Sei sulla pagina 1di 18

REVIEW ARTICLE

published: 19 January 2012


doi: 10.3389/fnins.2011.00136

Effects and mechanisms of 3α,5α,-THP on emotion,


motivation, and reward functions involving pregnane
xenobiotic receptor
Cheryl A. Frye 1,2,3,4 *, J. J. Paris 1 , A. A. Walf 4 and J. C. Rusconi 2
1
Department of Psychology, The University at Albany-SUNY, Albany, NY, USA
2
Biological Sciences, The University at Albany-SUNY, Albany, NY, USA
3
The Centers for Neuroscience, The University at Albany-SUNY, Albany, NY, USA
4
Life Science Research, The University at Albany-SUNY, Albany, NY, USA

Edited by: Progestogens [progesterone (P4 ) and its products] play fundamental roles in the develop-
Kazuyoshi Tsutsui, Waseda University,
ment and/or function of the central nervous system during pregnancy. We, and others,
Japan
have investigated the role of pregnane neurosteroids for a plethora of functional effects
Reviewed by:
Lan Ma, Fudan University, China beyond their pro-gestational processes. Emerging findings regarding the effects, mech-
Steven King, Texas Tech University anisms, and sources of neurosteroids have challenged traditional dogma about steroid
Health Sciences Center, USA action. How the P4 metabolite and neurosteroid, 3α-hydroxy-5α-pregnan-20-one (3α,5α-
*Correspondence: THP), influences cellular functions and behavioral processes involved in emotion/affect,
Cheryl A. Frye, The University at
motivation, and reward, is the focus of the present review. To further understand these
Albany-SUNY, Life Sciences Research
Building 01058, 1400 Washington processes, we have utilized an animal model assessing the effects, mechanisms, and
Avenue, Albany, NY 12222, USA. sources of 3α,5α-THP. In the ventral tegmental area (VTA), 3α,5α-THP has actions to facil-
e-mail: cafrye@albany.edu itate affective, and motivated, social behaviors through non-traditional targets, such as
GABA, glutamate, and dopamine receptors. 3α,5α-THP levels in the midbrain VTA both
facilitate, and/or are enhanced by, affective and social behavior. The pregnane xenobiotic
receptor (PXR) mediates the production of, and/or metabolism to, various neurobiological
factors. PXR is localized to the midbrain VTA of rats. The role of PXR to influence 3α,5α-THP
production from central biosynthesis, and/or metabolism of peripheral P4 , in the VTA, as
well as its role to facilitate, or be increased by, affective/social behaviors is under inves-
tigation. Investigating novel behavioral functions of 3α,5α-THP extends our knowledge of
the neurobiology of progestogens, relevant for affective/social behaviors, and their connec-
tions to systems that regulate affect and motivated processes, such as those important for
stress regulation and neuropsychiatric disorders (anxiety, depression, schizophrenia, drug
dependence).Thus, further understanding of 3α,5α-THP’s role and mechanisms to enhance
affective and motivated processes is essential.
Keywords: allopregnanolone, anxiety, lordosis, paced mating, progesterone, sex differences, stress, ventral
tegmental area

INTRODUCTION behavioral response to appropriate stimuli. We now know that


Steroids play fundamental roles in the development and/or func- steroid levels can change in response to environmental and/or
tion of the central nervous system (CNS) and exert both orga- behavioral situations and, thereby, influence the likelihood of sub-
nizational and activational effects. During pregnancy, levels of sequent steroid-mediated processes occurring. There is now a
progestogens [i.e., progesterone (P4 ) and its metabolites] are high greater understanding of subtle, and dynamic changes in steroids
and have well-known effects to maintain pregnancy and may alter to mediate homeostatic processes [e.g., hypothalamic-pituitary–
brain development of the gestating offspring. We, and others, have adrenal (HPA) axis function], which may exert proximate and
investigated the role of pregnane neurosteroids for a plethora of discrete effects on physiological and/or behavioral processes. Sec-
functional effects beyond their pro-gestational processes in adult- ond, the classic mechanisms of steroids are considered to involve
hood. Some of these effects are those involved in emotion/affect, their binding to cognate, intracellular steroid receptors, which are
motivation, and reward, which will be the focus of this review. present throughout the brain in hypothalamic and limbic regions
Emerging findings regarding the effects, mechanisms, and (Shughrue et al., 1997; Osterlund et al., 2000), and modulate tran-
sources of steroids have challenged traditional dogma and revealed scription and translation (Pfaff et al., 1976), a process which takes
non-traditional actions of hormones to influence cellular func- 10 min to days. Steroids can also act in the CNS via membrane
tions and behavioral processes. First, some of the most salient targets or rapid-signaling actions, which occur within seconds to
effects of steroids are to change the threshold for a biological or minutes. Neuro(active) steroids produce rapid effects on neuronal

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 1


Frye et al. 3α,5α-THP and PXR motivated behaviors

excitability and synaptic function that involve direct or indi- robust behavioral effects, such as enhancing affect and facili-
rect modulation of ion-gated or other neurotransmitter receptors tating reproductive and other motivated behaviors (Frye et al.,
and transporters, rather than classic nuclear hormone receptors. 2006a; Frye, 2009). For example, central infusions of 3α,5α-THP
Third, steroids are typically thought to be secreted from periph- to VTA (but not to nearby regions, such as central gray, raphe
eral endocrine glands into circulation, whereupon they can exert nucleus, substantia nigra) of non-sexually receptive rats signifi-
effects at target sites in the body and brain that are distant from cantly enhances affective and social behavior to levels commen-
the endocrine gland source. However, the brain, like the gonads, surate with those observed in sexually receptive rats (Frye and
adrenals, and placenta, can be considered an endocrine organ. Rhodes, 2006a, 2007a,b, 2008). The VTA is largely devoid of P4 ’s
That is, the brain requires coordinated actions of steroidogenic traditional cognate steroid receptor targets, progestin receptors
enzymes in neurons and glia to metabolize peripheral steroids (PRs). 3α,5α-THP has lower affinity for PRs than it does for γ-
to products that act in the brain (“neuroactive steroids”), or to aminobutyric acid (GABAA ), glutamatergic, and dopaminergic
produce steroids de novo in the brain independent of periph- receptor targets, which are highly expressed in the VTA (Frye and
eral gland secretion (“neurosteroids”). In this view, steroids exert Walf, 2008a). As well, blocking 3α,5α-THP targets, such as GABAA
intracrine effects to mediate intracellular events, and paracrine, receptors, in the VTA attenuates anti-anxiety and social behav-
or neurotransmitter-like, effects to induce biological responses in ior among sexually receptive females (Frye et al., 2006b,c; Frye
adjacent cells. This review will focus its discussion on the effects, and Paris, 2009). This is not observed when blockers are adminis-
mechanisms, and sources of the P4 metabolite and neurosteroid, tered to nearby missed sites, such as the substantia nigra or central
5α-pregnan-3α-ol-20-one (3α,5α-THP, a.k.a. allopregnanolone), gray (Frye and Paris, 2009). As such, actions of 3α,5α-THP in the
for affective, motivated, and reward processes. As discussed as VTA to enhance anti-anxiety and pro-social motivated behaviors
follows, we examine effects, sources, and mechanisms of progesto- may be specific to the VTA and its connections. Enzymes, such
gens in rats, which have demonstrated similar effects and patterns as 5α-reductase and 3α-hydroxysteroid dehydrogenase (3α-HSD),
of progestogen secretion as is seen in people (Holzbauer, 1975, that are necessary for the metabolism of P4 to 3α,5α-THP, and
1976; Holzbauer et al., 1985; Frye and Bayon, 1999), and, thus, can de novo synthesis of 3α,5α-THP, are highly expressed in the VTA
provide insight into progestogens’ role in these processes. (Li et al., 1997; Frye, 2001a,b), suggesting that this is a region
to investigate to further understand the sources of progestogens.
MODEL SYSTEM TO INVESTIGATE PROGESTOGENS’ Indeed, P4 , from central or peripheral sources, is readily metabo-
EFFECTS, MECHANISMS, AND SOURCES FOR AFFECTIVE lized to 5α-pregnane-3,20-dione-dihydroprogesterone (DHP), by
AND MOTIVATED BEHAVIORS actions of 5α-reductase, and 3α,5α-THP, by actions of 3α-HSD, in
We have utilized an animal model to elucidate the effects, mech- the VTA. Blocking P4 ’s metabolism to 3α,5α-THP in the VTA, or
anisms, and sources of progestogens to influence affective and blocking de novo production, or neurosteroidogenesis, of 3α,5α-
motivated behaviors of rodents. Progestogens may have a role in THP in the VTA, attenuates affective and social behavior among
the etiology, expression, and/or therapeutic treatment of anxiety, sexually receptive rats (Frye and Petralia, 2003a,b; Frye et al.,
stress, and/or mood (dys)regulation, as discussed below. Mating 2008b). Reinstating 3α,5α-THP concentrations via enhancement
is a sexually dimorphic and progestogen-mediated behavior. To of neurosteroidogenesis, or 3α,5α-THP add-back, reinstates these
be able to determine necessary mechanisms for this behavior, we behaviors (Petralia et al., 2005; Frye et al., 2009). Thus, we can use
have utilized lordosis (the posture that female rodents assume to behavioral endpoints of female rodents to ascertain the sources,
enable mating to occur, which can be considered a consummatory effects, and mechanisms of progestogens in the midbrain VTA,
aspect of mating for the female) as an ethologically relevant bioas- and determine the extent to which these actions are relevant in
say for progestogens’ actions. In this regard, we have examined other brain regions and systems. What follows is a discussion of
progestogens’ physiological and ethological role to mediate lordo- findings from our laboratory, and others, regarding the effects,
sis, as well as neuroendocrine and behavioral processes relevant for mechanisms, and sources of 3α,5α-THP for affect, motivation,
social interactions and/or affect, which can be considered appet- and reward processes.
itive aspects of mating. In this model, lordosis is both a sensitive
measure of progestogens’ effects as well as an experiential factor in EFFECTS OF 3α,5α-THP
the rodent’s life that can be manipulated to alter subsequent neu- GENDER DIFFERENCES FOR AFFECTIVE AND MOTIVATED PROCESSES
roendocrine and behavioral responses. As such, the directionality Depression and anxiety are serious and wide-spread disorders,
of the effects of progestogen production and affective and moti- with effects on emotion as well as motivation- and reward-related
vated responding can be examined. Thus, investigating behaviors processes. Incidence and expression of these disorders are greater
commonly disrupted in neuropsychiatric disorders (affect, social, among women compared to men, and may be related to fluctu-
and reproductive endocrine function), using an ethologically rel- ations in P4 and 3α,5α-THP. Over their lifetime, mature women
evant model of rodent behavior, can elucidate the functional role experience greater variations in, and higher levels of, progesto-
of progestogens, such as 3α,5α-THP, for mental health. gens than do men, and they are more susceptible to depres-
In this model system, we have focused to date on actions of sion and/or anxiety disorders (Weissman and Klerman, 1977;
progestogens in the midbrain ventral tegmental area (VTA). The Kessler et al., 1994; Seeman, 1997; Wittchen and Hoyer, 2001).
VTA is a target of interest for several reasons including its role in the During the follicular phase of the menstrual cycle, circulating
mesolimbic dopamine system. Natural fluctuations in progesto- progestogen levels of women are low (similar to men); how-
gens, and administration of progestogens to the VTA, produce ever, luteal phase circulating levels are two to fourfold higher

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 2


Frye et al. 3α,5α-THP and PXR motivated behaviors

(2–4 nmol/l) than follicular phase levels (∼1 nmol/l; Genaz- 2005; Walf et al., 2006; Frye, 2009). Thus, 3α,5α-THP may have
zani et al., 1998; Purdy et al., 1990; Sundström and Bäck- homeostatic effects by restoring parasympathetic tone following
ström, 1998a,b; Wang et al., 1996). During pregnancy, circulat- stressor exposure.
ing levels of progestogens rapidly increase to peak in the third
trimester (50–160 nmol/l), and reach nadir within a day post- STRESS ALTERS 3α,5α-THP
parturition (Sundström et al., 1999; Luisi et al., 2000; Herbison, Stressor exposure has salient effects to alter 3α,5α-THP through-
2001). The onset and/or expression of depression and/or anxi- out the lifespan. Brain levels of 3α,5α-THP are measurable as early
ety can be mediated by some of these changes in progestogen as embryonic day 14 in rats (Kellogg and Frye, 1999). By postna-
levels over natural cycles. In support, premenstrual syndrome, tal day 6, 3α,5α-THP increases in the brain of rats in response to
premenstrual dysphoric disorder (PMDD), post-partum depres- maternal separation stress (Kehoe et al., 2000; McCormick et al.,
sion, and menopause, are associated with negative affect/mood, 2002). In adult rodents, 3α,5α-THP increases in response to a
and occur when progestogen levels are low (Glick and Bennett, variety of acute stressors (cold-water swim, restraint, foot-shock,
1981; Angold et al., 1999; Endicott et al., 1999; Chaudron et al., loud noise, carbon dioxide inhalation, ether exposure, or admin-
2001; Girdler et al., 2001; Soares and Cohen, 2001; Freeman istration of anxiogenic drugs) occur in intact, gonadectomized
et al., 2002, 2004; Rapkin et al., 2002; Bäckström et al., 2003; (GDX), and/or ADX rodents (Erskine and Kornberg, 1992; Paul
Markou et al., 2005; Pearlstein et al., 2005). Moreover, substance and Purdy, 1992; Barbaccia et al., 1996, 1997; Frye, 2001a,b; Serra
abuse disorder is often co-morbid with depression and anxi- et al., 2002). Stimuli that can be considered more subtle than these
ety (Regier et al., 1990), especially among women compared to aforementioned acute stressors, such as social challenge and/or
men (Brady and Randall, 1999). Natural fluctuations in progesto- mating, alters production of 3α,5α-THP (as described in further
gens across the menstrual cycle alter responses to drugs of abuse, detail below; Frye, 2001a,b; Miczek et al., 2003; Frye and Rhodes,
such as alcohol, cocaine, and nicotine (Sofuoglu et al., 1999; 2006a). Increases in 3α,5α-THP produced by such experiences are
Pomerleau et al., 2000; Nyberg et al., 2004). Understanding the conserved across species, enhance GABA function, increase anxi-
effects, mechanisms, and sources of neurosteroids, such as 3α,5α- olysis, and reduce HPA responses (Paul and Purdy, 1992; Patchev
THP, may provide insight into gender/sex differences, etiology, and Almeida, 1996; Barbaccia et al., 2001; Frye, 2001a,b, 2009;
expression, progression, and/or treatment of some mental health Reddy, 2003). Thus, 3α,5α-THP is expressed early in development
disorders. and can be altered by stress at this time and during adulthood.
In addition to gender/sex differences for affective and motivated SEX/PROGESTOGEN EFFECTS ON STRESS
processes as discussed above, there may are gender/sex differ- There are sex differences in the 3α,5α-THP response to stres-
ences in normal stress responding of males and females. Males sors. Maternal separation stress produces greater increases in brain
may be more likely to cope by mounting a “fight-or-flight” type 3α,5α-THP levels of male, compared to female, pups between post-
response toward stressors, whereas females may cope better with a natal day 7 and 10 (Kehoe et al., 2000; McCormick et al., 2002).
“tend-and-befriend” response (Taylor et al., 2000). Although many In male rats, non-stress, basal 3α,5α-THP levels are higher at an
neurobiological factors clearly differ between males and females, early juvenile age (postnatal day 14), whereas in females, basal
and likely contribute to these differences in pathophysiological and 3α,5α-THP levels are higher at postpubertal ages (42 and 60 days).
normative responding, the focus of this review is on how 3α,5α- Although it is currently unclear what role higher 3α,5α-THP levels
THP has such actions. Thus, findings of 3α,5α-THP’s role in stress, in males at early juvenile ages, or in females at late adolescent/early
affect, and motivated processes follow. adult ages, may have in adults, evidence indicates that progesto-
gens modify stress responses. Post-partum women, who have
3α,5α-THP ALTERS THE HPA AXIS increased estradiol (E2 ), but decreased progestogens, have greater
3α,5α-THP has agonist-like actions at inhibitory GABA recep- HPA response to stressors (Altemus et al., 2001). Stress responses
tors and can dampen stress-induced HPA activity, which may of women may be increased when progestogens are decreased post-
mitigate parasympathetic tone. 3α,5α-THP’s anesthetic proper- menopause (De Leo et al., 1998). In premenopausal women, oral
ties have been recognized for some time (Selye, 1941), related contraceptives, or progestogens, decrease cortisol levels (Hellman
to its potency at enhancing GABA function (Majewska et al., et al., 1976; Jacobs et al., 1989). Thus, there are gender/sex differ-
1986). Administration of 3α,5α-THP reduces adrenocorticotropin ences in stress responding, and stress responses can be modulated
secretion in response to acute stress, and blocks adrenalec- by progestogens.
tomy (ADX)-induced increases in corticotrophin releasing factor
mRNA (Patchev et al., 1994, 1996). When 3α,5α-THP levels are 3α,5α-THP AND DEPRESSION
elevated, during proestrus or pregnancy, or when ovariectomized Neurosteroids, such as 3α,5α-THP, may play a role in depression.
(OVX) rats are administered 3α,5α-THP, there are robust anti- Stressful life events can precipitate depression (Brown et al., 1994).
anxiety and anti-stress effects (Harrison and Simmonds, 1984; Individuals with depression often have difficulties in coping with
Majewska et al., 1986; Lambert et al., 2003; Belelli and Lam- stress. Increased levels of corticotrophin releasing factor and cor-
bert, 2005; Martín-García and Pallarès, 2005; Frye et al., 2006a; tisol, and/or impaired glucocorticoid feedback to dexamethasone,
Frye, 2009). Blocking formation of 3α,5α-THP, or its actions are observed in depression (Carroll et al., 1976, 1981; Halbreich
at GABAA receptors, prevents anti-anxiety and anti-depressant et al., 1985; Rubin et al., 1987; Nemeroff et al., 1988; Young et al.,
behavior, as well as glucocorticoid secretion following stressor 2000). Depression is a common side-effect of treatment of alope-
exposure (Rhodes and Frye, 2001; Reddy, 2002; Verleye et al., cia or benign prostate hyperplasia with finasteride, a 5α-reductase

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 3


Frye et al. 3α,5α-THP and PXR motivated behaviors

inhibitor, which decreases neurosteroids, such as 3α,5α-THP and et al., 2003; Walf and Frye, 2006). Ovariectomy increases anxiety
its androgen equivalent, androstanediol (Altomare and Capella, behavior of female rodents (Frye and Walf, 2002; Walf et al., 2006)
2002; Clifford and Farmer, 2002; Townsend and Marlowe, 2004). and P4 or 3α,5α-THP reverses this (Frye and Walf, 2004; Frye et al.,
Thus, 3α,5α-THP inhibition can precipitate depression among 2004a; Walf et al., 2006), unless 3α,5α-THP formation is compro-
some individuals. mised or 3α,5α-THP withdrawal occurs (Rhodes and Frye, 2001;
Findings from animal models also support a role of progesto- Frye and Walf, 2002, 2004). Thus, some of progestogens’ effects for
gens in depressive behaviors. For example, when rodents are in anxiety-related behaviors may be due to 3α,5α-THP.
the proestrous phase of the estrous cycle and have high levels
of progestogens, they show less depressive behavior compared to
3α,5α-THP AND MOOD DYSREGULATION
rodents that are in low progestogen phases of the estrous cycle
An example of a disorder in which mood changes may be altered by
(Becker and Cha, 1989; Bitran et al., 1991; Frye et al., 2000b;
3α,5α-THP is PMDD. PMDD is characterized by both debilitating
Frye and Walf, 2002; Gulinello et al., 2003). Pregnant rats, which
physical symptoms and negative mood state, which occurs when
have sustained, higher levels of progestogens, show less depressive
E2 and progestogen levels fluctuate during the luteal phase (Bäck-
behavior in the forced swim test than do post-parturient rats, with
ström et al., 1983; Sanders et al., 1983; Hammarbäck et al., 1989,
lower levels of progestogens (Frye and Walf, 2004; Walf and Frye,
1991; Ramcharan et al., 1992; Endicott et al., 1999; Sveindóttir
2006). Ovariectomy, removal of the primary source of endoge-
and Bäckström, 2000). Some women with PMDD report greater
nous progestogens, increases depressive behavior among female
positive, and negative, mood with ovarian suppression and admin-
rats in the forced swim test (Frye and Wawrzycki, 2003; Walf et al.,
istration of E2 and/or P4 , respectively (Schmidt et al., 1998).
2006). Administration of P4 and/or 3α,5α-THP can reverse these
Among women with PMDD, there are also conflicting reports
effects (Frye and Walf, 2002; Hirani et al., 2002; Frye et al., 2004a).
that mood is more negative (Hammarbäck et al., 1989; Seippel
The anti-depressant effects of P4 are reduced when drugs that
and Bäckström, 1998), or improved (Wang et al., 1996; Girdler
block conversion of P4 to 3α,5α-THP, such as finasteride, are co-
et al., 2001), when hormone levels are greater during the luteal
administered (Frye and Walf, 2002; Hirani et al., 2002; Walf et al.,
phase. There is greater consensus that there are few differences
2006). Similarly, the anti-depressive effects of P4 in the forced swim
among absolute levels of E2 , P4 , and/or 3α,5α-THP of women with
test are attenuated among 5α-reductase knockout mice that cannot
PMDD and those that do not have PMDD (Rubinow et al., 1988;
readily convert P4 to 3α,5α-THP (Frye et al., 2004a). In another
Halbreich et al., 1993; Schmidt et al., 1994; Wang et al., 1996, 2001;
animal model of depression, social isolation of mice, lower lev-
Sundström and Bäckström, 1998a; Epperson et al., 2002). How-
els of 3α,5α-THP have been reported (Dong et al., 2001; Guidotti
ever, lower (Rapkin et al., 1997; Bicikova et al., 1998; Monteleone
et al., 2001). Thus, some of progestogens’ anti-depressive effects in
et al., 2000), and higher (Girdler et al., 2001), concentrations
rodents may be due to actions of 3α,5α-THP.
of 3α,5α-THP have been reported among women with PMDD.
There is evidence for differences in 3α,5α-THP among women
3α,5α-THP AND ANXIETY
with PMDD treated with selective serotonin reuptake inhibitors
3α,5α-THP’s role in various anxiety disorders have been investi-
(SSRI). In support, there is clinical improvement among those
gated. Baseline plasma levels of 3α,5α-THP are normal in patients
with stabilized 3α,5α-THP levels following SSRI treatment (Free-
with generalized anxiety disorders and social phobia (Le Mellédo
man et al., 2002; Gracia et al., 2009). One explanation for some of
and Baker, 2002). However, 3α,5α-THP levels are higher than nor-
the heterogeneity in these findings is that there may be bimodal
mal among people with panic disorder (Brambilla et al., 2003;
responses associated with differences in sensitivity of GABA recep-
Ströhle et al., 2003), and are reduced by infusions of sodium
tors (Sundström et al., 1997a,b, 1998). Together, these data suggest
lactate or cholecystokinin tetrapeptide, which can induce panic
that 3α,5α-THP may underlie some effects on mood processes
attacks (Ströhle et al., 2003). In people without a history of panic
among women.
attacks, levels of neurosteroids are either not affected, or greater,
following administration of panic-inducing agents (Tait et al.,
2002; Zwanzger et al., 2004; Eser et al., 2005). In women with 3α,5α-THP AND SCHIZOPHRENIA
panic disorder, perimenstrual, but not midluteal, 3α,5α-THP levels 3α,5α-THP may play a role in the pathophysiology of schiz-
were significantly higher than controls, and correlated with their ophrenia, which is characterized by deficits in social, affective,
panic–phobic symptoms (Brambilla et al., 2003). With Dr. John and cognitive functioning (Shirayama et al., 2002). Addition-
Casada, we found that among men with post-traumatic stress dis- ally, schizophrenia is characterized by dysregulation of the HPA
order, those with higher 3α,5α-THP levels have lower state anxiety axis (Lukoff et al., 1984; Malla et al., 1990; Norman and Malla,
following exposure to trauma-related stimuli (Frye, 2009). Thus, 1993; Myin-Germeys et al., 2001; Read et al., 2001) and psy-
there is some evidence for differences in 3α,5α-THP among those chosis can be precipitated by stress (Read et al., 2001). Those
with anxiety disorders. with schizophrenia may have altered stress-induced 3α,5α-THP
Animal models also support a role of progestogens in anxiety production, which is supported by a novel polymorphism in
behavior (Guidotti and Costa, 1998; Bitran et al., 2000; Jain et al., the gene sequence encoding for enzymes involved in 3α,5α-THP
2005). When P4 and 3α,5α-THP levels are high, such as during biosynthesis and may create a predisposition to over-sensitivity
proestrus and pregnancy, anxiety behavior is lower among female to stress (Kurumaji et al., 2000; Myin-Germeys et al., 2001; Read
rodents compared to when levels are declining or are low (Becker et al., 2001). Moreover, women have higher levels of 3α,5α-THP
and Cha, 1989; Bitran et al., 1991; Frye et al., 2000b; Gulinello and are more likely to have later onset schizophrenia, better

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 4


Frye et al. 3α,5α-THP and PXR motivated behaviors

prognosis, and therapeutic response to lower dosages of anti- effects of cocaine (Russo et al., 2008) and administration of 3α,5α-
psychotics than do men (Usall et al., 2007; Abel et al., 2010). THP reduces cocaine self-administration (Anker et al., 2010).
When 3α,5α-THP levels are low perimenstrually, greater psy- As well, administration of 3α,5α-THP, and to a lesser extent
chotic episodes and more negative schizophrenia symptoms are P4 , reduced cocaine reinstatement following abstinence among
reported (Hallonquist et al., 1993; Hendrick et al., 1996; Huber female, but not male rats (Anker et al., 2009). Effects of P4 were
et al., 2001). Thus, schizophrenia may involve a reduced capacity attenuated with co-administration of the 5α-reductase inhibitor,
to synthesize 3α,5α-THP in the brain, which may increase sen- finasteride, which blocks P4 ’s metabolism to 3α,5α-THP (Anker
sitivity to stress and, thereby, vulnerability to psychosis in this et al., 2009). Together, these data suggest a role of progestogens for
population. drug reward.

3α,5α-THP AND ACTIONS OF THERAPEUTICS MECHANISMS OF 3α,5α-THP FOR AFFECT AND MOTIVATED
3α,5α-THP may underlie some of the effects of therapeutics BEHAVIORS
for stress-related, neuropsychiatric disorders. Anxiolytic effects Neurosteroids, such as 3α,5α-THP, can have more immediate,
of etifoxine, correspond with increased levels of 3α,5α-THP in rapid-signaling effects through ion channel-associated membrane
sham-operated and GDX/ADX rats (Verleye et al., 2005). An atyp- receptors within milliseconds to seconds than steroids secreted
ical anti-psychotic drug, olanzapine, enhances social functioning by peripheral glands that act through classic nuclear steroid recep-
and increases 3α,5α-THP levels (Marx et al., 2000, 2003; Frye tors. The most extensively investigated actions of neurosteroids are
and Seliga, 2003a,b). Fluoxetine increases the affinity of 3α-HSD those at synaptic and extrasynaptic GABAA receptors, as described
for DHP, which elevates 3α,5α-THP (Griffin and Mellon, 1999). below. 3α,5α-THP can also have actions through other non-
Some patients with depression have reduced plasma concentra- steroidal, ligand-gated, ion channels, and/or G-protein-coupled
tions and/or cerebrospinal fluid levels of 3α,5α-THP (Romeo et al., receptors. Those that we have focused our investigations on to
1998; Uzunova et al., 1998). Anti-depressants, such as fluoxetine or date for affect, motivation, and reward are glutamate, dopamine,
fluvoxamine, normalize decreased 3α,5α-THP levels concomitant and membrane PRs (Rupprecht and Holsboer, 1999; Zhu et al.,
with reducing depressive symptomology (Uzunova et al., 2004, 2003; Frye and Walf, 2008a; Frye, 2011). A brief description of
2006; Dubrovsky, 2006). Other treatments of depression, such as some of progestogens’ actions at these non-traditional targets is
sleep deprivation (Schüle et al., 2003), electroconvulsive therapy described as follows. For further discussion, the reader is referred
(Baghai et al., 2005), and transcranial magnetic stimulation (Pad- to other recent reviews (e.g., see others in this special issue; Frye,
berg et al., 2002), modestly alter neurosteroids. Common features 2009, 2011).
of these therapeutic treatments include changes in steroid biosyn-
thesis and HPA function that may mitigate core symptoms of these P4 HAS NON-PR ACTIONS IN THE VTA
neuropsychiatric disorders described above (Dubrovsky, 2006). Among rodents, E2 and P4 initiate the facilitation of lordosis, in
Thus, 3α,5α-THP may underlie some actions of therapeutics. part, through actions to increase PR expression in the ventrome-
dial nucleus (Schwartz-Giblin and Pfaff, 1986). In the VTA, P4
3α,5α-THP AND DRUG ABUSE mediates the duration and intensity of lordosis. Receptor bind-
Recent investigations assessing the mechanisms of reward asso- ing and immunocytochemistry experiments conducted in our lab,
ciated with drugs of abuse have revealed a role for progestogens. and others, demonstrated that there are very few intracellular PRs
There is evidence for menstrual cycle effects for measures related to in the VTA of adult rodents (Warembourg, 1978; MacLusky and
drug abuse, such as subjective feelings and craving and withdrawal McEwen, 1980; Blaustein et al., 1994; Frye and Vongher, 1999).
following abstinence. In support, women in the luteal phase report Further, the few PRs that exist in the VTA are not inducible by
lower rating for feeling high following smoking of cocaine than did E2 , and reducing PR expression via infusion of antisense ODNs
women in the follicular phase of the menstrual cycle (Sofuoglu to the VTA does not affect lordosis, as is the case with infusions
et al., 1999). Among cocaine-dependent women, circulating lev- to the ventromedial nucleus (Frye and Vongher, 1999; Frye et al.,
els of progesterone were associated with cocaine craving, such 2000a; Frye, 2001a). Intravenous or intra-VTA infusions of P4 to
that those with high progesterone had lower stress- and cocaine hamsters or rats enhance firing of VTA neurons within 60 s (Rose,
cue-induced cravings for cocaine and reported less anxiety (Sinha 1990; Frye et al., 2000b) and facilitates lordosis (Pleim et al., 1991;
et al., 2007). Among women, there are menstrual cycle-related DeBold and Frye, 1994). This effect occurs even when P4 ’s actions
differences in craving and withdrawal symptoms with nicotine are relegated to cell membranes because it has been bound to
abstinence, which may be particularly strong among women with a macromolecule like bovine serum albumin and is too large to
severe menstrual symptomatology and/or co-morbid neuropsy- enter the cell (Frye et al., 1992; Frye and Gardiner, 1996). These
chiatric disorders (Pomerleau et al., 2000; Carpenter et al., 2006). data suggest that progestogens’ actions in the VTA for lordosis
There are also effects of progestogen administration. For exam- do not require traditional actions at PRs. Of interest is that E2 -
ple, oral P4 to women reduces self-reported pleasurable effects of stimulated neurosteroidogenesis of progesterone, via membrane
cocaine (Sofuoglu et al., 2002, 2004). Animal models show sup- E2 signaling, was required in OVX/ADX for proceptive, rather than
port for a role of progestogens in drug reward. There are sex and receptive, behavior (Micevych et al., 2008; Micevych and Dewing,
estrous cycle differences in behavioral effects and metabolism of 2011). Together, these studies suggest that there are non-genomic,
P4 to 3α,5α-THP following cocaine administration to rats (Frye, or non-traditional, targets of neurosteroids, like 3α,5α-THP for
2007; Quiñones-Jenab et al., 2008; Kohtz et al., 2010). Moreover, some of its functional effects. Some of these targets are described
systemic P4 administration to female rats blocks the rewarding as follows.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 5


Frye et al. 3α,5α-THP and PXR motivated behaviors

3α,5α-THP ACTS AT GABAA RECEPTORS and Frye, 2004, 2006a,b; Sumida et al., 2005; Frye and Walf, 2007,
In nanomolar concentrations, 3α,5α-THP directly activates 2008a,b,c). Moreover, increased progestogen levels of rodents are
GABAA and is a positive, allosteric modulator. 3α,5α-THP associated with increases in levels of cyclic adenosine monophos-
increases chloride channel currents and lowers neuronal excitabil- phate (cAMP) in the cortex (Frye, 2001a,b; Frye and Walf, 2008a).
ity with 20- and 200-fold higher efficacy than benzodiazepines Thus, progestogens’ actions in the VTA require activation of these
or barbiturates, respectively (Morrow et al., 1987; Gee et al., 1995; signal transduction molecules.
Brot et al., 1997; Lambert et al., 2003; Reddy, 2004; Weir et al., 2004;
Belelli and Lambert, 2005). In the VTA, 3α,5α-THP facilitates lor- SOURCES OF 3α,5α-THP
dosis in part through its agonist-like actions at GABAA receptors. Beyond an understanding of the various effects of 3α,5α-THP and
Pharmacological blockade of GABAA receptors attenuates lordosis the mechanisms for such effects, a critical question is the sources of
of proestrous hamsters (or rats) over vehicle when administered to 3α,5α-THP for these effects. Progestogen concentrations in brain
the VTA, but not other regions outside the VTA, such as the ventro- may be due to gonadal, adrenal, and central sources. One of the
medial hypothalamus, central gray, or substantia nigra (Frye et al., rate-limiting factors in understanding more about the functional
1993; Frye and Vongher, 1999; Frye, 2001a,b; Frye and Paris, 2009, significance of steroids lies in the challenge of parsing out the rel-
2011a). Conversely, muscimol, which has agonist-like actions at ative contributions of central versus peripheral endocrine glands.
GABAA receptors, enhances P4 -facilitated lordosis of hamsters or Neurosteroids are synthesized in the CNS and/or peripheral ner-
rats, when infused to the VTA (Frye and DeBold, 1992; Frye and vous system (PNS), rather than the gonads, adrenals, and/or pla-
Gardiner, 1996). Thus, 3α,5α-THP has actions in the midbrain centa (Baulieu, 1980, 1991). Levels of neurosteroids are typically
VTA in part through its agonist-like actions at GABAA receptors. greater in the CNS and PNS than in circulation. Enzymes involved
in peripheral gland steroidogenesis have been identified in the
3α,5α-THP ACTS AT N -METHYL-D-ASPARTATE RECEPTORS CNS and PNS (Li et al., 1997; Furukawa et al., 1998; Compagnone
Many of the GABAergic neurons that exist in this region also and Mellon, 2000). As well, high CNS and PNS levels of neu-
express N -methyl-d-aspartate receptors (NMDARs; Steffensen rosteroids persist after extirpation of peripheral glands (i.e., GDX
et al., 1998). Autoradiography studies indicate OVX rats admin- and/or ADX; Baulieu, 1980, 1991; Majewska, 1992; Paul and Purdy,
istered P4 and/or E2 have reduced NMDAR binding in cortex 1992; Mellon, 1994). Of continued interest are the factors that are
(Wu et al., 1991; Cyr et al., 2000). Neurosteroids, such as 3α,5α- involved in neurosteroid formation.
THP, have actions involving NMDARs (Korinek et al., 2011). The translocator protein (18 kDa TSPO; formally known as
Antagonizing NMDARs via intra-VTA infusions of MK-801, a the peripheral-type benzodiazepine receptor/recognition site)
non-competitive NMDAR antagonist, enhances P4 -facilitated lor- binds cholesterol in nanomolar affinities and is essential for
dosis (Frye, 2001a,b; Petralia et al., 2007; Frye et al., 2008a; Frye neurosteroidogenesis. In 1977, the TSPO was first identified as
and Paris, 2011b). Thus, 3α,5α-THP in the midbrain VTA may act the binding site for diazepam in peripheral tissues. The most
in part through its antagonist-like actions at NMDARs. extensively investigated functions of TSPOs are their role in
biosynthesis of steroids. The TSPO is a high affinity choles-
3α,5α-THP’s ACTIONS THROUGH DOPAMINE SIGNALING terol binding protein that imports cholesterol into the mito-
The VTA is also a site of dopaminergic activity, and actions of chondria (Papadopoulos et al., 2006). The steroidogenic acute
3α,5α-THP for socially relevant behavior. In support, dopamine regulatory (StAR) protein is also involved in the importing of
agonists can facilitate lordosis of rodents via phosphorylation of cholesterol, but it is unclear if TSPO and StAR work together
PRs (Mani, 2003). We have investigated the role of D1 receptors (King et al., 2004). After its importation into the mitochondria,
in the VTA for progestogen-facilitated lordosis. D1 receptors are cholesterol is then oxidized to pregnenolone by the cytochrome
localized to the VTA (Boyson et al., 1986). As well, in the VTA, P450-dependent side chain cleavage enzyme (P450scc; Mellon
where there are few PRs, infusions of D1 agonists and antago- and Deschepper, 1993). Pregnenolone, the precursor of all neu-
nists enhance and inhibit lordosis of E2 - and progestogen-primed rosteroids, is metabolized to P4 by the 3β-hydroxysteroid dehy-
rodents, respectively (Frye et al., 2004b, 2006b,c,d; Petralia and drogenase (3β-HSD), which can then be metabolized to form
Frye, 2004, 2006a,b; Sumida et al., 2005). Thus, it may be that DHP and 3α,5α-THP (Mellon and Deschepper, 1993). The CNS
D1 activation downstream of GABAA receptors in the VTA (Lavi- expresses all of these enzymes that initiates steroidogenesis and
olette and van der Kooy, 2001; Laviolette et al., 2004; Frye et al., can be considered rate-limiting steps in 3α,5α-THP biosynthe-
2006a) underlies some of the rewarding effects of social responding sis (Braestrup and Squires, 1977; Benavides et al., 1983; Li et al.,
among rodents. 1997; Furukawa et al., 1998; Gavish et al., 1999; Compagnone
and Mellon, 2000; Chen and Guilarte, 2008). Some of the regions
RAPID ACTIONS OF 3α,5α-THP VIA GABA, NMDA, AND D1 RECEPTORS with the highest expression of StAR, 3β-HSD, 5α-reductase, and
REQUIRE ACTIVATION OF SIGNAL TRANSDUCTION CASCADES 3α-HSD, all enzymes required for 3α,5α-THP formation, are
Progestogens’ actions in the VTA involve activation of signal midbrain, cortical, and limbic regions and the cerebellum (Li
transduction pathways. In brief, infusions of adenylyl cyclase, G- et al., 1997; Furukawa et al., 1998; Compagnone and Mellon,
proteins, protein kinase A (PKA), phospholipase C (PLC), or pro- 2000), which are involved in the affective and motivated processes
tein kinase C (PKC) inhibitors to the VTA attenuates the enhancing of focus in this review. Indeed, the reader is referred to other
effects of GABAA or D1 agonists for 3α,5α-THP-facilitated lor- interesting and recent reviews on the role of neurosteroidoge-
dosis (Fáncsik et al., 2000; Frye et al., 2004b, 2006b,d; Petralia nesis for functional processes (this volume; Rupprecht et al.,

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 6


Frye et al. 3α,5α-THP and PXR motivated behaviors

2010; Luchetti et al., 2011; Papadopoulos, 2011; Schüle et al., infusions and assessment in the test battery. The test battery con-
2011). sisted of rats being assessed in the open field and elevated plus maze
for anxiety-like behavior, the social interaction and social choice
P4 ’s EFFECTS IN THE VTA REQUIRE FORMATION OF 3α,5α-THP tasks for social behavior, and paced mating for reproductive behav-
In the VTA, P4 facilitates lordosis subsequent to formation of ior. Infusions of 3α,5α-THP to the VTA of diestrous rats enhanced
3α,5α-THP. Mating in rodents coincides with ovulation, when exploratory, anti-anxiety, social, and lordosis akin to that of proe-
circulating and midbrain levels of P4 and 3α,5α-THP are ele- strous rats infused with vehicle (Frye and Rhodes, 2008). 3α,5α-
vated. The frequency of lordosis is greater among rats that have THP infusions to the VTA of OVX rats enhanced exploratory,
higher 3α,5α-THP in the midbrain, such as those in proestrus anti-anxiety, and social behavior, independent of E2 , whereas lor-
or OVX rats administered subcutaneous injections of E2 with P4 dosis required E2 -priming for its full expression (Frye et al., 2006a,
or 3α,5α-THP, compared to those in diestrus, older 12- to 14- 2008b; Frye and Rhodes, 2007a). Interestingly, infusions of 3α,5α-
month-old rats, or OVX rats administered vehicle or E2 + P4 and THP to the VTA resulted in higher levels of 3α,5α-THP in the
an inhibitor of 3α,5α-THP formation (Frye and Leadbetter, 1994; midbrain VTA, as well as the hippocampus, cortex, and striatum.
Frye and Gardiner, 1996; Frye and Vongher, 1999; Frye, 2001a,b; Infusions of 3α,5α-THP to nearby sites (substantia nigra, central
Frye and Walf, 2004; Frye et al., 2004b; Petralia et al., 2005; Walf gray) neither significantly altered behaviors, nor 3α,5α-THP lev-
et al., 2011). Moreover, findings from a mutant mouse model sup- els, in these other regions. Thus, manipulating 3α,5α-THP in the
port these data. Female wild-type, or 5α-reductase knockout mice, midbrain VTA enhances exploratory, anxiety, and social respond-
which have perturbed function of the type II isoform of the 5α- ing, and elicits progestogen biosynthesis in other areas which may
reductase enzyme, were OVX, E2 -primed, and administered P4 modulate some of these functional changes, independent of E2.
(125, 250, or 500 μg, SC) or vehicle oil and assessed for social
responding. P4 dose-dependently enhanced lordosis among wild- DYNAMIC CONSEQUENCES OF AFFECTIVE AND SOCIAL RESPONDING
type, but not 5α-reductase knockout, mice (Figure 1). A similar ON ENDOGENOUS 3α,5α-THP
effect was observed for anxiety behavior among proestrous mice Beyond being necessary to alter affective and motivated responses,
or those that were hormone-primed (Koonce et al., 2012). These 3α,5α-THP levels in the midbrain are particularly dynamic and
data support the notion that P4 ’s metabolism to 3α,5α-THP is increase with challenges, such as social responding. In sup-
critical for normative affective and motivated behaviors among port, following mating, midbrain 3α,5α-THP levels are increased
female rodents. over those of non-mated, naturally receptive, or E2 + P4 -primed
rodents (Frye, 2001a,b). The rapidity of this increase in mid-
3α,5α-THP, INDEPENDENT OF E2 , IS NECESSARY AND SUFFICIENT IN brain 3α,5α-THP, and independence of secretion from the
THE VTA TO MEDIATE AFFECTIVE AND MOTIVATED BEHAVIORS ovaries and/or adrenals, suggests that biosynthesis and, subse-
To address whether actions of 3α,5α-THP in the midbrain VTA quent, metabolism, of central, rather than peripheral, progesto-
can influence exploratory, affective, and social behavior of rats, gens underlie these increases that we have observed among rats,
proestrous, or diestrous rats were infused with a physiological mice, and hamsters (Frye, 2001a,b). Increases in midbrain 3α,5α-
regimen of 3α,5α-THP (100 ng/μl) or β-cyclodextrin vehicle to THP with E2 , P4 , and/or social responding suggest that 3α,5α-THP
the VTA (or nearby missed sites, the substantia nigra, and central in varying concentrations, or when derived from peripheral versus
gray) and were assessed in a test battery. Additionally, the role of central prohormones, may influence sexually dimorphic processes,
E2 , given that E2 typically co-varies with progestogens and can such as affective behavior, as well as social and sexual behavior.
enhance 3α,5α-THP biosynthesis (Cheng and Karavolas, 1973),
was assessed among OVX rats that were E2 -primed or not before ESTABLISHMENT OF WHETHER PACED MATING UNDERLIES
ENHANCEMENT OF 3α,5α-THP IN THE BRAIN
In order to elucidate the role of behavioral processes on central
levels of 3α,5α-THP, proestrous, or diestrous rats were allowed
to behave in the entire battery of tasks with or without engag-
ing in paced mating. This experiment revealed the dynamic role
3α,5α-THP plays in the midbrain. While proestrous rats had
greater 3α,5α-THP levels in serum and all brain regions stud-
ied than diestrous rats, irrespective of mating condition, only
diestrous rats that engaged in paced mating had enhanced 3α,5α-
THP levels in midbrain compared to non-mated diestrous rats
(Figure 2, top; Frye and Rhodes, 2006b). In follow-up experi-
ments, proestrous rats were allowed to engage in portions of the
battery (exploratory/anxiety tasks only or social tasks only or no
tasks), or engaged in only one task in the battery, with or with-
FIGURE 1 | Lordosis is enhanced dose-dependently when out paced mating. Irrespective of portion (Figure 2, middle), or
progesterone (P4 ) is administered (SC) to wild-type mice but not those
individual task engaged in (Figure 2, bottom), only paced mat-
deficient in 5α-reductase (KO). ∗ Indicates p < 0.05 differences from
wild-type mice administered vehicle. ing was associated with 3α,5α-THP enhancement in every brain
region (but not serum) examined (Frye et al., 2007). As such, these

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 7


Frye et al. 3α,5α-THP and PXR motivated behaviors

FIGURE 2 | 3α,5α-THP levels are greater among proestrous rats in serum, social (partner preference and social interaction) tasks compared to
midbrain, hippocampus, diencephalon, and cortex than diestrous rats non-mated proestrous rats (middle). Among all tasks, only paced mating
irrespective of mating condition (top). 3α,5α-THP levels are enhanced in all enhances 3α,5α-THP levels of proestrous rats compared to non-mated
brain regions examined among proestrous rats that engage in paced mating proestrous rats. Line indicates performance of non-mated diestrous (top) or
irrespective of exposure to exploratory (open field and elevated plus maze) or proestrous (middle, bottom) control. ∗ Indicates p < 0.05.

experiments revealed that engaging in paced mating uniquely pro- rewarding and can be utilized to condition a place preference
duces progestogen biosynthesis in midbrain and hippocampus, among female rats (Frye et al., 1998; Martínez and Paredes, 2001).
and to a lesser extent the striatum and cortex (Frye and Rhodes, 3α,5α-THP has likewise been found to have hedonic effects. For
2007b). This complements findings that exposure to early life stres- instance, rats will preferentially self-administer 3α,5α-THP over
sors can alter neurosteroid formation in hippocampus and stress water (Sinnott et al., 2002). To further explore 3α,5α-THP’s effects
responses mediated by the hippocampus (Frye et al., 2006e). Thus, associated with rewarding processes, we assessed effects of cocaine
engaging in motivated, social responding can dynamically alter on progestogen concentrations in brain. We found that a sin-
progestogen synthesis in brain. gle injection (5 mg/kg intraperitoneal-IP) of cocaine to female
rats increased P4 and 3α,5α-THP concentrations in hippocampus,
3α,5α-THP FORMATION AND REWARD PROCESSES striatum, and circulation (Figure 3; Frye, 2007; Quiñones-Jenab
Central biosynthesis of 3α,5α-THP in response to environmental et al., 2008). These data support the notion that HPA-activating
stimuli may be a function of HPA regulation. 3α,5α-THP can act as stimuli may have rewarding effects associated with 3α,5α-THP
an endogenous modulator of the stress axis. 3α,5α-THP increases enhancement in brain.
in response to extreme stressors, such as foot-shock, ether expo-
sure, or cold-water swim (Purdy et al., 1991; Barbaccia et al., 1996). 3α,5α-THP BIOSYNTHESIS IN VTA IS ESSENTIAL FOR ENHANCED
3α,5α-THP has central actions to dampen HPA para/sympathetic AFFECTIVE AND MOTIVATED BEHAVIOR
physiological responses, in addition to its anxiolytic psychological Whether enhanced 3α,5α-THP in the VTA was due to central
effects (Patchev et al., 1994, 1996). Our data suggest that central biosynthesis, or dependent on ovarian sources, was of inter-
3α,5α-THP can be enhanced in response to a much more moderate est. Proestrous rats were infused with inhibitors of 3α,5α-THP
stressor, such as paced mating. Paced mating in particular is formation to the VTA. Rats received VTA infusions of PK11195

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 8


Frye et al. 3α,5α-THP and PXR motivated behaviors

CHANGES IN GENE EXPRESSION WITH MATING


Pharmacological studies discussed above are corroborated by
experiments examining differences in gene expression in the
midbrain of naturally receptive rats that underwent paced mat-
ing or did not have this social experience. Among mated rats,
genes that were upregulated in the midbrain were primarily
related to those substrates that our past pharmacological studies
have elucidated as targets for progestogens’ to influence lordo-
sis. First, of the approximately 40 genes that were upregulated in
mated rats, many are relevant to downstream intracellular sig-
naling pathways involved in non-genomic action (Paris et al.,
2011). For example, there was upregulation of three genes (Calb3-
increased 32.0-fold, Ascl1- increased 7.3-fold, DRd2- increased
2.0-fold) involved in regulating dopamine activity in midbrain.
Ascl1 encodes for a protein that mediates neurogenesis and dif-
ferentiation of tyrosine hydroxylase-containing neurons during
development. Calb3 encodes for calbindin 3 and, calbindin can
modulate depolarization of dopamine cells. Drd2 encodes the
dopamine type 2 receptor, which is a known autoreceptor that may
FIGURE 3 | Progesterone (P4 ; top) and 3α,5α-THP (bottom) levels are modulate activity of dopamine cell bodies in the VTA. There was
increased in serum, hippocampus, or diencephalon of female rats
administered cocaine (5 mg/kg, IP) compared to saline. ∗ Indicates
increased expression of genes that have implications for G-protein
p < 0.05. activity (i.e., guanosine-5 -diphosphate (GDP) and guanosine-
5 -triphosphate (GTP)- associated proteins), which substantiates
that G-protein activity in the VTA is involved in progestogens’
actions. Expression of two forms of ram, which encode for GTP
(400 ng/μl), which inhibits 3α,5α-THP formation from choles- binding proteins, were increased 2.3- and 2.0-fold and expres-
terol, or were infused with indomethacin (10 μg/μl), which blocks sion of RAB3d, which encodes the GDP/GTP exchange protein,
DHP metabolism to 3α,5α-THP, or received both inhibitors, or was 2.5 times greater in mated versus non-mated rats. Sec-
were infused with β-cyclodextrin vehicle and behaviorally assessed ond, mating induces 3α,5α-THP biosynthesis and many genes
in the affective and social responding battery. Infusions of any that were upregulated were those involved in steroid metabo-
combination of inhibitor significantly attenuated midbrain 3α,5α- lism (e.g., Fshb, Lhb, and Giot1). Third, genes involved in cell
THP levels of proestrous rats concomitant with reductions in proliferation and cell death were upregulated in the midbrain
exploratory, anti-anxiety, social, and reproductive behavior (Frye VTA of mated versus non-mated rats. These findings support
et al., 2008a). In order to assess whether central 3α,5α-THP is nec- a great deal of our prior research that has demonstrated a role
essary and sufficient for these effects, proestrous rats were infused of steroid biosynthesis and actions at GABAergic, glutamater-
with all combinations of inhibitors described, or vehicle, and were gic, dopaminergic substrates, and/or downstream signaling fac-
subsequently infused with a physiological dosage of 3α,5α-THP tors. Thus, mating alters expression of genes associated with
(100 ng/μl) to the VTA. Proestrous rats that were infused with steroid biosynthesis and non-traditional steroid actions in rat
3α,5α-THP subsequent to inhibitor infusions had a reinstatement midbrain.
of exploratory, anti-anxiety, and social behavior that was commen-
surate to that of vehicle-infused controls (Frye and Paris, 2009). In PXR, AN ENDOGENOUS TARGET OF 3α,5α-THP, MAY UNDERLIE
a follow-up study, effects of infusion of a neurosteroid enhancer BIOSYNTHESIS IN RESPONSE TO MATING
(FGIN 1,27; 5 μg/μl) following TSPO (PK11195, 400 ng/μl) or The data discussed to this point suggest that 3α,5α-THP plays
3α-HSD (indomethacin, 10 μg/μl) inhibitor infusion was assessed a causal role in mediating expression of affective and motivated
and revealed that enhancement of central biosynthesis via TSPO behaviors. Further, paced mating is one behavioral stimulus that
could overcome effects of 3α,5α-THP inhibitors on these behav- underlies 3α,5α-THP biosynthesis in brain regions, such as the
iors, as well as midbrain 3α,5α-THP levels (Frye et al., 2009). In midbrain VTA, cortex, hippocampus, and striatum, that are rele-
this study, it may be that FGIN 1,27 outcompeted PK11195 at vant for these behaviors. Other factors that remain to be elucidated
the TSPO in the VTA to overcome its inhibition and increase in this regulatory circuit within the midbrain VTA to modu-
3α,5α-THP levels. A question is whether FGIN 1,27 may have late affective and motivated behaviors are of great scientific and
had greater effects on DHP, compared to 3α,5α-THP, follow- clinical interest. The data that support involvement of a novel
ing indomethacin administration levels given cross-reactivity of pregnane mechanism, the pregnane xenobiotic receptor (PXR),
these steroids in the radioimmunoassay utilized. Despite these that has actions to modulate steroid synthesis within the brain are
considerations that need to be addressed, these data suggest that discussed as follows.
central biosynthesis of 3α,5α-THP in VTA is necessary and suffi- 3α,5α-THP is an endogenous positive activator of PXR. The
cient to enhance expression of affective/motivated responding in PXR receptor is a nuclear receptor that acts as a transcription fac-
proestrous rats. tor, and facilitates the expression of several major families of genes.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 9


Frye et al. 3α,5α-THP and PXR motivated behaviors

Genes of particular relevance are the cytochrome P450 (CYP) appropriate species-specific biotinylated 2˚ antibodies to deter-
enzymes (involved in steroid and neurosteroid biosynthesis, as mine the ideal concentration of antibodies. Blots were incubated
well as metabolism of a broad array of drugs), and of ATP-binding in Vector Duolux Reagent (Vector Labs), which binds to the sec-
cassette transporters, which also have broad functions in several ondary antibodies to produce a chemiluminescent peroxidase
sites, including the blood–brain barrier. The rodent PXR is analo- reaction that was observed following exposure to film. The rep-
gous to the steroid and xenobiotic receptor in humans, a.k.a. the resentative results of these dot blots are depicted in Figure 5;
human-PXR (Xie and Evans, 2002). Enhancing gestational levels Table 1.
of 3α,5α-THP can increase whole brain expression of PXR mRNA We observed expression of PXR, StAR, P450scc, 5α-reductase,
in offspring (Mellon et al., 2008). While PXR is most often studied and 3α-HSD protein in the midbrain. We have more recently
in the liver owing to its integral role in metabolism, PXR has been investigated whether there are differences in expression of PXR
found using real-time quantitative PCR (qPCR) and Western Blot- in diestrous and proestrous rats (Frye et al., 2010). These experi-
ting in the rabbit cortex, midbrain, and cerebellum (Marini et al., ments have shown that rats in proestrus have higher mRNA and
2007), rat brain capillaries (Bauer et al., 2004), and various regions protein expression of PXR in the midbrain than do diestrous
of the human brain (Lamba et al., 2004). Moreover, we have found rats (Frye et al., 2010). Indeed, mRNA and/or protein for PXR,
that PXR is expressed in the midbrain of rats, as described as StAR, P450scc, 3β-HSD, 5α-reductase, and 3α-HSD are present
follows. in the rat midbrain, and PXR expression is altered by hormonal
We have recently conducted microarray analyses on flash frozen status.
midbrain tissue of rats that were paced mated or not mated.
mRNA was isolated from midbrain tissues, reverse transcribed Manipulating PXR in the midbrain alters affective and motivated
into labeled cDNAs, hybridized, and used to probe the Affymetrix behaviors
GeneChip Rat 230 2.0 arrays per Affymetrix protocol. Analysis We have begun to assess the functional effects of PXR in the VTA
showed the presence of PXR gene in rat midbrain VTA (Table 1). for affective and motivated behaviors. In one study, we compared
We have gone on to confirm this microarray data and taken it a the effects of PXR ligands to the VTA of OVX rats. In this study,
step further to investigate whether PXR RNA and protein, as well OVX, E2 -primed rats were stereotaxically implanted with bilat-
as the protein and RNA from downstream metabolism enzymes eral guide cannulae aimed at the VTA. Rats were infused with
involved in 3α5α-THP formation that may be regulated by PXR, β-cyclodextrin vehicle or a positive modulator of PXR (3α,5α-
are present in the VTA. We have used qPCR to confirm the pres- THP, 3α,5β-THP, 3β,5α-THP, or RU-486) and then tested in the
ence of PXR, StAR, P450scc, 3α-HSD, and 5α-reductase seen in paced mating task 10 min later. Infusions of the PXR-positive
our microarray studies. We isolated total RNA from flash frozen modulators, compared to vehicle, increased lordosis responding
rat midbrain tissue using Tri-Reagent (Ambion). We then used (Frye, 2011).
the Ambion MicroPoly(A)Pure™(Ambion) and the iScript Select Although the data above imply that activating PXR in the mid-
RT (reverse transcriptase) kit (Bio-Rad) to generate cDNAs from brain VTA may facilitate lordosis, the effects of knocking down
mRNA. Primers for PCR were designed to differentiate between PXR in the VTA are of interest. To further assess the role of PXR
cDNAs and genomic contamination (we observed no contami- in the VTA for affective and motivated behavior, we infused OVX,
nation). We found that PXR as well as StAR, P450scc, 3α-HSD, E2 -primed (10 μg) rats with either a PXR antisense oligodeoxynu-
and 5α-reductase mRNAs are all present in the rat midbrain VTA cleotides (ODN; 5 CTTGCGGAAGGGGCACCTCA 3 ; 250 ng)
(Figure 4; Table 1). or a scrambled mis-sense ODN (5 CTCCGAAACGGACATCTGA
To see if these mRNAs are translated into protein, we used 3 ; 250 ng), or saline vehicle, bilaterally to the VTA. ODNs were
dot blots and antibody staining to look for protein expression infused 44, 24, and 0 h prior to testing in the elevated plus maze
in the rat midbrain VTA. For these studies, we homogenized and paced mating tasks. The site-specificity for the effects of
flash frozen rat midbrain VTA tissue in NuPAGE® sample buffer these manipulations was determined. Brains of OVX, E2 -primed
(1X; Invitrogen), at a concentration of 1 mg tissue/100 μl buffer. rats that had scrambled ODNs or PXR antisense ODNs infused
Samples were then dot blotted onto nitrocellulose by pipetting to the VTA were immediately collected after behavioral testing,
2 μl of homogenized sample in sample buffer onto the mem- flash frozen on dry ice, and stored at −80˚C until prepared
brane. We then probed the blots with different concentrations for western blotting analyses. Tissues have only been analyzed
(1:1000–1:5000) of 1˚ antibodies to PXR, StAR, P450scc, or 5α- to date for those with confirmed infusions to the VTA. Briefly,
reductase (all from Santa Cruz Biotechnology), and 3α-HSD, then tissues were dissected by one of two methods. First, the block

Table 1 | Expression confirmed in midbrain VTA of proestrous rats for pregnane xenobiotic receptor (PXR) and biosynthesis and metabolism
proteins/enzymes required for 3α,5α-THP formation [steroid acute regulatory protein (StAR), P450 side chain cleavage enzyme (P450scc),
5α-reductase, and 3α-hydroxysteroid dehydrogenase (3α-HSD)].

PXR StAR P450scc 5α-Reductase 3α-HSD

√ √ √ √ √
mRNA on microarray
√ √ √ √ √
mRNA confirmed with qPCR
√ √ √ √ √
Protein on westerns

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 10


Frye et al. 3α,5α-THP and PXR motivated behaviors

of midbrain tissue (inclusive of red nucleus, interpeduncular hypothesis that PXR is critical for 3α,5α-THP formation in this
nucleus, substantia nigra) was grossly dissected (typical weight region.
25–30 mg). Second, brains were sectioned anterior and poste- Rats infused with the antisense ODN spent significantly less
rior to the infusion site and midbrain VTA was “punched out” time on the open arms of the elevated plus maze, indicat-
(typical weight 2–3 mg of tissue). PXR expression was deter- ing less anti-anxiety behavior compared to controls (Figure 7).
mined in midbrain or VTA tissue via western blotting using the Additionally, rats infused with PXR antisense ODN to the VTA
same general techniques described above for tissue preparation, spent less time in social interaction with a conspecific and demon-
antibody incubation, blocking, and visualization. Specific to this strated less lordosis compared to controls, indicating less pro-
experiment, to optimize concentrations of primary and secondary social and motivated, and reproductive behavior among these
antibodies for PXR, dot blot analyses on positive control tis- rats (Figure 8). Infusions outside the VTA did not produce the
sues (liver) were conducted first. These blots were blocked in same effects (Table 2). Together with the western blotting data,
5% milk PBS-10% tween and then incubated in PXR mouse 1˚ these findings demonstrate that PXR knock-down can be achieved
(1:1000–1:5000; Santa Cruz Biotechnology) and HRP conjugated locally in the VTA and that reduction of PXR protein in this area
goat anti-mouse 2˚ (1:5000–1:20000; Bio-Rad) antibodies. This is sufficient to attenuate 3α,5α-THP-dependent anti-anxiety and
experiment determined that the ideal concentration for 1˚ PXR social behavior.
antibody was 1:3000 and 2˚ was 1:5000. These concentrations
were then used to determine PXR protein concentrations in mid-
brain tissue samples with typical gel electrophoresis separation
and transfer to nitrocellulose (as described in Frye, 2011). VTA
punches, but not gross midbrain dissections, demonstrated that
PXR expression was reduced following infusions of the PXR anti-
sense ODNs compared to scrambled control infusions (Figure 6).
These data support the notion that VTA is a central area within
the midbrain underlying neurosteroidogenesis of 3α,5α-THP and
that PXR’s effects on associated behaviors are site-specific and
relegated to the VTA. These findings are congruous with the

FIGURE 6 | Western blots of pregnane xenobiotic receptor (PXR)


expression (top) and β-actin control (bottom) in the whole midbrain
(left) and punch VTA infusion site (right) of rats administered
scrambled control or PXR antisense oligodeoxynucleotides (ODNs).

FIGURE 4 | Agarose gels to visualize bands from qPCR confirming that


mRNAs for pregnane xenobiotic receptor (PXR) and its possible
downstream effectors, steroidogenic acute regulatory protein (StAR),
P450 side chain cleavage enzyme (P450scc), 5α-reductase (5α-red), and
3α-hydroxysteroid dehydrogenase (3α-HSD) are expressed in the rat
midbrain. Note: gels depicted are those run separately for each of these
targets.

FIGURE 5 | Dot blots demonstrating the presence of pregnane


xenobiotic receptor (PXR), steroidogenic acute regulatory protein FIGURE 7 | Affective behavior in the elevated plus maze of E2 -primed
(StAR), P450 side chain cleavage enzymes (P450scc), 5α-reductase, and rats administered scrambled control or pregnane xenobiotic receptor
3α-hydroxysteroid dehydrogenase (3α-HSD) in the rat midbrain VTA. (PXR) antisense oligodeoxynucleotides (ODNs) to the midbrain VTA.
NP indicates no protein. ∗ Indicates different from all groups, p < 0.05.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 11


Frye et al. 3α,5α-THP and PXR motivated behaviors

of PXR antisense oligonucleotides were effective in knocking


down PXR protein and mRNA expression in the VTA (Frye
et al., 2010). Moreover, PXR antisense oligonucleotides to the
VTA of proestrous rats significantly reduced 3α,5α-THP lev-
els in the midbrain coincident with these behavioral changes.
Thus, PXR may be necessary for 3α,5α-THP formation in the
VTA, and its subsequent effects on affective and motivated
behaviors.
Together, these preliminary findings demonstrate that antisense
ODNs infused to the midbrain VTA knock-down PXR expres-
sion locally in the VTA and that this local PXR attenuation is
necessary for expression of anti-anxiety and motivated, social
behavior among OVX E-primed rats. Of continued interest in
the laboratory is the precise role of PXR. PXR expression studies
suggest that there may be positive feedback from gonadal hor-
mones and/or 3α,5α-THP on PXR in the midbrain, which in turn
regulates downstream enzymes required for 3α,5α-THP forma-
tion. Interestingly, these results suggest that there may be some
genomic signaling in the midbrain for non-traditional actions
of 3α,5α-THP, but that these occur through PXR, rather than
PRs (as discussed in see Mechanisms of 3α,5α-THP for Affect
and Motivated Behaviors). The extent to which these effects
are mediated by 3α,5α-THP, and the functional role of PXR
in this system, are currently being investigated further in our
laboratory.

SUMMARY
FIGURE 8 | Social behavior (top: social interaction; bottom: lordosis) of To summarize, the neurosteroid, 3α,5α-THP, has myriad effects,
E2 -primed rats administered scrambled control or pregnane xenobiotic mechanisms, and sources beyond the typically described actions
receptor (PXR) antisense oligodeoxynucleotides (ODNs) to the of progestogens. The VTA is a central target of progestogens for
midbrain VTA. ∗ Indicates different from all groups, p < 0.05.
their effects and mechanisms related to affective, motivated, and
reward processes. A compelling question is the source of progesto-
Table 2 | Data from missed-site controls do not demonstrate reliable gens in the VTA for these processes. A focus has been on the role
difference between rats infused with scrambled of PXR in the midbrain VTA. The midbrain VTA expresses RNA
oligodeoxynucleotides (ODN) versus antisense ODN in elevated plus and proteins for PXR and downstream metabolism enzymes that
maze (open arm time), social interaction (interaction time), or paced are rate-limiting factors for 3α,5α-THP production. Infusions of
mating (lordosis frequency). positive modulators of PXR to the VTA facilitate sexual behav-
ior. Infusions of antisense ODNs targeted against PXR to the VTA
Behavioral measure Missed-site infusate inhibit anti-anxiety and sexual behavior of rats. Together, these
data support the idea that PXR is expressed in the midbrain VTA
Vehicle Scrambled PXR and that it may have a functionally relevant role in the affective
ODN ASODN and social behaviors examined to date. Indeed, PXR may be a
Time on open arm (s ± SEM) 31 ± 7 26 ± 13 41 ± 2 powerful mechanism involved in the modulation of neurosteroid
Interaction time (s ± SEM) 115 ± 10 101 ± 20 99 ± 5 effects, which may mediate diverse human behaviors and clinical
Lordosis frequency (mean ± SEM) 30 ± 18 33 ± 13 27 ± 12 conditions (e.g., anxiety, depression, drug abuse). The previous
studies by our lab have elucidated a critical role of 3α,5α-THP in
the VTA in mediating a number of affective and social behaviors in
To further assess the importance of PXR in neurosteroid- rats. The ongoing focus of our lab is the conditions under which
enhanced lordosis, an experiment was conducted in diestrous and biosynthesis of 3α,5α-THP is initiated, and how these processes
proestrous rats infused with PXR antisense or control conditions result from environmental stimuli, behaviors and/or drugs, and
to the VTA, using the methods described above. In this exper- involve PXR.
iment, proestrous rats infused with antisense oligonucleotides
against PXR had reduced anti-anxiety and pro-social behavior, ACKNOWLEDGMENTS
compared to rats infused with control conditions (Frye et al., This research was supported by grants from the National Institute
2010). PXR antisense oligonucleotides to the VTA of diestrous of Mental Health (MH0676980; RMH067698B). Assistance pro-
rats did not alter these behavioral endpoints. Western blot analy- vided by Dr. Sridar Chittur, Alima DeVerteuil, Amy Kohtz, Carolyn
ses and qPCR demonstrated that these infusions to the VTA Koonce, and Jennifer Torgersen is appreciated.

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 12


Frye et al. 3α,5α-THP and PXR motivated behaviors

REFERENCES Barbaccia, M. L., Roscetti, G., Trabuc- ovarian steroid hormone-sensitive to 5α-pregnane-3,20-dione and 3α-
Abel, K. M., Drake, R., and Goldstein, J. chi, M., Purdy, R. H., Mostallino, M. neurons involved in female sexual hydroxy-5α-pregnan-20-one by rat
M. (2010). Sex differences in schiz- C., Concas, A., and Biggio, G. (1997). behavior. Psychoneuroendocrinology medical basal hypothalami and the
ophrenia. Int. Rev. Psychiatry 22, The effects of inhibitors of GABAer- 19, 505–516. effects of estradiol and stage of
417–428. gic transmission and stress on brain Boyson, S. J., McGonigle, P., and Moli- estrous cycle on the conversion.
Altemus, M., Redwine, L. S., Leong, and plasma allopregnanolone con- noff, P. B. (1986). Quantitative Endocrinology 93, 1157–1162.
Y. M., Frye, C. A., Porges, S. W., centrations. Br. J. Pharmacol. 120, autoradiographic localization of the Clifford, G. M., and Farmer, R. D.
and Carter, C. S. (2001). Responses 1582–15288. D1 and D2 subtypes of dopamine (2002). Drug or symptom-induced
to laboratory psychosocial stress Barbaccia, M. L., Serra, M., Purdy, R. receptors in rat brain. J. Neurosci. 6, depression in men treated with α1-
in postpartum women. Psychosom. H., and Biggio, G. (2001). Stress 3177–3188. blockers for benign prostatic hyper-
Med. 63, 814–821. and neuroactive steroids. Int. Rev. Brady, K. T., and Randall, C. L. (1999). plasia? A nested case-control study.
Altomare, G., and Capella, G. L. (2002). Neurobiol. 46, 243–272. Gender differences in substance use Pharmacoepidemiol. Drug Saf. 11,
Depression circumstantially related Bauer, B., Hartz, A. M., Fricker, G., disorders. Psychiatr. Clin. North Am. 55–61.
to the administration of finasteride and Miller, D. S. (2004). Preg- 22, 241–252. Compagnone, N. A., and Mellon, S. H.
for androgenetic alopecia. J. Derma- nane X receptor up-regulation of P- Braestrup, C., and Squires, R. F. (1977). (2000). Neurosteroids: biosynthesis
tol. 29, 665–669. glycoprotein expression and trans- Specific benzodiazepine receptors and function of these novel neuro-
Angold, A., Costello, E. J., Erkanli, port function at the blood-brain bar- in rat brain characterized by modulators. Front. Neuroendocrinol.
A., and Worthman, C. M. (1999). rier. Mol. Pharmacol. 66, 413–419. high-affinity [3H]diazepam bind- 21, 1–56.
Pubertal changes in hormone levels Baulieu, E. E. (1991). Neurosteroids: a ing. Proc. Natl. Acad. Sci. U.S.A. 74, Cyr, M., Ghribi, O., and Di Paolo, T.
and depression in girls. Psychol. Med. new function in the brain. Biol. Cell 3805–3809. (2000). Regional and selective effects
29, 1043–1053. 71, 3–10. Brambilla, F., Biggio, G., Pisu, M. G., of oestradiol and progesterone on
Anker, J. J., Holtz, N. A., Zlebnik, N., and Baulieu, E. E. (1980). Steroid hormone Bellodi, L., Perna, G., Bogdanovich- NMDA and AMPA receptors in the
Carroll, M. E. (2009). Effects of allo- receptors. Expos. Annu. Biochim. Djukic, V., Purdy, R. H., and Serra, rat brain. J. Neuroendocrinol. 12,
pregnanolone on the reinstatement Med. 34, 1–25. M. (2003). Neurosteroid secretion in 445–452.
of cocaine-seeking behavior in male Becker, J. B., and Cha, J. H. (1989). panic disorder. Psychiatry Res. 118, De Leo, V., la Marca, A., Talluri,
and female rats. Psychopharmacology Estrous cycle-dependent variation 107–116. B., D’Antona, D., and Morgante,
(Berl.) 203, 63–72. in amphetamine-induced behav- Brot, M. D.,Akwa,Y., Purdy, R. H., Koob, G. (1998). Hypothalamo-pituitary-
Anker, J. J., Zlebnik, N. E., and iors and striatal dopamine release G. F., and Britton, K. T. (1997). The adrenal axis and adrenal func-
Carroll, M. E. (2010). Differen- assessed with microdialysis. Behav. anxiolytic-like effects of the neu- tion before and after ovariectomy
tial effects of allopregnanolone on Brain Res. 35, 117–125. rosteroid allopregnanolone: interac- in premenopausal women. Eur. J.
the escalation of cocaine self- Belelli, D., and Lambert, J. J. (2005). tions with GABA(A) receptors. Eur. Endocrinol. 138, 430–435.
administration and sucrose intake Neurosteroids: endogenous regula- J. Pharmacol. 325, 1–7. DeBold, J. F., and Frye, C. A. (1994).
in female rats. Psychopharmacology tors of the GABAA receptor. Nat. Brown, G. W., Harris, T. O., and Hep- Genomic and non-genomic actions
(Berl.) 212, 419–429. Rev. Neurosci. 6, 565–575. worth, C. (1994). Life events and of progesterone in the control of
Bäckström, T., Andersson, A., Andreé, Benavides, J., Quarteronet, D., Imbault, endogenous depression. A puzzle female hamster sexual behavior.
L., Birzniece, V., Bixo, M., Björn, I., F., Malgouris, C., Uzan, A., Renault, reexamined. Arch. Gen. Psychiatry Horm. Behav. 28, 445–453.
Haage, D., Isaksson, M., Johansson, C., Dubroeucq, M. C., Gueremy, C., 51, 525–534. Dong, E., Matsumoto, K., Uzunova, V.,
I. M., Lindblad, C., Lundgren, P., and Le Fur, G. (1983). Labelling Carpenter, M. J., Upadhyaya, H. P., Sugaya, I., Takahata, H., Nomura,
Nyberg, S., Odmark, I. S., Strömberg, of “peripheral-type” benzodiazepine LaRowe, S. D., Saladin, M. E., and H., Watanabe, H., Costa, E., and
J., Sundström-Poromaa, I., Turk- binding sites in the rat brain by using Brady, K. T. (2006). Menstrual cycle Guidotti, A. (2001). Brain 5α-
men, S., Wahlström, G., Wang, M., [3H]PK 11195, an isoquinoline car- phase effects on nicotine withdrawal dihydroprogesterone and allopreg-
Wihlbäck, A. C., Zhu, D., and Zing- boxamide derivative: kinetic studies and cigarette craving: a review. Nico- nanolone synthesis in a mouse
mark, E. (2003). Pathogenesis in and autoradiographic localization. J. tine Tob. Res. 8, 627–638. model of protracted social isola-
menstrual cycle-linked CNS disor- Neurochem. 41, 1744–1750. Carroll, B. J., Curtis, G. C., and Mendels, tion. Proc. Natl. Acad. Sci. U.S.A. 98,
ders. Ann. N. Y. Acad. Sci. 1007, Bicikova, M., Putz, Z., Hill, M., Hampl, J. (1976). Neuroendocrine regula- 2849–2854.
42–53. R., Diebbelt, L., Tallova, J., and tion in depression. II. Discrimi- Dubrovsky, B. (2006). Neurosteroids,
Bäckström, T., Sanders, D., Leask, R., Starka, L. (1998). Serum levels of nation of depressed from nonde- neuroactive steroids, and symptoms
Davidson, D., Warner, P., and Ban- neurosteroid allopregnanolone in pressed patients. Arch. Gen. Psychi- of affective disorders. Pharmacol.
croft, J. (1983). Mood, sexuality, hor- patients with premenstrual syn- atry 33, 1051–1058. Biochem. Behav. 84, 644–655.
mones, and the menstrual cycle. II. drome and patients after thyroidec- Carroll, B. J., Greden, J. F., Feinberg, M., Endicott, J., Amsterdam, J., Eriksson, E.,
Hormone levels and their relation- tomy. Endocr. Regul. 32, 87–92. Lohr, N., James, N. M., Steiner, M., Frank, E., Freeman, E., Hirschfeld,
ship to the premenstrual syndrome. Bitran, D., Foley, M., Audette, D., Leslie, Haskett, R. F., Albala, A. A., DeVigne, R., Ling, F., Parry, B., Pearlstein,
Psychosom. Med. 45, 503–507. N., and Frye, C. A. (2000). Acti- J. P., and Tarika, J. (1981). Neuroen- T., Rosenbaum, J., Rubinow, D.,
Baghai, T. C., di Michele, F., Schüle, vation of peripheral mitochondr- docrine evaluation of depression in Schmidt, P., Severino, S., Steiner, M.,
C., Eser, D., Zwanzger, P., Pasini, ial benzodiazepine receptors in the borderline patients. Psychiatr. Clin. Stewart, D. E., and Thys-Jacobs, S.
A., Romeo, E., and Rupprecht, R. hippocampus stimulates allopreg- North Am. 4, 89–99. (1999). Is premenstrual dysphoric
(2005). Plasma concentrations of nanolone synthesis and produces Chaudron, L. H., Klein, M. H., Rem- disorder a distinct clinical entity? J.
neuroactive steroids before and after anxiolytic-like effects in the rat. Psy- ington, P., Palta, M., Allen, C., Womens Health Gend. Based Med. 8,
electroconvulsive therapy in major chopharmacology (Berl.) 151, 64–71. and Essex, M. J. (2001). Predictors, 663–679.
depression. Neuropsychopharmacol- Bitran, D., Hilvers, R. J., and Kel- prodromes and incidence of post- Epperson, C. N., Haga, K., Mason,
ogy 30, 1181–1186. logg, C. K. (1991). Anxiolytic effects partum depression. J. Psychosom. G. F., Sellers, E., Gueorguieva, R.,
Barbaccia, M. L., Roscetti, G., Trabuc- of 3α-hydroxy-5α[β]-pregnan-20- Obstet. Gynaecol. 22, 103–112. Zhang, W., Weiss, E., Rothman, D.
chi, M., Mostallino, M. C., Con- one: endogenous metabolites of Chen, M. K., and Guilarte, T. R. L., and Krystal, J. H. (2002). Corti-
cas, A., Purdy, R. H., and Biggio, G. progesterone that are active at the (2008). Translocator protein 18 kDa cal gamma-aminobutyric acid levels
(1996). Time-dependent changes in GABAA receptor. Brain Res. 561, (TSPO): molecular sensor of brain across the menstrual cycle in healthy
rat brain neuroactive steroid con- 157–161. injury, and repair. Pharmacol. Ther. women and those with premenstrual
centrations and GABAA receptor Blaustein, J. D., Tetel, M. J., Riccia- 118, 1–17. dysphoric disorder: a proton mag-
function after acute stress. Neuroen- rdi, K. H., Delville, Y., and Tur- Cheng, Y. J., and Karavolas, H. J. netic resonance spectroscopy study.
docrinology 63, 166–172. cotte, J. C. (1994). Hypothalamic (1973). Conversion of progesterone Arch. Gen. Psychiatry 59, 851–858.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 13


Frye et al. 3α,5α-THP and PXR motivated behaviors

Erskine, M. S., and Kornberg, E. 3α,5α-THP, enhance sexual motiva- does SKF38393. Psychobiology 28, are increased following paced
(1992). Stress and ACTH increase tion, receptivity, and proceptivity in 99–109. mating in midbrain, hippocam-
circulating concentrations of 3α- female rats. Brain Res. 808, 72–83. Frye, C. A., and Paris, J. J. (2009). pus, diencephalon, and cortex
androstanediol in female rats. Life Frye, C. A., and DeBold, J. F. (1992). Infusions of bicuculline to the ven- of rats in behavioral estrus, but
Sci. 51, 2065–2071. Muscimol facilitates sexual receptiv- tral tegmental area attenuates sexual, only in midbrain of diestrous
Eser, D., di Michele, F., Zwanzger, P., ity in hamsters when infused into exploratory, and anti-anxiety behav- rats. Neuroendocrinology 83,
Pasini, A., Baghai, T. C., Schüle, the ventral tegmentum. Pharmacol. ior of proestrous rats. Pharmacol. 336–347.
C., Rupprecht, R., and Romeo, Biochem. Behav. 42, 879–887. Biochem. Behav. 93, 474–481. Frye, C. A., and Rhodes, M. E. (2007a).
E. (2005). Panic induction with Frye, C. A., and Gardiner, S. G. (1996). Frye, C. A., and Paris, J. J. (2011a). Prog- “The role of midbrain 3α,5α-THP
cholecystokinin-tetrapeptide (CCK- Progestins can have a membrane- esterone turnover to its 5α-reduced in mediating exploration, anxiety,
4) Increases plasma concentrations mediated action in rat midbrain metabolites in the ventral tegmental social and reproductive behavior,”
of the neuroactive steroid 3α,5α- for facilitation of sexual receptivity. area of the midbrain is essential for in Neuroactive Steroids in Brain
tetrahydrodeoxycorticosterone (3α, Horm. Behav. 30, 682–691. initiating social and affective behav- Functions, Behavior, and Disorders:
5α-THDOC) in healthy volun- Frye, C. A., Koonce, C. J., Dacosta, D., ior and progesterone metabolism in Novel Strategies for Research and
teers. Neuropsychopharmacology 30, Torgersen, J., and Walf, A. A. (2010). female rats. J. Endocrinol. Invest. 34, Treatment, eds M. S. Ritsner and
192–195. “Knocking down expression of preg- 188–199. A. Weizman (Houten: Springer),
Fáncsik, A., Linn, D. M., and Tasker, nane xenobiotic receptor in the mid- Frye, C. A., and Paris, J. J. (2011b). 449–482.
J. G. (2000). Neurosteroid modu- brain ventral tegmental area attenu- Effects of neurosteroid actions at Frye, C. A., and Rhodes, M. E. (2007b).
lation of GABA IPSCs is phospho- ates estrous cycle variations in anx- N -methyl-d-aspartate and GABAA “Reciprocal effects of exploration,
rylation dependent. J. Neurosci. 20, iety, social, and reproductive behav- receptors in the midbrain ventral anxiety, social, and sexual behav-
67–75. ior of rats,” 2010 Neuroscience Meet- tegmental area for anxiety-like and iors and progestins in midbrain,
Freeman, E. W., Frye, C. A., Rickels, K., ing Planner (San Diego, CA: Society mating behavior of female rats. hippocampus, diencephalon, and
Martin, P. A., and Smith, S. S. (2002). for Neuroscience). Psychopharmacology (Berl.) 213, cortex,” in Evolutionary Molecu-
Allopregnanolone levels and symp- Frye, C. A., and Leadbetter, E. A. (1994). 93–103. lar Strategies and Plasticity, eds
tom improvement in severe premen- 5α-reduced progesterone metabo- Frye, C. A., Paris, J. J., and Rhodes, M. Canonaco and R. M. Fac-
strual syndrome. J. Clin. Psychophar- lites are essential in hamster VTA M. E. (2007). Engaging in paced ciolo (Kerala: Research Signpost),
macol. 22, 516–520. for sexual receptivity. Life Sci. 54, mating, but neither exploratory, 237–260.
Freeman, E. W., Sammel, M. D., Rin- 653–659. anti-anxiety, nor social behavior, Frye, C. A., and Rhodes, M. E.
audo, P. J., and Sheng, L. (2004). Pre- Frye, C. A., Marrone, J., and Walf, A. increases 5α-reduced progestin con- (2008). Infusions of 3α,5α-THP
menstrual syndrome as a predictor A. (2008a). Effects of manipulating centrations in midbrain, hippocam- to the VTA enhance exploratory,
of menopausal symptoms. Obstet. progesterone and NMDA receptors pus, striatum, and cortex. Reproduc- anti-anxiety, social, and sexual
Gynecol. 103, 960–966. in the ventral tegmental area for tion 133, 663–674. behavior and increase levels of
Frye, C. A. (2001a). The role of neu- lordosis of hamsters and rats. Psy- Frye, C. A., Paris, J. J., and Rhodes, 3α,5α-THP in midbrain, hippocam-
rosteroids and non-genomic effects chopharmacology (Berl.) 200, 71–80. M. E. (2009). Increasing 3α,5α- pus, diencephalon, and cortex of
of progestins in the ventral tegmen- Frye, C. A., Paris, J. J., and Rhodes, M. E. THP following inhibition of neu- female rats. Behav. Brain Res. 187,
tal area in mediating sexual recep- (2008b). Exploratory, anti-anxiety, rosteroid biosynthesis in the ven- 88–99.
tivity of rodents. Horm. Behav. 40, social, and sexual behaviors of rats in tral tegmental area reinstates anti- Frye, C. A., Rhodes, M. E., Petralia,
226–233. behavioral estrus is attenuated with anxiety, social, and sexual behavior S. M., Walf, A. A., Sumida, K.,
Frye, C. A. (2001b). The role of neuros- inhibition of 3α,5α-THP formation of naturally receptive rats. Reproduc- and Edinger, K. L. (2006a). 3α-
teroids and non-genomic effects of in the midbrain ventral tegmen- tion 137, 119–128. hydroxy-5α-pregnan-20-one in the
progestins and androgens in medi- tal area. Behav. Brain Res. 193, Frye, C. A., and Petralia, S. M. (2003a). midbrain ventral tegmental area
ating sexual receptivity of rodents. 269–276. Mitochondrial benzodiazepine mediates social, sexual, and affec-
Brain Res. Brain Res. Rev. 37, Frye, C. A., Mermelstein, P. G., and receptors in the ventral tegmental tive behaviors. Neuroscience 138,
201–222. DeBold, J. F. (1992). Evidence for area modulate sexual behaviour 1007–1014.
Frye, C. A. (2007). Progestins influence a non-genomic action of progestins of cycling or hormone-primed Frye, C. A., Walf, A. A., and Petralia,
motivation, reward, conditioning, on sexual receptivity in hamster ven- hamsters. J. Neuroendocrinol. 15, S. M. (2006b). Progestins’ effects
stress, and/or response to drugs of tral tegmental area but not hypothal- 677–686. on sexual behaviour of female rats
abuse. Pharmacol. Biochem. Behav. amus. Brain Res. 578, 87–93. Frye, C. A., and Petralia, S. M. (2003b). and hamsters involving D1 and
86, 209–219. Frye, C. A., Mermelstein, P. G., and Lordosis of rats is modified by GABA(A) receptors in the ventral
Frye, C. A. (2009). Neurosteroids-From DeBold, J. F. (1993). Bicuculline neurosteroidogenic effects of mem- tegmental area may be G-protein-
Basic Research to Clinical Perspec- infused into the hamster ventral brane benzodiazepine receptors in dependent. Behav. Brain Res. 2172,
tives, in Hormones/Behavior Rela- tegmentum inhibits, while sodium the ventral tegmental area. Neuroen- 286–293.
tions of Clinical Importance, eds R. valproate facilitates, sexual receptiv- docrinology 77, 71–82. Frye, C. A., Walf, A. A., and Petralia, S.
T. Rubin and D. W. Pfaff (San Diego: ity. Pharmacol. Biochem. Behav. 46, Frye, C. A., and Rhodes, M. E. (2006a). M. (2006c). In the ventral tegmen-
Academic Press), 395–416. 1–8. Infusions of 5α-pregnan-3α-ol-20- tal area, progestins have actions
Frye, C. A. (2011). Novel substrates Frye, C. A., Murphy, R. E., and Platek, one (3α,5α-THP) to the ventral at D1 receptors for lordosis of
for, and sources of, progestogens for S. M. (2000a). Anti-sense oligonu- tegmental area, but not the substan- hamsters and rats that involve
reproduction. J. Neuroendocrinol. cleotides, for progestin receptors tia nigra, enhance exploratory, anti- GABAA receptors. Horm. Behav. 50,
23, 961–973. in the VMH and glutamic acid anxiety, social and sexual behav- 332–337.
Frye, C. A., and Bayon, L. E. (1999). decarboxylase in the VTA, attenu- iours and concomitantly increase Frye, C. A., Walf, A. A., and Petralia,
Mating stimuli influence endoge- ate progesterone-induced lordosis in 3α,5α-THP concentrations in the S. M. (2006d). Progestin facilita-
nous variations in the neurosteroids hamsters and rats. Behav. Brain Res. hippocampus, diencephalon and tion of lordosis in rodents involves
3α,5α-THP and 3α-Diol. J. Neuroen- 115, 55–64. cortex of ovariectomised oestrogen- adenylyl cyclase activity in the ven-
docrinol. 11, 839–847. Frye, C. A., Bayon, L. E., and Vongher, primed rats. J. Neuroendocrinol. 18, tral tegmental area. Horm. Behav. 50,
Frye, C. A., Bayon, L. E., Pursnani, J. M. (2000b). Intravenous proges- 960–975. 237–244.
N. K., and Purdy, R. H. (1998). terone elicits a more rapid induc- Frye, C. A., and Rhodes, M. E. Frye, C. A., Rhodes, M. E., Raol,
The neurosteroids, progesterone and tion of lordosis in rats than (2006b). Progestin concentrations Y. H., and Brooks-Kayal, A. R.

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 14


Frye et al. 3α,5α-THP and PXR motivated behaviors

(2006e). Early postnatal stimulation Frye, C. A., Walf, A. A., and Sumida, Guidotti, A., and Costa, E. (1998). function during pregnancy and par-
alters pregnane neurosteroids in the K. (2004b). Progestins’ actions in Can the antidysphoric and anx- turition. Prog. Brain Res. 133, 39–47.
hippocampus. Psychopharmacology the VTA to facilitate lordosis involve iolytic profiles of selective sero- Hirani, K., Khisti, R. T., and Chopde,
(Berl.) 186. 343–350. dopamine-like Type 1 and 2 recep- tonin reuptake inhibitors be related C. T. (2002). Behavioral action of
Frye, C. A., and Seliga, A. M. (2003a). tors. Pharmacol. Biochem. Behav. 78, to their ability to increase brain ethanol in Porsolt’s forced swim
Effects of olanzapine infusions to the 405–418. 3α,5α-tetrahydroprogesterone (allo- test: modulation by 3α-hydroxy-
ventral tegmental area on lordosis Frye, C. A., and Wawrzycki, J. (2003). pregnanolone) availability? Biol. 5α-pregnan-20-one. Neuropharma-
and midbrain 3α,5α-THP concen- Effect of prenatal stress and gonadal Psychiatry 44, 865–873. cology 43, 1339–1350.
trations in rats. Psychopharmacology hormone condition on depressive Guidotti, A., Dong, E., Matsumoto, Holzbauer, M. (1975). Physiological
(Berl.) 170, 132–139. behaviors of female and male rats. K., Pinna, G., Rasmusson, A. M., variations in the ovarian produc-
Frye, C. A., and Seliga, A. M. Horm. Behav. 44, 319–326. and Costa, E. (2001). The socially- tion of 5α-pregnane derivatives with
(2003b). Olanzapine’s effects to Furukawa, A., Miyatake, A., Ohnishi, isolated mouse: a model to study the sedative properties in the rat. J.
reduce fear and anxiety and enhance T., and Ichikawa, Y. (1998). putative role of allopregnanolone Steroid Biochem. 6, 1307–1310.
social interactions coincide with Steroidogenic acute regulatory and 5α-dihydroprogesterone in Holzbauer, M. (1976). Proceedings:
increased progestin concentrations protein (StAR) transcripts con- psychiatric disorders. Brain Res. physiological aspects of the hypnotic
of ovariectomized rats. Psychoneu- stitutively expressed in the adult Brain Res. Rev. 37, 110–115. properties of steriod hormones. Br.
roendocrinology 28, 657–673. rat central nervous system: colo- Gulinello, M., Orman, R., and Smith, J. Pharmacol. 56, 382P–382P.
Frye, C. A., and Vongher, J. M. (1999). calization of StAR, cytochrome S. S. (2003). Sex differences in anx- Holzbauer, M., Birmingham, M. K., De
GABA(A), D1, and D5, but not prog- P-450SCC (CYP XIA1), and 3β- iety, sensorimotor gating and with- Nicola, A. F., Oliver, J. T. (1985).
estin receptor, antagonist and anti- hydroxysteroid dehydrogenase in drawal. Eur. J. Neurosci. 17, 641–648. In vivo secretion of 3α-hydroxy-
sense oligonucleotide infusions to the rat brain. J. Neurochem. 71, Halbreich, U., Asnis, G. M., Shin- 5α-pregnan-20-one, a potent anaes-
the ventral tegmental area of cycling 2231–22238. dledecker, R., Zumoff, B., and thetic steroid, by the adrenal gland
rats and hamsters attenuate lordosis. Gavish, M., Bachman, I., Shoukrun, Nathan, R. S. (1985). Cortisol secre- of the rat. J. Steroid Biochem. 22,
Behav. Brain Res. 103, 23–34. R., Katz, Y., Veenman, L., Weisinger, tion in endogenous depression. II. 97–102.
Frye, C. A., and Walf, A. A. (2002). G., and Weizman, A. (1999). Time-related functions. Arch. Gen. Huber, T. J., Rollnik, J., Wilhelms, J., von
Changes in progesterone metabo- Enigma of the peripheral benzodi- Psychiatry 42, 909–914. zur Mühlen, A., Emrich, H. M., and
lites in the hippocampus can mod- azepine receptor. Pharmacol. Rev. 51, Halbreich, U., Bancroft, J., Denner- Schneider, U. (2001). Estradiol levels
ulate open field and forced swim test 629–650. stein, L., Endicott, J., Faccinetti, F., in psychotic disorders. Psychoneu-
behavior of proestrous rats. Horm. Gee, K. W., McCauley, L. D., and Lan, Genazzani, A., Morse, C., Parry, roendocrinology 26, 27–35.
Behav. 41, 306–315. N. C. (1995). A putative receptor for B., Rubinow, D., Reid, R., Schiff, Jacobs, A. J., Odom, M. J., Word, R.
Frye, C. A., and Walf, A. A. (2004). neurosteroids on the GABAA recep- I., and Smith, S. (1993). Men- A., and Carr, B. R. (1989). Effect
Hippocampal 3α,5α-THP may alter tor complex: the pharmacological strually Related Disorders: points of oral contraceptives on adreno-
depressive behavior of pregnant and properties and therapeutic potential of consensus, debate, and disagree- corticotropin and growth hormone
lactating rats. Pharmacol. Biochem. of epalons. Crit. Rev. Neurobiol. 9, ment. Neuropsychopharmacology 9, secretion following CRH and GHRH
Behav. 78, 531–540. 207–227. 13–15. administration. Contraception 40,
Frye, C. A., and Walf, A. A. (2007). Genazzani, A. R., Petraglia, F., Bernardi, Hallonquist, J. D., Seeman, M. V., Lang, 691–699.
In the ventral tegmental area, F., Casarosa, E., Salvestroni, C., M., and Rector, N. A. (1993). Vari- Jain, N. S., Hirani, K., and Chopde,
the membrane-mediated actions of Tonetti, A., Nappi, R. E., Luisi, S., ation in symptom severity over the C. T. (2005). Reversal of caffeine-
progestins for lordosis of hormone- Palumbo, M., Purdy, R. H., and menstrual cycle of schizophrenics. induced anxiety by neurosteroid
primed hamsters involve phospho- Luisi, M. (1998). Circulating lev- Biol. Psychiatry 33, 207–209. 3-α-hydroxy-5-α-pregnane-20-one
lipase C and protein kinase C. J. els of allopregnanolone in humans: Hammarbäck, S., Damber, J. E., and in rats. Neuropharmacology 48,
Neuroendocrinol. 19, 717–724. gender, age, and endocrine influ- Bäckström, T. (1989). Relation- 627–638.
Frye, C. A., and Walf, A. A. (2008a). ences. J. Clin. Endocrinol. Metab. 83, ship between symptom severity and Kehoe, P., Mallinson, K., McCormick, C.
Membrane actions of progestins at 2099–2103. hormone changes in women with M., and Frye, C. A. (2000). Central
dopamine type 1-like and GABAA Girdler, S. S., Straneva, P. A., Light, K. premenstrual syndrome. J. Clin. allopregnanolone is increased in rat
receptors involve downstream signal C., Pedersen, C. A., and Morrow, A. Endocrinol. Metab. 68, 125–130. pups in response to repeated, short
transduction pathways. Steroids 73, L. (2001). Allopregnanolone levels Hammarbäck, S., Ekholm, U. B., and episodes of neonatal isolation. Brain
906–913. and reactivity to mental stress in pre- Bäckström, T. (1991). Spontaneous Res. Dev. Brain Res. 124, 133–136.
Frye, C. A., and Walf, A. A. (2008b). In menstrual dysphoric disorder. Biol. anovulation causing disappearance Kellogg, C. K., and Frye, C. A. (1999).
the ventral tegmental area, progesto- Psychiatry 49, 788–797. of cyclical symptoms in women with Endogenous levels of 5α-reduced
gens’ membrane mediated actions Glick, I. D., and Bennett, S. E. the premenstrual syndrome. Acta progestins and androgens in fetal
for lordosis of rats involve the (1981). Psychiatric complications of Endocrinol. 125, 132–137. vs. adult rat brains. Brain Res. Dev.
second-messenger phospholipase C. progesterone and oral contracep- Harrison, N. L., and Simmonds, M. A. Brain Res. 115, 17–24.
Brain Res. 1230, 218–223. tives. J. Clin. Psychopharmacol. 1, (1984). Modulation of the GABA Kessler, R. C., McGonagle, K. A., Zhao,
Frye, C. A., and Walf, A. A. (2008c). 350–367. receptor complex by a steroid anaes- S., Nelson, C. B., Hughes, M., Esh-
Activity of protein kinase C is Gracia, C. R., Freeman, E. W., Sammel, thetic. Brain Res. 323, 287–292. leman, S., Wittchen, H. U., and
important for 3α,5α-THP’s actions M. D., Lin, H., Sheng, L., and Frye, Hellman, L., Yoshida, K., Zumoff, Kendler, K. S. (1994). Lifetime and
at dopamine type 1-like and/or C. A. (2009). Allopregnanolone B., Levin, J., Kream, J., and 12-month prevalence of DSM-III-R
GABAA receptors in the ventral levels before and after selective Fukushima, D. K. (1976). The effect psychiatric disorders in the United
tegmental area for lordosis of rats. serotonin reuptake inhibitor treat- of medroxyprogesterone acetate on States. Results from the National
Brain Res. Bull. 77, 91–97. ment of premenstrual symptoms. the pituitary-adrenal axis. J. Clin. Comorbidity Survey. Arch. Gen. Psy-
Frye, C. A., Walf, A. A., Rhodes, J. Clin. Psychopharmacol. 29, Endocrinol. Metab. 42, 912–917. chiatry 51, 8–19.
M. E., and Harney, J. P. (2004a). 403–405. Hendrick, V., Altshuler, L. L., and Burt, King, S. R., Matassa, A. A., White, E.
Progesterone enhances motor, anx- Griffin, L. D., and Mellon, S. H. V. K. (1996). Course of psychiatric K., Walsh, L. P., Jo, Y., Rao, R. M.,
iolytic, analgesic, and antidepres- (1999). Selective serotonin reup- disorders across the menstrual cycle. Stocco, D. M., and Reyland, M. E.
sive behavior of wild-type mice, take inhibitors directly alter activ- Harv. Rev. Psychiatry 4, 200–207. (2004). Oxysterols regulate expres-
but not those deficient in type ity of neurosteroidogenic enzymes. Herbison, A. E. (2001). Physiological sion of the steroidogenic acute regu-
1 5α-reductase. Brain Res. 1004, Proc. Natl. Acad. Sci. U.S.A. 96, roles for the neurosteroid allopreg- latory protein. J. Mol. Endocrinol. 32,
116–124. 13512–13517. nanolone in the modulation of brain 507–517.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 15


Frye et al. 3α,5α-THP and PXR motivated behaviors

Kohtz, A. S., Paris, J. J., and Frye, C. Luchetti, S., Huitinga, I., and Swaab, D. Marx, C. E., Duncan, G. E., Gilmore, J. frontal cortex of suicide victims.
A. (2010). Low doses of cocaine F. (2011). Neurosteroid and GABA- H., Lieberman, J. A., and Morrow, A. Arch. Gen. Psychiatry 45, 577–579.
decrease, and high doses increase, A receptor alterations in Alzheimer’s L. (2000). Olanzapine increases allo- Norman, R. M., and Malla, A. K. (1993).
anxiety-like behavior and brain disease, Parkinson’s disease and mul- pregnanolone in the rat cerebral cor- Stressful life events and schizophre-
progestogen levels among intact rats. tiple sclerosis. Neuroscience 191, tex. Biol. Psychiatry 47, 1000–1004. nia. I: a review of the research. Br. J.
Horm. Behav. 57, 474–480. 6–21. Marx, C. E., VanDoren, M. J., Duncan, Psychiatry 162, 161–166.
Koonce, C. J., Walf, A. A., and Frye, C. Luisi, S., Petraglia, F., Benedetto, C., G. E., Lieberman, J. A., and Morrow, Nyberg, S., Wahlström, G., Bäck-
A. (2012). Type 1 5α-reductase may Nappi, R. E., Bernardi, F., Fadalti, A. L. (2003). Olanzapine and cloza- ström, T., and Sundström Poro-
be required for estrous cycle changes M., Reis, F. M., Luisi, M., and pine increase the GABAergic neu- maa, I. (2004). Altered sensitivity
in affective behaviors of female mice. Genazzani, A. R. (2000). Serum roactive steroid allopregnanolone in to alcohol in the late luteal phase
Behav. Brain. Res. 226, 376–380. allopregnanolone levels in pregnant rodents. Neuropsychopharmacology among patients with premenstrual
Korinek, M., Kapras, V., Vyklicky, women: changes during pregnancy, 28, 1–13. dysphoric disorder. Psychoneuroen-
V., Adamusova, E., Borovska, J., at delivery, and in hypertensive McCormick, C. M., Kehoe, P., docrinology 29, 767–777.
Vales, K., Stuchlik, A., Horak, patients. J. Clin. Endocrinol. Metab. Mallinson, K., Cecchi, L., and Osterlund, M. K., Gustafsson, J. A.,
M., Chodounska, H., and Vyklicky, 85, 2429–2433. Frye, C. A. (2002). Neonatal iso- Keller, E., and Hurd, Y. L. (2000).
L. Jr. (2011). Neurosteroid mod- Lukoff, D., Snyder, K., Ventura, J., lation alters stress hormone and Estrogen receptor β (ERβ) mes-
ulation of N -methyl-d-aspartate and Nuechterlein, K. H. (1984). mesolimbic dopamine release in senger ribonucleic acid (mRNA)
receptors: molecular mechanism Life events, familial stress, and cop- juvenile rats. Pharmacol. Biochem. expression within the human fore-
and behavioral effects. Steroids 76, ing in the developmental course of Behav. 73, 77–85. brain: distinct distribution pattern
1409–1418. schizophrenia. Schizophr. Bull. 10, Mellon, S. H. (1994). Neurosteroids: to ERβ mRNA. J. Clin. Endocrinol.
Kurumaji, A., Nomoto, H., Yoshikawa, 258–292. biochemistry, modes of action, and Metab. 85, 3840–3846.
T., Okubo, Y., and Toru, M. (2000). MacLusky, N. J., and McEwen, B. S. clinical relevance. J. Clin. Endocrinol. Padberg, F., di Michele, F., Zwanzger,
An association study between two (1980). Progestin receptors in rat Metab. 78, 1003–1008. P., Romeo, E., Bernardi, G., Schüle,
missense variations of the ben- brain: distribution and properties of Mellon, S. H., and Deschepper, C. C., Baghai, T. C., Ella, R., Pasini,
zodiazepine receptor (peripheral) cytoplasmic progestin-binding sites. F. (1993). Neurosteroid biosynthe- A., and Rupprecht, R. (2002).
gene and schizophrenia in a Japan- Endocrinology 106, 192–202. sis: genes for adrenal steroidogenic Plasma concentrations of neuroac-
ese sample. J. Neural Transm. 107, Majewska, M. D. (1992). Neuros- enzymes are expressed in the brain. tive steroids before and after repet-
491–500. teroids: endogenous bimodal modu- Brain Res. 629, 283–292. itive transcranial magnetic stimu-
Lamba, V., Yasuda, K., Lamba, J. lators of the GABAA receptor. Mech- Mellon, S. H., Gong, W., and Schone- lation (rTMS) in major depres-
K., Assem, M., Davila, J., Strom, anism of action and physiologi- mann, M. D. (2008). Endogenous sion. Neuropsychopharmacology 27,
S., and Schuetz, E. G. (2004). cal significance. Prog. Neurobiol. 38, and synthetic neurosteroids in treat- 874–878.
PXR (NR1I2): splice variants in 379–395. ment of Niemann-Pick Type C dis- Papadopoulos, V. (2011). From benzo-
human tissues, including brain, Majewska, M. D., Harrison, N. L., ease. Brain Res. Rev. 57, 410–420. diazepines to peripheral and brain
and identification of neurosteroids Schwartz, R. D., Barker, J. L., and Micevych, P., Soma, K. K., and Sinchak, steroid biosynthesis. Pharmacol. Res.
and nicotine as PXR activators. Paul, S. M. (1986). Steroid hor- K. (2008). Neuroprogesterone: key 64, 330–332.
Toxicol. Appl. Pharmacol. 199, mone metabolites are barbiturate- to estrogen positive feedback? Brain Papadopoulos, V., Baraldi, M., Guilarte,
251–265. like modulators of the GABA recep- Res. Rev. 57, 470–480. T. R., Knudsen, T. B., Lacapère, J.
Lambert, J. J., Belelli, D., Peden, D. tor. Science 232, 1004–1007. Micevych, P. E., and Dewing, P. (2011). J., Lindemann, P., Norenberg, M. D.,
R., Vardy, A. W., and Peters, J. A. Malla, A. K., Cortese, L., Shaw, T. S., Membrane-initiated estradiol sig- Nutt, D., Weizman, A., Zhang, M.
(2003). Neurosteroid modulation of and Ginsberg, B. (1990). Life events naling regulating sexual receptivity. R., and Gavish, M. (2006). Translo-
GABAA receptors. Prog. Neurobiol. and relapse in schizophrenia. A one Front. Endocrinol. (Lausanne) 2:26. cator protein (18kDa): new nomen-
71, 67–80. year prospective study. Soc. Psychia- doi:10.3389/fendo.2011.00026 clature for the peripheral-type ben-
Laviolette, S. R., Gallegos, R. A., Hen- try Psychiatr. Epidemiol. 25, 221–224. Miczek, K. A., Fish, E. W., and DeBold, zodiazepine receptor based on its
riksen, S. J., and van der Kooy, Mani, S. (2003). Signalling mechanisms J. F. (2003). Neurosteroids, GABAA structure and molecular function.
D. (2004). Opiate state controls in progesterone-neurotransmitter receptors, and escalated aggressive Trends Pharmacol. Sci. 27, 402–409.
bi-directional reward signaling via interactions. J. Mol. Endocrinol. 30, behavior. Horm. Behav. 44, 242–257. Paris, J. J., Chittur, S. V., Tine, J. A.,
GABAA receptors in the ventral 127–137. Monteleone, P., Luisi, S., Tonetti, A., and Frye, C. A. (2011). Paced mating
tegmental area. Nat. Neurosci. 7, Marini, S., Nannelli, A., Sodini, D., Bernardi, F., Genazzani, A. D., Luisi, enhances expression of hormonal
160–169. Dragoni, S., Valoti, M., Longo, V., M., Petraglia, F., and Genazzani, A. and trophic factors in the midbrain
Laviolette, S. R., and van der Kooy, and Gervasi, P. G. (2007). Expres- R. (2000). Allopregnanolone con- of female rats. Genes Brain Behav.
D. (2001). GABA(A) receptors sion, microsomal and mitochondr- centrations and premenstrual syn- Patchev, V. K., and Almeida, O. F.
in the ventral tegmental area ial activities of cytochrome P450 drome. Eur. J. Endocrinol. 142, (1996). Gonadal steroids exert facil-
control bidirectional reward sig- enzymes in brain regions from con- 269–273. itating and “buffering” effects on
nalling between dopaminergic and trol and phenobarbital-treated rab- Morrow, A. L., Suzdak, P. D., and glucocorticoid-mediated transcrip-
non-dopaminergic neural motiva- bits. Life Sci. 80, 910–917. Paul, S. M. (1987). Steroid hor- tional regulation of corticotropin-
tional systems. Eur. J. Neurosci. 13, Markou, A., Duka, T., and Prelevic, mone metabolites potentiate GABA releasing hormone and corticos-
1009–1015. G. M. (2005). Estrogens and brain receptor-mediated chloride ion flux teroid receptor genes in rat brain. J.
Le Mellédo, J. M., and Baker, G. B. function. Hormones 4, 9–17. with nanomolar potency. Eur. J. Neurosci. 16, 7077–7084.
(2002). Neuroactive steroids and Martínez, I., and Paredes, R. G. (2001). Pharmacol. 142, 483–485. Patchev, V. K., Hassan, A. H., Hols-
anxiety disorders. J. Psychiatry Neu- Only self-paced mating is rewarding Myin-Germeys, I., Nicolson, N. A., and boer, D. F., and Almeida, O. F.
rosci. 27, 161–165. in rats of both sexes. Horm. Behav. Delespaul, P. A. (2001). The context (1996). The neurosteroid tetrahy-
Li, X., Bertics, P. J., and Karavolas, 40, 510–517. of delusional experiences in the daily droprogesterone attenuates the
H. J. (1997). Regional distrib- Martín-García, E., and Pallarès, M. life of patients with schizophrenia. endocrine response to stress and
ution of cytosolic and partic- (2005). Intrahippocampal nicotine Psychol. Med. 31, 489–498. exerts glucocorticoid-like effects
ulate 5α-dihydroprogesterone 3α- and neurosteroids effects on the Nemeroff, C. B., Owens, M. J., Bis- on vasopressin gene transcrip-
hydroxysteroid oxidoreductases in anxiety-like behaviour in voluntary sette, G., Andorn, A. C., and Stanley, tion in the rat hypothalamus.
female rat brain. J. Steroid Biochem. and chronic alcohol-drinking rats. M. (1988). Reduced corticotropin Neuropsychopharmacology 15,
Mol. Biol. 60, 311–318. Behav. Brain Res. 164, 117–127. releasing factor binding sites in the 533–540.

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 16


Frye et al. 3α,5α-THP and PXR motivated behaviors

Patchev, V. K., Shoaib, M., Holsboer, F., menstrual phase and smoking absti- Catchment Area (ECA) Study. JAMA D. R. (1998). Differential behavioral
and Almeida, O. F. (1994). The neu- nence in smokers with and without a 264, 2511–2518. effects of gonadal steroids in women
rosteroid tetrahydroprogesterone history of major depressive disorder. Rhodes, M. E., and Frye, C. A. (2001). with and in those without premen-
counteracts corticotropin-releasing Addict. Behav. 25, 483–497. Inhibiting progesterone metabolism strual syndrome. N. Engl. J. Med. 33,
hormone-induced anxiety and Purdy, R. H., Moore, P. H. Jr, Rao, in the hippocampus of rats in behav- 209–216.
alters the release and gene P. N., Hagino, N., Yamaguchi, ioral estrus decreases anxiolytic Schmidt, P. J., Purdy, R. H., Moore,
expression of corticotropin- T., Schmidt, P., Rubinow, D. R., behaviors and enhances exploratory P. H. Jr., Paul, S. M., and Rubi-
releasing hormone in the rat Morrow, A. L., and Paul, S. M. and antinociceptive behaviors. Cogn. now, D. R. (1994). Circulating lev-
hypothalamus. Neuroscience 62, (1990). Radioimmunoassay of 3α- Affect. Behav. Neurosci. 1, 287–296. els of anxiolytic steroids in the
265–271. hydroxy-5α-pregnan-20-one in rat Romeo, E., Ströhle, A., Spalletta, G., luteal phase in women with premen-
Paul, S. M., and Purdy, R. H. (1992). and human plasma. Steroids 55, di Michele, F., Hermann, B., Hols- strual syndrome and in control sub-
Neuroactive steroids. FASEB J. 6, 290–296. boer, F., Pasini, A., and Rupprecht, jects. J. Clin. Endocrinol. Metab. 79,
2311–2322. Purdy, R. H., Morrow, A. L., Moore, P. R. (1998). Effects of antidepressant 1256–1260.
Pearlstein, T., Yonkers, K. A., Fayyad, H. Jr., and Paul, S. M. (1991). Stress- treatment on neuroactive steroids in Schüle, C., di Michele, F., Baghai, T.,
R., and Gillespie, J. A. (2005). induced elevations of gamma- major depression. Am. J. Psychiatry Romeo, E., Bernardi, G., Zwanzger,
Pretreatment pattern of symptom aminobutyric acid type A receptor- 155, 910–913. P., Padberg, F., Pasini, A., and
expression in premenstrual dys- active steroids in the rat brain. Rose, J. D. (1990). Tectoreticular neu- Rupprecht, R. (2003). Influence of
phoric disorder. J. Affect Disord. 85, Proc. Natl. Acad. Sci. U.S.A. 88, ronal activity associated with behav- sleep deprivation on neuroactive
275–282. 4553–4557. ior and progesterone action during steroids in major depression. Neu-
Petralia, S. M., DeBold, J. F., and Frye, Quiñones-Jenab, V., Minerly, A. C., Niy- the induction of lordosis responding ropsychopharmacology 28, 577–581.
C. A. (2007). MK-801 infusions omchia, T., Akahvan, A., Jenab, S., in hamsters. Abstr. Soc. Neurosci. 16, Schüle, C., Eser, D., Baghai, T. C.,
to the ventral tegmental area and and Frye, C. A. (2008). Progesterone 924. Nothdurfter, C., Kessler, J. S., and
ventromedial hypothalamus pro- and allopregnanolone are induced Rubin, R. T., Poland, R. E., Lesser, I. Rupprecht, R. (2011). Neuroactive
duce opposite effects on lordosis of by cocaine in serum and brain tissues M., Winston, R. A., and Blodgett, A. steroids in affective disorders: tar-
hormone-primed rats. Pharmacol. of male and female rats. Pharmacol. L. (1987). Neuroendocrine aspects get for novel antidepressant or
Biochem. Behav. 86, 377–385. Biochem. Behav. 89, 292–297. of primary endogenous depression. anxiolytic drugs? Neuroscience 191,
Petralia, S. M., and Frye, C. A. Ramcharan, S., Love, E. J., Fick, G. H., I. Cortisol secretory dynamics in 55–77.
(2006a). In the ventral tegmen- and Goldfien, A. (1992). The epi- patients and matched controls. Arch. Schwartz-Giblin, S., and Pfaff, D. W.
tal area, cyclic AMP mediates the demiology of premenstrual symp- Gen. Psychiatry 44, 328–336. (1986). Hypothalamic output con-
actions of progesterone at dopamine toms in a population-based sam- Rubinow, D. R., Hoban, M. C., Grover, trolling reticulospinal and vestibu-
type 1 receptors for lordosis of rats ple of 2650 urban women: attrib- G. N., Galloway, D. S., Roy-Byrne, lospinal systems important for emo-
and hamsters. J. Neuroendocrinol. 18, utable risk and risk factors. J. Clin. P., Andersen, R., and Merriam, G. tional behavior. Int. J. Neurol. 1986,
902–914. Epidemiol. 45, 377–392. R. (1988). Changes in plasma hor- 89–110.
Petralia, S. M., and Frye, C. A. Rapkin, A. J., Mikacich, J. A., Moatakef- mones across the menstrual cycle Seeman, M. V. (1997). Psychopathol-
(2006b). In the ventral tegmental Imani, B., and Rasgon, N. (2002). in patients with menstrually related ogy in women and men: focus on
area, G-proteins mediate proges- The clinical nature and formal mood disorder and in control sub- female hormones. Am. J. Psychiatry
terone’s actions at dopamine type diagnosis of premenstrual, postpar- jects. Am. J. Obstet. Gynecol. 158, 154, 1641–1647.
1 receptors for lordosis of rats tum, and perimenopausal affective 5–11. Seippel, L., and Bäckström, T. (1998).
and hamsters. Psychopharmacology disorders. Curr. Psychiatry Rep. 4, Rupprecht, R., and Holsboer, F. (1999). Luteal-phase estradiol relates to
(Berl.) 186, 133–142. 419–428. Neuroactive steroids: mechanisms of symptom severity in patients
Petralia, S. M., Jahagirdar, V., and Frye, Rapkin, A. J., Morgan, M., Gold- action and neuropsychopharmaco- with premenstrual syndrome.
C. A. (2005). Inhibiting biosynthe- man, L., Brann, D. W., Simone, logical perspectives. Trends Neurosci. J. Clin. Endocrinol. Metab. 83,
sis and/or metabolism of progestins D., and Mahesh, V. B. (1997). 22, 410–416. 1988–1992.
in the ventral tegmental area atten- Progesterone metabolite allopreg- Rupprecht, R., Papadopoulos, V., Selye, H. (1941). On the hormonal
uates lordosis of rats in behav- nanolone in women with premen- Rammes, G., Baghai, T. C., Fan, activity of a steroid compound. Sci-
ioural oestrus. J. Neuroendocrinol. strual syndrome. Obstet. Gynecol. 90, J., Akula, N., Groyer, G., Adams, ence 94, 94.
17, 545–552. 709–714. D., and Schumacher, M. (2010). Serra, M., Pisul, M. G., Dazzi, L., Purdy,
Petralia, S. M., and Frye, C. A. (2004). Read, J., Perry, B. D., Moskowitz, Translocator protein (18 kDa) R. H., and Biggio, G. (2002). Preven-
In the ventral tegmental area, G- A., and Connolly, J. (2001). The (TSPO) as a therapeutic target tion of the stress-induced increase
proteins and cAMP mediate the contribution of early traumatic for neurological and psychiatric in the concentration of neuroactive
neurosteroid 3α,5α-THP’s actions at events to schizophrenia in some disorders. Nat. Rev. Drug Discov. 9, steroids in rat brain by long-term
dopamine type 1 receptors for lor- patients: a traumagenic neurode- 971–988. administration of mirtazapine but
dosis of rats. Neuroendocrinology 80, velopmental model. Psychiatry 64, Russo, S. J., Sun, W. L., Minerly, A. C., not of fluoxetine. J. Psychopharma-
233–243. 319–345. Weierstall, K., Nazarian, A., Festa, col. 16, 133–138.
Pfaff, D. W., Gerlach, J. L., McEwen, Reddy, D. S. (2002). The clinical poten- E. D., Niyomchai, T., Akhavan, A., Shirayama, Y., Hashimoto, K., Suzuki,
B. S., Ferin, M., Carmel, P., and tials of endogenous neurosteroids. Luine, V., Jenab, S., and Quiñones- Y., and Higuchi, T. (2002). Corre-
Zimmerman, E. A. (1976). Autora- Drugs Today 38, 465–485. Jenab, V. (2008). Progesterone atten- lation of plasma neurosteroid levels
diographic localization of hormone- Reddy, D. S. (2003). Pharmacology uates cocaine-induced conditioned to the severity of negative symptoms
concentrating cells in the brain of of endogenous neuroactive steroids. place preference in female rats. Brain in male patients with schizophrenia.
the female rhesus monkey. J. Comp. Crit. Rev. Neurobiol. 15, 197–234. Res. 1189, 229–235. Schizophr. Res. 58, 69–74.
Neurol. 170, 279–293. Reddy, D. S. (2004). Pharmacology of Sanders, D., Warner, P., Bäckström, T., Shughrue, P. J., Lane, M. V., and
Pleim, E. T., Baumann, J., and Barfield, catamenial epilepsy. Methods Find and Bancroft, J. (1983). Mood, sex- Merchenthaler, I. (1997). Regu-
R. J. (1991). A contributory role for Exp. Clin. Pharmacol. 26, 547–561. uality, hormones and the menstrual lation of progesterone receptor
midbrain progesterone in the facil- Regier, D. A., Farmer, M. E., Rae, D. cycle. I. Changes in mood and phys- messenger ribonucleic acid in
itation of female sexual behavior in S., Locke, B. Z., Keith, S. J., Judd, ical state: description of subjects the rat medial preoptic nucleus
rats. Horm. Behav. 25, 19–28. L. L., and Goodwin, F. K. (1990). and method. Psychosom. Med. 45, by estrogenic and antiestrogenic
Pomerleau, C. S., Mehringer, A. M., Comorbidity of mental disorders 487–501. compounds: an in situ hybridiza-
Marks, J. L., Downey, K. K., and with alcohol and other drug abuse. Schmidt, P. J., Nieman, L. K., Danaceau, tion study. Endocrinology 138,
Pomerleau, O. F. (2000). Effects of Results from the Epidemiologic M. A., Adams, L. F., and Rubinow, 5476–5484.

www.frontiersin.org January 2012 | Volume 5 | Article 136 | 17


Frye et al. 3α,5α-THP and PXR motivated behaviors

Sinha, R., Fox, H., Hong, K. I., Sofuoglu, study. Psychoneuroendocrinology 22, receiving fluoxetine or fluvoxam- epidemiology of depression. Arch.
M., Morgan, P. T., and Bergquist, 25–38. ine. Proc. Natl. Acad. Sci. U.S.A. 95, Gen. Psychiatry 34, 98–111.
K. T. (2007). Sex steroid hormones, Sundström, I., Nyberg, S., and Bäck- 3239–3244. Wittchen, H. U., and Hoyer, J. (2001).
stress response, and drug crav- ström, T. (1997b). Patients with pre- Uzunova, V., Wrynn, A. S., Kin- Generalized anxiety disorder: nature
ing in cocaine-dependent women: menstrual syndrome have reduced nunen, A., Ceci, M., Kohler, C., and course. J. Clin. Psychiatry 62,
implications for relapse susceptibil- sensitivity to midazolam com- and Uzunov, D. P. (2004). Chronic 15–19.
ity. Exp. Clin. Psychopharmacol. 15, pared to control subjects. Neuropsy- antidepressants reverse cerebrocor- Wu, G., Kaper, J. M., and Jaspars, E.
445–452. chopharmacology 17, 370–381. tical allopregnanolone decline in the M. (1991). Replication of cucumber
Sinnott, R. S., Mark, G. P., and Finn, Sundström, I., and Bäckström, T. olfactory-bulbectomized rat. Eur. J. mosaic virus satellite RNA in vitro
D. A. (2002). Reinforcing effects of (1998a). Citalopram increases preg- Pharmacol. 486, 31–34. by an RNA-dependent RNA poly-
the neurosteroid allopregnanolone nanolone sensitivity in patients with Verleye, M., Akwa,Y., Liere, P., Ladurelle, merase from virus-infected tobacco.
in rats. Pharmacol. Biochem. Behav. premenstrual syndrome: an open N., Pianos, A., Eychenne, B., Schu- FEBS Lett. 292, 213–216.
72, 923–929. trial. Psychoneuroendocrinology 23, macher, M., and Gillardin, J. M. Xie, W., and Evans, R. M. (2002).
Soares, C. N., and Cohen, L. S. (2001). 73–88. (2005). The anxiolytic etifoxine acti- Pharmaceutical use of mouse mod-
The perimenopause, depressive dis- Sundström, I., and Bäckström, T. vates the peripheral benzodiazepine els humanized for the xenobiotic
orders, and hormonal variability. (1998b). Patients with premenstrual receptor and increases the neuros- receptor. Drug Discov. Today 7,
Sao Paulo Med. J. 119, 78–83. syndrome have decreased saccadic teroid levels in rat brain. Pharmacol. 509–515.
Sofuoglu, M., Babb, D. A., and Hat- eye velocity compared to con- Biochem. Behav. 82, 712–720. Young, E. A., Midgley, A. R., Carlson,
sukami, D. K. (2002). Effects of trol subjects. Biol. Psychiatry 44, Walf, A. A., and Frye, C. A. (2006). A N. E., and Brown, M. B. (2000).
progesterone treatment on smoked 755–764. review and update of mechanisms Alteration in the hypothalamic-
cocaine response in women. Phar- Sundström, I., Bäckström, T., Wang, M., of estrogen in the hippocampus and pituitary-ovarian axis in depressed
macol. Biochem. Behav. 72, 431–435. Olsson, T., Seippel, L., and Bixo, amygdala for anxiety and depression women. Arch. Gen. Psychiatry 57,
Sofuoglu, M., Dudish-Poulsen, S., Nel- M. (1999). Premenstrual syndrome, behavior. Neuropsychopharmacology 1157–1162.
son, D., Pentel, P. R., and Hat- neuroactive steroids and the brain. 31, 1097–1111. Zhu, Y., Bond, J., and Thomas, P.
sukami, D. K. (1999). Sex and men- Gynecol. Endocrinol. 13, 206–220. Walf, A. A., Paris, J. J., Llaneza, D. C., (2003). Identification, classification,
strual cycle differences in the subjec- Sveindóttir, H., and Bäckström, T. and Frye, C. A. (2011). I. Levels of and partial characterization of genes
tive effects from smoked cocaine in (2000). Prevalence of menstrual 5(-reduced progesterone metabolite in humans and other vertebrates
humans. Exp. Clin. Psychopharma- cycle symptom cyclicity and pre- in the midbrain account for vari- homologous to a fish membrane
col. 7, 274–283. menstrual dysphoric disorder in a ability in reproductive behavior of progestin receptor. Proc. Natl. Acad.
Sofuoglu, M., Mitchell, E., and Kosten, random sample of women using and middle-aged female rats. Brain Res. Sci. U.S.A. 100, 2237–2242.
T. R. (2004). Effects of progesterone not using oral contraceptives. Acta 1379, 137–148. Zwanzger, P., Eser, D., Padberg, F.,
treatment on cocaine responses in Obstet. Gynecol. Scand. 79, 405–413. Walf, A. A., Sumida, K., and Frye, C. A. Baghai, T. C., Schüle, C., Rupprecht,
male and female cocaine users. Phar- Tait, G. R., McManus, K., Bellavance, (2006). Inhibiting 5α-reductase in R., di Michele, F., Romeo, E., Pasini,
macol. Biochem. Behav. 78, 699–705. F., Lara, N., Chrapko, W., and Le the amygdala attenuates antianxiety A., and Ströhle, A. (2004). Neu-
Steffensen, S. C., Svingos, A. L., Pickel, V. Mellédo, J. M. (2002). Neuroac- and antidepressive behavior of roactive steroids are not affected
M., and Henriksen, S. J. (1998). Elec- tive steroid changes in response naturally receptive and hormone- by panic induction with 50 microg
trophysiological characterization of to challenge with the paγnicogenic primed ovariectomized rats. cholecystokinin-tetrapeptide (CCK-
GABAergic neurons in the ven- agent pentagastrin. Psychoneuroen- Psychopharmacology (Berl.) 186, 4) in healthy volunteers. J. Psychiatr.
tral tegmental area. J. Neurosci. 18, docrinology 27, 417–429. 302–311. Res. 38, 215–217.
8003–8015. Taylor, S. E., Klein, L. C., Lewis, B. Wang, M., Bäckström, T., Sundström,
Ströhle, A., Romeo, E., di Michele, P., Gruenewald, T. L., Gurung, R. I., Wahlström, G., Olsson, T., Zhu, Conflict of Interest Statement: The
F., Pasini, A., Hermann, B., Gajew- A., and Updegraff, J. A. (2000). D., Johansson, I. M., Björn, I., authors declare that the research was
sky, G., Holsboer, F., and Rup- Biobehavioral responses to stress and Bixo, M. (2001). Neuroactive conducted in the absence of any
precht, R. (2003). Induced panic in females: tend-and-befriend, not steroids and central nervous system commercial or financial relationships
attacks shift gamma-aminobutyric fight-or-flight. Psychol. Rev. 107, disorders. Int. Rev. Neurobiol. 46, that could be construed as a potential
acid type A receptor modulatory 411–429. 421–459. conflict of interest.
neuroactive steroid composition in Townsend, K. A., and Marlowe, K. Wang, M., Seippel, L., Purdy, R. H.,
patients with panic disorder: prelim- F. (2004). Relative safety and effi- and Bäckström, T. (1996). Rela- Received: 10 August 2011; paper pend-
inary results. Arch. Gen. Psychiatry cacy of finasteride for treatment of tionship between symptom sever- ing published: 25 August 2011; accepted:
60, 161–168. hirsutism. Ann. Pharmacother. 38, ity and steroid variation in women 25 November 2011; published online: 19
Sumida, K., Walf, A. A., and Frye, C. A. 1070–1073. with premenstrual syndrome: study January 2012.
(2005). Progestin-facilitated lordosis Usall, J., Suarez, D., Haro, J. M., and on serum pregnenolone, preg- Citation: Frye CA, Paris JJ, Walf AA
of hamsters may involve dopamine- SOHO Study Group. (2007). Gender nenolone sulfate, 5α-pregnane-3,20- and Rusconi JC (2012) Effects and
like type 1 receptors in the ven- differences in response to antipsy- dione and 3α-hydroxy-5α-pregnan- mechanisms of 3α,5α,-THP on emo-
tral tegmental area. Behav. Brain Res. chotic treatment in outpatients with 20-one. J. Clin. Endocrinol. Metab. tion, motivation, and reward func-
161, 1–7. schizophrenia. Psychiatry Res 153, 81, 1076–1082. tions involving pregnane xenobiotic
Sundström, I., Andersson, A., Nyberg, 225–231. Warembourg, M. (1978). Radioauto- receptor. Front. Neurosci. 5:136. doi:
S., Ashbrook, D., Purdy, R. H., Uzunova, V., Sampson, L., and Uzunov, graphic study of the rat brain, 10.3389/fnins.2011.00136
and Bäckström, T. (1998). Patients D. P. (2006). Relevance of endoge- uterus and vagina after [3H]R-5020 This article was submitted to Frontiers
with premenstrual syndrome have nous 3α-reduced neurosteroids injection. Mol. Cell. Endocrinol. 12, in Neuroendocrine Science, a specialty of
a different sensitivity to a neu- to depression and antidepressant 67–79. Frontiers in Neuroscience.
roactive steroid during the men- action. Psychopharmacology (Berl.) Weir, C. J., Ling, A. T., Belelli, D., Wild- Copyright © 2012 Frye, Paris, Walf
strual cycle compared to con- 186, 351–361. smith, J. A., Peters, J. A., and Lam- and Rusconi. This is an open-access
trol subjects. Neuroendocrinology 67, Uzunova, V., Sheline, Y., Davis, J. M., bert, J. J. (2004). The interaction of article distributed under the terms of
126–138. Rasmusson, A., Uzunov, D. P., Costa, anaesthetic steroids with recombi- the Creative Commons Attribution Non
Sundström, I., Ashbrook, D., and Bäck- E., and Guidotti, A.(1998). Increase nant glycine and GABAA receptors. Commercial License, which permits non-
ström, T. (1997a). Reduced ben- in the cerebrospinal fluid content Br. J. Anaesth. 92, 704–711. commercial use, distribution, and repro-
zodiazepine sensitivity in patients of neurosteroids in patients with Weissman, M. M., and Klerman, G. duction in other forums, provided the
with premenstrual syndrome: a pilot unipolar major depression who are L. (1977). Sex differences and the original authors and source are credited.

Frontiers in Neuroscience | Neuroendocrine Science January 2012 | Volume 5 | Article 136 | 18

Potrebbero piacerti anche