induced behavior in mice. Proc. Natl. Acad. Sci. USA 104, 18736–
Regarding endogenous neurosteroidogenesis, recently the 18741.
Akwa, Y., Purdy, R.H., Koob, G.F., Britton, K.T., 1999. The amygdala mediates the
translocator protein (18 kDa) (TSPO) has been identified as a anxiolytic-like effect of the neurosteroid allopregnanolone in rat. Behav. Brain
potential novel target. TSPO is the key element of the mitochon- Res. 106, 119–125.
drial import machinery facilitating the transport of cholesterol Amin, Z., Mason, G.F., Cavus, I., Krystal, J.H., Rothman, D.L., Epperson, C.N., 2006. The
interaction of neuroactive steroids and GABA in the development of neuropsy-
from the outer to the inner mitochondrial membrane and being
chiatric disorders in women. Pharmacol. Biochem. Behav. 84, 635–643.
responsible for supplying the substrate cholesterol to the first Backstrom, T., Andersson, A., Andree, L., Birzniece, V., Bixo, M., Bjorn, I., Haage, D.,
steroidogenic enzyme (P450scc), which transforms cholesterol Isaksson, M., Johansson, I.M., Lindblad, C., Lundgren, P., Nyberg, S., Odmark, I.S.,
into pregnenolone, the precursor of all neurosteroids (Papado- Stromberg, J., Sundstrom-Poromaa, I., Turkmen, S., Wahlstrom, G., Wang, M.,
Wihlback, A.C., Zhu, D., Zingmark, E., 2003. Pathogenesis in menstrual cycle-
poulos et al., 1997). TSPO ligands (e.g. XBD 173 or etifoxine) linked CNS disorders. Ann. N.Y. Acad. Sci. 1007, 42–53.
increase neurosteroidogenesis and are a target of novel anxiolytic Baghai, T.C., di Michele, F., Schule, C., Eser, D., Zwanzger, P., Pasini, A., Romeo, E.,
drugs producing anxiolytic effects without causing the side effects Rupprecht, R., 2005. Plasma concentrations of neuroactive steroids before and
after electroconvulsive therapy in major depression. Neuropsychopharmacol-
normally associated with conventional benzodiazepines such as ogy 30, 1181–1186.
sedation or tolerance (Nothdurfter et al., 2012a; Rupprecht et al., Belelli, D., Peden, D.R., Rosahl, T.W., Wafford, K.A., Lambert, J.J., 2005. Extrasynaptic
2009, 2010) (Fig. 2). As such, neuroactive steroids and/or GABAA receptors of thalamocortical neurons: a molecular target for hypnotics. J.
Neurosci. 25, 11513–11520.
Bicikova, M., Tallova, J., Hill, M., Krausova, Z., Hampl, R., 2000. Serum concentrations
cholesterol of some neuroactive steroids in women suffering from mixed anxiety-depres-
sive disorder. Neurochem. Res. 25, 1623–1627.
Biggio, G., Littera, M., Giliberto, G., Purdy, R.H., Serra, M., 1998. Carbamazepine
elevates plasma and brain content of neuroactive steroids. Soc. Neurosci. 24,
TSPO ligand TSPO outer
998.
Bitran, D., Hilvers, R.J., Kellogg, C.K., 1991. Anxiolytic effects of 3 alpha-hydroxy-5
mitochondrial alpha[beta]-pregnan-20-one: endogenous metabolites of progesterone that are
membrane active at the GABAA receptor. Brain Res. 561, 157–161.
Bloch, M., Schmidt, P.J., Danaceau, M., Murphy, J., Nieman, L., Rubinow, D.R., 2000.
Effects of gonadal steroids in women with a history of postpartum depression.
inner Am. J. Psychiatry 157, 924–930.
mitochondrial Brambilla, F., Biggio, G., Pisu, M.G., Bellodi, L., Perna, G., Bogdanovich-Djukic, V.,
membrane Purdy, R.H., Serra, M., 2003. Neurosteroid secretion in panic disorder. Psychiatry
Res. 118, 107–116.
Brambilla, F., Mellado, C., Alciati, A., Pisu, M.G., Purdy, R.H., Zanone, S., Perini, G.,
cholesterol Serra, M., Biggio, G., 2005. Plasma concentrations of anxiolytic neuroactive
steroids in men with panic disorder. Psychiatry Res. 135, 185–190.
Brinton, R.D., 2013. Neurosteroids as regenerative agents in the brain: therapeutic

NEUROSTEROIDOGENESIS implications. Nat. Rev. Endocrinol. 9, 241–250.


Darbra, S., Pallares, M., 2010. Alterations in neonatal neurosteroids affect explora-
tion during adolescence and prepulse inhibition in adulthood. Psychoneuroen-
docrinology 35, 525–535.
allopregnanolone Djebaili, M., Guo, Q., Pettus, E.H., Hoffman, S.W., Stein, D.G., 2005. The neurosteroids
progesterone and allopregnanolone reduce cell death, gliosis, and functional
deficits after traumatic brain injury in rats. J. Neurotrauma 22, 106–118.
Dong, E., Matsumoto, K., Uzunova, V., Sugaya, I., Takahata, H., Nomura, H., Wata-
nabe, H., Costa, E., Guidotti, A., 2001. Brain 5alpha-dihydroprogesterone and
Cl- allopregnanolone synthesis in a mouse model of protracted social isolation.
Proc. Natl. Acad. Sci. USA 98, 2849–2854.
Engin, E., Treit, D., 2007. The anxiolytic-like effects of allopregnanolone vary as a
allopregnanolone function of intracerebral microinfusion site: the amygdala, medial prefrontal
cortex, or hippocampus. Behav. Pharmacol. 18, 461–470.
Evans, J., Sun, Y., McGregor, A., Connor, B., 2012. Allopregnanolone regulates
neurogenesis and depressive/anxiety-like behaviour in a social isolation rodent
GABAA model of chronic stress. Neuropharmacology 63, 1315–1326.
Follesa, P., Biggio, F., Talani, G., Murru, L., Serra, M., Sanna, E., Biggio, G., 2006.
receptor Neurosteroids, GABAA receptors, and ethanol dependence. Psychopharmacol-
ogy (Berlin) 186, 267–280.
Frye, C.A., Paris, J.J., 2011. Progesterone turnover to its 5alpha-reduced metabolites
in the ventral tegmental area of the midbrain is essential for initiating social and
affective behavior and progesterone metabolism in female rats. J. Endocrinol.
Invest. 34, e188–e199.
Frye, C.A., Paris, J.J., Walf, A.A., Rusconi, J.C., 2011. Effects and mechanisms of
ANXIOLYSIS 3alpha,5alpha,-THP on emotion, motivation, and reward functions involving
pregnane xenobiotic receptor. Front. Neurosci. 5, 136.
Fig. 2. The role of TSPO in anxiolysis (Nothdurfter et al., 2012b). TSPO consists of a Frye, C.A., Rhodes, M.E., 2006. Infusions of 5alpha-pregnan-3alpha-ol-20-one
169-amino acid sequence arranged as a five transmembrane helix structure and is (3alpha,5alpha-THP) to the ventral tegmental area, but not the substantia nigra,
located in the outer mitochondrial membrane. TSPO promotes the transport of enhance exploratory, anti-anxiety, social and sexual behaviours and concomi-
tantly increase 3alpha,5alpha-THP concentrations in the hippocampus, dien-
cholesterol to the inner mitochondrial membrane, the rate-limiting step in
cephalon and cortex of ovariectomised oestrogen-primed rats. J.
neurosteroidogenesis. Within the mitochondrial matrix, cholesterol is converted to
Neuroendocrinol. 18, 960–975.
pregnenolone. After diffusion into the cytoplasm, pregnenolone is further processed
Gasior, M., Carter, R.B., Witkin, J.M., 1999. Neuroactive steroids: potential thera-
into other neurosteroids such as allopregnanolone, which is a positive allosteric peutic use in neurological and psychiatric disorders. Trends Pharmacol. Sci. 20,
GABAA receptor modulator, thereby exerting anxiolytic efficacy. TSPO ligands, such 107–112.
as XBD 173 or etifoxine, enhance the transport of cholesterol into the Ghoumari, A.M., Ibanez, C., El-Etr, M., Leclerc, P., Eychenne, B., O’Malley, B.W.,
mitochondrium via TSPO. Baulieu, E.E., Schumacher, M., 2003. Progesterone and its metabolites increase
86 C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87

myelin basic protein expression in organotypic slice cultures of rat cerebellum. Padberg, F., di Michele, F., Zwanzger, P., Romeo, E., Bernardi, G., Schule, C., Baghai,
J. Neurochem. 86, 848–859. T.C., Ella, R., Pasini, A., Rupprecht, R., 2002. Plasma concentrations of neuroac-
Girdler, S.S., Klatzkin, R., 2007. Neurosteroids in the context of stress: implications tive steroids before and after repetitive transcranial magnetic stimulation
for depressive disorders. Pharmacol. Ther. 116, 125–139. (rTMS) in major depression. Neuropsychopharmacology 27, 874–878.
Girdler, S.S., Lindgren, M., Porcu, P., Rubinow, D.R., Johnson, J.L., Morrow, A.L., 2012. Papadopoulos, V., Amri, H., Boujrad, N., Cascio, C., Culty, M., Garnier, M., Hardwick,
A history of depression in women is associated with an altered GABAergic M., Li, H., Vidic, B., Brown, A.S., Reversa, J.L., Bernassau, J.M., Drieu, K., 1997.
neuroactive steroid profile. Psychoneuroendocrinology 37, 543–553. Peripheral benzodiazepine receptor in cholesterol transport and steroidogene-
Griffin, L.D., Gong, W., Verot, L., Mellon, S.H., 2004. Niemann-Pick type C disease sis. Steroids 62, 21–28.
involves disrupted neurosteroidogenesis and responds to allopregnanolone. Papadopoulos, V., Baraldi, M., Guilarte, T.R., Knudsen, T.B., Lacapere, J.J., Lindemann,
Nat. Med. 10, 704–711. P., Norenberg, M.D., Nutt, D., Weizman, A., Zhang, M.R., Gavish, M., 2006.
Griffin, L.D., Mellon, S.H., 1999. Selective serotonin reuptake inhibitors directly alter Translocator protein (18 kDa): new nomenclature for the peripheral-type ben-
activity of neurosteroidogenic enzymes. Proc. Natl Acad. Sci. USA 96, 13512– zodiazepine receptor based on its structure and molecular function. Trends
13517. Pharmacol. Sci. 27, 402–409.
Hardoy, M.C., Serra, M., Carta, M.G., Contu, P., Pisu, M.G., Biggio, G., 2006. Increased Paul, S.M., Purdy, R.H., 1992. Neuroactive steroids. FASEB J. 6, 2311–2322.
neuroactive steroid concentrations in women with bipolar disorder or major Pearlstein, T., 2010. Premenstrual dysphoric disorder: out of the appendix. Arch.
depressive disorder. J. Clin. Psychopharmacol. 26, 379–384. Womens Ment. Health 13, 21–23.
Harrison, N.L., Simmonds, M.A., 1984. Modulation of the GABA receptor complex by Phan, K.L., Wager, T., Taylor, S.F., Liberzon, I., 2002. Functional neuroanatomy of
a steroid anaesthetic. Brain Res. 323, 287–292. emotion: a meta-analysis of emotion activation studies in PET and fMRI.
Herzog, A.G., 1995. Progesterone therapy in women with complex partial and Neuroimage 16, 331–348.
secondary generalized seizures. Neurology 45, 1660–1662. Pibiri, F., Nelson, M., Guidotti, A., Costa, E., Pinna, G., 2008. Decreased corticolimbic
Heydari, B., Le Melledo, J.M., 2002. Low pregnenolone sulphate plasma concentra- allopregnanolone expression during social isolation enhances contextual fear: a
tions in patients with generalized social phobia. Psychol. Med. 32, 929–933. model relevant for posttraumatic stress disorder. Proc. Natl. Acad. Sci. USA 105,
Jaworska-Feil, L., Budziszewska, B., Leskiewicz, M., Lason, W., 2000. Effects of some 5567–5572.
centrally active drugs on the allopregnanolone synthesis in rat brain. Pol. J. Picazo, O., Fernandez-Guasti, A., 1995. Anti-anxiety effects of progesterone and
Pharmacol. 52, 359–365. some of its reduced metabolites: an evaluation using the burying behavior test.
Khisti, R.T., Chopde, C.T., 2000. Serotonergic agents modulate antidepressant-like Brain Res. 680, 135–141.
effect of the neurosteroid 3alpha-hydroxy-5alpha-pregnan-20-one in mice. Pinna, G., Costa, E., Guidotti, A., 2006. Fluoxetine and norfluoxetine stereospecifi-
Brain Res. 865, 291–300. cally and selectively increase brain neurosteroid content at doses that are
Khisti, R.T., Chopde, C.T., Jain, S.P., 2000. Antidepressant-like effect of the neuro- inactive on 5-HT reuptake. Psychopharmacology (Berlin) 186, 362–372.
steroid 3alpha-hydroxy-5alpha-pregnan-20-one in mice forced swim test. Pinna, G., Dong, E., Matsumoto, K., Costa, E., Guidotti, A., 2003. In socially isolated
Pharmacol. Biochem. Behav. 67, 137–143. mice, the reversal of brain allopregnanolone down-regulation mediates the
Koonce, C.J., Walf, A.A., Frye, C.A., 2012. Type 1 5alpha-reductase may be required anti-aggressive action of fluoxetine. Proc. Natl. Acad. Sci. USA 100, 2035–2040.
for estrous cycle changes in affective behaviors of female mice. Behav. Brain Res. Pirker, S., Schwarzer, C., Wieselthaler, A., Sieghart, W., Sperk, G., 2000. GABA(A)
226, 376–380. receptors: immunocytochemical distribution of 13 subunits in the adult rat
Kumar, S., Porcu, P., Werner, D.F., Matthews, D.B., Diaz-Granados, J.L., Helfand, R.S., brain. Neuroscience 101, 815–850.
Morrow, A.L., 2009. The role of GABA(A) receptors in the acute and chronic Price, J.L., Drevets, W.C., 2010. Neurocircuitry of mood disorders. Neuropsycho-
effects of ethanol: a decade of progress. Psychopharmacology (Berlin) 205, 529– pharmacology 35, 192–216.
564. Purdy, R.H., Morrow, A.L., Moore Jr., P.H., Paul, S.M., 1991. Stress-induced elevations
Lambert, J.J., Belelli, D., Peden, D.R., Vardy, A.W., Peters, J.A., 2003. Neurosteroid of gamma-aminobutyric acid type A receptor-active steroids in the rat brain.
modulation of GABAA receptors. Prog. Neurobiol. 71, 67–80. Proc. Natl. Acad. Sci. USA 88, 4553–4557.
Luchetti, S., Huitinga, I., Swaab, D.F., 2011. Neurosteroid and GABA-A receptor Rapkin, A.J., Morgan, M., Goldman, L., Brann, D.W., Simone, D., Mahesh, V.B., 1997.
alterations in Alzheimer’s disease, Parkinson’s disease and multiple sclerosis. Progesterone metabolite allopregnanolone in women with premenstrual syn-
Neuroscience 191, 6–21. drome. Obstet. Gynecol. 90, 709–714.
Majewska, M.D., Harrison, N.L., Schwartz, R.D., Barker, J.L., Paul, S.M., 1986. Steroid Rasmusson, A.M., Pinna, G., Paliwal, P., Weisman, D., Gottschalk, C., Charney, D.,
hormone metabolites are barbiturate-like modulators of the GABA receptor. Krystal, J., Guidotti, A., 2006. Decreased cerebrospinal fluid allopregnanolone
Science 232, 1004–1007. levels in women with posttraumatic stress disorder. Biol. Psychiatry 60, 704–
Martin-Garcia, E., Pallares, M., 2005. Intrahippocampal nicotine and neurosteroids 713.
effects on the anxiety-like behaviour in voluntary and chronic alcohol-drinking Reddy, D.S., 2006. Physiological role of adrenal deoxycorticosterone-derived neu-
rats. Behav. Brain Res. 164, 117–127. roactive steroids in stress-sensitive conditions. Neuroscience 138, 911–920.
Marx, C.E., Bradford, D.W., Hamer, R.M., Naylor, J.C., Allen, T.B., Lieberman, J.A., Reddy, D.S., 2010. Neurosteroids: endogenous role in the human brain and thera-
Strauss, J.L., Kilts, J.D., 2011. Pregnenolone as a novel therapeutic candidate in peutic potentials. Prog. Brain Res. 186, 113–137.
schizophrenia: emerging preclinical and clinical evidence. Neuroscience 191, Reddy, D.S., 2011. Role of anticonvulsant and antiepileptogenic neurosteroids in the
78–90. pathophysiology and treatment of epilepsy. Front. Endocrinol. 2 , http://
Marx, C.E., Keefe, R.S., Buchanan, R.W., Hamer, R.M., Kilts, J.D., Bradford, D.W., dx.doi.org/10.3389/fendo.2011.00038, article 38.
Strauss, J.L., Naylor, J.C., Payne, V.M., Lieberman, J.A., Savitz, A.J., Leimone, L.A., Reddy, D.S., Kulkarni, S.K., 1997. Differential anxiolytic effects of neurosteroids in
Dunn, L., Porcu, P., Morrow, A.L., Shampine, L.J., 2009. Proof-of-concept trial the mirrored chamber behavior test in mice. Brain Res. 752, 61–71.
with the neurosteroid pregnenolone targeting cognitive and negative symp- Reddy, D.S., Kulkarni, S.K., 2000. Development of neurosteroid-based novel psy-
toms in schizophrenia. Neuropsychopharmacology 34, 1885–1903. chotropic drugs. Prog. Med. Chem. 37, 135–175.
Marx, C.E., Shampine, L.J., Khisti, R.T., Trost, W.T., Bradford, D.W., Grobin, A.C., Rodgers, R.J., Johnson, N.J., 1998. Behaviorally selective effects of neuroactive
Massing, M.W., Madison, R.D., Butterfield, M.I., Lieberman, J.A., Morrow, A.L., steroids on plus-maze anxiety in mice. Pharmacol. Biochem. Behav. 59, 221–
2006. Olanzapine and fluoxetine administration and coadministration increase 232.
rat hippocampal pregnenolone, allopregnanolone and peripheral deoxycorti- Romeo, E., Ströhle, A., Spalletta, G., di Michele, F., Hermann, B., Holsboer, F., Pasini,
costerone: implications for therapeutic actions. Pharmacol. Biochem. Behav. 84, A., Rupprecht, R., 1998. Effects of antidepressant treatment on neuroactive
609–617. steroids in major depression. Am. J. Psychiatry 155, 910–913.
Matsumoto, K., Pinna, G., Puia, G., Guidotti, A., Costa, E., 2005. Social isolation stress- Rupprecht, R., Reul, J.M., Trapp, T., van Steensel, B., Wetzel, C., Damm, K., Ziegl-
induced aggression in mice: a model to study the pharmacology of neuroster- gänsberger, W., Holsboer, F., 1993. Progesterone receptor-mediated effects of
oidogenesis. Stress 8, 85–93. neuroactive steroids. Neuron 11, 523–530.
Matsumoto, K., Puia, G., Dong, E., Pinna, G., 2007. GABA(A) receptor neurotransmis- Rupprecht, R., Holsboer, F., 1999. Neuroactive steroids: mechanisms of action and
sion dysfunction in a mouse model of social isolation-induced stress: possible neuropsychopharmacological perspectives. Trends Neurosci. 22, 410–416.
insights into a non-serotonergic mechanism of action of SSRIs in mood and Rupprecht, R., 2003. Neuroactive steroids: mechanisms of action and neuropsycho-
anxiety disorders. Stress 10, 3–12. pharmacological properties. Psychoneuroendocrinology 28, 139–168.
Mellon, S.H., Gong, W., Schonemann, M.D., 2008. Endogenous and synthetic neuro- Rupprecht, R., Rammes, G., Eser, D., Baghai, T.C., Schule, C., Nothdurfter, C., Troxler,
steroids in treatment of Niemann-Pick Type C disease. Brain Res. Rev. 57, 410– T., Gentsch, C., Kalkman, H.O., Chaperon, F., Uzunov, V., McAllister, K.H., Ber-
420. taina-Anglade, V., La Rochelle, C.D., Tuerck, D., Floesser, A., Kiese, B., Schuma-
Nappi, R.E., Petraglia, F., Luisi, S., Polatti, F., Farina, C., Genazzani, A.R., 2001. Serum cher, M., Landgraf, R., Holsboer, F., Kucher, K., 2009. Translocator protein (18 kD)
allopregnanolone in women with postpartum blues. Obstet. Gynecol. 97, 77–80. as target for anxiolytics without benzodiazepine-like side effects. Science 325,
Nelson, M., Pinna, G., 2011. S-norfluoxetine microinfused into the basolateral 490–493.
amygdala increases allopregnanolone levels and reduces aggression in socially Rupprecht, R., Papadopoulos, V., Rammes, G., Baghai, T.C., Fan, J., Akula, N., Groyer,
isolated mice. Neuropharmacology 60, 1154–1159. G., Adams, D., Schumacher, M., 2010. Translocator protein (18 kDa) (TSPO) as a
Nothdurfter, C., Rammes, G., Baghai, T.C., Schule, C., Schumacher, M., Papadopoulos, therapeutic target for neurological and psychiatric disorders. Nat. Rev. Drug
V., Rupprecht, R., 2012a. Translocator protein (18 kDa) as a target for novel Discov. 9, 971–988.
anxiolytics with a favourable side-effect profile. J. Neuroendocrinol. 24, 82–92. Sayeed, I., Guo, Q., Hoffman, S.W., Stein, D.G., 2006. Allopregnanolone, a progester-
Nothdurfter, C., Rupprecht, R., Rammes, G., 2012b. Recent developments in poten- one metabolite, is more effective than progesterone in reducing cortical infarct
tial anxiolytic agents targeting GABAA/BzR complex or the translocator protein volume after transient middle cerebral artery occlusion. Ann. Emerg. Med. 47,
(18 kDa) (TSPO). Curr. Top. Med. Chem. 12, 360–370. 381–389.
C. Schüle et al. / Progress in Neurobiology 113 (2014) 79–87 87

Schule, C., Baghai, T.C., Eser, D., Nothdurfter, C., Rupprecht, R., 2009. Lithium but not sition in patients with panic disorder before and during paroxetine treatment.
carbamazepine augments antidepressant efficacy of mirtazapine in unipolar Am. J. Psychiatry 159, 145–147.
depression: an open-label study. World J. Biol. Psychiatry 10, 390–399. Trauger, J.W., Jiang, A., Stearns, B.A., LoGrasso, P.V., 2002. Kinetics of allopregna-
Schule, C., Baghai, T.C., Eser, D., Zwanzger, P., Jordan, M., Buechs, R., Rupprecht, R., nolone formation catalyzed by human 3 alpha-hydroxysteroid dehydrogenase
2006a. Time course of hypothalamic-pituitary-adrenocortical axis activity type III (AKR1C2). Biochemistry 41, 13451–13459.
during treatment with reboxetine and mirtazapine in depressed patients. Uzunov, D.P., Cooper, T.B., Costa, E., Guidotti, A., 1996. Fluoxetine-elicited changes
Psychopharmacology (Berlin) 186, 601–611. in brain neurosteroid content measured by negative ion mass fragmentography.
Schule, C., di Michele, F., Baghai, T., Romeo, E., Bernardi, G., Zwanzger, P., Padberg, F., Proc. Natl. Acad. Sci. USA 93, 12599–12604.
Pasini, A., Rupprecht, R., 2003. Influence of sleep deprivation on neuroactive Uzunova, V., Ceci, M., Kohler, C., Uzunov, D.P., Wrynn, A.S., 2003. Region-specific
steroids in major depression. Neuropsychopharmacology 28, 577–581. dysregulation of allopregnanolone brain content in the olfactory bulbectomized
Schule, C., Eser, D., Baghai, T.C., Nothdurfter, C., Kessler, J.S., Rupprecht, R., 2011. rat model of depression. Brain Res. 976, 1–8.
Neuroactive steroids in affective disorders: target for novel antidepressant or Uzunova, V., Sheline, Y., Davis, J.M., Rasmusson, A., Uzunov, D.P., Costa, E., Guidotti,
anxiolytic drugs? Neuroscience 191, 55–77. A., 1998. Increase in the cerebrospinal fluid content of neurosteroids in patients
Schule, C., Romeo, E., Uzunov, D.P., Eser, D., di Michele, F., Baghai, T.C., Pasini, A., with unipolar major depression who are receiving fluoxetine or fluvoxamine.
Schwarz, M., Kempter, H., Rupprecht, R., 2006b. Influence of mirtazapine on Proc. Natl. Acad. Sci. USA 95, 3239–3244.
plasma concentrations of neuroactive steroids in major depression and on Uzunova, V., Wrynn, A.S., Kinnunen, A., Ceci, M., Kohler, C., Uzunov, D.P., 2004.
3alpha-hydroxysteroid dehydrogenase activity. Mol. Psychiatry 11, 261–272. Chronic antidepressants reverse cerebrocortical allopregnanolone decline in
Semeniuk, T., Jhangri, G.S., Le Melledo, J.M., 2001. Neuroactive steroid levels in the olfactory-bulbectomized rat. Eur. J. Pharmacol. 486, 31–34.
patients with generalized anxiety disorder. J. Neuropsychiatry Clin. Neurosci. van Wingen, G.A., van Broekhoven, F., Verkes, R.J., Petersson, K.M., Backstrom, T.,
13, 396–398. Buitelaar, J.K., Fernandez, G., 2008. Progesterone selectively increases amygdala
Serra, M., Mostallino, M.C., Talani, G., Pisu, M.G., Carta, M., Mura, M.L., Floris, I., reactivity in women. Mol. Psychiatry 13, 325–333.
Maciocco, E., Sanna, E., Biggio, G., 2006. Social isolation-induced increase in van Wingen, G., van Broekhoven, F., Verkes, R.J., Petersson, K.M., Backstrom, T.,
alpha and delta subunit gene expression is associated with a greater efficacy of Buitelaar, J., Fernandez, G., 2007. How progesterone impairs memory for
ethanol on steroidogenesis and GABA receptor function. J. Neurochem. 98, 122– biologically salient stimuli in healthy young women. J. Neurosci. 27, 11416–
133. 11423.
Serra, M., Pisu, M.G., Littera, M., Papi, G., Sanna, E., Tuveri, F., Usala, L., Purdy, R.H., Vivian, J.A., Barros, H.M., Manitiu, A., Miczek, K.A., 1997. Ultrasonic vocalizations
Biggio, G., 2000. Social isolation-induced decreases in both the abundance of in rat pups: modulation at the gamma-aminobutyric acidA receptor complex
neuroactive steroids and GABA(A) receptor function in rat brain. J. Neurochem. and the neurosteroid recognition site. J. Pharmacol. Exp. Ther. 282,
75, 732–740. 318–325.
Shirayama, Y., Muneoka, K., Fukumoto, M., Tadokoro, S., Fukami, G., Hashimoto, K., Wang, J.M., Johnston, P.B., Ball, B.G., Brinton, R.D., 2005. The neurosteroid allopreg-
Iyo, M., 2011. Infusions of allopregnanolone into the hippocampus and amyg- nanolone promotes proliferation of rodent and human neural progenitor cells
dala, but not into the nucleus accumbens and medial prefrontal cortex, produce and regulates cell-cycle gene and protein expression. J. Neurosci. 25, 4706–
antidepressant effects on the learned helplessness rats. Hippocampus 21, 1105– 4718.
1113. Wang, J.M., Singh, C., Liu, L., Irwin, R.W., Chen, S., Chung, E.J., Thompson, R.F.,
Singh, C., Liu, L., Wang, J.M., Irwin, R.W., Yao, J., Chen, S., Henry, S., Thompson, R.F., Brinton, R.D., 2010. Allopregnanolone reverses neurogenic and cognitive def-
Brinton, R.D., 2012. Allopregnanolone restores hippocampal-dependent learn- icits in mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 107,
ing and memory and neural progenitor survival in aging 3xTgAD and nonTg 6498–6503.
mice. Neurobiol. Aging 33, 1493–1506. Wieland, S., Belluzzi, J.D., Stein, L., Lan, N.C., 1995. Comparative behavioral charac-
Sripada, R.K., Marx, C.E., King, A.P., Rampton, J.C., Ho, S.S., Liberzon, I., 2013. terization of the neuroactive steroids 3 alpha-OH,5 alpha-pregnan-20-one and
Allopregnanolone elevations following pregnenolone administration are asso- 3 alpha-OH,5 beta-pregnan-20-one in rodents. Psychopharmacology (Berlin)
ciated with enhanced activation of emotion regulation neurocircuits. Biol. 118, 65–71.
Psychiatry 73, 1045–1053. Zimmerberg, B., Brunelli, S.A., Hofer, M.A., 1994. Reduction of rat pup ultrasonic
Ströhle, A., Romeo, E., di Michele, F., Pasini, A., Hermann, B., Gajewsky, G., Holsboer, vocalizations by the neuroactive steroid allopregnanolone. Pharmacol. Bio-
F., Rupprecht, R., 2003. Induced panic attacks shift gamma-aminobutyric acid chem. Behav. 47, 735–738.
type A receptor modulatory neuroactive steroid composition in patients with Zwanzger, P., Eser, D., Padberg, F., Baghai, T.C., Schule, C., Rupprecht, R., di Michele,
panic disorder: preliminary results. Arch. Gen. Psychiatry 60, 161–168. F., Romeo, E., Pasini, A., Strohle, A., 2004. Neuroactive steroids are not affected
Strohle, A., Romeo, E., di Michele, F., Pasini, A., Yassouridis, A., Holsboer, F., by panic induction with 50 microg cholecystokinin-tetrapeptide (CCK-4) in
Rupprecht, R., 2002. GABA(A) receptor-modulating neuroactive steroid compo- healthy volunteers. J. Psychiatr. Res. 38, 215–217.

Potrebbero piacerti anche