You are on page 1of 24

Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Contents lists available at ScienceDirect

Frontiers in Neuroendocrinology
journal homepage: www.elsevier.com/locate/yfrne

Review

Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis


Robert J. Handa a,⇑, Michael J. Weiser b
a
Department of Basic Medical Science, The University of Arizona College of Medicine, Phoenix, AZ 85004, United States
b
DSM Nutritional Products Ltd., R&D Human Nutrition and Health, Boulder, CO 80301, United States

a r t i c l e i n f o a b s t r a c t

Article history: The hypothalamo–pituitary–adrenal (HPA) axis represents a complex neuroendocrine feedback loop
Available online xxxx controlling the secretion of adrenal glucocorticoid hormones. Central to its function is the paraventricular
nucleus of the hypothalamus (PVN) where neurons expressing corticotropin releasing factor reside. These
Keywords: HPA motor neurons are a primary site of integration leading to graded endocrine responses to physical
Androgen receptor and psychological stressors. An important regulatory factor that must be considered, prior to generating
Estrogen receptor an appropriate response is the animal’s reproductive status. Thus, PVN neurons express androgen and
Glucocorticoid
estrogen receptors and receive input from sites that also express these receptors. Consequently, changes
CRF
Sex difference
in reproduction and gonadal steroid levels modulate the stress response and this underlies sex
HPA differences in HPA axis function. This review examines the make up of the HPA axis and hypothal-
HPG amo–pituitary–gonadal (HPG) axis and the interactions between the two that should be considered when
Dihydrotestosterone exploring normal and pathological responses to environmental stressors.
Estradiol Ó 2013 Elsevier Inc. All rights reserved.
Testosterone

1. Introduction Selye. It was Cannon who originally coined the term, ‘‘fight or
flight’’, when referring to the physiological responses to acute
The origins of the study of stress physiology are rooted in the stressors (Cannon, 1915). He later described the concept of homeo-
contributions of the early physiologists, Walter Cannon and Hans stasis as a steady state condition that requires active mechanisms
to maintain (Cannon, 1932). These pioneering concepts were fur-
ther explored by Hans Selye who examined the effect of chronic
Abbreviations: 3ß-diol, 5alpha-androstane-3beta, 17beta-diol; 3a-diol, 5alpha- stressors on an organism’s physiology. Selye’s studies into the
androstane-3alpha, 17beta-diol; 5-HT, serotonin; 5aR, 5-alpha-reductase; ACTH, body’s reactions to chronic stressors led to his development of
adrenocortcotropin releasing hormone; AR, androgen receptor; AVP, arginine
the general adaptation syndrome (GAS), a set of nonspecific re-
vasopressin; BnST, bed nucleus of the stria terminalis; BSA, bovine serum albumin;
CBP, CREB binding protein; CORT, corticosterone; CRE, cyclic adenosine monophos- sponses to a stressor which could give rise to pathology after con-
phate response element; CREB, CRE binding protein; CRF, corticotropin releasing tinuous, unrelieved stress.
factor; DES, diethylstilbesterol; DEX, dexamethasone; DHT, dihydrotestosterone; The pioneering work of Cannon and Selye were closely followed
DPN, diarylproprionitrile; ER, estrogen receptor; ERE, estrogen response element; by studies focused on teasing out the biological mechanisms
FSH, follicle stimulating hormone; FSL, flinders sensitive line; GABA, gamma-
underlying the stress response. These included the demonstration
aminobutyric acid; GAS, general adaptation syndrome; GH, growth hormone;
GLP-1, glucagon-like peptide 1; GnRH, gonadotropin releasing hormone; GPER, that an anterior pituitary hormone (i.e. adrenocorticotropic hor-
G-protein coupled estrogen receptor; GR, glucocorticoid receptor; HPA, hypothal- mone; ACTH) can stimulate adrenal glucocorticoid release (Sayers,
amo–pituitary–adrenal; HPG, hypothalamo–pituitary–gonadal; HRE, hormone 1950) and the development of the postulate that pituitary gland
respone element; HSD, hydroxysteroid dehydrogenase; ICV, intracerebroventricu-
function was under neural control (Harris, 1951a,b). The latter
lar; ir, immunoreactive; ISH, in situ hybridization; LH, luteinizing hormone; LHRH,
luteinizing hormone releasing hormone; MPOA, medial preoptic area; MR,
hypothesis was based on Harris’ observations of the capillary sys-
mineralocorticoid receptor; NTS, nucleus of the solitary tract; PPT, propylpyrazo- tem that existed connecting the ventral hypothalamus with the
letriol; PR, progesterone receptor; PVN, paraventricular nucleus; RAR, retinoic acid anterior lobe of the pituitary. In these pioneering studies, Harris
receptors; SCN, suprachiasmatic nucleus; SRC, steroid receptor coactivators; T, demonstrated that disrupting blood flow from the hypothalamus
testosterone; Tfm, testicular feminizing mutation; THR, thyroid hormone receptors;
to the pituitary by pituitary stalk section would impair ACTH re-
TPH, tryptophan hydroxylase; TRH, thyrotropin releasing hormone; TSH, thyroid
stimulating hormone. lease (Fortier et al., 1957), whereas electrical stimulation of the
⇑ Corresponding author. Address: Department of Basic Medical Sciences, Univer- rabbit hypothalamus triggered the release of ACTH into the general
sity of Arizona College of Medicine, 425N. 5th Street, Phoenix, AZ 85004, United circulation (De Groot and Harris, 1950). Harris also showed that
States. Fax: +1 602 827 2130.
pituitary explants were non-functional, yet viable (Harris and
E-mail address: rhanda@arizona.edu (R.J. Handa).

0091-3022/$ - see front matter Ó 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.yfrne.2013.11.001

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
2 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Jacobsohn, 1952). Such studies confirmed the importance of the the short term as they permit the fight or flight response. By con-
hypothalamus in controlling anterior pituitary function and helped trast, these same beneficial responses can turn detrimental when
establish the field of neuroendocrinology as a discipline. Further the stressor is maintained over long periods of time. Indeed,
studies by McCann (1953) and Porter (1953) demonstrated that chronic activation of the HPA axis results in deleterious effects
hypothalamic lesions prevented ACTH release thereby establishing on immune, cardiovascular, metabolic and neural functions and
the hypothalamus as the source of the alleged ‘releasing factors’ may decrease the viability of neurons and glia to subsequent neu-
that affected pituitary function. rotoxic insults (Jauregui-Huerta et al., 2010; McEwen, 1998;
The initial attempts to isolate the putative corticotropin releas- Rajkowska and Miguel-Hidalgo, 2007).
ing factor (CRF) proved difficult, although initially, the ability of an
alternate ‘‘CRF’’, vasopressin, to induce ACTH release was described
(McCann and Brobeck, 1954). Hypothalamic releasing factors, such 2.2. Anatomy of the paraventricular nucleus (PVN)
as thyrotropin releasing hormone (TRH), and luteinizing hormone
releasing hormone (LHRH, or gonadotropin releasing hormone, The neurons responsible for controlling HPA axis activity reside
GnRH, Schally et al., 1971a) were initially isolated in the late ‘60s in the paraventricular nucleus of the hypothalamus (PVN). The PVN
and early ‘70s (Amoss et al., 1971; Burgus et al., 1970; Nair et al., represents a collection of neurons in the rostral hypothalamus that
1970; Schally et al., 1971b,c), yet, it was a decade later when Vale is positioned to coordinate neuroendocrine, autonomic and behav-
et al. (1981) isolated, characterized, and described the biological ioral responses to stressors as well as to maintain energy and water
activity of a hypothalamic peptide that caused the release of ACTH balance (Herman et al., 2008; Levy and Tasker, 2012). The PVN has
from the anterior pituitary gland. Since then, the 41 amino acid been most comprehensively studied in the rat brain and reportedly
CRF, has been shown to be expressed in many brain areas and consists of approximately 100,000 neurons in a volume of about
has been implicated in a wide variety of behaviors and neurobio- 0.5 mm3. The PVN is arranged in a wing shape structure along
logical functions (Bale and Vale, 2004). We now know that CRF the dorsal portion of the third ventricle in the anterior region of
and vasopressin can be co-localized in some neurons of the para- the hypothalamus. The initial description of the cytoarchitectural
ventricular nucleus of the hypothalamus (Sawchenko et al., 1984; organization of the anterior hypothalamus was made by Gurdjian
Whitnall and Gainer, 1988), that they can be co-released, and that (1927). Based upon Nissl stained material, he described a medial
vasopressin acts to enhance the secretogogue properties of CRF at group of neurons with small cell bodies, a dense lateral group with
the anterior pituitary gland (Bilezikjian and Vale, 1987; Gillies medium to large cell bodies, and a dorsal group with unique Nissl
et al., 1982; Rivier and Vale, 1983). staining properties in the area that was later included in the hypo-
Of importance for this discussion are the findings that a number thalamic paraventricular nucleus. Bargmann (1949) and Bargmann
of human neuropsychiatric disorders are accompanied by a dysreg- and Scharrer (1951) later identified the densely packed neurons
ulation of the HPA axis and that many of these disorders exhibit with large cell bodies (i.e. magnocells) as projecting to the poster-
profound sex differences in risk implicating a modulatory role for ior pituitary gland.
gonadal steroid hormones (Kessler et al., 1993; Kessler, 2003). Later studies described the PVN based on cytoarchitecture and
For example, the incidence of major depressive disorder is at least chemoarchitecture using results from tract tracing, Golgi impreg-
two fold greater in women than in men (Angold and Worthman, nation, and immunocytochemical approaches (Armstrong et al.,
1993; Kessler et al., 1993; Weissman et al., 1993) and this is asso- 1980; Swanson and Kuypers, 1980). Based on such parameters,
ciated with enhanced HPA activity associated with a reduced abil- the neurons of the rodent PVN were grouped into subdivisions
ity to feedback regulate the system (Ising et al., 2007; Strohle and with each subdivision associated with specific functions (Biag
Holsboer, 2003). In this review, we first examine the HPA axis and et al., 2012). Furthermore, while there is some controversy regard-
its regulatory elements and follow with a discussion of pre-clinical ing the parcellation of the PVN in humans, it seems likely that sim-
studies showing sex differences in the function of the HPA axis and ilar subdivisions also exist based on criteria used for the rat brain
the well-described role of gonadal steroid hormones in modulating (Koutcherov et al., 2000). Moreover, the neurons of the PVN can
HPA axis responsivity to stress and stress-related behaviors in be further divided by function into three main types. (1) Neurose-
adulthood. cretory parvocellular neurons which send their axons to the exter-
nal zone of the median eminence where they secret releasing
factors (e.g. CRF, vasopressin, thyrotropin releasing hormone
2. The hypothalamo–pituitary–adrenal (HPA) axis (TSH), somatostatin) into the hypothalamo–hypophyseal portal
vasculature to control the secretion of anterior pituitary hormones
2.1. An overview of the HPA axis such as ACTH, TSH and growth hormone (GH). (2) Neurosecretory
magnocellular neurons which send projections to fenestrated capil-
Animals respond to real or perceived threats to their welfare by laries in the posterior pituitary where they secrete hormones (e.g.
activating neurons that control neuroendocrine responses (e.g. the oxytocin, vasopressin) directly into the general circulation. (3)
HPA axis) and the sympathetic autonomic response. For HPA axis Long-projecting neurons which send their axons to brainstem and
activation, the net response is the secretion of glucocorticoids from spinal cord regions involved in controlling autonomic and somato-
the adrenal cortex into the general circulation. In humans and sensory function. Moreover, although PVN neurons are largely clas-
many mammals, the main adrenal glucocorticoid is cortisol, sified by output, they can further be subdivided by phenotype,
whereas corticosterone is the primary glucocorticoid in most ro- afferent input, cell size, density and dendritic morphology
dents. Circulating corticosteroids act on a variety of tissues to (Armstrong et al., 1980; Ju et al., 1986; Kiss et al., 1991; Rho and
mobilize energy stores, induce lipolysis and proteolysis, potentiate Swanson, 1989; Swanson et al., 1986; van den Pol, 1982).
vasoconstriction driven by the autonomic nervous system, sup- The magnocellular neurons of the PVN are largely distributed
press reproductive function, and alter a number of stress related into two distinct but adjoining areas. The medial magnocellular
behaviors; all in an attempt to maintain homeostasis (Herman division lies anteromedially within the PVN and contains mostly
et al., 2008; Papadimitriou and Priftis, 2009). It is generally thought oxytocin expressing neurons. The lateral magnocellular division is
that many of the responses to acute elevations in glucocorticoids comprised largely of a sphere shaped mass of vasopressin express-
that occur following stressors, such as enhanced cognition and ing neurons that is surrounded by a loop of oxytocin neurons. The
metabolism and inhibition of immune function, are beneficial in majority of these neurons project to the neurohypophysis and are

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 3

neurosecretory (Rhodes et al., 1981; Swanson and Sawchenko, (Aguilera et al., 2004). ACTH in turn stimulates the biosynthesis
1983; Vandesande and Dierickx, 1975). and release of glucocorticoids (cortisol in human, corticosterone
The parvocellular group of small to medium sized neurons in in rat) by the adrenal cortex (Axelrod and Reisine, 1984; de Kloet,
the PVN send axonal projections to the median eminence. These 1984). Ultimately, glucocorticoids bind to their receptors (gluco-
neurons reside in two main areas of the PVN. The anterior parvocel- corticoid receptors, GR) in various tissues including the tissues reg-
lular division extends from the rostral boundary of the PVN to the ulating the HPA axis, thereby reducing the secretion of CRF and
rostral boundary of the medial magnocellular division, just lateral ACTH. This negative feedback mechanism (see below) is essential
to the periventricular area. The medial parvocellular division lies for keeping the balance of the HPA axis activity during basal con-
lateral to the periventricular area and medial to the medial magno- ditions and in response to stress (Tsigos and Chrousos, 2002).
cellular division. Neurons in the anterior and medial parvocellular Moreover, the changes in glucocorticoids that arise following envi-
groups project to the median eminence or other hypothalamic and ronmental perturbations can also impact numerous behaviors
extrahypothalamic regions, with the majority of the CRF peptide through actions at other brain regions (Weiser and Handa, 2009,
found in the median eminence originating from parvocellular for review see: Shansky and Lipps, 2013).
PVN neurons (Koegler-Muly et al., 1993; Lind et al., 1985). CRF is the primary regulator of ACTH secretion by the anterior
Whether in the anterior or medial groups, parvocellular neu- pituitary gland. Nonetheless, supporting roles of vasopressin and
rons are chemically very diverse and have been shown to express oxytocin as co-secretagogues have been also demonstrated (Her-
a large list of neuropeptides including, but not limited to: oxytocin, man et al., 1990; Ring, 2005). Although vasopressin has been orig-
vasopressin, TRH, CRF, angiotensin II, cholecystokinin, cocaine and inally described as a regulator of osmotic balance, and oxytocin has
amphetamine-regulated transcript, enkephalin, galanin, and been considered a principal hormone for parturition, both of these
somatostain (Ceccatelli et al., 1989; Healy and Printz, 1984; Kiss, peptides have been now shown to co-localize with CRF in discrete
1985; Plotsky and Sawchenko, 1987; Suzuki et al., 2001b; Vrang PVN neuronal populations and are co-released with CRF (Bondy
et al., 1999). Although arginine vasopressin (AVP) expression is rel- et al., 1989; Raadsheer et al., 1993; Whitnall and Gainer, 1988)
atively low in the parvocellular PVN, compared to its expression in thereby potentiating CRF’s secretogogue activity at the level of
magnocellular neurons, it is reportedly co-localized in a majority of the corticotroph (Rivier and Vale, 1983; Schlosser et al., 1994).
CRF neurons, especially after adrenalectomy (Kiss et al., 1984; Saw- Nonetheless, both vasopressin and oxytocin can stimulate ACTH
chenko et al., 1984; Whitnall and Gainer, 1988) secretion even in the absence of CRF (Gillies et al., 1982; Schlosser
Most non-neurosecretory neurons of the PVN are found in three et al., 1994) through the activation of the V1b receptor expressed
main regions: the dorsomedial cap, the ventral PVN, and the pos- by corticotrophs (Schlosser et al., 1994). In contrast, when applied
terior PVN (Blair et al., 1996; Ferguson et al., 2008; Swanson, directly to the PVN, or following intracerebroventricular injection,
1977; Swanson and Kuypers, 1980). These neurons largely project oxytocin and vasopressin inhibit HPA responses (Landgraf and
to autonomic preganglionic neurons and associated nuclei. The Neumann, 2004; Neumann et al., 2000a; Windle et al., 1997),
dorsomedial cap sends the majority of its fibers to the lateral gray thereby indicating that these neuropeptides can modify PVN func-
horn of the spinal column (intermediolateral cell columns), tion in a paracrine action, perhaps through local release. This is
whereas the ventral and posterior regions project to a wide array thought to occur through dendritic release of the peptide (Neu-
of brainstem and spinal cord regions including the dorsal vagal mann, 2007) resulting in regulatory effects on the PVN that are
motor nucleus, nucleus of the solitary tract, periaqueductal gray, very different from their actions on the anterior pituitary.
dorsal raphe, locus coeruleus, parabrachial nucleus and ventrolat- Recent studies have also demonstrated that other regulatory
eral reticular nucleus (Hosoya et al., 1991; Luiten et al., 1985; Py- factors are expressed by PVN neurons that can modulate CRF re-
ner and Coote, 2000; Saper et al., 1976; Shafton et al., 1998; sponses. For example, the endocannabinoids, endogenous arachid-
Shapiro and Miselis, 1985). These descending connections utilize onate-like lipids, are ligands for cannabinoid receptors (CB1 and
glutamate, gamma-aminobutyric acid (GABA) or the various neu- CB2) that are expressed in brain (Bellocchio et al., 2008). Studies
ropeptides for communication. Of note, the non-neurosecretory have demonstrated that the CB1 receptor is involved in modulating
neurons of the PVN express a number of neuropeptides including HPA axis function (Cota et al., 2007). The CB1 receptor is expressed
CRF, AVP and oxytocin (Jansen et al., 1995; Milner et al., 1993; on axon terminals of glutamatergic, GABAergic and monoaminer-
Sawchenko, 1987b; Sofroniew and Schrell, 1982). More CRF than gic neurons and its activation causes suppression of neurotrans-
AVP neurons of the PVN are long projecting neurons (Sawchenko, mitter release to the synapse (Freund et al., 2003). Within the
1987b) when detected following colchicine treatment to inhibit PVN, glucocorticoids cause the release of endocannabinoids which
axonal transport, and substantially more oxytocin than vasopres- suppress the excitatory inputs to CRF neurons, and this can be
sin cells were found with long-projecting projections (Sawchenko blocked with CB1 receptor antagonists (Evanson et al., 2010). Be-
and Swanson, 1982). Moreover, although the presence of interneu- cause the endocannabinoids are widely distributed throughout
rons within the PVN have not been described, axon collaterals from the hypothalamic and limbic circuitries (see below), they occupy
PVN neurons terminate within the PVN and project to other fore- a unique niche to regulate both excitatory and inhibitory transmit-
brain areas (van den Pol, 1982). Recurrent collaterals from CRF ter release, the net result being constraint on HPA activity (Hill
neurons could explain the existence of CRF positive synapses on et al., 2010). Of particular importance for this discussion, endocan-
neurons of the medial parvocellular region and as well as magno- nabinoid systems also overlap considerably with receptors for go-
cellular and periventricular areas (Hisano et al., 1993; Liposits nadal steroid hormones (for review see: Gorzalka and Dang, 2012)
et al., 1985; Silverman et al., 1989), however, the PVN also receives raising the possibility that this represents an important neuromod-
a dense CRF innervation from neurons in the bed nucleus of the ulatory system that can be utilized for sex steroid hormone regula-
stria terminalis (BnST) (Dong et al., 2001b). tion of HPA axis function.

2.3. Regulation of the HPA axis 2.4. Stress integration: neurocircuitry of the HPA axis

Acute stress exposure activates the HPA axis resulting in the The hypophysiotrophic neurons of the parvocellular PVN re-
secretion of corticotropin releasing factors from the PVN (Dunn ceive stress-related neuronal input from a variety of sources and
and Berridge, 1990). CRF causes the release of pituitary ACTH integrate that information to produce a relevant response. In
(Antoni, 1986) by binding to CRF-R1 receptors on corticotrophs addition to neural inputs, it is important to note that these neurons

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
4 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

can also respond to the systemic hormonal milieu. The PVN is one (Cummings and Seybold, 1988; Itoi et al., 1994; Khan et al.,
of the most vascularized regions of the brain (Herman et al., 2005; 2007; Kiss and Aguilera, 1992; Szafarczyk et al., 1987; Whitnall
Menendez and Alvarez-Uria, 1987; van den Pol, 1982) and the neu- et al., 1993; Windle et al., 1997). The NTS is critical for mediating
rovascular development of this area may be influenced by GABA reflex control of the cardiovascular system and in relaying infor-
signaling (Frahm et al., 2012). Changes in the neurovascular com- mation on visceral illness and systemic infection (Ericsson et al.,
ponent of the PVN may reflect functional changes underlying al- 1994; Lawrence and Jarrott, 1996; Seeley et al., 2000). Such pertur-
tered neuronal responses to blood-born substances that could bations are thought to also recruit the secretion of glucocorticoids
modulate autonomic and neuroendocrine function (Tobet et al., to assist in restoring homeostasis.
2013). Indeed, it has been shown that glucocorticoid exposure In addition, glucagon-like peptide-1 (GLP-1) neurons from the
can alter angiogenesis in areas such as the hippocampus and pre- NTS have recently been shown to participate in direct activation
frontal cortex (Ekstrand et al., 2008; Neigh et al., 2010). Further, of parvocellular CRF neurons. These GLP-1 synthesizing neurons
the terminals of PVN neurosecretory neurons are also in place to in the NTS make direct and dense synaptic connections with a
sample the hypophyseal portal vasculature that is thought to be majority of parvocellular CRF neurons (Sarkar et al., 2003; Tauchi
blood brain barrier deficient (Antoni, 1986; Whitnall, 1993). In et al., 2008), and the GLP-1 receptor is highly expressed in the par-
contrast, the dendritic arbors of neurons of the PVN are confined vocellular PVN (Gu et al., 2013; Merchenthaler et al., 1999; Shugh-
to the boundaries of the PVN (Rho and Swanson, 1989; van den rue et al., 1996b). Interestingly, the psychogenic stressor, restraint,
Pol, 1982), thereby restricting the synaptic regulation of PVN neu- causes a rapid and substantial decrease in GLP-1 fiber immunore-
rons to within the PVN whereas the influence of blood-born sub- activity in the PVN, indicating recent GLP-1 release (Zhang et al.,
stances may be more widespread than that of synaptic sampling 2009). Intracerebroventricular (ICV) administration of GLP-1 re-
alone. sults in the rapid induction of c-Fos immunoreactivity in a majority
Although PVN neurons receive a rather limited number of direct (>80%) of CRF neurons in the PVN, and a corresponding robust in-
inputs, they receive substantial inputs from neurons in the imme- crease in plasma corticosterone concentrations (Larsen et al.,
diate surround (peri-PVN) and it is this design that may allow 1997). This effect is blocked with ICV pretreatment with a selective
greater integration potential. The shell of neurons in the peri- GLP-1 antagonist. Furthermore, microinfusion of GLP-1 directly
PVN contain substantial numbers of PVN projecting GABA neurons into the PVN results in a dose-dependent increase in plasma corti-
(Roland and Sawchenko, 1993). This architecture raises the possi- costerone to levels commonly observed with potent stressors (Kin-
bility that inputs to the PVN are first processed, or filtered, locally zig et al., 2003). Recently, GLP-1 has been shown to induce CRF and
prior to contact with the PVN neurons themselves. Application of AVP gene expression in the hypothalamic 4B cell line (Kageyama
glutamate to the peri-PVN region results in GABA-dependent inhi- et al., 2012). These data suggest that stress-activated GLP-1 inputs
bition of PVN neurons (Boudaba et al., 1996) indicating that these from the NTS may induce CRF and AVP release from the parvocell-
neurons are inhibitory to CRF neurons. Similarly, the microinjec- uar PVN although this important correlative phenomenon has not
tion of glutamate receptor antagonists to the peri-PVN increases yet been directly tested (for a recent review of the role of GLP-1 in
the corticosterone response to restraint (Ziegler and Herman, stress regulation see Ghosal et al., 2013). Other brainstem regions
2000). Peri-PVN neurons show robust c-fos mRNA expression fol- that are critical in autonomic integration and send direct projec-
lowing restraint stress indicating that they are active during the tions to the PVN include the parabrachial nucleus (cardiovascular)
stress response and likely represent an important gating mecha- and periaqueductal grey (visceral pain) (Behbehani, 1995; Saper,
nism for signals originating in forebrain and brainstem regions 1995; Sawchenko and Swanson, 1983).
(Herman et al., 2002). The median and dorsal raphe nuclei of the brainstem also send
There are a select group of direct inputs to the parvocellular direct dense serotonergic projections to the parvocellular PVN
PVN from brainstem neurons that are integral to initiating re- (Sawchenko et al., 1983). Serotonin (5-HT) receptors are expressed
sponses to systemic stressors. The parvocellular PVN is densely by parvocellular neurons (Zhang et al., 2002) and serotonin’s ef-
innervated with noradrenergic and adrenergic fibers (Cunningham fects are largely stimulatory to PVN output (Van de Kar and Blair,
et al., 1990; Hornby and Piekut, 1989; Kitazawa et al., 1987; Lipos- 1999). The 5-HT 2C receptor has been shown to be necessary for
its et al., 1986; Mezey et al., 1984; Plotsky et al., 1989; Sawchenko 5-HT induced activation of the HPA axis (Heisler et al., 2007). How-
and Swanson, 1981) and many of these arise from the locus coeru- ever, activation of 5-HT1A receptors in the PVN also increases
leus, nucleus of the solitary tract (NTS) and ventrolateral medulla. ACTH secretion (Rossi et al., 2010), and this effect is desensitized
CRF neurons of the medial parvocellular PVN receive dense norad- by estradiol treatment. Restraint induced elevations in corticoste-
renergic innervation primarily from A2 adrenergic cell groups in rone and ACTH can also be inhibited by blocking the 5HT-7 recep-
the NTS (Hornby and Piekut, 1989; Kitazawa et al., 1987; Liposits tor (Garcia-Iglesias et al., 2013). A network of serotonin fibers
et al., 1986; Plotsky et al., 1989). A wide array of stressful stimuli surrounds the PVN as well, and serotonin has been shown to inhi-
increase norepinephrine (NE) release in the PVN, a phenomenon bit GABAergic synaptic transmission in the PVN (Lee et al., 2008).
that is negatively correlated with CRF content of the median emi- Therefore, the effect of serotonin on PVN neuron function likely
nence (indicative of increased CRF release) and positively corre- varies depending on where afferent fibers terminate as well as
lated with ACTH plasma levels (Chen et al., 2004; McIntyre et al., the presence or absence of gonadal steroid hormones.
1999; Pacak et al., 1993, 1995; Pacak, 2000; Terrazzino et al., The parvocellular PVN receives substantial input from other
1995). Furthermore, catecholaminergic denervation of the PVN re- limbic areas. The bed nucleus of the stria terminalis (BnST), a com-
sults in an attenuated ACTH response to numerous stressors (Gail- plex group of several related subnuclei, have extensive projections
let et al., 1991; Gibson et al., 1986). Accordingly, direct to the PVN (Cullinan et al., 1993; Dong et al., 2001b). These neu-
microinfusion of NE into the PVN results in rapid induction of rons also express estrogen and androgen receptors (Simerly,
the CRF gene (Cole and Sawchenko, 2002; Itoi et al., 1994, 1999; 1993) and may be critically involved in the modulation of HPA
Khan et al., 2007). Expression of adrenergic receptors in the PVN function by sex steroid hormones (vide infra). The majority of these
is limited to alpha adrenergic receptors, and both alpha(1) and al- PVN-projecting neurons are GABAergic (Cullinan et al., 1993). Le-
pha(2) receptors are co-expressed with CRF in the medial parvocel- sion studies indicate that these projections inhibit CRF mRNA lev-
lular PVN (Cummings and Seybold, 1988; Day et al., 1997, 1999; els and corticosterone responses to stress (Dunn, 1987; Herman
Sands and Morilak, 1999). However, it appears that alpha(1a) et al., 1994). However, not all of the BnST neurons send inhibitory
receptors are responsible for the stimulatory effects of NE on CRF projections since selective lesions of the anterior or lateral portions

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 5

of the BnST decrease ACTH secretion (Gray et al., 1993; Herman binding, mRNA and protein expression throughout the adult and
et al., 1994). The neurons of the BnST also receive collateral affer- developing rat brain. In the adult rat brain, both GR mRNA and pro-
ents from neurons in the PVN (Dabrowska et al., 2011) thus raising tein are expressed in the cortex, hippocampus, thalamus, hypothal-
the possibility of extensive feedback loops between the PVN and amus, amygdala, septum, striatum and cerebellum (Ahima and
BnST. Indeed, there is a significant population of CRF producing Harlan, 1990; Cintra et al., 1994; McGimsey et al., 1991; Morimoto
neurons in the BnST that send projections to the PVN (Dong et al., 1996). The cortex and hippocampus both express the highest
et al., 2001b; Dong and Swanson, 2006). Moreover, these CRF pro- levels of GR mRNA and protein. Within the adult rat cortex, GR
jections are functional since they have been shown to enhance mRNA and protein expressing neurons are concentrated in the
GABAergic neurotransmission through the activation of CRF recep- anterior cingulate cortex and retrosplenial cortex (also known as
tor 1 in the bed nucleus of the stria terminalis (Kash and Winder, the posterior cingulate cortex). The cingulate cortex laminar layers
2006). II, III, and VI have been reported to have the greatest concentration
Most limbic brain regions such as the hippocampus, prefrontal of GR with more than 70% of the cells expressing either the mRNA
cortex, medial amygdala and lateral septum that have been shown or protein. (Ahima and Harlan, 1990; Aronsson et al., 1988; Cintra
to modulate HPA axis activity do not directly innervate parvocellu- et al., 1994; Morimoto et al., 1996). In the adult rat hippocampus,
lar PVN neurons and must act through an intermediary synapse. In GR is expressed the highest in the hippocampal regions CA1 and
general, these areas project to areas like the BnST and peri-PVN CA2 and to a lesser extent in the granule cell layer of the dentate
which have direct GABAergic input to the PVN (Dong et al., gyrus, with lowest levels in CA3 (Ahima and Harlan, 1990; Arons-
2001a; Prewitt and Herman, 1998). Thus, the largely glutamatergic son et al., 1988; Cintra et al., 1994; McGimsey et al., 1991; Morim-
output of the hippocampus and prefrontal cortex (Walaas and Fon- oto et al., 1996). Neurons in the PVN also express GR, perhaps at
num, 1980) is translated into an inhibitory tone on the HPA axis the highest level of all nuclei in the hypothalamus. Importantly,
(Diorio et al., 1993; Figueiredo et al., 2003; Herman et al., 1998). GR expressing PVN neurons have been shown to express CRF, vaso-
On the other hand, amygdaloid projections to the BnST and peri- pressin, and most other neuropeptides (Ceccatelli et al., 1989;
PVN are largely GABAergic and can work to decrease the inhibition, Liposits et al., 1987; Sawchenko, 1987a; Uht et al., 1988).
essentially turning up the gain of the system (Swanson and Petro- The subcellular localization of GR protein has also been estab-
vich, 1998). By parsing incoming information through direct inputs lished in neurons of the rat brain. Immunoreactive GR predomi-
or by funneling through inhibitory interneurons, the ability to dis- nantly localizes to the cell nucleus with weak cytoplasmic
tinguish and grade levels of information in a spatial and context localization (Ahima and Harlan, 1990; Lawson et al., 1991; Morim-
dependent fashion appears to be an effective method of integration oto et al., 1996). However, following adrenalectomy, GR is localized
by the PVN. to the cytoplasm (Ahima and Harlan, 1990; Morimoto et al., 1996).
These localization patterns indicate that circulating corticosterone
in the intact animal causes receptor trafficking of the ligand-bound
3. Mineralocorticoid and glucocorticoid receptors
GR to the nucleus whereas, in the absence of ligand, as in the adre-
nalectomized animal, the unbound GR is found in the cytoplasm.
Following secretion from the adrenal cortex, corticosteroids
regulate numerous functions throughout the body, including those
3.2. Mineralocorticoid receptor localization
of the central nervous system. The actions of glucocorticoids are
mediated by two receptors, the type I corticosteroid receptor or
The distribution of MR mRNA and protein in the developing and
mineralocorticoid receptor (MR; also designated NR3C2), and the
adult rat brain overlaps with GR expression (Ahima et al., 1991;
type II corticosteroid receptor or glucocorticoid receptor (GR, also
Lawson et al., 1991; van Eekelen et al., 1991). In vivo binding stud-
designated NR3C1). Both of these receptor types are expressed in
ies using the MR selective ligand, H3-aldosterone suggested that a
multiple regions of the mature and developing rat brain (Ahima
majority of MR binding sites in the adult rat brain are found in the
et al., 1991; Ahima and Harlan, 1990; Cintra et al., 1994; Lawson
hippocampus (Coirini et al., 1985). Similar to GR, MR protein is
et al., 1991; McGimsey et al., 1991; Morimoto et al., 1996; van
highly expressed in the adult hippocampus, in regions CA1 and
Eekelen et al., 1991). Glucocorticoid receptor and MR actions in
CA2 and to a much lesser extent in the CA3 and dentate gyrus.
the adult and postnatal brain are often in opposition, an example
Curiously, the greatest expression of MR is found in CA2 neurons.
of this can be seen when examining corticosteroid effects on cell
Furthermore, MR protein is also expressed at moderate levels in
death mechanisms in the CNS. In both rodents (Almeida et al.,
the cortex, including the cingulate cortex, the hypothalamus and
2000) and in primary cultures of hippocampal neurons (Croche-
subcortical regions. Despite the apparent complete overlap in MR
more et al., 2005) the activation of the GR has been shown to cause
and GR protein in the cingulate cortex, studies have shown that
a neuroendangering phenotype (a situation where cells are put at
MR protein is confined to only the region of the cingulate cortex lo-
risk for subsequent insults that might further impair metabolism),
cated dorsal to the triangular septal nucleus (Ahima et al., 1991).
whereas MR activation has been shown to be neuroprotective. Typ-
In addition to the overlap in GR and MR expression within hip-
ical of all members of the nuclear receptor superfamily, the classi-
pocampus and cortex, GR and MR can be co-expressed within indi-
cal mechanisms employed by the GR and MR to alter cellular
vidual cells (van Steensel et al., 1996). Further, the brain regions or
physiology are through the regulation of gene transcription. Since
cells that express both receptor types are capable of mediating var-
these two receptors share common ligands, the relative binding
ied responses to the endogenous glucocorticoid. Finally, the
affinities for the various natural and synthetic glucocorticoids con-
expression levels of MR and GR are autologously regulated and
fer a portion of receptor specificity for the initiation of downstream
are subject to further modulation by gonadal steroid receptors.
events (Coirini et al., 1985; Sutanto and De Kloet, 1987), in addition
For example, removal of corticosterone by adrenalectomy in-
to location and circuit specific effects due to distinct receptor
creases expression of both MR and GR mRNA in the rat CA1 and
localization.
dentate gyrus regions of the hippocampus (Burgess and Handa,
1993). The adrenalectomy-induced changes in receptor expression
3.1. Glucocorticoid receptor localization can subsequently be modified by concomitant treatment with the
MR selective agonist aldosterone or with estrogen but not andro-
Studies employing autoradiographic binding, in situ hybridiza- gen treatment (Burgess and Handa, 1993; Kerr et al., 1996).
tion (ISH) and immunohistochemical analyses have identified GR Additionally, GR activation by the selective agonist, RU28362, also

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
6 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

attenuates the adrenalectomy induced expression of GR and MR of MR to GR may be important for regulating HPA reactivity as well
mRNA in the CA1 region, but not the dentate gyrus (Chao et al., as stress related behaviors.
1998). The relatively lower affinity of GR for corticosterone (compared
The main endogenous ligands for GR and MR are the glucocor- to MR) is thought to direct its actions toward negative feedback
ticoids, cortisol and corticosterone. Both of these hormones bind following stressors. The GR appears to be largely unoccupied dur-
MR preferentially to GR. The dissociation constants (Kd) of cortico- ing basal glucocorticoid conditions, but rapidly becomes occupied
sterone for the MR are in the <0.5 nM range whereas the Kd for GR when glucocorticoid levels increase following stress (Reul and de
are 4–10 times higher (indicative of a lower affinity) than that of Kloet, 1985; Reul et al., 1990). Therefore, GR activation allows
MR (Reul and de Kloet, 1985; Sutanto and De Kloet, 1987). The the return of HPA activity to baseline following high amplitude
GR and MR are also both able to bind the synthetic glucocorticoid, excursions in corticosteroids, such as following a stressor. Similar
dexamethasone (DEX). However, unlike the endogenous glucocor- to MR, the hippocampal neurons express high concentrations of
ticoids, DEX has greater preference for binding for the GR over the GR, as does the PVN and the anterior pituitary gland (Ahima and
MR (Allen et al., 1988; Brinton and McEwen, 1988; Burgess and Harlan, 1990; Cintra et al., 1994; McGimsey et al., 1991; Morimoto
Handa, 1992). RU28362 is another synthetic glucocorticoid, but it et al., 1996). HPA axis regulation following a stressor is thought to
is a selective agonist for the GR, with no apparent binding affinity be mediated through GR in the hippocampus (Sapolsky et al.,
for the MR (Coirini et al., 1985; Philibert and Moguilewsky, 1983; 1984), amygdala (Shepard et al., 2003) and the hypothalamus
Quirk and Funder, 1988), and a Kd for the GR in the low nanomolar (Feldman and Weidenfeld, 2002), with the stress-related increase
range. in corticosterone acting predominantly upon vasopressin neurons
of the PVN to initiate the negative feedback regulation of the
3.3. Negative feedback regulation of the HPA axis HPA axis (Kovacs et al., 2000). The importance of GR in negative
feedback regulation and behaviors is further emphasized by stud-
The HPA axis is governed by a closed-loop negative feedback ies using transgenic mice. Conditional knockout of GR in the fore-
system typical of most neuroendocrine axes. Glucocorticoid- brain of mice that spare the PVN, causes an increase in basal and
dependent negative feedback control is essential for the termina- stress-responsive corticosterone levels (Kolber and Muglia, 2009)
tion of the stress response and reduces the potential for deleterious indicating the importance of GR in forebrain sites such as the hip-
high amplitude swings in circulating glucocorticoids. Normal HPA pocampus. In contrast, GR overexpression in forebrain did not alter
function is dependent upon glucocorticoid-mediated negative basal ACTH and corticosterone levels, nor their response to mild
feedback which is dose and duration dependent (Abe and stress, thus the level of forebrain GR alone is not the only require-
Critchlow, 1980; Sapolsky et al., 2000). Removal of negative feed- ment setting the basal tone of the HPA axis (Wei et al., 2004).
back by adrenalectomy, for example, results in higher PVN neuro- Selective genetic disruption of GR in the PVN causes enhanced
peptide expression and secretion in both the basal and stimulated CRF immunoreactivity in the PVN and correspondingly increased
states (Imaki et al., 1991; Kovacs et al., 1986; Sawchenko, 1987b). levels of ACTH and corticosterone (Jeanneteau et al., 2012), which
Furthermore, negative feedback can inhibit the system by acting at would be predicted since GR is involved in inhibitory feedback.
the level of the CRF and AVP neurons of the PVN, through cortico- Glucocorticoid actions on HPA function occur in several time
trophs of the anterior pituitary, or indirectly through GR and MR domains. Fast feedback can occur within seconds to minutes,
containing brain regions that project to the PVN (Akana et al., whereas delayed feedback occurs in the minutes to hours time-
1986; Bradbury et al., 1994; Dallman et al., 1987a; Sawchenko, frame. The fast feedback component occurs independent of protein
1987a). The influence of corticosteroids on the anterior pituitary synthesis and is mediated at the cell membrane level and may in-
secretion of ACTH is variable and dependent upon the ligand volve endocannabinoids (Dallman, 2005; Hill and Tasker, 2012;
examined. Corticosteroid effects may further be minimized by Tasker and Herman, 2011) whereas delayed negative feedback is
the presence of a corticosteroid-binding globulin-like molecule driven by changes in gene expression through classical actions of
present in corticotrophs (de Kloet et al., 1977), whereas for some GR (Pecoraro et al., 2006). This latter type of feedback has been
synthetic glucocorticoids, the action is largely at the pituitary shown to modulate Ca2+ dynamics through DNA dependent alter-
(Meijer et al., 1998). ation in voltage gated Ca2+ channels (Joels et al., 2009, 2012; Joels
Since circulating glucocorticoids can bind either GR or MR, both and Karst, 2012). Moreover, in areas like the hippocampus, cortico-
have been implicated in the negative feedback regulation of the steroids can influence both excitatory and inhibitory neurotrans-
HPA axis, where differential sensitivity to a common ligand ap- mission. For example, following corticosterone treatment, CA1
pears to allow selective actions. MR has a particularly high affinity hippocampal neurons have been shown to respond within minutes
for endogenous glucocorticoids and these receptors are predomi- by increasing miniature excitatory postsynaptic current (mEPSC)
nantly bound under baseline levels of corticosterone (Reul and frequency (Karst and Joels, 2005) suggesting spontaneous release
de Kloet, 1985; Reul et al., 1990). The hypothesis that MR regulates of glutamate. These effects of corticosterone appear to involve an
HPA axis activity during the non-stressed state, is supported by MR rather than a GR (for review see Joels et al., 2012) and are
studies showing that adrenalectomy increases basal CRF and ACTH linked to the ERK1/2 pathway (Olijslagers et al., 2008).
levels (Dallman et al., 1985, 1987b; Rabadan-Diehl et al., 1997)
whereas corticosterone replacement at doses that selectively bind 3.4. Circadian influences on the HPA axis
MR returns ACTH levels to normal (Bradbury et al., 1994). More-
over, the hippocampus expresses MR at high levels (Coirini et al., The HPA axis operates under a circadian timing mechanism that
1985), an observation that suggests that this is a predominant site is both diurnal and ultradian in nature. Basal levels of corticoste-
for HPA regulation during the basal state. Consistent with this, MR roids undergo daily fluctuations with peak levels attained near
antagonists administered directly to the hippocampus elevated the time of lights out in rodents, thereby predicting the onset of
basal ACTH and corticosterone levels in a fashion similar to adre- activity and feeding (Kalsbeek et al., 2012). At its peak, the diurnal
nalectomy (Van Haarst et al., 1997). In contrast, in transgenic mice elevations of corticosterone can reach levels that are 5–10 times
that overexpress MR in the forebrain there is a reduction in the cor- that found at its nadir. In rodents and other nocturnal animals,
ticosterone response to restraint stress and concomitant decreases the nadir in corticosterone secretion is found in the morning, after
in anxiety like behaviors in both males and females (Rozeboom lights on, whereas this is reversed in diurnal animals and humans.
et al., 2007). Thus, collectively, these results suggest that the ratio Of importance, ACTH shows a similar, but lower amplitude rhythm

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 7

throughout the day that can vary by up to several fold when mea- Merchenthaler et al., 1980), send processes to the median emi-
sured from trough to peak (Akana et al., 1986; Dallman et al., 1978; nence to release GnRH into the hypothalamo–hypophyseal portal
Kalsbeek et al., 1996). It is thought that this dampened rhythm of vasculature where it ultimately acts upon the gonadotropes of
ACTH compared to corticosterone is partly due to a rhythm in adre- the anterior pituitary to stimulate the release of the gonadotropins,
nal sensitivity to ACTH (Dallman et al., 1978; Kaneko et al., 1980, luteinizing hormone (LH) and follicle stimulating hormone (FSH).
1981). The diurnal rise in glucocorticoids is allowed through a GnRH secretion is pulsatile in nature and this is mirrored by the
reduction in inhibitory tone from the suprachiasmatic nuclei pulsatile release of luteinizing hormone from the gonadotrophs
through projections to the PVN (Kalsbeek et al., 1996, 2012; Sza- of the anterior pituitary (Levine et al., 1982, 1991) but this pattern
farczyk et al., 1983), coupled with an increased adrenal sensitivity is different from that of ACTH since it possesses an interpulse inter-
to ACTH driven by autonomic inputs, thereby enhancing cortico- val of approximately 30 min. in the rat (Levine and Duffy, 1988)
sterone secretion (Oster et al., 2006). Indeed, this concept is sup- compared to 1 h for ACTH (Sarabdjitsingh et al., 2010). The pulsa-
ported by studies showing that splanchnic nerve transection can tile pattern of GnRH is controlled through the upstream actions of a
alter adrenal sensitivity to ACTH (Jasper and Engeland, 1997; Ul- group of neurons that reside in the arcuate nucleus and express the
rich-Lai and Engeland, 2002). neuropeptide, kisspeptin (Li et al., 2009), Kisspeptin has been
Evidence that CRF plays a role in the diurnal rhythm of cortico- shown to be a potent activator of GnRH secretion affecting both
sterone comes from studies showing that the afternoon increase in the surge and pulse modes of secretion (Maeda et al., 2010). The
corticosterone is absent in CRF-deficient mice (Muglia et al., 1997). pulsatile nature of GnRH is essential for normal gonadotrope func-
However, the rhythm is dependent upon the suprachiasmatic nu- tion since studies show that the continuous administration of
cleus (SCN) since lesions of the SCN eliminate the daily rhythm GnRH is unable to stimulate LH secretion (Lincoln et al., 1986;
in corticosterone (Cascio et al., 1987; Ibuka and Kawamura, Southworth et al., 1991).
1975). Projections from the SCN that allow it to regulate diurnal In both sexes, circulating LH stimulates the secretion of steroid
function of the neuroendocrine and pre-autonomic neurons in hormones from the gonads, although the pulses of LH do not nec-
the PVN are thought to be through a vasopressinergic phenotype essarily correspond to elevations in steroid on a one-to-one basis
to the PVN and peri-PVN regions. However, this is still somewhat (Ellis and Desjardins, 1982). In the testis, the receptors for LH are
controversial since AVP gene deletion does not seem to affect the found on Leydig cells which synthesize testosterone (T) following
daily corticosterone rhythm (Abel and Majzoub, 2005). However, LH stimulation.
vasopressin from the SCN inhibits corticosterone release in a noc- In the ovary, steroidogenic cells of the follicle are primarily un-
turnal species (rat) whereas it stimulated corticosterone in a diur- der control of pituitary LH and FSH, which act by binding to their
nal rodent (Kalsbeek et al., 2008). This may be through differential respective high affinity receptors on granulosa and thecal cell
targets of SCN efferent projections carrying vasopressin. Thus, membranes. FSH actions are restricted to the granulosa cells where
although there is a close association between the SCN and PVN, FSH receptors work to activate the aromatase enzyme and allow
the exact mechanisms underlying these interactions have not been synthesis of estrogens from aromatizable androgens (testosterone
completely teased apart. and androstenedione). However, granulosa cells rely upon the the-
Glucocorticoids are also secreted in a pulsatile fashion through- cal cell, whose primary mission appears to be the production of
out the day with an interpulse interval of approximately 1 h (Light- androgens that act as a substrate for estrogen biosynthesis by
man et al., 2000). However, these ultradian secretory patterns do the granulosa cells. Thecal cell production of androgens is under
not appear to be dependent on the circadian pacemaker since pul- regulatory control by LH and this forms the basis of the ‘two cell,
satile secretion is maintained under constant light conditions or two gonadotropin’’ theory of estrogen secretion by the follicle
after SCN lesions (Waite et al., 2012). Nonetheless, the ultradian (Gore-Langton and Armstrong, 1994)
pattern is also thought to be dependent upon hypothalamic CRF.
In this case, CRF activates a pituitary–adrenal network responsible
5. Gonadal steroid receptors
for the oscillations. Corresponding pulses of CRF are not necessary
for the ultradian pattern and elevated levels of CRF appear to dis-
Classical gonadal steroid receptors belong to a ‘‘superfamily’’ of
rupt the network. Thus, unlike other neuroendocrine systems, it
intracellular receptors that act as ligand-activated transcription
is the level, not the pattern of CRF secretion that sets the pulsatile
factors (Evans, 1988). The steroid/thyroid hormone receptor super-
dynamics of glucocorticoid secretion (Walker et al., 2012). In turn,
family consists of three main classes (for reviews see Evans, 1988;
the GR has been shown to have an extremely rapid association and
Mangelsdorf et al., 1995). Class 3 comprises the estrogen receptor
dissociation from DNA following activation (Conway-Campbell
(ER), androgen receptor (AR), progesterone receptor (PR), GR, and
et al., 2007) and its loss from the nucleus reflects the timing of
MR. In addition, gonadal steroids have rapid effects through classic
the 1h pulse of corticosteroid secretion, whereas that of the MR
and non-classic receptors at the membrane or in the cytosol to
did not. Thus, this MR and GR difference further provides for differ-
influence second messenger pathways and ion channel function
ent patterns of MR and GR binding to sites on DNA and allows mul-
which in turn regulates neuronal excitability, regulates cell death,
tiple cell specific responses (Conway-Campbell et al., 2012).
and influences transcriptional activity (Foradori et al., 2008;
Vasudevan and Pfaff, 2008) (Fig. 1).
4. Overview of the hypothalamo–pituitary–gonadal (HPG) axis
5.1. Androgen receptors
Reproduction, the physiological process that is all-important for
the survival of the species, is regulated by a neuroendocrine axis Androgen receptors are responsible for many of the peripheral
that is parallel to the HPA axis and involves the hypothalamus, and central actions of testosterone and its 5-alpha reduced metab-
anterior pituitary gland and gonads. This hypothalamo–pituitary– olite, dihydrotestosterone (DHT).
gonadal (HPG) axis is comprised, at its most fundamental element, The androgen receptor (NR3C4: Nuclear Receptor subfamily 3,
of GnRH expressing neurons that, in the rodent, are located in the group C, member 4) is a ligand-activated transcription factor that
rostral forebrain (medial septum, diagonal band of broca, medial belongs to the nuclear receptor superfamily. It is activated after
preoptic area, anteroventral preoptic (Jennes and Conn, 1994)). binding by either testosterone or DHT, although DHT binds with
These neurons, through several routes (King et al., 1982; greater affinity and has a more powerful transcription activation

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
8 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Fig. 1. Diagrammatic representation of the various intracellular actions of steroid hormone receptors. (1) Steroids can freely diffuse through the plasma membrane lipid
bilayer and bind to a steroid hormone receptor (SHR) associated with heat shock protein (HSP90) in the cytoplasm or nucleus. Steroid binding leads to the release of HSP90
and translocation of the SHR to chromatin DNA. (2) SHR homo- or heterodimers recruit coregulatory proteins such as the p160 and p300 family of coactivators. These
coactivators have intrinsic histone acetyl transferase (HAT) activity and can acetylate histones associated with inactive chromatin to uncoil DNA and expose regulatory
regions and transcription initiation sites of target genes. The SHR dimer then binds to a regulatory region (e.g., hormone response element, HRE) and in association with these
coregulatory protein factors (e.g., SRC, CBP) can stimulate transcription through activation of the pre-initiation complex (PIC). Transcription can subsequently be inhibited by
the activity of histone deacetylases (HDACs), which act to remove acetyl groups from histones and promote chromatin recoiling. (3) Steroid-bound receptor can also act as a
coactivator or corepressor and affect gene expression by tethering to other DNA bound transcription factors (e.g. fos and jun at an AP-1 response element). (4) Steroids have
effects attributable to actions at a membrane-associated steroid hormone receptor (mSHR). These receptors can be classic SHRs tethered to the membrane, G-protein coupled
receptors, or ion channels. These effects are diverse and include alterations in membrane potential and second messenger pathways. AP-1 RE, activator protein 1 response
element; Ca2+, calcium; CBP, cyclic adenosine monophosphate response element binding protein (CREB) binding protein; ERK, extracellular regulated kinase; HAT, histone
acetyl transferase; HDAC, histone deacetylase; HRE, hormone response element; HSP90, heat shock protein 90; MAPK, mitogen-activated protein kinase; mSHR, membrane-
associated steroid hormone receptor; NOS, nitric oxide synthase; PIC, pre-initiation complex; PKA, protein kinase A; PKB, protein kinase B; PKC, protein kinase C; PLC,
phospholipase C; SHR, steroid hormone receptor; SRC, steroid receptor coactivator; TATA, core promoter sequence.

function (Brinkmann, 2011). Subfamily 3 also includes the estro- 5.2. Androgen receptor distribution in brain
gen, glucocorticoid and progesterone receptors as well. Receptors
in this subfamily are characterized by their binding to hormone re- Early studies utilized in vivo autoradiographic approaches with
sponse elements (HREs) on DNA consisting of two half-sites sepa- radiolabeled T and DHT to demonstrate specific binding sites in the
rated by a three nucleotide spacer. The two half-sites represent brain of rodents following administration of a bolus of radiola-
inverted repeats, or palindromes, of each other (Denayer et al., belled hormone to the animal. These autoradiographic approaches
2010). Androgen receptors have also been shown to recognize were able to distinguish high affinity, selective binding sites in the
other non-classical androgen response elements (Denayer et al., hypothalamus which corresponded with androgen’s ability to reg-
2010). ulate reproductive neuroendocrine function and behaviors (Sar and
In the unbound state, androgen receptors are found in the cyto- Stumpf, 1973, 1977). Later studies using binding assays (Handa
plasm of cells (Tyagi et al., 2000), and in neurons they have been de- et al., 1986, 1987; McGinnis et al., 1983; Roselli, 1991) and
scribed in other cellular locations as well (DonCarlos et al., 2003, in situ hybridization (Simerly et al., 1990) or immunocytochemis-
2006; Sarkey et al., 2008). Upon binding to ligand, the cytoplasmic try (Kritzer, 2004) demonstrated a unique distribution for AR in the
androgen receptor translocates to the nucleus and can be identified limbic brain. Highest levels of expression are found in hypotha-
in discrete subnuclear compartments, similar to that described for lamic areas involved in regulating reproduction, such as the medial
estrogen receptors (Htun et al., 1999; Stenoien et al., 2000). preoptic area, ventromedial n. and arcuate n. Moreover, AR was

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 9

also found in extrahypothalamic regions with implications for reg- an anti-inflammatory property (Saijo et al., 2011; Wu et al., 2013).
ulating neuroendocrine stress responses, stress-related behaviors Thus, the widespread distribution of ERs within limbic brain
and autonomic function, such as the BnST, lateral septum, medial regions suggests the possibility that they can interact in a number
amygdala, hippocampus and cortex. Indeed, a role for androgens of ways with neuropeptide and glucocorticoid receptors in the
has been shown for a number of non-reproductive behaviors regulation of the HPA axis.
including cognition and mood (Hawley et al., 2013) as well as mod-
ulating HPA axis reactivity to stress. 5.5. G-protein coupled sex steroid receptors

5.3. Estrogen receptors In addition to its actions in regulating transcription, it has be-
come increasingly clear that estradiol has rapid cellular effects that
The genomic actions of estrogens are mediated by two distinct are mediated by interactions with the cell membrane. To date,
intracellular receptors that function as ligand activated transcrip- although rapid effects of estrogens have been explored in repro-
tion factors. The classical estrogen receptors have been shown to ductive systems, the examination of membrane mediated gonadal
exist in two types, termed estrogen receptor alpha (ERa, NR3A1) steroid signaling related to the HPA axis is largely missing from the
and estrogen receptor beta (ERb, NR3A2). These two estrogen literature. The effects of estradiol to induce rapid changes in sec-
receptor subtypes are encoded by different genes (designated ond messenger pathways and changes in intracellular Ca2+ concen-
ESR1 and ESR2, respectively), and they are classified together with trations have been described (Chaban et al., 2011; Kelly and
glucocorticoid and mineralocorticoid receptors based on their sig- Ronnekleiv, 2012) and these non-genomic actions may actually
naling pathways through DNA binding. work to potentiate the transcriptional effects of estradiol (Vasud-
Similar to other receptors in subfamily 3, the occupancy by evan et al., 2001). Evidence for membrane sites of estradiol action
estradiol causes estrogen receptor dimerization and subsequent come from studies showing rapid effects of estradiol that are pres-
binding to estrogen response elements (ERE) on DNA. ERa and ent even when it is made membrane impermeable by conjugation
ERb share similarity in the DNA binding domains (96% homology) to bovine serum albumin (BSA) (Schmidt et al., 2000; Vasudevan
and ligand binding domains (56% homology) and bind to the same and Pfaff, 2008). Estradiol treatment has also been shown to poten-
response elements on DNA (Kuiper et al., 1996). In addition, non- tiate NMDA receptor excitation in hypothalamic neurons (Kow
classical mechanisms have been described where ERs can enhance et al., 2005), augment kainate-induced inward currents (Gu and
transcription through tethering to other transcription factors. An Moss, 1996) and initiate Src/ERK signaling pathway cascades in
example of this is the differential interactions of ERa and ERb with hippocampal neurons in vitro (Wu et al., 2005). Such data demon-
c-Fos, a protein that modulates transcription through the Activator strate that estradiol has actions too rapid to be dependent upon de
Protein complex (AP-1). Using a simple promoter containing multi- novo protein synthesis, a concept first pioneered by Szego and Da-
ple EREs, Paech et al. (1997) were able to show that ERa can acti- vis (1967) in uterine tissues and later by Kelly et al. (1977) for
vate promoters through an adjacent AP-1 site following binding by hypothalamus.
ER agonists such as estradiol and diethylstilbestrol (DES). In con- The identity of the estrogen receptors that mediate the rapid ef-
trast, after estradiol binding, ERb inhibited transcription through fects of estradiol are currently being elucidated. A membrane local-
the AP-1 site. Furthermore, when bound by an antagonist, ERb ization of the classical estrogen receptors has been demonstrated
was a potent transcriptional activator at an AP-1 site. For ERb, this by some investigators (Levin, 2005) and the non-selective ER
is also a function of the splice variant expressed (Handa et al., antagonist, ICI 182,780, can block estradiol’s rapid induction of cal-
2012; Price et al., 2001). Thus, the selectivity of the ER signal is modulin/kinase activity in hippocampal neurons (Chaban et al.,
dependent, not only upon receptor subtype, but also the ligand 2004). Further evidence that classical estrogen receptors can have
and DNA element(s) involved. Similar effects have been shown membrane actions comes from studies using a double ER knockout,
(vide infra) for endogenous promoters such as CRF (Chen et al., which do not express ERa or ERb, and which lose the ability to re-
2008; Lalmansingh and Uht, 2008; Miller et al., 2004), vasopressin spond to estradiol with the phosphorylation of ERK (Abraham
(Pak et al., 2007), GnRH (Pak et al., 2006), and Oxytocin (Hiroi et al., et al., 2004). However, these types of responses in knockout ani-
2013). mals should be interpreted cautiously since ERs control the expres-
sion of a number of genes that may help initiate the membrane
5.4. Estrogen receptor distribution in brain responses. Lastly, recent evidence has also demonstrated that
ERa can be physically associated with metabotropic glutamate
Estrogen receptors are expressed throughout the rostral–caudal receptors (Micevych and Mermelstein, 2008). ERs are organized
extent of the brain and spinal cord (Simerly and Young, 1991). ERa with metabotropic glutamate receptors into membrane signaling
and ERb possess overlapping patterns of expression. Brain regions domains via caveolin proteins in hippocampus (Luoma et al.,
important for HPA and autonomic circuitry that express both forms 2008). Estradiol binding to these membrane ERs can thereby affect
of ER include the BnST, medial and cortical amygdaloid nuclei, metabotropic glutamate signaling independent of glutamate
preoptic area, arcuate n., parabrachial nucleus, locus ceruleus, (Grove-Strawser et al., 2010; Meitzen and Mermelstein, 2011;
nucleus of the solitary tract. Striking differences in expression Mermelstein, 2009) thus providing additional evidence for mem-
patterns between ERa and ERb are seen in other brain areas. For brane sites of estrogen action.
example, ERa alone is found in the subfornical organ and ERa is That estradiol can also have effects on homeostatic mechanisms
more abundant in the arcuate n. and ventromedial n. In contrast, through modulation of G-protein coupled receptors (GPCRs) has
ERb is found exclusively in the supraoptic n., paraventricular n., been suggested. Effects of estradiol on autonomic responses, such
suprachiasmatic n., and pineal gland. ERb appears to be greater as control of feeding and core body temperature, can be mimicked
in the hippocampus and adult cortex (Chu and Fuller, 1997; by Gaq-signaling ligands like the diphenylacrylamide compound,
Laflamme et al., 1998; Shughrue et al., 1996a). Several studies have STX, further implicating specific estradiol effects through G-pro-
also demonstrated that astrocytes and oligodendrocytes can ex- tein coupled receptors in controlling hypothalamic function (for
press ERa and ERb (Azcoitia et al., 1999; Mhyre and Dorsa, 2006; review see Kelly and Ronnekleiv, 2013).
Platania et al., 2003; Santagati et al., 1994; Zhang et al., 2004) Studies have also described an orphan member of the G-protein
although the function of these receptors remain unknown. Recent coupled receptor family, GPR-30 or G-protein coupled estrogen
studies examining ERb in microglia and astrocytes have suggested receptor (GPER) that is an integral membrane protein with a high

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
10 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

affinity for estradiol (Brailoiu et al., 2007; Filardo et al., 2002). GPER stress-related neural input with current endogenous glucocorti-
has been shown to couple with Gas to activate adenylyl cyclase coid levels and reproductive status of the animal.
(Prossnitz et al., 2008; Thomas et al., 2005). GPER has also been In addition to these direct effects of estrogen on HPA function,
localized to the endoplasmic reticulum which puts it into an excel- the stress response can be indirectly influenced through changes
lent location for regulating intracellular calcium mobilization and in the expression of circulating proteins that modulate corticoste-
synthesis of phosphatidylinositol 3,4,5-triphosphate (Revankar roid bioavailability following secretion by the adrenal gland. Corti-
et al., 2005). GPR30 agonists can mimic the desensitizing effects costeroid binding globulin (CBG) is a glycoprotein produced in
of estradiol on the corticosterone response to the 5-HT1A receptor liver. It has a relatively high binding affinity for corticosteroids
agonist (+)8-OH-DPAT, and knockdown of GPR30 prevents the and as a result, most corticosteroids in the circulation are found
estradiol induced decreases in 5-HT1A protein in PVN neurons that bound to CBG (Westphal, 1983). Only free unbound hormone is
accompany this response (McAllister et al., 2012). available for binding the intracellular receptor or subject to hepatic
Androgens have also been reported to have rapid membrane degradation (Rosner, 1990). Thus, a reported sex difference in the
mediated actions on a number of neuronal cell phenotypes (Fora- levels of CBG, where females have greater CBG concentrations than
dori et al., 2008; Tabori et al., 2005), and a membrane associated males (Westphal, 1971) could act to modulate tissue availability of
AR similar to the classic AR has been reported in the GT1–7 hypo- corticosteroids to brain and thereby influence HPA axis function.
thalamic cell line (Shakil et al., 2002). Androgens and androgen
metabolites, such as 3a-diol have been shown to rapidly affect 6.2. HPA axis function across the reproductive cycle
some behaviors, although whether this is through an androgen
receptor mediated mechanism is currently debatable (Frye et al., Early studies examining sex differences in HPA axis function in
2010). At present, there is only sporadic evidence for a membrane rodents described elevated levels of corticosterone in females com-
androgen receptor in brain, whereas a membrane androgen recep- pared to males (Critchlow et al., 1963). This difference is in part
tor has been described in prostate, colon and breast tumor cells (Gu due to the presence of gonadal steroid hormones since there are
et al., 2011; Papadopoulou et al., 2009), and BSA conjugated DHT differences in HPA axis function across the estrous cycle of female
has been shown to have a number of actions on the ERK/MAPK rats where the ACTH and corticosterone responses are greatest on
and Akt signaling pathways in these tissues as well as in brain proestrus, when estradiol levels are greatest, compared with estrus
(Gatson and Singh, 2007; Sato et al., 2010). Thus, evidence exists and diestrus (Viau and Meaney, 1991; Walker et al., 2001).
for rapid membrane-initiated signals that may be mediated by a Moreover, estradiol and estradiol plus progesterone treated
membrane AR (Schmidt et al., 2012) but the protein has yet to be
clearly elucidated (for review, see Foradori et al., 2008).

6. Gonadal steroid control of the HPA axis

6.1. Sex difference in HPA axis function

Numerous reports have indicated that the function of the HPA


axis is different between the sexes, although the direction of this
difference is sometimes dependent upon the species being exam-
ined. In rodents, basal and stress-induced adrenal glucocorticoid
secretion has been reported to be greater in females than in males
(Critchlow et al., 1963; Handa et al., 1994a; Kitay, 1963). Activation
of the stress response and of PVN neurons is reported to be higher
in females than males (Larkin et al., 2010; Seale et al., 2004; Viau
et al., 2005), and this could be due to variations in estradiol levels
that occur across the estrous cycle of females (Iwasaki-Sekino
et al., 2009; Rhodes et al., 2004; Viau and Meaney, 1991). This
has been further supported in studies examining the pulsatile pat-
terns of corticosterone secretion, where females show a higher
amplitude, frequency and number of corticosteroid pulses com-
pared to males (Seale et al., 2004). Moreover, these sex differences
appear to be modulated, not only by the adult gonadal steroid
environment, but also after neonatal treatment with testosterone
suggesting an organizational effect of gonadal steroids on the neu- Fig. 2. Distribution of gonadal and adrenal steroid hormone receptors in relation-
ship to inputs to HPA circuitry. Gonadal and adrenal steroid hormone receptors
ral substrate controlling corticosteroid pulses (Seale et al., have considerable overlap in expression within brain regions that have direct inputs
2005a,b). To date, the mechanism(s) by which gonadal steroid hor- to the paraventricular nucleus (PVN) and peri-PVN. The brain regions involved and
mones may act to influence HPA function have not been com- the overall influence on the output of the PVN depends upon stressor modality
pletely resolved. Evidence for androgens and estrogens acting in (psychogenic, limbic; homeostatic, brain stem) and gonadal steroid hormone levels
derived from systemic and/or local de novo sources. Green solid arrows indicate
part, by modulating adrenal (Kitay, 1965), anterior pituitary (Coy-
excitatory glutamate connections. Red dashed arrows indicate inhibitory gamma-
ne and Kitay, 1969, 1971; Viau and Meaney, 2004) and hypotha- aminobutyric acid (GABA) connections. Black arrow indicates serotonergic connec-
lamic functions (Handa et al., 1994b; Viau and Meaney, 1996; tions. Blue arrow indicates mixed norepinephrine and glucagon-like peptide 1
Viau et al., 2001) have been shown. This is exemplified by the con- connections. Purple arrow indicates mixed GABA and arginine vasopressin
siderable overlap in gonadal and adrenal steroid hormone receptor connections. AMY, amygdala; AR, androgen receptor; BNST, bed nucleus of the
stria terminalis; ERa, estrogen receptor alpha; ERß, estrogen receptor beta; GR,
expression within the neural circuitry of the PVN (Fig. 2). Its prox- glucocorticoid receptor; MPOA, medial preoptic area; MR, mineralocorticoid
imity to the third ventricle, dense vascularization, and expression receptor; NTS, nucleus of the solitary tract; PFC, prefrontal cortex; PVN, paraven-
of adrenal and gonadal steroid receptors enable the PVN to weigh tricular nucleus; Raphe, dorsal raphe nucleus; SCN, suprachiasmatic nucleus.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 11

animals secreted more corticosterone after stress termination of the function of the PVN by estradiol can occur through diverse
(Viau and Meaney, 1991). Therefore, it appears that enhanced re- pathways.
sponses to stress are most prevalent in the early part of proestrus The role for ERb in HPA axis regulation has been explored by the
when estradiol levels are greatest. Progesterone may reduce the ef- use of ER subtype selective compounds. Treatment of ovariecto-
fects of estradiol on HPA axis function as illustrated by studies mized females with the ERb selective agonist, diarylpropionitrile
showing that it prevents E-induced increases in CRF mRNA in the (DPN), significantly decreases the CORT and ACTH response to
PVN of female monkeys (Roy et al., 1999) and inhibits ACTH release stressors (Lund et al., 2005). This effect can also be seen when
in ewes (Keller-Wood, 1998). Moreover, the neurosteroid, tetrahy- DPN is placed directly adjacent to the PVN (Lund et al., 2006), an
droprogesterone can act to prevent the adrenalectomy (ADX)-in- effect that is blocked by the ER antagonist, tamoxifen, thus sug-
duced upregulation of AVP mRNA in the PVN of rats (Patchev gesting a direct action through ERb containing neurons in the
et al., 1996). Thus, it appears that progesterone functions to inhibit PVN. The effects of systemic administered DPN on the CORT and
HPA function at certain times of the cycle. Whether or not these ef- ACTH response to stress are also present in wild type, but not in
fects are mediated by progesterone receptors, or are neurosteroid- ERb knockout female mice (Oyola et al., 2012), and the effects of
like effects of progesterone metabolites (Brunton and Russell, DPN can be replicated following treatment with other ERb selective
2011) remains to be determined. agonists (Weiser et al., 2009). Taken together, these data demon-
strate that the actions of ERb activation are to reduce the gain of
the HPA axis response to an acute stressor.
6.3. Estrogen regulation of the HPA axis and mechanisms of action It is also worthwhile noting that adrenal steroids can influence
the expression of ERb, thereby altering the effect of ERb signaling
In female rodents, ovariectomy reduces stress-induced CORT on the HPA axis in a parallel feedback fashion. For example, dexa-
and ACTH secretion and this effect is reversed by estradiol treat- methasone treatment of ovariectomized rats increases ERb expres-
ment (Burgess and Handa, 1992; Handa et al., 1994a; Serova sion in the PVN (Suzuki and Handa, 2004). Similarly, removal of
et al., 2010; Suzuki et al., 2001a; Weiser and Handa, 2009). Consis- endogenous glucocorticoids by adrenalectomy reduces ERb mRNA
tent with this, implants of a specific estrogen receptor antagonist levels in the PVN of female rats, and CORT replacement reverses
reduce levels of CORT following restraint stress (Isgor et al., this effect (Isgor et al., 2003). However, up-regulation of ERb mRNA
2003), whereas infusion of estradiol into the brain increases corti- was observed only during proestrous when estradiol levels are
costerone responses (Liu et al., 2012; Lund et al., 2006). However, high. These data suggest that elevated adrenal steroid levels fol-
this effect of estrogen is not always consistent and several groups lowing a stressor may act to increase ERb expression within the
have reported that estrogen can inhibit neuroendocrine responses PVN thereby dampening HPA axis reactivity to a subsequent
to stress (Figueiredo et al., 2002; Ochedalski et al., 2007; Young stressor.
et al., 2001) or have no effect (Babb et al., 2013). Such variation In contrast to the effects of ERb agonists on stress-induced glu-
is likely due to the dose and duration of estradiol treatment or cocorticoid secretion, treatment with the ERa agonist, propylpy-
the length of time the animal is ovariectomized prior to estradiol razoletriol (PPT) increases the CORT and ACTH response to
treatment (Young et al., 2001). Finally, recent studies indicate that restraint stressors (Liu et al., 2012; Lund et al., 2005, 2006; Serova
the effect of estradiol can also be influenced by the ER signaling et al., 2010; Weiser and Handa, 2009). Similarly, treatment with
pathway involved since ERa and ERb can have opposing actions PPT increases stress-induced c-fos mRNA expression in the PVN,
in regulating HPA axis function (vide infra). an effect that is also blocked by concomitant treatment with
Insight into the mechanism whereby estradiol modulates HPA tamoxifen (Lund et al., 2006). The effects of ERa activation on
axis activity can be deduced by identifying the locations and phe- HPA axis activity have been explored in much less detail than that
notypes of neurons that express the various forms of ER. ERb is ex- of ERb. However, the results of studies by Weiser and Handa
pressed in some populations of CRF, vasopressin, and oxytocin (2009), indicate that ERa is found in GABAergic peri-PVN neurons
containing neurons in the hypothalamus (Hrabovszky et al., and that estradiol can work at the level of the peri-PVN to block the
2004; Laflamme et al., 1998; Suzuki and Handa, 2004). Approxi- negative feedback regulation of the HPA axis by dexamethasone.
mately 85% of oxytocin immunoreactive (-ir) neurons in the PVN These data raise the intriguing possibility that ERa acts to inhibit
have been shown to co-express ERb (Hrabovszky et al., 2004; Suzu- the negative tone on HPA axis that is provided by GABAergic neu-
ki and Handa, 2004). ERb is also expressed by a smaller population rons in the peri-PVN, thereby effectively enhancing the gain of the
of CRF-ir neurons in the medial parvocellular PVN and in much lar- HPA axis. Such a mechanism (reduction of GABAergic inhibition) is
ger numbers of CRF expressing neurons in the caudolateral PVN consistent with the mechanism proposed for the increase in hippo-
(60–80%; Laflamme et al., 1998). Most of these CRF/ERb expressing campal spine density seen following estradiol treatment (Murphy
neurons appear to be pre-autonomic neurons, however. ERb is also et al., 1998).
expressed in vasopressin-ir neurons of the PVN (Sladek and Som-
ponpun, 2004; Somponpun and Sladek, 2002; Suzuki and Handa, 6.4. Androgen regulation of the HPA axis and mechanisms of action
2004). Such data implicate ERb as a cellular mediator of the direct
actions of estradiol on PVN function. Evidence from physiological studies using gonadectomized and
In contrast to ERb, ERa is not expressed at very high levels by hormone replaced male rodents indicates that androgens provide
PVN neurons (Laflamme et al., 1998; Simonian and Herbison, an inhibitory influence on HPA axis activity (Handa et al., 1994b;
1997; Suzuki and Handa, 2005; Weiser and Handa, 2009), but Viau and Meaney, 2004). Gonadectomy of male rats results in ele-
rather is found at high levels in brain regions that send direct vated stress induced CORT and ACTH secretion and this effect is
and indirect projections to the PVN such as the peri-PVN, BnST, reversible with testosterone or DHT treatment (Handa et al.,
medial preoptic area, lateral septum, and hippocampus (Shughrue 1994b; Viau and Meaney, 2004). Androgen treatment has consis-
et al., 1998; Suzuki and Handa, 2005). The limbic distribution of tently been shown to inhibit HPA axis reactivity in rodents, mon-
ERa overlaps considerably with ERb. Moreover, under certain cir- keys and humans (Handa et al., 1994b; Kalil et al., 2013;
cumstances, such as following chronic food restriction, immunore- Rubinow et al., 2005; Toufexis and Wilson, 2012; Williamson and
active ER (presumably ERa) has been shown to be expressed by Viau, 2008). Further, treatment with the non-aromatizable andro-
some PVN neurons (Estacio et al., 1996). Therefore, the differing gen, DHT, can also inhibit stress-induced activation of c-fos mRNA
distribution patterns between ERa and ERb suggest that regulation in the PVN (Lund et al., 2004b; Seale et al., 2004) suggesting that

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
12 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

androgens reduce the gain of the system. The changes in restraint- the synthesis of DHT from 3b-diol is thought to be minimal be-
induced CORT secretion are not accompanied by changes in pitui- cause 3b-diol is either irreversibly hydroxylated at C-6 and/or C-
tary sensitivity to CRF (Handa et al., 1994b) nor are there changes 7 positions or is oxidized to (epi) androsterone (Becker et al.,
in circulating corticosteroid binding globulin (Lund et al., 2004a), 1973; Gemzik et al., 1992; Ofner et al., 1983), although a recent re-
thus indicating that these effects of androgen are centrally port indicates that this retro conversion may occur in prostate can-
mediated. cer cells (Ishizaki et al., 2013). Interestingly, 3b-diol exhibits very
Accompanying the suppression of HPA axis reactivity by weak affinity for the AR (T. Lund and R. Handa, unpublished), yet
androgens is a reduction in CRF-immunoreactivity (ir) in the selectively binds and transactivates ERb (Kuiper et al., 1997).
PVN (Bingaman et al., 1994). Again, by focusing on the distribu- Importantly, it has been suggested that 3b-diol is the endogenous
tion of androgen receptors (AR) one can rule in or out some po- ligand for ERb in the male since studies in prostate gland show that
tential sites of androgen action. Androgen receptors are not it inhibits growth in a way that counteracts the growth promoting
localized in neuroendocrine neurons within the PVN (Bingaman actions of its precursor, DHT (Weihua et al., 2002).
et al., 1994; Bingham et al., 2006), but rather, AR-ir neurons are The conversion of DHT to 3b-diol involves any one of several en-
found in the dorsal and the ventral medial parvocellular parts zymes, 3a-HSD, 3b-hydroxysteroid dehydrogenase (3b-HSD), 17b-
of the PVN, which are non-neuroendocrine neurons that project hydroxysteroid dehydrogenase (17b-HSD), 20a-hydroxysteroid
to spinal cord and brainstem pre-autonomic nuclei (Bingham dehydrogenase (20a-HSD) (Gangloff et al., 2003; Jin and Penning,
et al., 2006). Studies show that the implantation of testosterone 2001; Steckelbroeck et al., 2004; Torn et al., 2003; Weihua et al.,
(T) into the medial preoptic area (MPOA) and BnST can reduce 2002). Of interest, the expression of the mRNAs for many of the en-
the CORT response to acute stress (Viau and Meaney, 1996; Viau, zymes in this pathway, including 5aR and cyp7b1 (the enzyme
2002; Williamson and Viau, 2008). Because these brain regions responsible for the metabolism of 3b-diol), have been demon-
provide afferent input to the PVN, or the peri-PVN, it has been strated in the PVN (Lund et al., 2006). This raises the unique possi-
postulated that androgens act through androgen receptors to reg- bility that biological effects, including effects on HPA axis activity,
ulate PVN neuropeptide expression and secretion through a trans- once attributed to DHT binding to the AR may in part be mediated
synaptic mechanism. In support of this, retrograde tracing studies through conversion to 3b-diol and binding to ERb. Hence, the local
show that AR-ir are found in neurons of the BnST, MPOA and regulation of ligand selection prior to receptor binding and activa-
anteroventral periventricular n. that project to the PVN (Suzuki tion become important control points for determining which
et al., 2001a; Williamson and Viau, 2007). However, these areas receptor pathways will be activated.
may not be the only brain site(s) mediating androgen’s inhibitory Similar to the actions of DHT, 3b-diol is a potent inhibitor of
effect on HPA reactivity. Androgen treatment has been reported HPA axis reactivity (Lund et al., 2004b, 2006). Treatment of gonad-
to attenuate the serotonergic activation of the HPA axis perhaps ectomized mice or rats with 3b-diol can reduce plasma CORT and
through modulation of hippocampus or lateral septum (Goel ACTH responses to stress (Lund et al., 2004a). This effect is not
et al., 2011). Moreover, stereotaxic application of DHT to a region present in ERb null mice (M. Oyola, S. Mani, unpublished observa-
just above the PVN (to prevent mechanical disruption of the PVN) tion). This effect of 3b-diol on HPA activity can also be seen after
was as effective as peripherally administered DHT in inhibiting central administration to areas adjacent to the PVN (Lund et al.,
HPA function (Lund et al., 2006). Such results raise the possibility 2006). In further support of an ER-mediated mechanism, Lund
that androgens can work at multiple brain sites to regulate the et al. (2006) showed that the effects of 3b-diol and DHT are not
gain of the HPA axis. blocked by the AR antagonist flutamide, but rather, are blocked
with tamoxifen, an ER antagonist. Thus, these studies provide an
6.5. Steroid metabolism and pre-receptor regulation of HPA axis interesting twist to the emerging story concerning the regulatory
function mechanisms whereby gonadal steroids influence HPA axis gain.
Adjusting the ligand identity prior to receptor binding can work
It is now well established that testosterone, the principle circu- to direct activity through different receptor pathways and thereby
lating androgen in males, can be intracellularly converted to estra- fine tune hormone signaling events.
diol in brain tissue by the aromatase enzyme (Roselli et al., 1985, Additionally, 3a- and 3b-diol may have biological actions other
1997), or to DHT by the 5a-reductase enzyme (5aR; Lephart, than through activation of steroid hormone receptors. In particular,
1993). Although both testosterone and DHT bind the AR with high and similar to other 3a-tetrahydrosteroids, such as 3a-hydroxy-
affinity, DHT has classically been used in studies of androgen ac- pregnane neurosteroids, 3a-diol may be an allosteric mediator of
tion as it is considered to be a more potent and selective agonist the ionotropic GABAA receptor (Belelli and Lambert, 2005; Lambert
for ARs and is not a substrate for aromatization to estradiol. How- et al., 2001). These effects of 3a-tetrahydrosteroids are not shared
ever, whether the actions of androgen require such pre-receptor by the 3b-diastereomers, such as the 3b-hydroxypregnanes, which
selection of ligand to allow receptor specific actions remains to may be direct noncompetitive blockers of GABAA receptors (Wang
be determined. 5a reduction of testosterone to DHT is thought to et al., 2002). Whether these latter effects hold true for 3b-diol, and
be a necessary step for the inhibitory effects of androgens on are relevant for its actions in regulating HPA axis activity, remains
HPA function since inhibition of 5a-reductase with finasteride to be determined.
treatment can increase the HPA reactivity to stress in intact males
in a fashion similar to gonadectomy. Furthermore, central adminis-
tration of finasteride can block the effects of testosterone treat- 7. Molecular mechanism for gonadal steroids in controlling the
ment on HPA axis reactivity (Handa et al., 2013), suggesting HPA Axis
testosterone’s effect is mediated centrally by DHT.
Under most conditions, DHT is intracellularly reduced to 5a The subfamily of nuclear receptors that contain the steroid hor-
androstane-3a,17b-diol (3a-diol) or 5a-androstane-3b,17b-diol mone receptors are characterized by their ability to regulate tran-
(3b-diol) (Jin and Penning, 2006; Rizner et al., 2003; Steckelbroeck scription by interacting with select DNA response elements. The
et al., 2004). Animal studies show that oxidative 3a-hydroxyster- classically described DNA element is the inverted repeat, a nucleo-
oid dehydrogenase (3a-HSD) activity can convert 3a-diol back to tide sequence that is the reversed complement of another down-
DHT and this represents an alternative pathway for DHT synthesis stream sequence, in which both sequences are separated by a
(Ishizaki et al., 2013; Shaw et al., 2006). However, unlike 3a-diol, variable number of nucleotides. Alternatively, steroid hormone

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 13

receptors can act via composite elements, which consist of a hor- 3b-diol potentiates ERb mediated AVP promoter activity (Pak
mone response element half-site and a half-site for a monomer et al., 2007), and 3b-diol induced activity may be greater than that
of another transcription factor (Malkoski et al., 1997). This type of estradiol. In contrast to 3b-Diol, DHT has been shown to sup-
of element has been implicated in the negative glucocorticoid reg- press the AVP promoter through an androgen receptor dependent
ulation of the CRF promoter (Malkoski and Dorin, 1999). Finally, an mechanism. Thus, the gonadal steroid regulation of the AVP pro-
‘alternative’ pathway involves a third type of element, a tethering moter is dependent upon the type of steroid receptor present in
element, by which one transcription factor regulates, through pro- the neuron (e.g. AR, ERa, ERb) as well as the identity of the ligand
tein:protein interactions, the activity of another transcription fac- involved (i.e. DHT, 3b diol, E2), further emphasizing pre-receptor
tor that is bound to its DNA-binding site. A good example of this regulatory mechanisms as important factors to consider when
mechanism is seen when ER associates with an AP-1 or SP-1 bound examining receptor mediated control of neuronal function.
transcription factor (Kushner et al., 2000; Paech et al., 1997; Safe
and Kim, 2008) to alter its specificity. All three of these interactions 7.3. Oxytocin regulation by estrogen receptors
can be implicated in the estrogen regulation of neuroendocrine
neuropeptides (for review see: Handa et al., 2011) that are in- Increasing evidence shows that oxytocin is anxiolytic and inhib-
volved in the control of HPA axis reactivity. its the activity of the HPA axis both in females and males (Neu-
mann et al., 2000b; Windle et al., 1997, 2004) when acting at the
7.1. CRF regulation by estrogen receptors level of the PVN. Oxytocin treatment also reduces activation of par-
vocellular PVN neurons in a similar pattern to that shown for ERb
The molecular mechanisms for the regulation of CRF by estra- agonists (Lund et al., 2006). For example, ICV administration of
diol was initially described by Vamvakopoulos and Chrousos oxytocin, decreased anxiety related behaviors, the CORT and ACTH
(1993) who showed that the human CRF promoter is devoid of response to stress, and the induction of c-Fos expression in neu-
classical EREs but ERE half-sites are present. Moreover, earlier rons of the PVN (Ochedalski et al., 2007; Windle et al., 2004,
studies by Seasholtz et al. (1988) demonstrated that CRF expres- 2006). Oxytocin knockout mice show enhanced CORT secretion
sion is dependent upon a cAMP response element (CRE) in the (Mantella et al., 2004) and increased CRF expression in the PVN
proximal promoter. The cAMP regulatory element binding protein (Mantella et al., 2004; Nomura et al., 2003) and central blockade
(CREB) and steroid receptor coactivators (SRC) were found to inter- of OTR with des Gly-NH(2) d(CH(2))(5) [Tyr(Me)(2),Thr(4)] OVT,
act with CREB binding protein (CBP) (McKenna et al., 1999) to reg- enhances HPA reactivity (Neumann et al., 2000a,b). Together these
ulate transcription through a CRE. Based on these discoveries, ER studies provide evidence that oxytocin is involved in the central
regulation of gene expression was shown to involve the formation regulation of the HPA axis and thus, oxytocin neurons may be a tar-
of an ER:SRC:CBP complex (Kushner et al., 2000) and this mecha- get for estrogen action.
nism remains a possibility for the estrogenic regulation of the It has long been known that the oxytocin promoter is under
CRF promoter as well. estrogen regulation (Burbach et al., 1994; Richard and Zingg,
By using a CRF promoter:reporter gene construct, it was shown 1990, for review see Burbach et al., 2001). The primary site of
that the CRF promoter could be activated through estradiol binding estrogen regulation appears to lie in a complex composite response
to ERb (Miller et al., 2004). Importantly, the anti-estrogen, tamox- element that exists between nucleotides 72 to 148 in the rat OT
ifen could also activate the promoter through the wild type ERb, promoter (Adan et al., 1993; Richard and Zingg, 1990). This com-
raising the possibility that CRF promoter regulation involves an posite response element is consistent across many species and it
alternate pathway (Miller et al., 2004). Both ERa and ERb occu- contains a variant of an ERE but with no spacing (Burbach et al.,
pancy the region of the CRE within the CRF promoter increase very 1998). Both the human and rat oxytocin promoters can be stimu-
quickly following E2 treatment and this corresponded with SRC-1 lated through this site by ERs, thyroid hormone receptors (THRs)
and CBP occupancy (Lalmansingh and Uht, 2008). Estradiol and and retinoic acid receptors (RARs; Adan et al., 1993; Burbach
DPN have also been shown to increase CRF promoter reporter gene et al., 1998; Richard and Zingg, 1990, 1991). Koohi et al. (2005)
activity in hypothalamic cells whereas the ERa agonist, PPT did not showed that E2 and ER antagonists bound ERa and activated the
(Ogura et al., 2008). Evidence for regulation of the CRF promoter by bovine OT promoter but not through direct DNA binding, suggest-
other ligands that bind ERb has also been demonstrated (Huang ing an alternate pathway. Furthermore, ERa and ERb are differen-
et al., 2008). 3b-diol also increases CRF promoter activity in a fash- tially recruited to the OT promoter in a ligand dependent fashion.
ion equivalent to that of estradiol suggesting an ERb mechanism. Using chromatin immunoprecipitation, Sharma et al. (2012)
These effects could be blocked by tamoxifen treatment thus con- showed that both 3b-diol and E2 increased ERb occupancy of the
firming ER involvement. Together, these data demonstrate that composite response element, but with differing time courses that
the regulation of the CRF promoter can be activated by ERb and corresponded with their relative efficacy in increasing oxytocin
that this may involve an alternate pathway of gene regulation. transcript expression. Promoter occupancy by ERa was not appar-
One caveat to this mechanism is the finding that many CRF ent after 3b-diol and E2 treatment. This pattern of receptor occu-
expressing neurons in the PVN do not contain ERa or ERb, thus pancy of the oxytocin promoter is consistent with the ability of
the significance of these findings is partly tempered by the some- ERb to increase oxytocin expression in the rat PVN (Hiroi et al.,
what restricted overlap between the receptors in question and 2013) and activate a human oxytocin promoter/reporter gene
the neuropeptide targets (Laflamme et al., 1998; Suzuki and Handa, construct (Hiroi et al., 2013). The effect of ERb on the oxytocin pro-
2005). moter is mediated through the composite response element at
approximately 160 since targeted mutation of the response
7.2. Vasopressin regulation by gonadal steroid receptors element eliminated the response. Moreover, 3b-diol is slightly
more effective than estradiol in increasing oxytocin promoter
The differential regulation of the vasopressin promoter by ERa activity through ERb even though it has a relative binding affinity
and ERb were originally examined by Shapiro et al. (2000) who that is much less than estradiol. Thus, the pattern of promoter acti-
showed ERa and ERb activation through an upstream ERE. ERb also vation by estradiol and 3b-diol is consistent with the neuroana-
possesses constitutive activity upon the AVP promoter which is not tomical findings showing that ERb is expressed highly in
shared by ERa, although the degree to which this occurs is still oxytocin neurons of the PVN (Hrabovszky et al., 2004; Suzuki
arguable (Pak et al., 2007; Shapiro et al., 2000). Treatment with and Handa, 2005). Although ERa may also drive the OT promoter,

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
14 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

the absence of its expression in OT neurons (Hrabovszky et al., report some expression of ERa in 5-HT neurons of the rostral dorsal
2004; Suzuki and Handa, 2005), indicates that this may be a lit- raphe nucleus of mice. These findings suggest that estradiol acts
tle–utilized pathway, at least for the rodent PVN. Together, these predominantly through ERb to modulate serotonin neurotransmis-
findings support the contention that ERb may be involved in regu- sion in the dorsal raphe to regulate behaviors, and this may also be
lating PVN function and this may underlie the effects of ERb ago- reflected by changes in HPA activity.
nists on HPA reactivity. Studies utilizing subtype specific ER agonists have generally
The actions of OT are mediated by the OT-receptor (OTR), a supported the initial observations using knockout animals. In the
member of the G-protein-coupled receptor super-family. OTRs open field test, ovariectomized females show more anxiolytic
are closely related to the AVP receptors and together are classified behaviors in the open field test and elevated plus maze following
as V1a, V1b, V2 or OTR. The V1b receptor and OTR are found exten- treatment with the ERb agonists, DPN and WAY200070 (Lund
sively in brain and modulate behaviors and HPA axis functions et al., 2005; Walf and Frye, 2005; Weiser et al., 2009). That these
(Vaccari et al., 1998). Of importance, estradiol treatment has been effects of ERb agonists on behavior are not found in bERKO mice,
shown to upregulate oxytocin receptors in some brain areas further support the role of ERb dependent pathways in regulating
(Champagne et al., 2001), but not the PVN (Patisaul et al., 2003). anxiety- and depressive-like behaviors (Oyola et al., 2012). Fur-
This effect of estradiol is mediated by ERa (Young et al., 1998), thermore, the Flinders Sensitive Line (FSL) of rat, a strain selec-
and therefore, because ERa is expressed by few PVN neurons (Su- tively bred for showing depressive-like behaviors, exhibit
zuki and Handa, 2005), it appears that estradiol may not act to alter decreased immobility in the forced swim test and increased social
OT sensitivity at the level of the PVN. interactions following DPN treatment, both anti-depressive signals
(Overstreet et al., 2006). The ERb agonist, WAY200070 has also
been shown to have anti-depressant actions in the tail suspension
8. Gonadal steroid regulation of stress-related behaviors test in male mice (Hughes et al., 2008). Thus, similarities seen in
multiple tests of anxiety and depressive-like behaviors and across
8.1. Effects of estrogens on anxiety and depressive-like behaviors multiple ERb agonists consistently implicate ERb as being a posi-
tive modulator for mood.
Studies examining anxiety and depressive-like behaviors in ERa By contrast to ERb agonists, treatment with the ERa agonist, PPT
and ERb knockout (bERKO) mice have generally concluded that ERb has been shown to be anxiogenic in the elevated plus maze and
is involved in dampening anxiety and stress related behaviors. The open field apparatus and to enhance depressive-like behaviors in
initial clue that ERb might be involved in regulating anxiety-like the forced swim test (Lund et al., 2005; Walf and Frye, 2006; Weis-
behaviors came from studies by Krezel et al. (2001) who demon- er et al., 2009). This was consistent with its actions in enhancing
strated elevated anxiety-like behaviors in female ERb null mutant the neuroendocrine response to restraint stress (Lund et al.,
mice in the open field arena and the elevated plus maze. Corre- 2006). Correspondingly, animals with elevated levels of ERa seem
spondingly ERb null mice show increased expression of 5-HT1a to respond to estradiol with anxiogenic types of responses (Spiteri
receptors, and, as one of the principal 5-HT receptors, the 5-HT1a et al., 2012). Taken together, these opposing actions of ERa and ERb
receptor is a demonstrated regulator of GABA function (Levkovitz agonists on anxiety- and depressive-like behaviors in rodent mod-
and Segal, 1997). Alterations in behaviors in the elevated plus els may help explain previous reports of both anxiogenic and anx-
maze were also shown in the bERKO mouse by Imwalle et al. iolytic effects following estradiol (Leret et al., 1994; Palermo-Neto
(2005), who also demonstrated corresponding reductions in 5-HT and Dorce, 1990). A more thorough description of effects of estra-
content within the BnST, preoptic area and hippocampus. These diol on a variety of behaviors and mechanisms of regulation can be
findings are supported by those of Donner and Handa (2009) found in Gasbarri et al. (2012) and Luine and Frankfurt (2012).
who showed that implants of an ERb agonist into the dorsal raphe,
the main nuclear group synthesizing serotonin in the midbrain, re- 8.2. Effects of androgens on anxiety- and depressive-like behaviors
sulted in an upregulation of tryptophan hydroxylase 2 (TPH-2)
mRNA, a brain specific variant of TPH, the rate-limiting enzyme Although the effects of estradiol on measures of mood have
in 5-HT synthesis. A recent study has also demonstrated that ERb been well studied in rodent models, the role of androgens in
agonists can also increase TPH-1 mRNA (Clark et al., 2012). Thus stress-related behaviors is less well understood. Gonadectomy of
the serotonergic system of the brain is a principal target of ERb male rats, to remove the source of circulating androgens, causes in-
and this may be one mechanism where estradiol can exert actions creased anxiety- and depressive-like behaviors that are reversed
to modify anxiety- and depressive-like behaviors. by systemic testosterone treatment (Adler et al., 1999; Frye and
Depressive-like behaviors, such as the amount of time strug- Seliga, 2001; Slob et al., 1981). Treatment with the non-aromatiz-
gling, or time to immobility in the forced swim test (Porsolt able androgen, dihydrotestosterone, can also reduce anxiety- and
et al., 1977) are also modulated by estradiol, but the effects of depressive-like behaviors (Edinger and Frye, 2005; Frye and
estradiol are lost in bERKO mice further supporting the concept Wawrzycki, 2003). Further evidence that this effect is mediated
that estradiol’s antidepressant actions are mediated through ERb by the androgen receptor comes from studies of mice with the tes-
(Rocha et al., 2005). Similarly, ERb agonist treatment, whether ticular feminizing mutation (Tfm). In these animals, a mutation in
administered peripherally or by implants directly into the raphe the AR gene renders the AR protein nonfunctional and they display
nuclei, has been shown to reduce depressive-like behaviors in increased anxiety-like behaviors in the elevated plus maze and
the forced swim test (Clark et al., 2012; Donner and Handa, open field arena that cannot be reversed by androgen treatment
2009) in a fashion similar to treatment following estradiol (Estra- (Rizk et al., 2005; Zuloaga et al., 2008). Similar responses are seen
da-Camarena et al., 2006; Kandi and Hayslett, 2011). Neuroana- in men following chemical castration for prostate cancer treatment
tomical evidence also supports a role for ERb in regulating 5-HT (Almeida et al., 2004) or during the decline in androgen levels that
synthesis. More than 90% of ERb expressing neurons in the dorsal is associated with aging (Amore, 2005). Taken together, these data
raphe and periaqueductal gray co-express TPH in the mouse (Nom- suggest that androgens are predominantly anxiolytic in nature.
ura et al., 2005), Guinea pig (Lu et al., 1999) and the primate The molecular mechanisms underlying androgen’s effect on
(Gundlah et al., 2001). By contrast, few ERa expressing neurons mood are not well described in the literature but may involve
colocalize with markers of 5-HT synthesis (Lu et al., 1999) in gui- CRF. A dysregulation of CRF signaling has been suggested in the
nea pig although others (Nomura et al., 2005; Sheng et al., 2004) development of depression and anxiety (Arborelius et al., 1999;

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 15

Heuser et al., 1998; Reul and Holsboer, 2002). The receptors for Ahima, R., Krozowski, Z., Harlan, R., 1991. Type I corticosteroid receptor-like
immunoreactivity in the rat CNS: distribution and regulation by corticosteroids.
CRF, designated CRFR1 and CRFR2 have integral roles in regulating
J. Comp. Neurol. 313, 522–538.
stress sensitivity and alterations in receptor expression can be Akana, S.F., Cascio, C.S., Du, J.Z., Levin, N., Dallman, M.F., 1986. Reset of feedback in
linked to behavioral disorders (for review see Bale and Vale, the adrenocortical system: an apparent shift in sensitivity of
2004). Specifically, CRFR2 has been implicated in regulating anxi- adrenocorticotropin to inhibition by corticosterone between morning and
evening. Endocrinology 119, 2325–2332.
ety type behaviors and it is also expressed in stress responsive re- Allen, B.D., Sutanto, W., Jones, M.T., 1988. A correlative study of RU38486
gions of the rodent brain, many of which also express androgen biopotency and competition with [3H]dexamethasone for receptors in the rat
receptor (Van Pett et al., 2000). Moreover, the CRFR2 upstream pro- central nervous system. J. Steroid Biochem. 30, 411–415.
Almeida, O.F., Conde, G.L., Crochemore, C., Demeneix, B.A., Fischer, D., Hassan, A.H.,
moter contains EREs and AREs suggesting the possibility of an Meyer, M., Holsboer, F., Michaelidis, T.M., 2000. Subtle shifts in the ratio
interaction (Catalano et al., 2003). Examination of androgen recep- between pro- and antiapoptotic molecules after activation of corticosteroid
tor dependent increases in CRFR2 mRNA and binding in the male receptors decide neuronal fate. FASEB J. 14, 779–790.
Almeida, O.P., Waterreus, A., Spry, N., Flicker, L., Martins, R.N., 2004. One year
rat forebrain showed that DHT treatment increased CRFR2 mRNA follow-up study of the association between chemical castration, sex hormones,
in specific brain regions. The effect of DHT on both receptor bind- beta-amyloid, memory and depression in men. Psychoneuroendocrinology 29,
ing and mRNA levels was found in the lateral septum. Androgen 1071–1081.
Amore, M., 2005. Partial androgen deficiency and neuropsychiatric symptoms in
treatment could also increase CRFR2 expression in primary hippo- aging men. J. Endocrinol. Invest. 28, 49–54.
campal cell cultures, and this could be blocked with the AR antag- Amoss, M., Burgus, R., Blackwell, R., Vale, W., Fellows, R., Guillemin, R., 1971.
onist flutamide (Weiser et al., 2008). These data suggest the Purification, amino acid composition and N-terminus of the hypothalamic
luteinizing hormone releasing factor (LRF) of ovine origin. Biochem. Biophys.
possibility that androgen regulation of CRFR2 expression may be
Res. Commun. 44, 205–210.
a potential mechanism whereby ARs modulate stress and stress- Angold, A., Worthman, C.W., 1993. Puberty onset of gender differences in rates of
related behaviors. Lastly, because the HPA and HPG axis interact depression: a developmental, epidemiologic and neuroendocrine perspective. J.
considerably, androgen regulation of other behaviors such as Affect. Disord. 29, 145–158.
Antoni, F.A., 1986. Hypothalamic control of adrenocorticotropin secretion: advances
aggression, may also influence HPA function (Cunningham et al., since the discovery of 41-residue corticotropin-releasing factor. Endocr. Rev. 7,
2012). 351–378.
Arborelius, L., Owens, M.J., Plotsky, P.M., Nemeroff, C.B., 1999. The role of
corticotropin-releasing factor in depression and anxiety disorders. J.
9. Summary and conclusions Endocrinol. 160, 1–12.
Armstrong, W.E., Warach, S., Hatton, G.I., McNeill, T.H., 1980. Subnuclei in the rat
hypothalamic paraventricular nucleus: a cytoarchitectural, horseradish
It is becoming apparent that the ability of the hypothalamus to peroxidase and immunocytochemical analysis. Neuroscience 5, 1931–1958.
oversee normal physiology and make rapid adjustments in re- Aronsson, M., Fuxe, K., Dong, Y., Agnati, L.F., Okret, S., Gustafsson, J.A., 1988.
sponse to shifts in the environment is swayed by the reproductive Localization of glucocorticoid receptor mRNA in the male rat brain by in situ
hybridization. Proc. Natl. Acad. Sci. USA 85, 9331–9335.
status of the animal. Hence, the hypothalamus monitors reproduc- Axelrod, J., Reisine, T.D., 1984. Stress hormones: their interaction and regulation.
tive state through neurons that express receptors for gonadal ste- Science 224, 452–459.
roid hormones. The identification and function of these androgen Azcoitia, I., Sierra, A., Garcia-Segura, L.M., 1999. Localization of estrogen receptor
beta-immunoreactivity in astrocytes of the adult rat brain. Glia 26, 260–267.
and estrogen receptor containing neurons and the neuroanatomi- Babb, J.A., Masini, C.V., Day, H.E., Campeau, S., 2013. Sex differences in activated
cal and molecular pathways that they utilize to influence neuroen- corticotropin-releasing factor neurons within stress-related neurocircuitry and
docrine stress reactivity and autonomic function and behaviors are hypothalamic–pituitary–adrenocortical axis hormones following restraint in
rats. Neuroscience 234, 40–52.
an emergent area of research. Sex differences in homeostatic Bale, T.L., Vale, W.W., 2004. CRF and CRF receptors: role in stress responsivity and
mechanisms and autonomic responses are the result of develop- other behaviors. Annu. Rev. Pharmacol. Toxicol. 44, 525–557.
mental programming events as well as differing adult levels of go- Bargmann, W., 1949. Uber die neurosekretorische verknupfung von hypothlamus
und neurohypophyse. Z. Zellforsch. Mikrosk. Anat. 34, 610–634.
nadal steroid hormones. These can profoundly impact, not only
Bargmann, W., Scharrer, E., 1951. The site of origin of the hormones of the posterior
normal physiology, but also the development of stress-related neu- pituitary. Am. Sci. 39, 255–259.
ropathology. The body of work tying the changes in gonadal steroid Becker, H., Grabosch, E., Hoffmann, C., Voigt, K.D., 1973. Metabolism and mode of
action of androgens in target tissues of male rats. 3. Metabolism of 5 alpha-
hormones across the lifetime to the efficacy of mounting a stress
androstane-3,17-dione, of 5 alpha-androstane-3 beta,17 beta-diol and of 5
response to environmental challenges, whether actual or per- alpha-androstane-3 beta,17 beta-diol in target organs and peripheral tissues.
ceived, is still incomplete in animals and in humans. Nonetheless, Acta Endocrinol. (Copenh) 73, 407–416.
it is clear that genetic sex and gonadal steroid hormone levels Behbehani, M.M., 1995. Functional characteristics of the midbrain periaqueductal
gray. Prog. Neurobiol. 46, 575–605.
should be taken into account when considering normal homeo- Belelli, D., Lambert, J.J., 2005. Neurosteroids: endogenous regulators of the GABA(A)
static responses as well as therapeutic approaches to combat receptor. Nat. Rev. Neurosci. 6, 565–575.
stress-related illnesses. Bellocchio, L., Cervino, C., Pasquali, R., Pagotto, U., 2008. The endocannabinoid
system and energy metabolism. J. Neuroendocrinol. 20, 850–857.
Biag, J., Huang, Y., Gou, L., Hintiryan, H., Askarinam, A., Hahn, J.D., Toga, A.W., Dong,
References H.W., 2012. Cyto- and chemoarchitecture of the hypothalamic paraventricular
nucleus in the C57BL/6J male mouse: a study of immunostaining and multiple
fluorescent tract tracing. J. Comp. Neurol. 520, 6–33.
Abe, K., Critchlow, V., 1980. Delayed feedback inhibition of stress-induced
Bilezikjian, L.M., Vale, W.W., 1987. Regulation of ACTH secretion from
activation of pituitary-adrenal function: effects of varying dose, rate and
corticotrophs: the interaction of vasopressin and CRF. Ann. N. Y. Acad. Sci.
duration of corticosterone administration and of telencephalon removal.
512, 85–96.
Neuroendocrinology 31, 349–354.
Bingaman, E.W., Magnuson, D.J., Gray, T.S., Handa, R.J., 1994. Androgen inhibits
Abel, K., Majzoub, J.A., 2005. Molecular biology of the HPA axis. In: Steckler, T., Kalin,
the increases in hypothalamic corticotropin-releasing hormone (CRH) and
N.H., Reul, J.M. (Eds.), Handbook of Stress and the Brain. Elsevier, pp. 79–94.
CRH-immunoreactivity following gonadectomy. Neuroendocrinology 59, 228–
Abraham, I.M., Todman, M.G., Korach, K.S., Herbison, A.E., 2004. Critical in vivo roles
234.
for classical estrogen receptors in rapid estrogen actions on intracellular
Bingham, B., Williamson, M., Viau, V., 2006. Androgen and estrogen receptor-beta
signaling in mouse brain. Endocrinology 145, 3055–3061.
distribution within spinal-projecting and neurosecretory neurons in the
Adan, R.A., Cox, J.J., Beischlag, T.V., Burbach, J.P., 1993. A composite hormone
paraventricular nucleus of the male rat. J. Comp. Neurol. 499, 911–923.
response element mediates the transactivation of the rat oxytocin gene by
Blair, M.L., Piekut, D., Want, A., Olschowka, J.A., 1996. Role of the hypothalamic
different classes of nuclear hormone receptors. Mol. Endocrinol. 7, 47–57.
paraventricular nucleus in cardiovascular regulation. Clin. Exp. Pharmacol.
Adler, A., Vescovo, P., Robinson, J.K., Kritzer, M.F., 1999. Gonadectomy in adult life
Physiol. 23, 161–165.
increases tyrosine hydroxylase immunoreactivity in the prefrontal cortex and
Bondy, C.A., Whitnall, M.H., Brady, L.S., Gainer, H., 1989. Coexisting peptides in
decreases open field activity in male rats. Neuroscience 89, 939–954.
hypothalamic neuroendocrine systems: some functional implications. Cell. Mol.
Aguilera, G., Nikodemova, M., Wynn, P.C., Catt, K.J., 2004. Corticotropin releasing
Neurobiol. 9, 427–446.
hormone receptors: two decades later. Peptides 25, 319–329.
Boudaba, C., Szabo, K., Tasker, J.G., 1996. Physiological mapping of local inhibitory
Ahima, R.S., Harlan, R.E., 1990. Charting of type II glucocorticoid receptor-like
inputs to the hypothalamic paraventricular nucleus. J. Neurosci. 16, 7151–7160.
immunoreactivity in the rat central nervous system. Neuroscience 39, 579–604.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
16 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Bradbury, M.J., Akana, S.F., Dallman, M.F., 1994. Roles of type I and II corticosteroid Cole, R.L., Sawchenko, P.E., 2002. Neurotransmitter regulation of cellular activation
receptors in regulation of basal activity in the hypothalamo–pituitary–adrenal and neuropeptide gene expression in the paraventricular nucleus of the
axis during the diurnal trough and the peak: evidence for a nonadditive effect of hypothalamus. J. Neurosci. 22, 959–969.
combined receptor occupation. Endocrinology 134, 1286–1296. Conway-Campbell, B.L., McKenna, M.A., Wiles, C.C., Atkinson, H.C., de Kloet, E.R.,
Brailoiu, E., Dun, S.L., Brailoiu, G.C., Mizuo, K., Sklar, L.A., Oprea, T.I., Prossnitz, E.R., Lightman, S.L., 2007. Proteasome-dependent down-regulation of activated
Dun, N.J., 2007. Distribution and characterization of estrogen receptor G nuclear hippocampal glucocorticoid receptors determines dynamic responses
protein-coupled receptor 30 in the rat central nervous system. J. Endocrinol. to corticosterone. Endocrinology 148, 5470–5477.
193, 311–321. Conway-Campbell, B.L., Pooley, J.R., Hager, G.L., Lightman, S.L., 2012. Molecular
Brinkmann, A.O., 2011. Molecular mechanisms of androgen action – a historical dynamics of ultradian glucocorticoid receptor action. Mol. Cell. Endocrinol. 348,
perspective. Methods Mol. Biol. 776, 3–24. 383–393.
Brinton, R.E., McEwen, B.S., 1988. Regional distinctions in the regulation of type I Cota, D., Steiner, M.A., Marsicano, G., Cervino, C., Herman, J.P., Grubler, Y., Stalla, J.,
and type II adrenal steroid receptors in the central nervous system. Neurosci. Pasquali, R., Lutz, B., Stalla, G.K., Pagotto, U., 2007. Requirement of cannabinoid
Res. Commun. 2, 37–45. receptor type 1 for the basal modulation of hypothalamic–pituitary–adrenal
Brunton, P.J., Russell, J.A., 2011. Allopregnanolone and suppressed hypothalamo– axis function. Endocrinology 148, 1574–1581.
pituitary–adrenal axis stress responses in late pregnancy in the rat. Stress 14, 6– Coyne, M.D., Kitay, J.I., 1969. Effect of ovariectomy on pituitary secretion of ACTH.
12. Endocrinology 85, 1097–1102.
Burbach, J.P., Lopes da Silva, S., Cox, J.J., Adan, R.A., Cooney, A.J., Tsai, M.J., Tsai, S.Y., Coyne, M.D., Kitay, J.I., 1971. Effect of orchiectomy on pituitary secretion of ACTH.
1994. Repression of estrogen-dependent stimulation of the oxytocin gene by Endocrinology 89, 1024–1028.
chicken ovalbumin upstream promoter transcription factor I. J. Biol. Chem. 269, Critchlow, V., Liebelt, R.A., Bar-Sela, M., Mountcastle, W., Lipscomb, H.S., 1963. Sex
15046–15053. difference in resting pituitary–adrenal function in the rat. Am. J. Physiol. 205,
Burbach, J.P., van Schaick, H., Lopes da Silva, S., Asbreuk, C.H., Smidt, M.P., 1998. 807–815.
Hypothalamic transcription factors and the regulation of the hypothalamo– Crochemore, C., Lu, J., Wu, Y., Liposits, Z., Sousa, N., Holsboer, F., Almeida, O.F., 2005.
neurohypophysial system. Adv. Exp. Med. Biol. 449, 29–37. Direct targeting of hippocampal neurons for apoptosis by glucocorticoids is
Burbach, J.P., Luckman, S.M., Murphy, D., Gainer, H., 2001. Gene regulation in the reversible by mineralocorticoid receptor activation. Mol. Psychiatry 10, 790–
magnocellular hypothalamo–neurohypophysial system. Physiol. Rev. 81, 1197– 798.
1267. Cullinan, W.E., Herman, J.P., Watson, S.J., 1993. Ventral subicular interaction with
Burgess, L.H., Handa, R.J., 1992. Chronic estrogen-induced alterations in the hypothalamic paraventricular nucleus: evidence for a relay in the bed
adrenocorticotropin and corticosterone secretion, and glucocorticoid receptor- nucleus of the stria terminalis. J. Comp. Neurol. 332, 1–20.
mediated functions in female rats. Endocrinology 131, 1261–1269. Cummings, S., Seybold, V., 1988. Relationship of alpha-1- and alpha-2-adrenergic-
Burgess, L.H., Handa, R.J., 1993. Estrogen-induced alterations in the regulation of binding sites to regions of the paraventricular nucleus of the hypothalamus
mineralocorticoid and glucocorticoid receptor messenger RNA expression in the containing corticotropin-releasing factor and vasopressin neurons.
female rat anterior pituitary gland and brain. Mol. Cell. Neurosci. 4, 191–198. Neuroendocrinology 47, 523–532.
Burgus, R., Dunn, T.F., Desiderio, D., Ward, D.N., Vale, W., Guillemin, R., 1970. Cunningham Jr., E.T., Bohn, M.C., Sawchenko, P.E., 1990. Organization of adrenergic
Characterization of ovine hypothalamic hypophysiotropic TSH-releasing factor. inputs to the paraventricular and supraoptic nuclei of the hypothalamus in the
Nature 226, 321–325. rat. J. Comp. Neurol. 292, 651–667.
Cannon, W.B., 1915. Bodily Changes in Pain, Hunger, Fear and Rage. Applegate and Cunningham, R.L., Lumia, A.R., McGinnis, M.Y., 2012. Androgen receptors, sex
Co., New York. behavior, and aggression. Neuroendocrinology 96, 131–140.
Cannon, W.B., 1932. Wisdom of the Body. Norton, New York. Dabrowska, J., Hazra, R., Ahern, T.H., Guo, J.D., McDonald, A.J., Mascagni, F., Muller,
Cascio, C.S., Shinsako, J., Dallman, M.F., 1987. The suprachiasmatic nuclei stimulate J.F., Young, L.J., Rainnie, D.G., 2011. Neuroanatomical evidence for reciprocal
evening ACTH secretion in the rat. Brain Res. 423, 173–178. regulation of the corticotrophin-releasing factor and oxytocin systems in the
Catalano, R.D., Kyriakou, T., Chen, J., Easton, A., Hillhouse, E.W., 2003. Regulation of hypothalamus and the bed nucleus of the stria terminalis of the rat:
corticotropin-releasing hormone type 2 receptors by multiple promoters and implications for balancing stress and affect. Psychoneuroendocrinology 36,
alternative splicing: identification of multiple splice variants. Mol. Endocrinol. 1312–1326.
17, 395–410. Dallman, M.F., 2005. Fast glucocorticoid actions on brain: back to the future. Front.
Ceccatelli, S., Cintra, A., Hokfelt, T., Fuxe, K., Wikstrom, A.C., Gustafsson, J.A., 1989. Neuroendocrinol. 26, 103–108.
Coexistence of glucocorticoid receptor-like immunoreactivity with Dallman, M.F., Engeland, W.C., Rose, J.C., Wilkinson, C.W., Shinsako, J., Siedenburg,
neuropeptides in the hypothalamic paraventricular nucleus. Exp. Brain Res. F., 1978. Nycthemeral rhythm in adrenal responsiveness to ACTH. Am. J. Physiol.
78, 33–42. 235, R210–R218.
Chaban, V.V., Lakhter, A.J., Micevych, P., 2004. A membrane estrogen receptor Dallman, M.F., Makara, G.B., Roberts, J.L., Levin, N., Blum, M., 1985. Corticotrope
mediates intracellular calcium release in astrocytes. Endocrinology 145, 3788– response to removal of releasing factors and corticosteroids in vivo.
3795. Endocrinology 117, 2190–2197.
Chaban, V., Li, J., McDonald, J.S., Rapkin, A., Micevych, P., 2011. Estradiol attenuates Dallman, M.F., Akana, S.F., Cascio, C.S., Darlington, D.N., Jacobson, L., Levin, N.,
the adenosine triphosphate-induced increase of intracellular calcium through 1987a. Regulation of ACTH secretion: variations on a theme of B. Recent Prog.
group II metabotropic glutamate receptors in rat dorsal root ganglion neurons. J. Horm. Res. 43, 113–173.
Neurosci. Res. 89, 1707–1710. Dallman, M.F., Akana, S.F., Jacobson, L., Levin, N., Cascio, C.S., Shinsako, J., 1987b.
Champagne, F., Diorio, J., Sharma, S., Meaney, M.J., 2001. Naturally occurring Characterization of corticosterone feedback regulation of ACTH secretion. Ann.
variations in maternal behavior in the rat are associated with differences in N. Y. Acad. Sci. 512, 402–414.
estrogen-inducible central oxytocin receptors. Proc. Natl. Acad. Sci. USA 98, Day, H.E., Campeau, S., Watson, S.J., Akil, H., 1997. Distribution of alpha 1a-, alpha
12736–12741. 1b- and alpha 1d-adrenergic receptor mRNA in the rat brain and spinal cord. J.
Chao, H.M., Ma, L.Y., McEwen, B.S., Sakai, R.R., 1998. Regulation of glucocorticoid Chem. Neuroanat. 13, 115–139.
receptor and mineralocorticoid receptor messenger ribonucleic acids by Day, H.E., Campeau, S., Watson, S.J., Akil, H., 1999. Expression of alpha(1b)
selective agonists in the rat hippocampus. Endocrinology 139, 1810–1814. adrenoceptor mRNA in corticotropin-releasing hormone-containing cells of
Chen, X.Q., Du, J.Z., Wang, Y.S., 2004. Regulation of hypoxia-induced release of the rat hypothalamus and its regulation by corticosterone. J. Neurosci. 19,
corticotropin-releasing factor in the rat hypothalamus by norepinephrine. 10098–10106.
Regul. Pept. 119, 221–228. De Groot, J., Harris, G.W., 1950. Hypothalmic control of the anterior pituitary gland
Chen, X.N., Zhu, H., Meng, Q.Y., Zhou, J.N., 2008. Estrogen receptor-alpha and -beta and blood lymphocytes. J. Physiol. 111, 335–346.
regulate the human corticotropin-releasing hormone gene through similar de Kloet, E.R., 1984. Adrenal steroids as modulators of nerve cell function. J. Steroid
pathways. Brain Res. 1223, 1–10. Biochem. 20, 175–181.
Chu, S., Fuller, P.J., 1997. Identification of a splice variant of the rat estrogen receptor de Kloet, E.R., Burbach, P., Mulder, G.H., 1977. Localization and role of transcortin-
beta gene. Mol. Cell. Endocrinol. 132, 195–199. like molecules in the anterior pituitary. Mol. Cell. Endocrinol. 7, 261–273.
Cintra, A., Bhatnagar, M., Chadi, G., Tinner, B., Lindberg, J., Gustafsson, J.A., Agnati, Denayer, S., Helsen, C., Thorrez, L., Haelens, A., Claessens, F., 2010. The rules of DNA
L.F., Fuxe, K., 1994. Glial and neuronal glucocorticoid receptor immunoreactive recognition by the androgen receptor. Mol. Endocrinol. 24, 898–913.
cell populations in developing, adult, and aging brain. Ann. N. Y. Acad. Sci. 746, Diorio, D., Viau, V., Meaney, M.J., 1993. The role of the medial prefrontal cortex
42–61, discussion 61-3. (cingulate gyrus) in the regulation of hypothalamic–pituitary–adrenal
Clark, J.A., Alves, S., Gundlah, C., Rocha, B., Birzin, E.T., Cai, S.J., Flick, R., Hayes, E., Ho, responses to stress. J. Neurosci. 13, 3839–3847.
K., Warrier, S., Pai, L., Yudkovitz, J., Fleischer, R., Colwell, L., Li, S., Wilkinson, H., DonCarlos, L.L., Garcia-Ovejero, D., Sarkey, S., Garcia-Segura, L.M., Azcoitia, I., 2003.
Schaeffer, J., Wilkening, R., Mattingly, E., Hammond, M., Rohrer, S.P., 2012. Androgen receptor immunoreactivity in forebrain axons and dendrites in the
Selective estrogen receptor-beta (SERM-beta) compounds modulate raphe rat. Endocrinology 144, 3632–3638.
nuclei tryptophan hydroxylase-1 (TPH-1) mRNA expression and cause DonCarlos, L.L., Sarkey, S., Lorenz, B., Azcoitia, I., Garcia-Ovejero, D., Huppenbauer,
antidepressant-like effects in the forced swim test. Neuropharmacology 63, C., Garcia-Segura, L.M., 2006. Novel cellular phenotypes and subcellular sites for
1051–1063. androgen action in the forebrain. Neuroscience 138, 801–807.
Coirini, H., Magarinos, A.M., De Nicola, A.F., Rainbow, T.C., McEwen, B.S., 1985. Dong, H.W., Swanson, L.W., 2006. Projections from bed nuclei of the stria terminalis,
Further studies of brain aldosterone binding sites employing new anteromedial area: cerebral hemisphere integration of neuroendocrine,
mineralocorticoid and glucocorticoid receptor markers in vitro. Brain Res. autonomic, and behavioral aspects of energy balance. J. Comp. Neurol. 494,
361, 212–216. 142–178.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 17

Dong, H.W., Petrovich, G.D., Swanson, L.W., 2001a. Topography of projections from Gatson, J.W., Singh, M., 2007. Activation of a membrane-associated androgen
amygdala to bed nuclei of the stria terminalis. Brain Res. Brain Res. Rev. 38, receptor promotes cell death in primary cortical astrocytes. Endocrinology 148,
192–246. 2458–2464.
Dong, H.W., Petrovich, G.D., Watts, A.G., Swanson, L.W., 2001b. Basic organization of Gemzik, B., Jacob, S., Jennings, S., Veltman, J., Parkinson, A., 1992. Species differences
projections from the oval and fusiform nuclei of the bed nuclei of the stria in 5 alpha-androstane-3 beta,17 beta-diol hydroxylation by rat, monkey, and
terminalis in adult rat brain. J. Comp. Neurol. 436, 430–455. human prostate microsomes. Arch. Biochem. Biophys. 296, 374–383.
Donner, N., Handa, R.J., 2009. Estrogen receptor beta regulates the expression of Ghosal, S., Myers, B., Herman, J.P., 2013. Role of central glucagon-like peptide-1 in
tryptophan-hydroxylase 2 mRNA within serotonergic neurons of the rat dorsal stress regulation. Physiol. Behav. 122, 201–207.
raphe nuclei. Neuroscience 163, 705–718. Gibson, A., Hart, S.L., Patel, S., 1986. Effects of 6-hydroxydopamine-induced lesions
Dunn, J.D., 1987. Plasma corticosterone responses to electrical stimulation of the of the paraventricular nucleus, and of prazosin, on the corticosterone response
bed nucleus of the stria terminalis. Brain Res. 407, 327–331. to restraint in rats. Neuropharmacology 25, 257–260.
Dunn, A.J., Berridge, C.W., 1990. Is corticotropin-releasing factor a mediator of stress Gillies, G.E., Linton, E.A., Lowry, P.J., 1982. Corticotropin releasing activity of the new
responses? Ann. N. Y. Acad. Sci. 579, 183–191. CRF is potentiated several times by vasopressin. Nature 299, 355–357.
Edinger, K.L., Frye, C.A., 2005. Testosterone’s anti-anxiety and analgesic effects may Goel, N., Plyler, K.S., Daniels, D., Bale, T.L., 2011. Androgenic influence on
be due in part to actions of its 5alpha-reduced metabolites in the hippocampus. serotonergic activation of the HPA stress axis. Endocrinology 152, 2001–2010.
Psychoneuroendocrinology 30, 418–430. Gore-Langton, R.E., Armstrong, D.T., 1994. Follicular steroidogenesis and its control.
Ekstrand, J., Hellsten, J., Tingstrom, A., 2008. Environmental enrichment, exercise In: The Physiology of Reproduction. Raven Press Ltd., NY, NY, pp. 571–627.
and corticosterone affect endothelial cell proliferation in adult rat hippocampus Gorzalka, B.B., Dang, S.S., 2012. Minireview: endocannabinoids and gonadal
and prefrontal cortex. Neurosci. Lett. 442, 203–207. hormones: bidirectional interactions in physiology and behavior.
Ellis, G.B., Desjardins, C., 1982. Male rats secrete luteinizing hormone and Endocrinology 153, 1016–1024.
testosterone episodically. Endocrinology 110, 1618–1627. Gray, T.S., Piechowski, R.A., Yracheta, J.M., Rittenhouse, P.A., Bethea, C.L., Van de Kar,
Ericsson, A., Kovacs, K.J., Sawchenko, P.E., 1994. A functional anatomical analysis of L.D., 1993. Ibotenic acid lesions in the bed nucleus of the stria terminalis
central pathways subserving the effects of interleukin-1 on stress-related attenuate conditioned stress-induced increases in prolactin, ACTH and
neuroendocrine neurons. J. Neurosci. 14, 897–913. corticosterone. Neuroendocrinology 57, 517–524.
Estacio, M.A., Yamada, S., Tsukamura, H., Hirunagi, K., Maeda, K., 1996. Effect of Grove-Strawser, D., Boulware, M.I., Mermelstein, P.G., 2010. Membrane estrogen
fasting and immobilization stress on estrogen receptor immunoreactivity in the receptors activate the metabotropic glutamate receptors mGluR5 and mGluR3
brain in ovariectomized female rats. Brain Res. 717, 55–61. to bidirectionally regulate CREB phosphorylation in female rat striatal neurons.
Estrada-Camarena, E., Lopez-Rubalcava, C., Fernandez-Guasti, A., 2006. Facilitating Neuroscience 170, 1045–1055.
antidepressant-like actions of estrogens are mediated by 5-HT1A and estrogen Gu, Q., Moss, R.L., 1996. 17 beta-Estradiol potentiates kainate-induced currents via
receptors in the rat forced swimming test. Psychoneuroendocrinology 31, 905– activation of the cAMP cascade. J. Neurosci. 16, 3620–3629.
914. Gu, S., Papadopoulou, N., Nasir, O., Foller, M., Alevizopoulos, K., Lang, F., Stournaras,
Evans, R.M., 1988. The steroid and thyroid hormone receptor superfamily. Science C., 2011. Activation of membrane androgen receptors in colon cancer inhibits
240, 889–895. the prosurvival signals Akt/bad in vitro and in vivo and blocks migration via
Evanson, N.K., Tasker, J.G., Hill, M.N., Hillard, C.J., Herman, J.P., 2010. Fast feedback vinculin/actin signaling. Mol. Med. 17, 48–58.
inhibition of the HPA axis by glucocorticoids is mediated by endocannabinoid Gu, G., Roland, B., Tomaselli, K., Dolman, C.S., Lowe, C., Heilig, J.S., 2013. Glucagon-
signaling. Endocrinology 151, 4811–4819. like peptide-1 in the rat brain: distribution of expression and functional
Feldman, S., Weidenfeld, J., 2002. Further evidence for the central effect of implication. J. Comp. Neurol. 521, 2235–2261.
dexamethasone at the hypothalamic level in the negative feedback Gundlah, C., Lu, N.Z., Mirkes, S.J., Bethea, C.L., 2001. Estrogen receptor beta (ERbeta)
mechanism. Brain Res. 958, 291–296. mRNA and protein in serotonin neurons of macaques. Brain Res. Mol. Brain Res.
Ferguson, A.V., Latchford, K.J., Samson, W.K., 2008. The paraventricular nucleus of 91, 14–22.
the hypothalamus – a potential target for integrative treatment of autonomic Gurdjian, E.S., 1927. The diencephalon of the albino rat. J. Comp. Neurol. 43, 1–114.
dysfunction. Expert Opin. Ther. Targets 12, 717–727. Handa, R.J., Reid, D.L., Resko, J.A., 1986. Androgen receptors in brain and pituitary of
Figueiredo, H.F., Dolgas, C.M., Herman, J.P., 2002. Stress activation of cortex and female rats: cyclic changes and comparisons with the male. Biol. Reprod. 34,
hippocampus is modulated by sex and stage of estrus. Endocrinology 143, 293–303.
2534–2540. Handa, R.J., Roselli, C.E., Horton, L., Resko, J.A., 1987. The quantitative distribution of
Figueiredo, H.F., Bodie, B.L., Tauchi, M., Dolgas, C.M., Herman, J.P., 2003. Stress cytosolic androgen receptors in microdissected areas of the male rat brain:
integration after acute and chronic predator stress: differential activation of effects of estrogen treatment. Endocrinology 121, 233–240.
central stress circuitry and sensitization of the hypothalamo–pituitary– Handa, R.J., Burgess, L.H., Kerr, J.E., O’Keefe, J.A., 1994a. Gonadal steroid hormone
adrenocortical axis. Endocrinology 144, 5249–5258. receptors and sex differences in the hypothalamo–pituitary–adrenal axis.
Filardo, E.J., Quinn, J.A., Frackelton Jr., A.R., Bland, K.I., 2002. Estrogen action via the Horm. Behav. 28, 464–476.
G protein-coupled receptor, GPR30: stimulation of adenylyl cyclase and cAMP- Handa, R.J., Nunley, K.M., Lorens, S.A., Louie, J.P., McGivern, R.F., Bollnow, M.R.,
mediated attenuation of the epidermal growth factor receptor-to-MAPK 1994b. Androgen regulation of adrenocorticotropin and corticosterone
signaling axis. Mol. Endocrinol. 16, 70–84. secretion in the male rat following novelty and foot shock stressors. Physiol.
Foradori, C.D., Weiser, M.J., Handa, R.J., 2008. Non-genomic actions of androgens. Behav. 55, 117–124.
Front. Neuroendocrinol. 29, 169–181. Handa, R.J., Sharma, D., Uht, R., 2011. A role for the androgen metabolite, 5alpha
Fortier, C., Harris, G.W., McDonald, I.R., 1957. The effect of pituitary stalk section on androstane 3beta, 17beta diol (3beta-diol) in the regulation of the
the adrenocortical response to stress in the rabbit. J. Physiol. 136, 344–363. hypothalamo–pituitary–adrenal axis. Front. Endocrinol. (Lausanne) 2, 65.
Frahm, K.A., Schow, M.J., Tobet, S.A., 2012. The vasculature within the Handa, R.J., Ogawa, S., Wang, J.M., Herbison, A.E., 2012. Roles for oestrogen receptor
paraventricular nucleus of the hypothalamus in mice varies as a function of beta in adult brain function. J. Neuroendocrinol. 24, 160–173.
development, subnuclear location, and GABA signaling. Horm. Metab. Res. 44, Handa, R.J., Kudwa, A.E., Donner, N.C., McGivern, R.F., Brown, R., 2013. Central 5-
619–624. alpha reduction of testosterone is required for testosterone’s inhibition of the
Freund, T.F., Katona, I., Piomelli, D., 2003. Role of endogenous cannabinoids in hypothalamo–pituitary–adrenal axis response to restraint stress in adult male
synaptic signaling. Physiol. Rev. 83, 1017–1066. rats. Brain Res. 1529, 74–82.
Frye, C.A., Seliga, A.M., 2001. Testosterone increases analgesia, anxiolysis, and Harris, G.W., 1951a. Neural control of the pituitary gland. I. The neurohypophysis.
cognitive performance of male rats. Cogn. Affect. Behav. Neurosci. 1, 371– Br. Med. J. 2, 559–564.
381. Harris, G.W., 1951b. Neural control of the pituitary gland. II. The adenohypophysis,
Frye, C.A., Wawrzycki, J., 2003. Effect of prenatal stress and gonadal hormone with special reference to the secretion of A.C.T.H. Br. Med. J. 2, 627–634.
condition on depressive behaviors of female and male rats. Horm. Behav. 44, Harris, G.W., Jacobsohn, D., 1952. Functional grafts of the anterior pituitary gland.
319–326. Proc. Roy. Soc. Lond. B Biol. Sci. 139, 263–276.
Frye, C.A., Edinger, K.L., Lephart, E.D., Walf, A.A., 2010. 3alpha-androstanediol, but Hawley, W.R., Grissom, E.M., Martin, R.C., Halmos, M.B., Bart, C.L., Dohanich, G.P.,
not testosterone, attenuates age-related decrements in cognitive, anxiety, and 2013. Testosterone modulates spatial recognition memory in male rats. Horm.
depressive behavior of male rats. Front. Aging Neurosci. 2, 15. Behav. 63, 559–565.
Gaillet, S., Lachuer, J., Malaval, F., Assenmacher, I., Szafarczyk, A., 1991. The Healy, D.P., Printz, M.P., 1984. Distribution of immunoreactive angiotensin II,
involvement of noradrenergic ascending pathways in the stress-induced angiotensin I, angiotensinogen and renin in the central nervous system of intact
activation of ACTH and corticosterone secretions is dependent on the nature and nephrectomized rats. Hypertension 6, I130–1136.
of stressors. Exp. Brain Res. 87, 173–180. Heisler, L.K., Pronchuk, N., Nonogaki, K., Zhou, L., Raber, J., Tung, L., Yeo, G.S.,
Gangloff, A., Shi, R., Nahoum, V., Lin, S.X., 2003. Pseudo-symmetry of C19 steroids, O’Rahilly, S., Colmers, W.F., Elmquist, J.K., Tecott, L.H., 2007. Serotonin activates
alternative binding orientations, and multispecificity in human estrogenic the hypothalamic–pituitary–adrenal axis via serotonin 2C receptor stimulation.
17beta-hydroxysteroid dehydrogenase. FASEB J. 17, 274–276. J. Neurosci. 27, 6956–6964.
Garcia-Iglesias, B.B., Mendoza-Garrido, M.E., Gutierrez-Ospina, G., Rangel-Barajas, Herman, J.P., Wiegand, S.J., Watson, S.J., 1990. Regulation of basal corticotropin-
C., Noyola-Diaz, M., Terron, J.A., 2013. Sensitization of restraint-induced releasing hormone and arginine vasopressin messenger ribonucleic acid
corticosterone secretion after chronic restraint in rats: involvement of 5-HT expression in the paraventricular nucleus: effects of selective hypothalamic
receptors. Neuropharmacology 71C, 216–227. deafferentations. Endocrinology 127, 2408–2417.
Gasbarri, A., Tavares, M.C., Rodrigues, R.C., Tomaz, C., Pom pili, A., 2012. Estrogen, Herman, J.P., Cullinan, W.E., Watson, S.J., 1994. Involvement of the bed nucleus of
cognitive functions and emotion: an overview on humans, non-human primates the stria terminalis in tonic regulation of paraventricular hypothalamic CRH and
and rodents in reproductive years. Rev. Neurosci. 23, 587–606. AVP mRNA expression. J. Neuroendocrinol. 6, 433–442.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
18 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Herman, J.P., Dolgas, C.M., Carlson, S.L., 1998. Ventral subiculum regulates Jasper, M.S., Engeland, W.C., 1997. Splanchnicotomy increases adrenal sensitivity to
hypothalamo–pituitary–adrenocortical and behavioural responses to cognitive ACTH in nonstressed rats. Am. J. Physiol. 273, E363–E368.
stressors. Neuroscience 86, 449–459. Jauregui-Huerta, F., Ruvalcaba-Delgadillo, Y., Gonzalez-Castaneda, R., Garcia-
Herman, J.P., Cullinan, W.E., Ziegler, D.R., Tasker, J.G., 2002. Role of the Estrada, J., Gonzalez-Perez, O., Luquin, S., 2010. Responses of glial cells to
paraventricular nucleus microenvironment in stress integration. Eur. J. stress and glucocorticoids. Curr. Immunol. Rev. 6, 195–204.
Neurosci. 16, 381–385. Jeanneteau, F.D., Lambert, W.M., Ismaili, N., Bath, K.G., Lee, F.S., Garabedian, M.J.,
Herman, J.P., Mueller, N.K., Figueiredo, H., Cullinan, W.E., 2005. Neurocircuit Chao, M.V., 2012. BDNF and glucocorticoids regulate corticotrophin-releasing
regulation of the hypothalamic–pituitary–adrenocortical stress response – an hormone (CRH) homeostasis in the hypothalamus. Proc. Natl. Acad. Sci. USA
overview. In: Steckler, T., Kalin, N.H., Reul, J.M. (Eds.), Handbook of Stress and 109, 1305–1310.
the Brain. Elsevier, pp. 405–418. Jennes, L., Conn, P.M., 1994. Gonadotropin-releasing hormone and its receptors in
Herman, J.P., Flak, J., Jankord, R., 2008. Chronic stress plasticity in the hypothalamic rat brain. Front. Neuroendocrinol. 15, 51–77.
paraventricular nucleus. Prog. Brain Res. 170, 353–364. Jin, Y., Penning, T.M., 2001. Steroid 5alpha-reductases and 3alpha-hydroxysteroid
Heuser, I., Bissette, G., Dettling, M., Schweiger, U., Gotthardt, U., Schmider, J., dehydrogenases: key enzymes in androgen metabolism. Best Pract. Res. Clin.
Lammers, C.H., Nemeroff, C.B., Holsboer, F., 1998. Cerebrospinal fluid Endocrinol. Metab. 15, 79–94.
concentrations of corticotropin-releasing hormone, vasopressin, and Jin, Y., Penning, T.M., 2006. Multiple steps determine the overall rate of the
somatostatin in depressed patients and healthy controls: response to reduction of 5alpha-dihydrotestosterone catalyzed by human type 3 3alpha-
amitriptyline treatment. Depress Anxiety 8, 71–79. hydroxysteroid dehydrogenase: implications for the elimination of androgens.
Hill, M.N., Tasker, J.G., 2012. Endocannabinoid signaling, glucocorticoid-mediated Biochemistry 45, 13054–13063.
negative feedback, and regulation of the hypothalamic–pituitary–adrenal axis. Joels, M., Karst, H., 2012. Corticosteroid effects on calcium signaling in limbic
Neuroscience 204, 5–16. neurons. Cell Calcium 51, 277–283.
Hill, M.N., Patel, S., Campolongo, P., Tasker, J.G., Wotjak, C.T., Bains, J.S., 2010. Joels, M., Krugers, H.J., Lucassen, P.J., Karst, H., 2009. Corticosteroid effects on
Functional interactions between stress and the endocannabinoid system: from cellular physiology of limbic cells. Brain Res. 1293, 91–100.
synaptic signaling to behavioral output. J. Neurosci. 30, 14980–14986. Joels, M., Sarabdjitsingh, R.A., Karst, H., 2012. Unraveling the time domains of
Hiroi, R., Lacagnina, A.F., Hinds, L.R., Carbone, D.G., Uht, R.M., Handa, R.J., 2013. The corticosteroid hormone influences on brain activity: rapid, slow, and chronic
androgen metabolite, 5alpha-androstane-3beta,17beta-diol (3beta-diol), modes. Pharmacol. Rev. 64, 901–938.
activates the oxytocin promoter through an estrogen receptor-beta pathway. Ju, G., Liu, S., Tao, J., 1986. Projections from the hypothalamus and its adjacent areas
Endocrinology 154, 1802–1812. to the posterior pituitary in the rat. Neuroscience 19, 803–828.
Hisano, S., Fukui, Y., Chikamori-Aoyama, M., Aizawa, T., Shibasaki, T., 1993. Kageyama, K., Yamagata, S., Akimoto, K., Sugiyama, A., Murasawa, S., Suda, T., 2012.
Reciprocal synaptic relations between CRF-immunoreactive- and TRH- Action of glucagon-like peptide 1 and glucose levels on corticotropin-releasing
immunoreactive neurons in the paraventricular nucleus of the rat factor and vasopressin gene expression in rat hypothalamic 4B cells. Mol. Cell.
hypothalamus. Brain Res. 620, 343–346. Endocrinol. 362, 221–226.
Hornby, P.J., Piekut, D.T., 1989. Opiocortin and catecholamine input to CRF- Kalil, B., Leite, C.M., Carvalho-Lima, M., Anselmo-Franci, J.A., 2013. Role of sex
immunoreactive neurons in rat forebrain. Peptides 10, 1139–1146. steroids in progesterone and corticosterone response to acute restraint stress in
Hosoya, Y., Sugiura, Y., Okado, N., Loewy, A.D., Kohno, K., 1991. Descending input rats: sex differences. Stress.
from the hypothalamic paraventricular nucleus to sympathetic preganglionic Kalsbeek, A., van Heerikhuize, J.J., Wortel, J., Buijs, R.M., 1996. A diurnal rhythm of
neurons in the rat. Exp. Brain Res. 85, 10–20. stimulatory input to the hypothalamo–pituitary–adrenal system as revealed by
Hrabovszky, E., Kallo, I., Steinhauser, A., Merchenthaler, I., Coen, C.W., Petersen, S.L., timed intrahypothalamic administration of the vasopressin V1 antagonist. J.
Liposits, Z., 2004. Estrogen receptor-beta in oxytocin and vasopressin neurons Neurosci. 16, 5555–5565.
of the rat and human hypothalamus: immunocytochemical and in situ Kalsbeek, A., Verhagen, L.A., Schalij, I., Foppen, E., Saboureau, M., Bothorel, B., Buijs,
hybridization studies. J. Comp. Neurol. 473, 315–333. R.M., Pevet, P., 2008. Opposite actions of hypothalamic vasopressin on circadian
Htun, H., Holth, L.T., Walker, D., Davie, J.R., Hager, G.L., 1999. Direct visualization of corticosterone rhythm in nocturnal versus diurnal species. Eur. J. Neurosci. 27,
the human estrogen receptor alpha reveals a role for ligand in the nuclear 818–827.
distribution of the receptor. Mol. Biol. Cell 10, 471–486. Kalsbeek, A., van der Spek, R., Lei, J., Endert, E., Buijs, R.M., Fliers, E., 2012. Circadian
Huang, Q., Zhu, H., Fischer, D.F., Zhou, J.N., 2008. An estrogenic effect of 5alpha- rhythms in the hypothalamo–pituitary–adrenal (HPA) axis. Mol. Cell.
androstane-3beta, 17beta-diol on the behavioral response to stress and on CRH Endocrinol. 349, 20–29.
regulation. Neuropharmacology 54, 1233–1238. Kandi, P., Hayslett, R.L., 2011. Nicotine and 17beta-estradiol produce an
Hughes, Z.A., Liu, F., Platt, B.J., Dwyer, J.M., Pulicicchio, C.M., Zhang, G., Schechter, antidepressant-like effect in female ovariectomized rats. Brain Res. Bull. 84,
L.E., Rosenzweig-Lipson, S., Day, M., 2008. WAY-200070, a selective agonist of 224–228.
estrogen receptor beta as a potential novel anxiolytic/antidepressant agent. Kaneko, M., Hiroshige, T., Shinsako, J., Dallman, M.F., 1980. Diurnal changes in
Neuropharmacology 54, 1136–1142. amplification of hormone rhythms in the adrenocortical system. Am. J. Physiol.
Ibuka, N., Kawamura, H., 1975. Loss of circadian rhythm in sleep–wakefulness cycle 239, R309–R316.
in the rat by suprachiasmatic nucleus lesions. Brain Res. 96, 76–81. Kaneko, M., Kaneko, K., Shinsako, J., Dallman, M.F., 1981. Adrenal sensitivity to
Imaki, T., Nahan, J.L., Rivier, C., Sawchenko, P.E., Vale, W., 1991. Differential adrenocorticotropin varies diurnally. Endocrinology 109, 70–75.
regulation of corticotropin-releasing factor mRNA in rat brain regions by Karst, H., Joels, M., 2005. Corticosterone slowly enhances miniature excitatory
glucocorticoids and stress. J. Neurosci. 11, 585–599. postsynaptic current amplitude in mice CA1 hippocampal cells. J. Neurophysiol.
Imwalle, D.B., Gustafsson, J.A., Rissman, E.F., 2005. Lack of functional estrogen 94, 3479–3486.
receptor beta influences anxiety behavior and serotonin content in female mice. Kash, T.L., Winder, D.G., 2006. Neuropeptide Y and corticotropin-releasing factor bi-
Physiol. Behav. 84, 157–163. directionally modulate inhibitory synaptic transmission in the bed nucleus of
Isgor, C., Cecchi, M., Kabbaj, M., Akil, H., Watson, S.J., 2003. Estrogen receptor beta in the stria terminalis. Neuropharmacology 51, 1013–1022.
the paraventricular nucleus of hypothalamus regulates the neuroendocrine Keller-Wood, M., 1998. ACTH responses to hypotension and feedback inhibition of
response to stress and is regulated by corticosterone. Neuroscience 121, 837– ACTH increased by chronic progesterone treatment. Am. J. Physiol. 274, R81–
845. R87.
Ishizaki, F., Nishiyama, T., Kawasaki, T., Miyashiro, Y., Hara, N., Takizawa, I., Naito, Kelly, M.J., Ronnekleiv, O.K., 2012. Membrane-initiated actions of estradiol that
M., Takahashi, K., 2013. Androgen deprivation promotes intratumoral synthesis regulate reproduction, energy balance and body temperature. Front.
of dihydrotestosterone from androgen metabolites in prostate cancer. Sci. Rep. Neuroendocrinol. 33, 376–387.
3, 1528. Kelly, M.J., Ronnekleiv, O.K., 2013. A selective membrane estrogen receptor agonist
Ising, M., Horstmann, S., Kloiber, S., Lucae, S., Binder, E.B., Kern, N., Kunzel, H.E., maintains autonomic functions in hypoestrogenic states. Brain Res..
Pfennig, A., Uhr, M., Holsboer, F., 2007. Combined dexamethasone/corticotropin Kelly, M.J., Moss, R.L., Dudley, C.A., 1977. The effects of microelectrophoretically
releasing hormone test predicts treatment response in major depression – a applied estrogen, cortisol and acetylcholine on medial preoptic-septal unit
potential biomarker? Biol. Psychiatry 62, 47–54. activity throughout the estrous cycle of the female rat. Exp. Brain Res. 30, 53–
Itoi, K., Suda, T., Tozawa, F., Dobashi, I., Ohmori, N., Sakai, Y., Abe, K., Demura, H., 64.
1994. Microinjection of norepinephrine into the paraventricular nucleus of the Kerr, J.E., Beck, S.G., Handa, R.J., 1996. Androgens selectively modulate C-fos
hypothalamus stimulates corticotropin-releasing factor gene expression in messenger RNA induction in the rat hippocampus following novelty.
conscious rats. Endocrinology 135, 2177–2182. Neuroscience 74, 757–766.
Itoi, K., Helmreich, D.L., Lopez-Figueroa, M.O., Watson, S.J., 1999. Differential regulation Kessler, R.C., 2003. Epidemiology of women and depression. J. Affect. Disord. 74, 5–
of corticotropin-releasing hormone and vasopressin gene transcription in the 13.
hypothalamus by norepinephrine. J. Neurosci. 19, 5464–5472. Kessler, R.C., McGonagle, K.A., Swartz, M., Blazer, D.G., Nelson, C.B., 1993. Sex and
Iwasaki-Sekino, A., Mano-Otagiri, A., Ohata, H., Yamauchi, N., Shibasaki, T., 2009. depression in the National Comorbidity Survey. I: lifetime prevalence,
Gender differences in corticotropin and corticosterone secretion and chronicity and recurrence. J. Affect. Disord. 29, 85–96.
corticotropin-releasing factor mRNA expression in the paraventricular nucleus Khan, A.M., Ponzio, T.A., Sanchez-Watts, G., Stanley, B.G., Hatton, G.I., Watts, A.G.,
of the hypothalamus and the central nucleus of the amygdala in response to 2007. Catecholaminergic control of mitogen-activated protein kinase signaling
footshock stress or psychological stress in rats. Psychoneuroendocrinology 34, in paraventricular neuroendocrine neurons in vivo and in vitro: a proposed role
226–237. during glycemic challenges. J. Neurosci. 27, 7344–7360.
Jansen, A.S., Wessendorf, M.W., Loewy, A.D., 1995. Transneuronal labeling of CNS King, J.C., Tobet, S.A., Snavely, F.L., Arimura, A.A., 1982. LHRH immunopositive cells
neuropeptide and monoamine neurons after pseudorabies virus injections into and their projections to the median eminence and organum vasculosum of the
the stellate ganglion. Brain Res. 683, 1–24. lamina terminalis. J. Comp. Neurol. 209, 287–300.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 19

Kinzig, K.P., D’Alessio, D.A., Herman, J.P., Sakai, R.R., Vahl, T.P., Figueiredo, H.F., Lawson, A., Ahima, R., Krozowski, Z., Harlan, R., 1991. Postnatal development of
Murphy, E.K., Seeley, R.J., 2003. CNS glucagon-like peptide-1 receptors mediate corticosteroid receptor immunoreactivity in the rat hippocampus. Brain Res.
endocrine and anxiety responses to interoceptive and psychogenic stressors. J. Dev. Brain Res. 62, 69–79.
Neurosci. 23, 6163–6170. Lee, K.S., Han, T.H., Jo, J.Y., Kang, G., Lee, S.Y., Ryu, P.D., Im, J.H., Jeon, B.H., Park, J.B.,
Kiss, J.Z., 1985. Anatomical studies of cholecystokinin in neurons and pathways 2008. Serotonin inhibits GABA synaptic transmission in presympathetic
involved in neuroendocrine regulation. Ann. N. Y. Acad. Sci. 448, 144–151. paraventricular nucleus neurons. Neurosci. Lett. 439, 138–142.
Kiss, A., Aguilera, G., 1992. Participation of alpha 1-adrenergic receptors in the Lephart, E.D., 1993. Brain 5alpha-reductase: cellular, enzymatic, and molecular
secretion of hypothalamic corticotropin-releasing hormone during stress. perspectives and implications for biological function. Mol. Cell. Neurosci. 4,
Neuroendocrinology 56, 153–160. 473–484.
Kiss, J.Z., Mezey, E., Skirboll, L., 1984. Corticotropin-releasing factor- Leret, M.L., Molina-Holgado, F., Gonzalez, M.I., 1994. The effect of perinatal
immunoreactive neurons of the paraventricular nucleus become vasopressin exposure to estrogens on the sexually dimorphic response to novelty. Physiol.
positive after adrenalectomy. Proc. Natl. Acad. Sci. USA 81, 1854–1858. Behav. 55, 371–373.
Kiss, J.Z., Martos, J., Palkovits, M., 1991. Hypothalamic paraventricular nucleus: a Levin, E.R., 2005. Integration of the extranuclear and nuclear actions of estrogen.
quantitative analysis of cytoarchitectonic subdivisions in the rat. J. Comp. Mol. Endocrinol. 19, 1951–1959.
Neurol. 313, 563–573. Levine, J.E., Duffy, M.T., 1988. Simultaneous measurement of luteinizing hormone
Kitay, J.I., 1963. Pituitary–adrenal function in the rat after gonadectomy and (LH)-releasing hormone, LH, and follicle-stimulating hormone release in intact
gonadal hormone replacement. Endocrinology 73, 253–260. and short-term castrate rats. Endocrinology 122, 2211–2221.
Kitay, J.I., 1965. Depression of adrenal corticosterone production in Levine, J.E., Pau, K.Y., Ramirez, V.D., Jackson, G.L., 1982. Simultaneous measurement
oophorectomized rats. Endocrinology 77, 1048–1052. of luteinizing hormone-releasing hormone and luteinizing hormone release in
Kitazawa, S., Shioda, S., Nakai, Y., 1987. Catecholaminergic innervation of neurons unanesthetized, ovariectomized sheep. Endocrinology 111, 1449–1455.
containing corticotropin-releasing factor in the paraventricular nucleus of the Levine, J.E., Bauer-Dantoin, A.C., Besecke, L.M., Conaghan, L.A., Legan, S.J., Meredith,
rat hypothalamus. Acta Anat. (Basel) 129, 337–343. J.M., Strobl, F.J., Urban, J.H., Vogelsong, K.M., Wolfe, A.M., 1991. Neuroendocrine
Koegler-Muly, S.M., Owens, M.J., Ervin, G.N., Kilts, C.D., Nemeroff, C.B., 1993. regulation of the luteinizing hormone-releasing hormone pulse generator in the
Potential corticotropin-releasing factor pathways in the rat brain as determined rat. Recent Prog. Horm. Res. 47, 97–151, discussion 151-3.
by bilateral electrolytic lesions of the central amygdaloid nucleus and the Levkovitz, Y., Segal, M., 1997. Serotonin 5-HT1A receptors modulate hippocampal
paraventricular nucleus of the hypothalamus. J. Neuroendocrinol. 5, 95–98. reactivity to afferent stimulation. J. Neurosci. 17, 5591–5598.
Kolber, B.J., Muglia, L.J., 2009. Defining brain region-specific glucocorticoid action Levy, B.H., Tasker, J.G., 2012. Synaptic regulation of the hypothalamic–pituitary–
during stress by conditional gene disruption in mice. Brain Res. 1293, 85–90. adrenal axis and its modulation by glucocorticoids and stress. Front. Cell
Koohi, M.K., Ivell, R., Walther, N., 2005. Transcriptional activation of the oxytocin Neurosci. 6, 24.
promoter by oestrogens uses a novel non-classical mechanism of oestrogen Li, X.F., Kinsey-Jones, J.S., Cheng, Y., Knox, A.M., Lin, Y., Petrou, N.A., Roseweir, A.,
receptor action. J. Neuroendocrinol. 17, 197–207. Lightman, S.L., Milligan, S.R., Millar, R.P., O’Byrne, K.T., 2009. Kisspeptin
Koutcherov, Y., Mai, J.K., Ashwell, K.W., Paxinos, G., 2000. Organization of the signalling in the hypothalamic arcuate nucleus regulates GnRH pulse
human paraventricular hypothalamic nucleus. J. Comp. Neurol. 423, 299–318. generator frequency in the rat. PLoS ONE 4, e8334.
Kovacs, K., Kiss, J.Z., Makara, G.B., 1986. Glucocorticoid implants around the Lightman, S.L., Windle, R.J., Julian, M.D., Harbuz, M.S., Shanks, N., Wood, S.A.,
hypothalamic paraventricular nucleus prevent the increase of corticotropin- Kershaw, Y.M., Ingram, C.D., 2000. Significance of pulsatility in the HPA axis.
releasing factor and arginine vasopressin immunostaining induced by Novartis Found. Symp. 227, 244–257, discussion 257-60.
adrenalectomy. Neuroendocrinology 44, 229–234. Lincoln, G.A., Fraser, H.M., Abbott, M.P., 1986. Blockade of pulsatile LH, FSH and
Kovacs, K.J., Foldes, A., Sawchenko, P.E., 2000. Glucocorticoid negative feedback testosterone secretion in rams by constant infusion of an LHRH agonist. J.
selectively targets vasopressin transcription in parvocellular neurosecretory Reprod. Fertil. 77, 587–597.
neurons. J. Neurosci. 20, 3843–3852. Lind, R.W., Swanson, L.W., Sawchenko, P.E., 1985. Anatomical evidence that neural
Kow, L.M., Easton, A., Pfaff, D.W., 2005. Acute estrogen potentiates excitatory circuits related to the subfornical organ contain angiotensin II. Brain Res. Bull.
responses of neurons in rat hypothalamic ventromedial nucleus. Brain Res. 15, 79–82.
1043, 124–131. Liposits, Z., Paull, W.K., Setalo, G., Vigh, S., 1985. Evidence for local corticotropin
Krezel, W., Dupont, S., Krust, A., Chambon, P., Chapman, P.F., 2001. Increased anxiety releasing factor (CRF)-immunoreactive neuronal circuits in the paraventricular
and synaptic plasticity in estrogen receptor beta-deficient mice. Proc. Natl. nucleus of the rat hypothalamus. An electron microscopic
Acad. Sci. USA 98, 12278–12282. immunohistochemical analysis. Histochemistry 83, 5–16.
Kritzer, M., 2004. The distribution of immunoreactivity for intracellular androgen Liposits, Z., Sherman, D., Phelix, C., Paull, W.K., 1986. A combined light and electron
receptors in the cerebral cortex of hormonally intact adult male and female microscopic immunocytochemical method for the simultaneous localization of
rats: localization in pyramidal neurons making corticocortical connections. multiple tissue antigens. Tyrosine hydroxylase immunoreactive innervation of
Cereb. Cortex 14, 268–280. corticotropin releasing factor synthesizing neurons in the paraventricular
Kuiper, G.G., Carlsson, B., Grandien, K., Enmark, E., Haggblad, J., Nilsson, S., nucleus of the rat. Histochemistry 85, 95–106.
Gustafsson, J.A., 1997. Comparison of the ligand binding specificity and Liposits, Z., Uht, R.M., Harrison, R.W., Gibbs, F.P., Paull, W.K., Bohn, M.C., 1987.
transcript tissue distribution of estrogen receptors alpha and beta. Ultrastructural localization of glucocorticoid receptor (GR) in hypothalamic
Endocrinology 138, 863–870. paraventricular neurons synthesizing corticotropin releasing factor (CRF).
Kuiper, G.G., Enmark, E., Pelto-Huikko, M., Nilsson, S., Gustafsson, J.A., 1996. Cloning Histochemistry 87, 407–412.
of a novel receptor expressed in rat prostate and ovary. Proc. Natl. Acad. Sci. USA Liu, J., Bisschop, P.H., Eggels, L., Foppen, E., Fliers, E., Zhou, J.N., Kalsbeek, A., 2012.
93, 5925–5930. Intrahypothalamic estradiol modulates hypothalamus–pituitary–adrenal-axis
Kushner, P.J., Agard, D.A., Greene, G.L., Scanlan, T.S., Shiau, A.K., Uht, R.M., Webb, P., activity in female rats. Endocrinology 153, 3337–3344.
2000. Estrogen receptor pathways to AP-1. J. Steroid Biochem. Mol. Biol. 74, Lu, N.Z., Shlaes, T.A., Gundlah, C., Dziennis, S.E., Lyle, R.E., Bethea, C.L., 1999. Ovarian
311–317. steroid action on tryptophan hydroxylase protein and serotonin compared to
Laflamme, N., Nappi, R.E., Drolet, G., Labrie, C., Rivest, S., 1998. Expression and localization of ovarian steroid receptors in midbrain of guinea pigs. Endocrine
neuropeptidergic characterization of estrogen receptors (ERalpha and ERbeta) 11, 257–267.
throughout the rat brain: anatomical evidence of distinct roles of each subtype. Luine, V.N., Frankfurt, M., 2012. Estrogens facilitate memory processing through
J. Neurobiol. 36, 357–378. membrane mediated mechanisms and alterations in spine density. Front.
Lalmansingh, A.S., Uht, R.M., 2008. Estradiol regulates corticotropin-releasing Neuroendocrinol. 33, 388–402.
hormone gene (crh) expression in a rapid and phasic manner that parallels Luiten, P.G., ter Horst, G.J., Karst, H., Steffens, A.B., 1985. The course of
estrogen receptor-alpha and -beta recruitment to a 30 ,50 -cyclic adenosine 50 - paraventricular hypothalamic efferents to autonomic structures in medulla
monophosphate regulatory region of the proximal crh promoter. Endocrinology and spinal cord. Brain Res. 329, 374–378.
149, 346–357. Lund, T.D., Munson, D.J., Haldy, M.E., Handa, R.J., 2004a. Dihydrotestosterone may
Lambert, J.J., Belelli, D., Harney, S.C., Peters, J.A., Frenguelli, B.G., 2001. Modulation of inhibit hypothalamo–pituitary–adrenal activity by acting through estrogen
native and recombinant GABA(A) receptors by endogenous and synthetic receptor in the male mouse. Neurosci. Lett. 365, 43–47.
neuroactive steroids. Brain Res. Brain Res. Rev. 37, 68–80. Lund, T.D., Munson, D.J., Haldy, M.E., Handa, R.J., 2004b. Androgen inhibits, while
Landgraf, R., Neumann, I.D., 2004. Vasopressin and oxytocin release within the oestrogen enhances, restraint-induced activation of neuropeptide neurones in
brain: a dynamic concept of multiple and variable modes of neuropeptide the paraventricular nucleus of the hypothalamus. J. Neuroendocrinol. 16, 272–
communication. Front. Neuroendocrinol. 25, 150–176. 278.
Larkin, J.W., Binks, S.L., Li, Y., Selvage, D., 2010. The role of oestradiol in sexually Lund, T.D., Rovis, T., Chung, W.C., Handa, R.J., 2005. Novel actions of estrogen
dimorphic hypothalamic–pituitary–adrena axis responses to receptor-beta on anxiety-related behaviors. Endocrinology 146, 797–807.
intracerebroventricular ethanol administration in the rat. J. Neuroendocrinol. Lund, T.D., Hinds, L.R., Handa, R.J., 2006. The androgen 5alpha-dihydrotestosterone
22, 24–32. and its metabolite 5alpha-androstan-3beta, 17beta-diol inhibit the
Larsen, P.J., Tang-Christensen, M., Jessop, D.S., 1997. Central administration of hypothalamo–pituitary–adrenal response to stress by acting through estrogen
glucagon-like peptide-1 activates hypothalamic neuroendocrine neurons in the receptor beta-expressing neurons in the hypothalamus. J. Neurosci. 26, 1448–
rat. Endocrinology 138, 4445–4455. 1456.
Lawrence, A.J., Jarrott, B., 1996. Neurochemical modulation of cardiovascular Luoma, J.I., Boulware, M.I., Mermelstein, P.G., 2008. Caveolin proteins and estrogen
control in the nucleus tractus solitarius. Prog. Neurobiol. 48, 21–53. signaling in the brain. Mol. Cell. Endocrinol. 290, 8–13.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
20 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Maeda, K., Ohkura, S., Uenoyama, Y., Wakabayashi, Y., Oka, Y., Tsukamura, H., Nair, R.M., Barrett, J.F., Bowers, C.Y., Schally, A.V., 1970. Structure of porcine
Okamura, H., 2010. Neurobiological mechanisms underlying GnRH pulse thyrotropin releasing hormone. Biochemistry 9, 1103–1106.
generation by the hypothalamus. Brain Res. 1364, 103–115. Neigh, G.N., Owens, M.J., Taylor, W.R., Nemeroff, C.B., 2010. Changes in the vascular
Malkoski, S.P., Dorin, R.I., 1999. Composite glucocorticoid regulation at a area fraction of the hippocampus and amygdala are induced by prenatal
functionally defined negative glucocorticoid response element of the human dexamethasone and/or adult stress. J. Cereb. Blood Flow Metab. 30, 1100–1104.
corticotropin-releasing hormone gene. Mol. Endocrinol. 13, 1629–1644. Neumann, I.D., 2007. Stimuli and consequences of dendritic release of oxytocin
Malkoski, S.P., Handanos, C.M., Dorin, R.I., 1997. Localization of a negative within the brain. Biochem. Soc. Trans. 35, 1252–1257.
glucocorticoid response element of the human corticotropin releasing Neumann, I.D., Kromer, S.A., Toschi, N., Ebner, K., 2000a. Brain oxytocin inhibits the
hormone gene. Mol. Cell. Endocrinol. 127, 189–199. (re)activity of the hypothalamo–pituitary–adrenal axis in male rats:
Mangelsdorf, D.J., Thummel, C., Beato, M., Herrlich, P., Schutz, G., Umesono, K., involvement of hypothalamic and limbic brain regions. Regul. Pept. 96, 31–38.
Blumberg, B., Kastner, P., Mark, M., Chambon, P., Evans, R.M., 1995. The nuclear Neumann, I.D., Wigger, A., Torner, L., Holsboer, F., Landgraf, R., 2000b. Brain
receptor superfamily: the second decade. Cell 83, 835–839. oxytocin inhibits basal and stress-induced activity of the hypothalamo–
Mantella, R.C., Vollmer, R.R., Rinaman, L., Li, X., Amico, J.A., 2004. Enhanced pituitary–adrenal axis in male and female rats: partial action within the
corticosterone concentrations and attenuated Fos expression in the medial paraventricular nucleus. J. Neuroendocrinol. 12, 235–243.
amygdala of female oxytocin knockout mice exposed to psychogenic stress. Am. Nomura, M., Saito, J., Ueta, Y., Muglia, L.J., Pfaff, D.W., Ogawa, S., 2003. Enhanced up-
J. Physiol. Regul. Integr. Comp. Physiol. 287, R1494–R1504. regulation of corticotropin-releasing hormone gene expression in response to
McAllister, C.E., Creech, R.D., Kimball, P.A., Muma, N.A., Li, Q., 2012. GPR30 is restraint stress in the hypothalamic paraventricular nucleus of oxytocin gene-
necessary for estradiol-induced desensitization of 5-HT1A receptor signaling in deficient male mice. J. Neuroendocrinol. 15, 1054–1061.
the paraventricular nucleus of the rat hypothalamus. Nomura, M., Akama, K.T., Alves, S.E., Korach, K.S., Gustafsson, J.A., Pfaff, D.W.,
Psychoneuroendocrinology 37, 1248–1260. Ogawa, S., 2005. Differential distribution of estrogen receptor (ER)-alpha and
McCann, S.M., 1953. Effect of hypothalamic lesions on the adrenal cortical response ER-beta in the midbrain raphe nuclei and periaqueductal gray in male mouse:
to stress in the rat. Am. J. Physiol. 175, 13–20. predominant role of ER-beta in midbrain serotonergic systems. Neuroscience
McCann, S.M., Brobeck, J.R., 1954. Evidence for a role of the supraopticohypophyseal 130, 445–456.
system in regulation of adrenocorticotrophin secretion. Proc. Soc. Exp. Biol. Ochedalski, T., Subburaju, S., Wynn, P.C., Aguilera, G., 2007. Interaction between
Med. 87, 318–324. oestrogen and oxytocin on hypothalamic–pituitary–adrenal axis activity. J.
McEwen, B.S., 1998. Protective and damaging effects of stress mediators. N. Engl. J. Neuroendocrinol. 19, 189–197.
Med. 338, 171–179. Ofner, P., Sousa, R.L., Vena, R.L., Krinsky-Feibush, P., LeQuesne, P.W., 1983.
McGimsey, W.C., Cidlowski, J.A., Stumpf, W.E., Sar, M., 1991. Immunocytochemical Metabolism of radiotestosterone to 3 beta,6 alpha- and 3 beta,7 alpha-
localization of the glucocorticoid receptor in rat brain, pituitary, liver, and dihydroxy 5 alpha-steroids by rat ventral, canine and human prostate in
thymus with two new polyclonal antipeptide antibodies. Endocrinology 129, organ culture. J. Steroid Biochem. 18, 415–423.
3064–3072. Ogura, E., Kageyama, K., Hanada, K., Kasckow, J., Suda, T., 2008. Effects of estradiol
McGinnis, M.Y., Davis, P.G., Meaney, M.J., Singer, M., McEwen, B.S., 1983. In vitro on regulation of corticotropin-releasing factor gene and interleukin-6
measurement of cytosol and cell nuclear androgen receptors in male rat brain production via estrogen receptor type beta in hypothalamic 4B cells. Peptides
and pituitary. Brain Res. 275, 75–82. 29, 456–464.
McIntyre, D.C., Kent, P., Hayley, S., Merali, Z., Anisman, H., 1999. Influence of Olijslagers, J.E., de Kloet, E.R., Elgersma, Y., van Woerden, G.M., Joels, M., Karst, H.,
psychogenic and neurogenic stressors on neuroendocrine and central 2008. Rapid changes in hippocampal CA1 pyramidal cell function via pre- as
monoamine activity in fast and slow kindling rats. Brain Res. 840, 65–74. well as postsynaptic membrane mineralocorticoid receptors. Eur. J. Neurosci.
McKenna, N.J., Xu, J., Nawaz, Z., Tsai, S.Y., Tsai, M.J., O’Malley, B.W., 1999. Nuclear 27, 2542–2550.
receptor coactivators: multiple enzymes, multiple complexes, multiple Oster, H., Damerow, S., Kiessling, S., Jakubcakova, V., Abraham, D., Tian, J., Hoffmann,
functions. J. Steroid Biochem. Mol. Biol. 69, 3–12. M.W., Eichele, G., 2006. The circadian rhythm of glucocorticoids is regulated by a
Meijer, O.C., de Lange, E.C., Breimer, D.D., de Boer, A.G., Workel, J.O., de Kloet, E.R., gating mechanism residing in the adrenal cortical clock. Cell Metab. 4, 163–173.
1998. Penetration of dexamethasone into brain glucocorticoid targets is Overstreet, D.H., Osterlund, M., Dahllund, J., Appelqvist, T., Lindstrom, E., Ryan, C.N.,
enhanced in mdr1A P-glycoprotein knockout mice. Endocrinology 139, 1789– Witt, M.R., 2006. Estrogen receptor beta agonists reduce exaggerated swim test
1793. immobility in a genetic animal model of depression. In: Abstract 476.11. Society
Meitzen, J., Mermelstein, P.G., 2011. Estrogen receptors stimulate brain region for Neuroscience 36th Annual Meeting, Atlanta, GA.
specific metabotropic glutamate receptors to rapidly initiate signal Oyola, M.G., Portillo, W., Reyna, A., Foradori, C.D., Kudwa, A., Hinds, L., Handa, R.J.,
transduction pathways. J. Chem. Neuroanat. 42, 236–241. Mani, S.K., 2012. Anxiolytic effects and neuroanatomical targets of estrogen
Menendez, A., Alvarez-Uria, M., 1987. The development of vascularization in the receptor-beta (ERbeta) activation by a selective ERbeta agonist in female mice.
postnatal rat paraventricular nucleus: a morphometric analysis. J. Hirnforsch. Endocrinology 153, 837–846.
28, 325–329. Pacak, K., 2000. Stressor-specific activation of the hypothalamic–pituitary–
Merchenthaler, I., Kovacs, G., Lavasz, G., Setalo, G., 1980. The preoptico-infundibular adrenocortical axis. Physiol. Res. 49 (Suppl. 1), S11–S17.
LH–RH tract of the rat. Brain Res. 198, 63–74. Pacak, K., Palkovits, M., Kvetnansky, R., Kopin, I.J., Goldstein, D.S., 1993. Stress-
Merchenthaler, I., Lane, M., Shughrue, P., 1999. Distribution of pre-pro-glucagon induced norepinephrine release in the paraventricular nucleus of rats with
and glucagon-like peptide-1 receptor messenger RNAs in the rat central brainstem hemisections: a microdialysis study. Neuroendocrinology 58, 196–
nervous system. J. Comp. Neurol. 403, 261–280. 201.
Mermelstein, P.G., 2009. Membrane-localised oestrogen receptor alpha and beta Pacak, K., Palkovits, M., Kopin, I.J., Goldstein, D.S., 1995. Stress-induced
influence neuronal activity through activation of metabotropic glutamate norepinephrine release in the hypothalamic paraventricular nucleus and
receptors. J. Neuroendocrinol. 21, 257–262. pituitary–adrenocortical and sympathoadrenal activity: in vivo microdialysis
Mezey, E., Kiss, J.Z., Skirboll, L.R., Goldstein, M., Axelrod, J., 1984. Increase of studies. Front. Neuroendocrinol. 16, 89–150.
corticotropin-releasing factor staining in rat paraventricular nucleus neurones Paech, K., Webb, P., Kuiper, G.G., Nilsson, S., Gustafsson, J., Kushner, P.J., Scanlan, T.S.,
by depletion of hypothalamic adrenaline. Nature 310, 140–141. 1997. Differential ligand activation of estrogen receptors ERalpha and ERbeta at
Mhyre, A.J., Dorsa, D.M., 2006. Estrogen activates rapid signaling in the brain: role of AP1 sites. Science 277, 1508–1510.
estrogen receptor alpha and estrogen receptor beta in neurons and glia. Pak, T.R., Chung, W.C., Roberts, J.L., Handa, R.J., 2006. Ligand-independent effects of
Neuroscience 138, 851–858. estrogen receptor beta on mouse gonadotropin-releasing hormone promoter
Micevych, P.E., Mermelstein, P.G., 2008. Membrane estrogen receptors acting activity. Endocrinology 147, 1924–1931.
through metabotropic glutamate receptors: an emerging mechanism of Pak, T., Chung, W., Handa, R., 2007. Estrogen receptor-beta mediates DHT-induced
estrogen action in brain. Mol. Neurobiol. 38, 66–77. stimulation of the arginine vasopressin promoter in neuronal cells.
Miller, W.J., Suzuki, S., Miller, L.K., Handa, R., Uht, R.M., 2004. Estrogen receptor Endocrinology.
(ER)beta isoforms rather than ERalpha regulate corticotropin-releasing Palermo-Neto, J., Dorce, V.A., 1990. Influences of estrogen and/or progesterone on
hormone promoter activity through an alternate pathway. J. Neurosci. 24, some dopamine related behavior in rats. Gen. Pharmacol. 21, 83–87.
10628–10635. Papadimitriou, A., Priftis, K.N., 2009. Regulation of the hypothalamic–pituitary–
Milner, T.A., Reis, D.J., Pickel, V.M., Aicher, S.A., Giuliano, R., 1993. Ultrastructural adrenal axis. NeuroImmunoModulation 16, 265–271.
localization and afferent sources of corticotropin-releasing factor in the rat Papadopoulou, N., Papakonstanti, E.A., Kallergi, G., Alevizopoulos, K., Stournaras, C.,
rostral ventrolateral medulla: implications for central cardiovascular 2009. Membrane androgen receptor activation in prostate and breast tumor
regulation. J. Comp. Neurol. 333, 151–167. cells: molecular signaling and clinical impact. IUBMB Life 61, 56–61.
Morimoto, M., Morita, N., Ozawa, H., Yokoyama, K., Kawata, M., 1996. Distribution Patchev, V.K., Hassan, A.H., Holsboer, D.F., Almeida, O.F., 1996. The neurosteroid
of glucocorticoid receptor immunoreactivity and mRNA in the rat brain: an tetrahydroprogesterone attenuates the endocrine response to stress and exerts
immunohistochemical and in situ hybridization study. Neurosci. Res. 26, 235– glucocorticoid-like effects on vasopressin gene transcription in the rat
269. hypothalamus. Neuropsychopharmacology 15, 533–540.
Muglia, L.J., Jacobson, L., Weninger, S.C., Luedke, C.E., Bae, D.S., Jeong, K.H., Majzoub, Patisaul, H.B., Scordalakes, E.M., Young, L.J., Rissman, E.F., 2003. Oxytocin, but not
J.A., 1997. Impaired diurnal adrenal rhythmicity restored by constant infusion oxytocin receptor, is regulated by oestrogen receptor beta in the female mouse
of corticotropin-releasing hormone in corticotropin-releasing hormone- hypothalamus. J. Neuroendocrinol. 15, 787–793.
deficient mice. J. Clin. Invest. 99, 2923–2929. Pecoraro, N., Dallman, M.F., Warne, J.P., Ginsberg, A.B., Laugero, K.D., la Fleur, S.E.,
Murphy, D.D., Cole, N.B., Greenberger, V., Segal, M., 1998. Estradiol increases Houshyar, H., Gomez, F., Bhargava, A., Akana, S.F., 2006. From Malthus to
dendritic spine density by reducing GABA neurotransmission in hippocampal motive: how the HPA axis engineers the phenotype, yoking needs to wants.
neurons. J. Neurosci. 18, 2550–2559. Prog. Neurobiol. 79, 247–340.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 21

Philibert, D., Moguilewsky, M., 1983. RU 28362, a useful tool for the Roselli, C.E., 1991. Sex differences in androgen receptors and aromatase activity in
characterization of glucocorticoid and mineralocorticoid receptors. In: 65th microdissected regions of the rat brain. Endocrinology 128, 1310–1316.
Annual Meeting of the Endocrine Soc. P335. Roselli, C.E., Horton, L.E., Resko, J.A., 1985. Distribution and regulation of aromatase
Platania, P., Laureanti, F., Bellomo, M., Giuffrida, R., Giuffrida-Stella, A.M., Catania, activity in the rat hypothalamus and limbic system. Endocrinology 117, 2471–
M.V., Sortino, M.A., 2003. Differential expression of estrogen receptors alpha 2477.
and beta in the spinal cord during postnatal development: localization in glial Roselli, C.E., Abdelgadir, S.E., Resko, J.A., 1997. Regulation of aromatase gene
cells. Neuroendocrinology 77, 334–340. expression in the adult rat brain. Brain Res. Bull. 44, 351–357.
Plotsky, P.M., Sawchenko, P.E., 1987. Hypophysial-portal plasma levels, median Rosner, W., 1990. The functions of corticosteroid-binding globulin and sex
eminence content, and immunohistochemical staining of corticotropin- hormone-binding globulin: recent advances. Endocr. Rev. 11, 80–91.
releasing factor, arginine vasopressin, and oxytocin after pharmacological Rossi, D.V., Dai, Y., Thomas, P., Carrasco, G.A., DonCarlos, L.L., Muma, N.A., Li, Q.,
adrenalectomy. Endocrinology 120, 1361–1369. 2010. Estradiol-induced desensitization of 5-HT1A receptor signaling in the
Plotsky, P.M., Cunningham Jr., E.T., Widmaier, E.P., 1989. Catecholaminergic paraventricular nucleus of the hypothalamus is independent of estrogen
modulation of corticotropin-releasing factor and adrenocorticotropin receptor-beta. Psychoneuroendocrinology 35, 1023–1033.
secretion. Endocr. Rev. 10, 437–458. Roy, B.N., Reid, R.L., Van Vugt, D.A., 1999. The effects of estrogen and progesterone
Porsolt, R.D., Le Pichon, M., Jalfre, M., 1977. Depression: a new animal model on corticotropin-releasing hormone and arginine vasopressin messenger
sensitive to antidepressant treatments. Nature 266, 730–732. ribonucleic acid levels in the paraventricular nucleus and supraoptic nucleus
Porter, R.W., 1953. Hypothalamic involvement in the pituitary–adrenocortical of the rhesus monkey. Endocrinology 140, 2191–2198.
response to stress stimuli. Am. J. Physiol. 172, 515–519. Rozeboom, A.M., Akil, H., Seasholtz, A.F., 2007. Mineralocorticoid receptor
Prewitt, C.M., Herman, J.P., 1998. Anatomical interactions between the central overexpression in forebrain decreases anxiety-like behavior and alters the
amygdaloid nucleus and the hypothalamic paraventricular nucleus of the rat: a stress response in mice. Proc. Natl. Acad. Sci. USA 104, 4688–4693.
dual tract-tracing analysis. J. Chem. Neuroanat. 15, 173–185. Rubinow, D.R., Roca, C.A., Schmidt, P.J., Danaceau, M.A., Putnam, K., Cizza, G.,
Price Jr., R.H., Butler, C.A., Webb, P., Uht, R., Kushner, P., Handa, R.J., 2001. A splice Chrousos, G., Nieman, L., 2005. Testosterone suppression of CRH-stimulated
variant of estrogen receptor beta missing exon 3 displays altered subnuclear cortisol in men. Neuropsychopharmacology 30, 1906–1912.
localization and capacity for transcriptional activation. Endocrinology 142, Safe, S., Kim, K., 2008. Nonclassical genomic ER/Sp and ER/AP-1 signaling pathways.
2039–2049. J. Mol. Endocrinol..
Prossnitz, E.R., Arterburn, J.B., Smith, H.O., Oprea, T.I., Sklar, L.A., Hathaway, H.J., Saijo, K., Collier, J.G., Li, A.C., Katzenellenbogen, J.A., Glass, C.K., 2011. An ADIOL-
2008. Estrogen signaling through the transmembrane G protein-coupled ERbeta-CtBP transrepression pathway negatively regulates microglia-mediated
receptor GPR30. Annu. Rev. Physiol. 70, 165–190. inflammation. Cell 145, 584–595.
Pyner, S., Coote, J.H., 2000. Identification of branching paraventricular neurons of Sands, S.A., Morilak, D.A., 1999. Expression of alpha1D adrenergic receptor messenger
the hypothalamus that project to the rostroventrolateral medulla and spinal RNA in oxytocin- and corticotropin-releasing hormone-synthesizing neurons in
cord. Neuroscience 100, 549–556. the rat paraventricular nucleus. Neuroscience 91, 639–649.
Quirk, S.J., Funder, J.W., 1988. Steroid receptors, and the generation of closely Santagati, S., Melcangi, R.C., Celotti, F., Martini, L., Maggi, A., 1994. Estrogen receptor
coupled/biphasic dose–response curves. J. Steroid Biochem. 30, 9–15. is expressed in different types of glial cells in culture. J. Neurochem. 63, 2058–
Raadsheer, F.C., Sluiter, A.A., Ravid, R., Tilders, F.J., Swaab, D.F., 1993. Localization of 2064.
corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus Saper, C.B., 1995. Central autonomic control. In: Paxinos, G. (Ed.), The Rat Nervous
of the human hypothalamus; age-dependent colocalization with vasopressin. System. Academic Press, San Diego, pp. 107–135.
Brain Res. 615, 50–62. Saper, C.B., Swanson, L.W., Cowan, W.M., 1976. The efferent connections of the
Rabadan-Diehl, C., Makara, G., Kiss, A., Zelena, D., Aguilera, G., 1997. Regulation of ventromedial nucleus of the hypothalamus of the rat. J. Comp. Neurol. 169,
pituitary corticotropin releasing hormone (CRH) receptor mRNA and CRH 409–442.
binding during adrenalectomy: role of glucocorticoids and hypothalamic Sapolsky, R.M., Krey, L.C., McEwen, B.S., 1984. Glucocorticoid-sensitive hippocampal
factors. J. Neuroendocrinol. 9, 689–697. neurons are involved in terminating the adrenocortical stress response. Proc.
Rajkowska, G., Miguel-Hidalgo, J.J., 2007. Gliogenesis and glial pathology in Natl. Acad. Sci. USA 81, 6174–6177.
depression. CNS Neurol. Disord.: Drug Targets 6, 219–233. Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids influence
Reul, J.M., de Kloet, E.R., 1985. Two receptor systems for corticosterone in rat brain: stress responses? Integrating permissive, suppressive, stimulatory, and
microdistribution and differential occupation. Endocrinology 117, 2505–2511. preparative actions. Endocr. Rev. 21, 55–89.
Reul, J.M., Holsboer, F., 2002. Corticotropin-releasing factor receptors 1 and 2 in Sar, M., Stumpf, W.E., 1973. Autoradiographic localization of radioactivity in the rat
anxiety and depression. Curr. Opin. Pharmacol. 2, 23–33. brain after the injection of 1,2-3H-testosterone. Endocrinology 92, 251–256.
Reul, J.M., de Kloet, E.R., van Sluijs, F.J., Rijnberk, A., Rothuizen, J., 1990. Binding Sar, M., Stumpf, W.E., 1977. Distribution of androgen target cells in rat forebrain and
characteristics of mineralocorticoid and glucocorticoid receptors in dog brain pituitary after [3H]-dihydrotestosterone administration. J. Steroid Biochem. 8,
and pituitary. Endocrinology 127, 907–915. 1131–1135.
Revankar, C.M., Cimino, D.F., Sklar, L.A., Arterburn, J.B., Prossnitz, E.R., 2005. A Sarabdjitsingh, R.A., Conway-Campbell, B.L., Leggett, J.D., Waite, E.J., Meijer, O.C., de
transmembrane intracellular estrogen receptor mediates rapid cell signaling. Kloet, E.R., Lightman, S.L., 2010. Stress responsiveness varies over the ultradian
Science 307, 1625–1630. glucocorticoid cycle in a brain-region-specific manner. Endocrinology 151,
Rho, J.H., Swanson, L.W., 1989. A morphometric analysis of functionally defined 5369–5379.
subpopulations of neurons in the paraventricular nucleus of the rat with Sarkar, S., Fekete, C., Legradi, G., Lechan, R.M., 2003. Glucagon like peptide-1 (7–36)
observations on the effects of colchicine. J. Neurosci. 9, 1375–1388. amide (GLP-1) nerve terminals densely innervate corticotropin-releasing
Rhodes, C.H., Morrell, J.I., Pfaff, D.W., 1981. Immunohistochemical analysis of hormone neurons in the hypothalamic paraventricular nucleus. Brain Res.
magnocellular elements in rat hypothalamus: distribution and numbers of cells 985, 163–168.
containing neurophysin, oxytocin, and vasopressin. J. Comp. Neurol. 198, 45– Sarkey, S., Azcoitia, I., Garcia-Segura, L.M., Garcia-Ovejero, D., DonCarlos, L.L., 2008.
64. Classical androgen receptors in non-classical sites in the brain. Horm. Behav. 53,
Rhodes, M.E., Kennell, J.S., Belz, E.E., Czambel, R.K., Rubin, R.T., 2004. Rat estrous 753–764.
cycle influences the sexual diergism of HPA axis stimulation by nicotine. Brain Sato, S.M., Johansen, J.A., Jordan, C.L., Wood, R.I., 2010. Membrane androgen
Res. Bull. 64, 205–213. receptors may mediate androgen reinforcement. Psychoneuroendocrinology 35,
Richard, S., Zingg, H.H., 1990. The human oxytocin gene promoter is regulated by 1063–1073.
estrogens. J. Biol. Chem. 265, 6098–6103. Sawchenko, P.E., 1987a. Evidence for a local site of action for glucocorticoids in
Richard, S., Zingg, H.H., 1991. Identification of a retinoic acid response element in inhibiting CRF and vasopressin expression in the paraventricular nucleus. Brain
the human oxytocin promoter. J. Biol. Chem. 266, 21428–21433. Res. 403, 213–223.
Ring, R.H., 2005. The central vasopressinergic system: examining the opportunities Sawchenko, P.E., 1987b. Evidence for differential regulation of corticotropin-
for psychiatric drug development. Curr. Pharm. Des. 11, 205–225. releasing factor and vasopressin immunoreactivities in parvocellular
Rivier, C., Vale, W., 1983. Interaction of corticotropin-releasing factor and arginine neurosecretory and autonomic-related projections of the paraventricular
vasopressin on adrenocorticotropin secretion in vivo. Endocrinology 113, 939– nucleus. Brain Res. 437, 253–263.
942. Sawchenko, P.E., Swanson, L.W., 1981. Central noradrenergic pathways for the
Rizk, A., Robertson, J., Raber, J., 2005. Behavioral performance of tfm mice supports integration of hypothalamic neuroendocrine and autonomic responses. Science
the beneficial role of androgen receptors in spatial learning and memory. Brain 214, 685–687.
Res. 1034, 132–138. Sawchenko, P.E., Swanson, L.W., 1982. Immunohistochemical identification of
Rizner, T.L., Lin, H.K., Peehl, D.M., Steckelbroeck, S., Bauman, D.R., Penning, T.M., neurons in the paraventricular nucleus of the hypothalamus that project to the
2003. Human type 3 3alpha-hydroxysteroid dehydrogenase (aldo-keto medulla or to the spinal cord in the rat. J. Comp. Neurol. 205, 260–272.
reductase 1C2) and androgen metabolism in prostate cells. Endocrinology Sawchenko, P.E., Swanson, L.W., 1983. The organization of forebrain afferents to the
144, 2922–2932. paraventricular and supraoptic nuclei of the rat. J. Comp. Neurol. 218, 121–144.
Rocha, B.A., Fleischer, R., Schaeffer, J.M., Rohrer, S.P., Hickey, G.J., 2005. 17 Beta- Sawchenko, P.E., Swanson, L.W., Steinbusch, H.W., Verhofstad, A.A., 1983. The
estradiol-induced antidepressant-like effect in the forced swim test is absent in distribution and cells of origin of serotonergic inputs to the paraventricular and
estrogen receptor-beta knockout (BERKO) mice. Psychopharmacology 179, supraoptic nuclei of the rat. Brain Res. 277, 355–360.
637–643. Sawchenko, P.E., Swanson, L.W., Vale, W.W., 1984. Co-expression of corticotropin-
Roland, B.L., Sawchenko, P.E., 1993. Local origins of some GABAergic projections to releasing factor and vasopressin immunoreactivity in parvocellular
the paraventricular and supraoptic nuclei of the hypothalamus in the rat. J. neurosecretory neurons of the adrenalectomized rat. Proc. Natl. Acad. Sci. USA
Comp. Neurol. 332, 123–143. 81, 1883–1887.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
22 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

Sayers, G., 1950. The adrenal cortex and homoestasis. Physiol. Rev. 30, 241–320. Simerly, R.B., Chang, C., Muramatsu, M., Swanson, L.W., 1990. Distribution of
Schally, A.V., Arimura, A., Kastin, A.J., Matsuo, H., Baba, Y., Redding, T.W., Nair, R.M., androgen and estrogen receptor mRNA-containing cells in the rat brain: an
Debeljuk, L., White, W.F., 1971a. Gonadotropin-releasing hormone: one in situ hybridization study. J. Comp. Neurol. 294, 76–95.
polypeptide regulates secretion of luteinizing and follicle-stimulating Simonian, S.X., Herbison, A.E., 1997. Differential expression of estrogen receptor
hormones. Science 173, 1036–1038. alpha and beta immunoreactivity by oxytocin neurons of rat paraventricular
Schally, A.V., Baba, Y., Nair, R.M., Bennett, C.D., 1971b. The amino acid sequence of a nucleus. J. Neuroendocrinol. 9, 803–806.
peptide with growth hormone-releasing activity isolated from porcine Sladek, C.D., Somponpun, S.J., 2004. Oestrogen receptor beta: role in
hypothalamus. J. Biol. Chem. 246, 6647–6650. neurohypophyseal neurones. J. Neuroendocrinol. 16, 365–371.
Schally, A.V., Nair, R.M., Redding, T.W., Arimura, A., 1971c. Isolation of the Slob, A.K., Bogers, H., van Stolk, M.A., 1981. Effects of gonadectomy and exogenous
luteinizing hormone and follicle-stimulating hormone-releasing hormone gonadal steroids on sex differences in open field behaviour of adult rats. Behav.
from porcine hypothalami. J. Biol. Chem. 246, 7230–7236. Brain Res. 2, 347–362.
Schlosser, S.F., Almeida, O.F., Patchev, V.K., Yassouridis, A., Elands, J., 1994. Sofroniew, M.V., Schrell, U., 1982. Evidence for a direct projection from oxitocin and
Oxytocin-stimulated release of adrenocorticotropin from the rat pituitary is vasopressin neurons in the hypothalamic paraventricular nucleus to the
mediated by arginine vasopressin receptors of the V1b type. Endocrinology 135, medulla oblongata: immunohistochemical visualization of both the
2058–2063. horseradish peroxidase transported and the peptide produced by the same
Schmidt, B.M., Gerdes, D., Feuring, M., Falkenstein, E., Christ, M., Wehling, M., 2000. neurons. Neurosci. Lett. 22, 211–217.
Rapid, nongenomic steroid actions: a new age? Front. Neuroendocrinol. 21, 57– Somponpun, S., Sladek, C.D., 2002. Role of estrogen receptor-beta in regulation of
94. vasopressin and oxytocin release in vitro. Endocrinology 143, 2899–2904.
Schmidt, E.M., Gu, S., Anagnostopoulou, V., Alevizopoulos, K., Foller, M., Lang, F., Southworth, M.B., Matsumoto, A.M., Gross, K.M., Soules, M.R., Bremner, W.J., 1991.
Stournaras, C., 2012. Serum- and glucocorticoid-dependent kinase-1-induced The importance of signal pattern in the transmission of endocrine information:
cell migration is dependent on vinculin and regulated by the membrane pituitary gonadotropin responses to continuous and pulsatile gonadotropin-
androgen receptor. FEBS J. 279, 1231–1242. releasing hormone. J. Clin. Endocrinol. Metab. 72, 1286–1289.
Seale, J.V., Wood, S.A., Atkinson, H.C., Harbuz, M.S., Lightman, S.L., 2004. Gonadal Spiteri, T., Ogawa, S., Musatov, S., Pfaff, D.W., Agmo, A., 2012. The role of the
steroid replacement reverses gonadectomy-induced changes in the estrogen receptor alpha in the medial preoptic area in sexual incentive
corticosterone pulse profile and stress-induced hypothalamic–pituitary– motivation, proceptivity and receptivity, anxiety, and wheel running in
adrenal axis activity of male and female rats. J. Neuroendocrinol. 16, female rats. Behav. Brain Res. 230, 11–20.
989–998. Steckelbroeck, S., Jin, Y., Gopishetty, S., Oyesanmi, B., Penning, T.M., 2004. Human
Seale, J.V., Wood, S.A., Atkinson, H.C., Harbuz, M.S., Lightman, S.L., 2005a. Postnatal cytosolic 3alpha-hydroxysteroid dehydrogenases of the aldo–keto reductase
masculinization alters the HPA axis phenotype in the adult female rat. J. Physiol. superfamily display significant 3beta-hydroxysteroid dehydrogenase activity:
563, 265–274. implications for steroid hormone metabolism and action. J. Biol. Chem. 279,
Seale, J.V., Wood, S.A., Atkinson, H.C., Lightman, S.L., Harbuz, M.S., 2005b. 10784–10795.
Organizational role for testosterone and estrogen on adult hypothalamic– Stenoien, D.L., Mancini, M.G., Patel, K., Allegretto, E.A., Smith, C.L., Mancini, M.A.,
pituitary–adrenal axis activity in the male rat. Endocrinology 146, 1973–1982. 2000. Subnuclear trafficking of estrogen receptor-alpha and steroid receptor
Seasholtz, A.F., Thompson, R.C., Douglass, J.O., 1988. Identification of a cyclic coactivator-1. Mol. Endocrinol. 14, 518–534.
adenosine monophosphate-responsive element in the rat corticotropin- Strohle, A., Holsboer, F., 2003. Stress responsive neurohormones in depression and
releasing hormone gene. Mol. Endocrinol. 2, 1311–1319. anxiety. Pharmacopsychiatry 36 (Suppl. 3), S207–S214.
Seeley, R.J., Blake, K., Rushing, P.A., Benoit, S., Eng, J., Woods, S.C., D’Alessio, D., 2000. Sutanto, W., De Kloet, E.R., 1987. Species-specificity of corticosteroid receptors in
The role of CNS glucagon-like peptide-1 (7–36) amide receptors in mediating hamster and rat brains. Endocrinology 121, 1405–1411.
the visceral illness effects of lithium chloride. J. Neurosci. 20, 1616–1621. Suzuki, S., Handa, R.J., 2004. Regulation of estrogen receptor-beta expression in the
Serova, L.I., Harris, H.A., Maharjan, S., Sabban, E.L., 2010. Modulation of responses to female rat hypothalamus: differential effects of dexamethasone and estradiol.
stress by estradiol benzoate and selective estrogen receptor agonists. J. Endocrinology 145, 3658–3670.
Endocrinol. 205, 253–262. Suzuki, S., Handa, R.J., 2005. Estrogen receptor-beta, but not estrogen receptor-
Shafton, A.D., Ryan, A., Badoer, E., 1998. Neurons in the hypothalamic alpha, is expressed in prolactin neurons of the female rat paraventricular and
paraventricular nucleus send collaterals to the spinal cord and to the rostral supraoptic nuclei: comparison with other neuropeptides. J. Comp. Neurol. 484,
ventrolateral medulla in the rat. Brain Res. 801, 239–243. 28–42.
Shakil, T., Hoque, A.N., Husain, M., Belsham, D.D., 2002. Differential regulation of Suzuki, S., Lund, T.D., Price, R.H., Handa, R.J. (Eds.), 2001a. Sex differences in the
gonadotropin-releasing hormone secretion and gene expression by androgen: Hypothalamo–Pituitary–Adrenal Axis: Novel Roles for Androgen and Estrogen
membrane versus nuclear receptor activation. Mol. Endocrinol. 16, 2592–2602. Receptors. Transworld Research Network, Trivandrum.
Shansky, R.M., Lipps, J., 2013. Stress-induced cognitive dysfunction: hormone- Suzuki, S., Solberg, L.C., Redei, E.E., Handa, R.J., 2001b. Prepro-thyrotropin releasing
neurotransmitter interactions in the prefrontal cortex. Front. Hum. Neurosci. 7, hormone 178–199 immunoreactivity is altered in the hypothalamus of the
123. Wistar-Kyoto strain of rat. Brain Res. 913, 224–233.
Shapiro, R.E., Miselis, R.R., 1985. The central neural connections of the area Swanson, L.W., 1977. Immunohistochemical evidence for a neurophysin-containing
postrema of the rat. J. Comp. Neurol. 234, 344–364. autonomic pathway arising in the paraventricular nucleus of the hypothalamus.
Shapiro, R.A., Xu, C., Dorsa, D.M., 2000. Differential transcriptional regulation of rat Brain Res. 128, 346–353.
vasopressin gene expression by estrogen receptor alpha and beta. Swanson, L.W., Kuypers, H.G., 1980. The paraventricular nucleus of the
Endocrinology 141, 4056–4064. hypothalamus: cytoarchitectonic subdivisions and organization of projections
Sharma, D., Handa, R.J., Uht, R.M., 2012. The ERbeta ligand 5alpha-androstane, to the pituitary, dorsal vagal complex, and spinal cord as demonstrated by
3beta,17beta-diol (3beta-diol) regulates hypothalamic oxytocin (Oxt) gene retrograde fluorescence double-labeling methods. J. Comp. Neurol. 194, 555–
expression. Endocrinology 153, 2353–2361. 570.
Shaw, G., Fenelon, J., Sichlau, M., Auchus, R.J., Wilson, J.D., Renfree, M.B., 2006. Role Swanson, L.W., Petrovich, G.D., 1998. What is the amygdala? Trends Neurosci. 21,
of the alternate pathway of dihydrotestosterone formation in virilization of the 323–331.
Wolffian ducts of the tammar wallaby, Macropus eugenii. Endocrinology 147, Swanson, L.W., Sawchenko, P.E., 1983. Hypothalamic integration: organization of
2368–2373. the paraventricular and supraoptic nuclei. Annu. Rev. Neurosci. 6, 269–324.
Sheng, Z., Kawano, J., Yanai, A., Fujinaga, R., Tanaka, M., Watanabe, Y., Shinoda, K., Swanson, L.W., Sawchenko, P.E., Lind, R.W., 1986. Regulation of multiple peptides in
2004. Expression of estrogen receptors (alpha, beta) and androgen receptor in CRF parvocellular neurosecretory neurons: implications for the stress response.
serotonin neurons of the rat and mouse dorsal raphe nuclei; sex and species Prog. Brain Res. 68, 169–190.
differences. Neurosci. Res. 49, 185–196. Szafarczyk, A., Ixart, G., Alonso, G., Malaval, F., Nouguier-Soule, J., Assenmacher, I.,
Shepard, J.D., Barron, K.W., Myers, D.A., 2003. Stereotaxic localization of 1983. CNS control of the circadian adrenocortical rhythm. J. Steroid Biochem.
corticosterone to the amygdala enhances hypothalamo–pituitary–adrenal 19, 1009–1015.
responses to behavioral stress. Brain Res. 963, 203–213. Szafarczyk, A., Malaval, F., Laurent, A., Gibaud, R., Assenmacher, I., 1987. Further
Shughrue, P.J., Komm, B., Merchenthaler, I., 1996a. The distribution of estrogen evidence for a central stimulatory action of catecholamines on
receptor-beta mRNA in the rat hypothalamus. Steroids 61, 678–681. adrenocorticotropin release in the rat. Endocrinology 121, 883–892.
Shughrue, P.J., Lane, M.V., Merchenthaler, I., 1996b. Glucagon-like peptide-1 Szego, C.M., Davis, J.S., 1967. Adenosine 30 ,50 -monophosphate in rat uterus: acute
receptor (GLP1-R) mRNA in the rat hypothalamus. Endocrinology 137, 5159– elevation by estrogen. Proc. Natl. Acad. Sci. USA 58, 1711–1718.
5162. Tabori, N.E., Stewart, L.S., Znamensky, V., Romeo, R.D., Alves, S.E., McEwen, B.S.,
Shughrue, P.J., Scrimo, P.J., Merchenthaler, I., 1998. Evidence for the colocalization of Milner, T.A., 2005. Ultrastructural evidence that androgen receptors are located
estrogen receptor-beta mRNA and estrogen receptor-alpha immunoreactivity in at extranuclear sites in the rat hippocampal formation. Neuroscience 130, 151–
neurons of the rat forebrain. Endocrinology 139, 5267–5270. 163.
Silverman, A.J., Hou-Yu, A., Chen, W.P., 1989. Corticotropin-releasing factor Tasker, J.G., Herman, J.P., 2011. Mechanisms of rapid glucocorticoid feedback
synapses within the paraventricular nucleus of the hypothalamus. inhibition of the hypothalamic–pituitary–adrenal axis. Stress 14, 398–406.
Neuroendocrinology 49, 291–299. Tauchi, M., Zhang, R., D’Alessio, D.A., Stern, J.E., Herman, J.P., 2008. Distribution of
Simerly, R.B., 1993. Distribution and regulation of steroid hormone receptor gene glucagon-like peptide-1 immunoreactivity in the hypothalamic paraventricular
expression in the central nervous system. Adv. Neurol. 59, 207–226. and supraoptic nuclei. J. Chem. Neuroanat. 36, 144–149.
Simerly, R.B., Young, B.J., 1991. Regulation of estrogen receptor messenger Terrazzino, S., Perego, C., De Simoni, M.G., 1995. Effect of development of
ribonucleic acid in rat hypothalamus by sex steroid hormones. Mol. habituation to restraint stress on hypothalamic noradrenaline release and
Endocrinol. 5, 424–432. adrenocorticotropin secretion. J. Neurochem. 65, 263–267.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx 23

Thomas, P., Pang, Y., Filardo, E.J., Dong, J., 2005. Identity of an estrogen membrane Walaas, I., Fonnum, F., 1980. Biochemical evidence for glutamate as a transmitter in
receptor coupled to a G protein in human breast cancer cells. Endocrinology hippocampal efferents to the basal forebrain and hypothalamus in the rat brain.
146, 624–632. Neuroscience 5, 1691–1698.
Tobet, S.A., Handa, R.J., Goldstein, J.M., 2013. Sex-dependent pathophysiology as Walf, A.A., Frye, C.A., 2005. ERbeta-selective estrogen receptor modulators produce
predictors of comorbidity of major depressive disorder and cardiovascular antianxiety behavior when administered systemically to ovariectomized rats.
disease. Pflugers Arch.. Neuropsychopharmacology 30, 1598–1609.
Torn, S., Nokelainen, P., Kurkela, R., Pulkka, A., Menjivar, M., Ghosh, S., Coca-Prados, Walf, A.A., Frye, C.A., 2006. Administration of estrogen receptor beta-specific
M., Peltoketo, H., Isomaa, V., Vihko, P., 2003. Production, purification, and selective estrogen receptor modulators to the hippocampus decrease anxiety
functional analysis of recombinant human and mouse 17beta-hydroxysteroid and depressive behavior of ovariectomized rats. Pharmacol. Biochem. Behav..
dehydrogenase type 7. Biochem. Biophys. Res. Commun. 305, 37–45. Walker, Q.D., Francis, R., Cabassa, J., Kuhn, C.M., 2001. Effect of ovarian hormones
Toufexis, D.J., Wilson, M.E., 2012. Dihydrotestosterone differentially modulates the and estrous cycle on stimulation of the hypothalamo–pituitary–adrenal axis by
cortisol response of the hypothalamic–pituitary–adrenal axis in male and cocaine. J. Pharmacol. Exp. Ther. 297, 291–298.
female rhesus macaques, and restores circadian secretion of cortisol in females. Walker, J.J., Spiga, F., Waite, E., Zhao, Z., Kershaw, Y., Terry, J.R., Lightman, S.L., 2012.
Brain Res. 1429, 43–51. The origin of glucocorticoid hormone oscillations. PLoS Biol. 10, e1001341.
Tsigos, C., Chrousos, G.P., 2002. Hypothalamic–pituitary–adrenal axis, Wang, M., He, Y., Eisenman, L.N., Fields, C., Zeng, C.M., Mathews, J., Benz, A., Fu, T.,
neuroendocrine factors and stress. J. Psychosom. Res. 53, 865–871. Zorumski, E., Steinbach, J.H., Covey, D.F., Zorumski, C.F., Mennerick, S., 2002.
Tyagi, R.K., Lavrovsky, Y., Ahn, S.C., Song, C.S., Chatterjee, B., Roy, A.K., 2000. Dynamics 3beta-hydroxypregnane steroids are pregnenolone sulfate-like GABA(A)
of intracellular movement and nucleocytoplasmic recycling of the ligand- receptor antagonists. J. Neurosci. 22, 3366–3375.
activated androgen receptor in living cells. Mol. Endocrinol. 14, 1162–1174. Wei, Q., Lu, X.Y., Liu, L., Schafer, G., Shieh, K.R., Burke, S., Robinson, T.E., Watson, S.J.,
Uht, R.M., McKelvy, J.F., Harrison, R.W., Bohn, M.C., 1988. Demonstration of Seasholtz, A.F., Akil, H., 2004. Glucocorticoid receptor overexpression in
glucocorticoid receptor-like immunoreactivity in glucocorticoid-sensitive forebrain: a mouse model of increased emotional lability. Proc. Natl. Acad.
vasopressin and corticotropin-releasing factor neurons in the hypothalamic Sci. USA 101, 11851–11856.
paraventricular nucleus. J. Neurosci. Res. 19, 405–411, 468–469. Weihua, Z., Lathe, R., Warner, M., Gustafsson, J.A., 2002. An endocrine pathway in
Ulrich-Lai, Y.M., Engeland, W.C., 2002. Adrenal splanchnic innervation modulates the prostate, ERbeta, AR, 5alpha-androstane-3beta,17beta-diol, and CYP7B1,
adrenal cortical responses to dehydration stress in rats. Neuroendocrinology 76, regulates prostate growth. Proc. Natl. Acad. Sci. USA 99, 13589–13594.
79–92. Weiser, M.J., Handa, R.J., 2009. Estrogen impairs glucocorticoid dependent negative
Vaccari, C., Lolait, S.J., Ostrowski, N.L., 1998. Comparative distribution of vasopressin feedback on the hypothalamic–pituitary–adrenal axis via estrogen receptor
V1b and oxytocin receptor messenger ribonucleic acids in brain. Endocrinology alpha within the hypothalamus. Neuroscience 159, 883–895.
139, 5015–5033. Weiser, M.J., Goel, N., Sandau, U.S., Bale, T.L., Handa, R.J., 2008. Androgen regulation
Vale, W., Spiess, J., Rivier, C., Rivier, J., 1981. Characterization of a 41-residue ovine of corticotropin-releasing hormone receptor 2 (CRHR2) mRNA expression and
hypothalamic peptide that stimulates secretion of corticotropin and beta- receptor binding in the rat brain. Exp. Neurol..
endorphin. Science 213, 1394–1397. Weiser, M.J., Wu, T.J., Handa, R.J., 2009. Estrogen receptor-beta agonist
Vamvakopoulos, N.C., Chrousos, G.P., 1993. Evidence of direct estrogenic regulation diarylpropionitrile: biological activities of R- and S-enantiomers on behavior
of human corticotropin-releasing hormone gene expression. Potential and hormonal response to stress. Endocrinology 150, 1817–1825.
implications for the sexual dimophism of the stress response and immune/ Weissman, M.M., Bland, R., Joyce, P.R., Newman, S., Wells, J.E., Wittchen, H.U., 1993.
inflammatory reaction. J. Clin. Invest. 92, 1896–1902. Sex differences in rates of depression: cross-national perspectives. J. Affect.
Van de Kar, L.D., Blair, M.L., 1999. Forebrain pathways mediating stress-induced Disord. 29, 77–84.
hormone secretion. Front. Neuroendocrinol. 20, 1–48. Westphal, U., 1971. Steroid–protein interactions. Monogr. Endocrinol. 4, 1–567.
van den Pol, A.N., 1982. The magnocellular and parvocellular paraventricular Westphal, U., 1983. Corticosteroid-binding globulin. A review of some recent
nucleus of rat: intrinsic organization. J. Comp. Neurol. 206, 317–345. aspects. Mol. Cell. Biochem. 55, 145–157.
van Eekelen, J.A., Rots, N.Y., Sutanto, W., Oitzl, M.S., de Kloet, E.R., 1991. Brain Whitnall, M.H., 1993. Regulation of the hypothalamic corticotropin-releasing
corticosteroid receptor gene expression and neuroendocrine dynamics during hormone neurosecretory system. Prog. Neurobiol. 40, 573–629.
aging. J. Steroid Biochem. Mol. Biol. 40, 679–683. Whitnall, M.H., Gainer, H., 1988. Major pro-vasopressin-expressing and pro-
Van Haarst, A.D., Oitzl, M.S., de Kloet, E.R., 1997. Facilitation of feedback inhibition vasopressin-deficient subpopulations of corticotropin-releasing hormone
through blockade of glucocorticoid receptors in the hippocampus. Neurochem. neurons in normal rats. Differential distributions within the paraventricular
Res. 22, 1323–1328. nucleus. Neuroendocrinology 47, 176–180.
Van Pett, K., Viau, V., Bittencourt, J.C., Chan, R.K., Li, H.Y., Arias, C., Prins, G.S., Perrin, M., Whitnall, M.H., Kiss, A., Aguilera, G., 1993. Contrasting effects of central alpha-1-
Vale, W., Sawchenko, P.E., 2000. Distribution of mRNAs encoding CRF receptors in adrenoreceptor activation on stress-responsive and stress-nonresponsive
brain and pituitary of rat and mouse. J. Comp. Neurol. 428, 191–212. subpopulations of corticotropin-releasing hormone neurosecretory cells in the
van Steensel, B., van Binnendijk, E.P., Hornsby, C.D., van der Voort, H.T., Krozowski, rat. Neuroendocrinology 58, 42–48.
Z.S., de Kloet, E.R., van Driel, R., 1996. Partial colocalization of glucocorticoid and Williamson, M., Viau, V., 2007. Androgen receptor expressing neurons that project
mineralocorticoid receptors in discrete compartments in nuclei of rat to the paraventricular nucleus of the hypothalamus in the male rat. J. Comp.
hippocampus neurons. J. Cell Sci. 109 (Pt. 4), 787–792. Neurol. 503, 717–740.
Vandesande, F., Dierickx, K., 1975. Identification of the vasopressin producing and of Williamson, M., Viau, V., 2008. Selective contributions of the medial preoptic
the oxytocin producing neurons in the hypothalamic magnocellular nucleus to testosterone-dependant regulation of the paraventricular nucleus of
neurosecretroy system of the rat. Cell Tissue Res. 164, 153–162. the hypothalamus and the HPA axis. Am. J. Physiol. Regul. Integr. Comp. Physiol.
Vasudevan, N., Pfaff, D.W., 2008. Non-genomic actions of estrogens and their 295, R1020–R1030.
interaction with genomic actions in the brain. Front. Neuroendocrinol. 29, 238– Windle, R.J., Shanks, N., Lightman, S.L., Ingram, C.D., 1997. Central oxytocin
257. administration reduces stress-induced corticosterone release and anxiety
Vasudevan, N., Kow, L.M., Pfaff, D.W., 2001. Early membrane estrogenic effects behavior in rats. Endocrinology 138, 2829–2834.
required for full expression of slower genomic actions in a nerve cell line. Proc. Windle, R.J., Kershaw, Y.M., Shanks, N., Wood, S.A., Lightman, S.L., Ingram, C.D.,
Natl. Acad. Sci. USA 98, 12267–12271. 2004. Oxytocin attenuates stress-induced c-fos mRNA expression in specific
Viau, V., 2002. Functional cross-talk between the hypothalamic–pituitary–gonadal forebrain regions associated with modulation of hypothalamo–pituitary–
and –adrenal axes. J. Neuroendocrinol. 14, 506–513. adrenal activity. J. Neurosci. 24, 2974–2982.
Viau, V., Meaney, M.J., 1991. Variations in the hypothalamic–pituitary–adrenal Windle, R.J., Gamble, L.E., Kershaw, Y.M., Wood, S.A., Lightman, S.L., Ingram, C.D.,
response to stress during the estrous cycle in the rat. Endocrinology 129, 2503– 2006. Gonadal steroid modulation of stress-induced hypothalamo–pituitary–
2511. adrenal activity and anxiety behavior: role of central oxytocin. Endocrinology
Viau, V., Meaney, M.J., 1996. The inhibitory effect of testosterone on hypothalamic– 147, 2423–2431.
pituitary–adrenal responses to stress is mediated by the medial preoptic area. J. Wu, T.W., Wang, J.M., Chen, S., Brinton, R.D., 2005. 17Beta-estradiol induced Ca2+
Neurosci. 16, 1866–1876. influx via L-type calcium channels activates the Src/ERK/cyclic-AMP response
Viau, V., Meaney, M.J., 2004. Testosterone-dependent variations in plasma and element binding protein signal pathway and BCL-2 expression in rat
intrapituitary corticosteroid binding globulin and stress hypothalamic– hippocampal neurons: a potential initiation mechanism for estrogen-induced
pituitary–adrenal activity in the male rat. J. Endocrinol. 181, 223–231. neuroprotection. Neuroscience 135, 59–72.
Viau, V., Soriano, L., Dallman, M.F., 2001. Androgens alter corticotropin releasing hormone Wu, W.F., Tan, X.J., Dai, Y.B., Krishnan, V., Warner, M., Gustafsson, J.A., 2013.
and arginine vasopressin mRNA within forebrain sites known to regulate activity in Targeting estrogen receptor beta in microglia and T cells to treat experimental
the hypothalamic–pituitary–adrenal axis. J. Neuroendocrinol. 13, 442–452. autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 110, 3543–3548.
Viau, V., Bingham, B., Davis, J., Lee, P., Wong, M., 2005. Gender and puberty interact Young, L.J., Wang, Z., Donaldson, R., Rissman, E.F., 1998. Estrogen receptor alpha is
on the stress-induced activation of parvocellular neurosecretory neurons and essential for induction of oxytocin receptor by estrogen. NeuroReport 9, 933–
corticotropin-releasing hormone messenger ribonucleic acid expression in the 936.
rat. Endocrinology 146, 137–146. Young, E.A., Altemus, M., Parkison, V., Shastry, S., 2001. Effects of estrogen
Vrang, N., Larsen, P.J., Clausen, J.T., Kristensen, P., 1999. Neurochemical antagonists and agonists on the ACTH response to restraint stress in female
characterization of hypothalamic cocaine–amphetamine-regulated transcript rats. Neuropsychopharmacology 25, 881–891.
neurons. J. Neurosci. 19, RC5. Zhang, Y., Damjanoska, K.J., Carrasco, G.A., Dudas, B., D’Souza, D.N., Tetzlaff, J.,
Waite, E.J., McKenna, M., Kershaw, Y., Walker, J.J., Cho, K., Piggins, H.D., Lightman, Garcia, F., Hanley, N.R., Scripathirathan, K., Petersen, B.R., Gray, T.S., Battaglia, G.,
S.L., 2012. Ultradian corticosterone secretion is maintained in the absence of Muma, N.A., Van de Kar, L.D., 2002. Evidence that 5-HT2A receptors in the
circadian cues. Eur. J. Neurosci. 36, 3142–3150.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001
24 R.J. Handa, M.J. Weiser / Frontiers in Neuroendocrinology xxx (2013) xxx–xxx

hypothalamic paraventricular nucleus mediate neuroendocrine responses to (- Ziegler, D.R., Herman, J.P., 2000. Local integration of glutamate signaling in the
)DOI. J. Neurosci. 22, 9635–9642. hypothalamic paraventricular region: regulation of glucocorticoid stress
Zhang, Z., Cerghet, M., Mullins, C., Williamson, M., Bessert, D., Skoff, R., 2004. responses. Endocrinology 141, 4801–4804.
Comparison of in vivo and in vitro subcellular localization of estrogen receptors Zuloaga, D.G., Morris, J.A., Jordan, C.L., Breedlove, S.M., 2008. Mice with the
alpha and beta in oligodendrocytes. J. Neurochem. 89, 674–684. testicular feminization mutation demonstrate a role for androgen receptors in
Zhang, R., Packard, B.A., Tauchi, M., D’Alessio, D.A., Herman, J.P., 2009. the regulation of anxiety-related behaviors and the hypothalamic–pituitary–
Glucocorticoid regulation of preproglucagon transcription and RNA stability adrenal axis. Horm. Behav..
during stress. Proc. Natl. Acad. Sci. USA 106, 5913–5918.

Please cite this article in press as: Handa, R.J., Weiser, M.J. Gonadal steroid hormones and the hypothalamo–pituitary–adrenal axis. Front. Neuroendocrinol.
(2013), http://dx.doi.org/10.1016/j.yfrne.2013.11.001

You might also like