You are on page 1of 13

REVIEW ARTICLE

published: 11 May 2012


CELLULAR NEUROSCIENCE doi: 10.3389/fncel.2012.00024

Synaptic regulation of the hypothalamicpituitaryadrenal


axis and its modulation by glucocorticoids and stress
Benjamin H. Levy1 and Jeffrey G. Tasker1,2*
1
Neuroscience Program, Tulane University, New Orleans, LA, USA
2
Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA

Edited by: Dysregulation of the hypothalamicpituitaryadrenal (HPA) axis has been implicated in
Harmen J. Krugers, Universiteit van a range of affective and stress-related disorders. The regulatory systems that control
Amsterdam, Netherlands
HPA activity are subject to modulation by environmental inuences, and stressful life
Reviewed by:
Yang Dan, University of California
events or circumstances can promote subsequent HPA dysregulation. The brain is a
Berkeley, USA major regulator of the HPA axis, and stress-induced plasticity of the neural circuitry
Corette Wierenga, Utrecht University, involved in HPA regulation might constitute an etiological link between stress and the
Netherlands development of HPA dysregulation. This review focuses on the synaptic regulation of
*Correspondence: neuroendocrine corticotropin-releasing hormone (CRH) neurons of the hypothalamic par-
Jeffrey G. Tasker, Department of Cell
and Molecular Biology, Tulane
aventricular nucleus, which are the cells through which the brain predominantly exerts its
University, 2000 Percival Stern Hall, inuence on the HPA axis. CRH neuronal activity is largely orchestrated by three neuro-
New Orleans, LA 70118-5221, USA. transmitters: GABA, glutamate, and norepinephrine. We discuss our current understanding
e-mail: tasker@tulane.edu of the neural circuitry through which these neurotransmitters regulate CRH cell activity, as
well as the plastic changes in this circuitry induced by acute and chronic stress and the
resultant changes in HPA function.
Keywords: corticosteroid, glutamate, GABA, norepinephrine, neural circuits, depression, synaptic plasticity,
paraventricular nucleus

INTRODUCTION Sarkhel et al., 2011), and dysregulation of the HPA axis may be an
Glucocorticoids are released in response to physical, emotional, etiological link between stress and the subsequent development
and/or metabolic stress, and many of the effects of glucocorti- of pathology.
coids are thought to serve as adaptive responses to stressful events The physiological origins of stress-induced variation in HPA
or circumstances. Physiological levels of glucocorticoids are reg- regulation have remained largely unknown, although rodent mod-
ulated by the hypothalamicpituitaryadrenal (HPA) axis, which els have shed some light on the matter, largely by providing insight
is responsible for a cascade of hormone signals that begins in the into the ways that the brain regulates HPA activity. In fact, the
brain and ends with glucocorticoid secretion from the adrenal cor- neural circuitry that regulates the HPA axis has been found to be
tex. An additional component of the HPA axis is a glucocorticoid- highly plastic, and that plasticity is induced by both stress and
mediated negative feedback mechanism, wherein glucocorticoids changes in glucocorticoid levels. It is possible that exposure to
act on the anterior pituitary and in the brain to suppress HPA stress and/or the resultant uctuations in circulating glucocorti-
activity. Given the association between glucocorticoids and stress, coids results in pathology by evoking long-lasting alterations in
it is not surprising that abnormal regulation of the HPA axis is the circuitry that regulates the HPA axis. Here, we will review
commonly associated with a range of affective and stress-related what is known about the synaptic regulation of the HPA axis and
disorders. Research into these phenomena has dealt mostly with the plasticity of the circuitry regulating the HPA axis induced
hypersecretion of glucocorticoid in patients with depressive ill- under stress conditions. We will focus on the neuroendocrine
ness (Gillespie and Nemeroff, 2005), although there has also been corticotropin-releasing hormone (CRH) cells, which are located
interest in the hyposecretion of glucocorticoids found in some in the paraventricular nucleus (PVN) of the hypothalamus and
cases of post-traumatic stress disorder (Yehuda, 2006) and depres- are situated at the apex of the HPA axis. The CRH cells are of
sion (Morphy et al., 1985; Penninx et al., 2007; Ahrens et al., 2008). particular interest because it is principally through the regulation
Dysregulation of the HPA axis has also been associated with panic of CRH release into the portal circulation that the brain exerts
disorder (Abelson et al., 2007), chronic fatigue syndrome (Van its inuence on the HPA axis. As with all neurons, CRH cells
Houdenhove et al., 2009), obsessive-compulsive disorder (Gustafs- are stimulated by depolarization of their cell membrane, which
son et al., 2008), bromyalgia (Tanriverdi et al., 2007), generalized stimulates action potentials and triggers the release of peptide
anxiety disorder (Lenze et al., 2011), and bipolar disorder (Daban from their axon terminals. However, unlike with classical neurons,
et al., 2005). Importantly, stressful life events have been impli- these terminals do not form synapses with postsynaptic neurons;
cated in the onset of each of these disorders (Kendler et al., 1995; but rather, they are incorporated into a region at the base of the
Hatcher and House, 2003; Gupta and Silman, 2004; Brawman- brain, the median eminence, where released neuropeptide accesses
Mintzer et al., 2005; Garno et al., 2005; Goodwin et al., 2005; the pituitary portal circulation via fenestrated portal arterioles.

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 1

fncel-06-00024 2012/5/9 20:17 page 1 #1


Levy and Tasker Synaptic regulation of the HPA axis

In addition to CRH, the CRH cells can synthesize and release electrophysiological studies (Figure 1). Magnocellular neuroen-
vasopressin (VP). CRH and VP in the portal circulation bind to docrine cells generate a robust A-type voltage-dependent K+
CRH and VP receptors on a subset of cells, the corticotropes, of the current, which causes a prominent transient outward rectication
anterior pituitary to stimulate the secretion of adrenocorticotropic that delays action potential generation (Tasker and Dudek, 1991).
hormone (ACTH) into the systemic circulation. The ACTH then Most parvocellular preautonomic neurons generate a T-type Ca2+
stimulates the synthesis and systemic secretion of glucocorticoids current, which causes a small low-threshold spike and clustering
from the adrenal cortex. Hypothalamic CRH neuronal activity is of Na+ -dependent action potentials (Stern, 2001; Luther et al.,
largely orchestrated by three neurotransmitters: GABA, glutamate, 2002). Parvocellular neuroendocrine cells can be distinguished
and norepinephrine. In this review we discuss our current under- from both magnocellular neuroendocrine cells and parvocellular
standing of how these neurotransmitter systems regulate CRH preautonomic cells by the absence of both the transient outward
neuron activity (and consequently HPA activity), and the mod- rectication and the low-threshold Ca2+ spike (Luther and Tasker,
ulation of these neurotransmitter systems by glucocorticoids and 2000; Luther et al., 2002; Figure 1).
stress. One aim of this review is to provide a framework to help While useful for narrowing down the eld of the three cell
guide future investigations of the synaptic regulation of CRH cells subtypes, these anatomical and electrophysiological approaches
that have the potential to advance our understanding of the neu- do not allow investigators to specically distinguish CRH neu-
rological bases for HPA dysfunction. It is also an aim of this paper rons from the other PVN parvocellular neuroendocrine cells,
to inform those who study stress-induced abnormalities in HPA such as thyrotropin releasing hormone and somatostatin cells.
activity, but who may not be familiar with studies of synaptic Electron and confocal microscopy immunohistochemical stud-
transmission, in the hope of making these studies more accessi- ies using selective antibodies for CRH and for neurotransmitters
ble for the purpose of enhancing communication and promoting or vesicular neurotransmitter transporters have been useful for
interdisciplinary investigations. characterizing the anatomical innervation of CRH neurons and
its plasticity in response to stress and adrenalectomy (Flak et al.,
METHODS OF IDENTIFYING CRH CELLS 2009). Because basal CRH peptide expression is not robust,
Inasmuch as CRH cells play such a central role here, before dis- attempts to identify CRH neurons during electrophysiological
cussing the synaptic regulation of the CRH neurons, we will briey recordings with a combination of intracellular dye injection and
address the challenges in identifying the CRH cells in the course post hoc immunostaining for CRH have been largely unsuccess-
of physiological experiments. The PVN can be divided into three ful. One method that has been useful for specically targeting
major cell types: the magnocellular neuroendocrine cells, which CRH cells for synaptic analysis is intracellular recording com-
project their axons into the posterior lobe of the pituitary, the bined with single-cell, reverse transcriptionPCR analysis (Di
parvocellular neuroendocrine cells, which send their axons to the et al., 2003). This technique allows the correlation of molecular
median eminence, and the parvocellular preautonomic neurons, expression proles, including CRH mRNA expression, with elec-
which project to the brain stem and spinal cord. The CRH neu- trophysiological properties. Transgenic mice that express green
rons fall into the parvocellular neuroendocrine cell type. The uorescent protein under the transcriptional control of the CRH
three cell types can be distinguished with reasonable reliability promoter have recently become available (Alon et al., 2009) and
on the basis of their respective somatic sizes and shapes, den- offer the greatest promise for contributing signicantly to the
dritic morphologies, and positions in the PVN. Magnocellular study of the synaptic regulation of identied CRH neurons
neuroendocrine cells tend to have larger, more rounded somata and the synaptic plasticity of CRH neurons under conditions of
and fewer dendritic branches, often bipolar in morphology, com- stress.
pared to the parvocellular neuroendocrine cells, which tend to
be smaller, fusiform, and with multipolar dendritic arbors. The GABA INPUTS
magnocellular neuroendocrine cells are concentrated in the lat- Origins of GABA inputs to the PVN have been identied through
eral portion of the PVN, while the parvocellular neuroendocrine the use of tract tracing in conjunction with immunohistochemical
cells are located mainly in the medial region of the PVN. The staining, as well as through in vitro electrophysiological analy-
dorsal-most and ventral-most regions of the PVN tend to be ses. A retrograde tracing-immunohistochemistry study revealed
occupied by the parvocellular preautonomic neurons, which are four discrete origins of signicant GABAergic innervation of the
intermediate in somatic size between the parvocellular neuroen- PVN, which are: the area surrounding the supraoptic nucleus, the
docrine cells and the magnocellular neuroendocrine cells, and have anterior perifornical region, the anterior hypothalamic area, and
multipolar dendritic arbors. While this morphological and topo- the anterior one-third of the PVN itself (Roland and Sawchenko,
graphical organization of the PVN can be used to identify enriched 1993). There is also evidence from tract tracing studies for a
populations of the different subtypes of PVN neurons, there is prominent GABAergic projection from the bed nucleus of the
fairly extensive overlap of the anatomical characteristics of the cell stria terminalis (BNST) to the PVN that relays inhibitory sig-
types within the PVN (Simmons and Swanson, 2009), such that naling to CRH cells from the prefrontal cortex (Radley et al.,
it does not provide for a strict distillation of the three neuron 2009) and the hippocampus (Radley and Sawchenko, 2011). Stud-
subpopulations. ies employing in vivo ibotenic acid lesions of subnuclei of the
Distinct electrical properties have also been characterized in the BNST have implicated specically the posterior medial region
three PVN cell types and have been used to assign electrical nger- of the bed nucleus as an inhibitory relay from limbic struc-
prints for the reliable identication of individual cells in in vitro tures to the PVN (Choi et al., 2007, 2008). An in vitro brain

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 2

fncel-06-00024 2012/5/9 20:17 page 2 #2


Levy and Tasker Synaptic regulation of the HPA axis

FIGURE 1 | Distinct electrophysiological fingerprints of PVN neuroendocrine cells fail to generate a transient outward rectication and
magnocellular and parvocellular neurons. (A) Magnocellular delay to spiking (arrow) in response to a similar current injection protocol.
neuroendocrine cells recorded in current-clamp mode generate an A-type K+ (C) Parvocellular preautonomic cells do not display a transient outward
current-mediated transient outward rectication (arrow) that is generated by rectication, but generate a low-threshold spike (arrow), mediated by a T-type
depolarization from a hyperpolarized holding membrane potential and delays Ca2+ current, and clustered action potentials in response to a similar current
the onset of spiking. Lower traces: current injection protocol. (B) Parvocellular injection protocol. Modied from Luther and Tasker (2000).

slice electrophysiological study identied robust local GABAer-


gic innervation of both parvocellular and magnocellular neurons
of the PVN that originates in the perinuclear area surrounding the
PVN (Boudaba et al., 1996). The perinuclear PVN area has also
been postulated to serve as an inhibitory relay into the PVN from
upstream limbic structures, such as the hippocampus and septum
(Ziegler and Herman, 2002).
The inhibitory effect of GABA on CRH cells is mediated pri-
marily by GABAA receptors. In situ hybridization assays detected
the expression of the GABAA receptor 2, 1, and 3 subunits in
nearly all the CRH cells of the PVN, and expression of 1 and
2 in about half of the CRH cells (Cullinan, 2000). Interestingly,
microinjection of a GABAA receptor antagonist into the PVN is
sufcient to activate CRH cells and elicit a surge in glucocorticoid
secretion (Cole and Sawchenko, 2002), revealing a tonic inhibitory
GABAergic input in the PVN that constrains CRH neuronal activ-
ity under basal conditions. The tonic inhibition of CRH neurons
is preserved in organotypic hypothalamic slice cultures contain-
ing the PVN (Bali and Kovacs, 2003), which suggests that local
GABA neuronal circuitry (i.e., GABA circuitry retained through
the slicing procedure) plays a central role in this mechanism. The
source of local GABAergic innervation of the PVN CRH neurons FIGURE 2 | Model of noradrenergic regulation of PVN CRH neuron
is predominantly extranuclear (Figure 2), including a robust input activity. Noradrenergic inputs (NE) originate in the brainstem and regulate
from the peri-PVN region (Boudaba et al., 1996). CRH neuronal activity by modulating glutamate and GABA release.
Norepinephrine both suppresses and enhances GABAergic inhibition of
CRH neuron activity, suppressing GABA release via 2 -adrenoceptor
STRESS PLASTICITY OF GABA INPUTS activation at presynaptic terminals and promoting GABA release by
activating 1 -adrenoceptors on upstream GABAergic somata/dendrites.
Acute stress plasticity of GABA inputs
Noradrenergic facilitation of glutamate release onto CRH neurons is
The GABAergic synaptic innervation of PVN neurons undergoes spike-dependent and is mediated by 1 -adrenoceptor activation.
signicant plastic changes in response to acute stress, which alters Preliminary evidence suggests that the facilitatory effect on glutamate
the excitability of the parvocellular neurons and activation of the release may be mediated by the release of a retrograde messenger that
stimulates upstream local glutamatergic circuits (dashed arrow). 3V, thrid
HPA axis. A 3060 min acute restraint stress is enough to cause a ventricle; OT, optic tract.
signicant shift in the Cl gradient across the PVN parvocellular
neuron membrane via downregulation of the membrane K+ Cl
co-transporter KCC2. This shift in the Cl gradient causes a post- expression, although KCC2 expression has been found to be sub-
synaptic attenuation of the inhibitory GABAergic inputs to these ject to steroidal modulation by sex hormones in substantia nigra
cells and leads to the disinhibition of the HPA axis (Hewitt et al., neurons wherein, at postnatal day 15, GABAA receptors are hyper-
2009). It is not known whether glucocorticoids modulate KCC2 polarizing in females and depolarizing in males (Galanopoulou

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 3

fncel-06-00024 2012/5/9 20:17 page 3 #3


Levy and Tasker Synaptic regulation of the HPA axis

and Moshe, 2003). Interestingly, a similar phenomenon has been provided a physiological corroboration with the nding that puta-
discovered in the magnocellular neurons of the PVN, in which tive parvocellular PVN neurons undergo a signicant increase
the GABAA reversal potential shifts positive and the GABA signals in inhibitory synaptic inputs in brain slices from adrenalec-
are transformed into excitatory signals in oxytocinergic magno- tomized rats, and paired-pulse analysis suggested that this effect of
cellular neurons following dehydration, a chronic physiological adrenalectomy was due to an increase in the number of GABAergic
stress (Kim et al., 2011); GABA was shown to be excitatory in synapses (Verkuyl and Joels, 2003). Glucocorticoid replacement
VP cells, but not in oxytocin cells, under normal conditions in this study conrmed that the effect of adrenalectomy was
(Haam et al., 2012). due to the loss of endogenous glucocorticoids. Finally, binding
Acute stress also appears to induce a long-term presynap- of radiolabeled muscimol, a selective GABAA receptor agonist,
tic plasticity in the GABAergic inhibitory innervation of PVN was found to increase in the hypothalamus following adrenalec-
parvocellular neurons via a transient elevation in circulating glu- tomy, suggesting an increase in postsynaptic expression or mem-
cocorticoids. Thus, a 30-min acute restraint stress was found to brane localization of GABAA receptors following adrenalectomy
cause a persistent reduction in inhibitory synaptic inputs to puta- (Majewska et al., 1985).
tive parvocellular neurons in recordings performed in brain slices Whereas the loss of endogenous glucocorticoids by adrenalec-
in vitro up to 5 h later, and this effect was mimicked by a pre- tomy was shown to increase the number of GABAergic synapses
vious in vivo subcutaneous injection of glucocorticoid (Verkuyl on CRH cells, chronic stress and chronically high glucocorticoid
et al., 2005). These changes in inhibitory inputs were not spike- levels were reported to suppress GABAergic inhibitory synaptic
dependent, suggestive of plasticity in the GABA release probability, inputs to PVN parvocellular neurons. In vitro electrophysiologi-
which was corroborated with paired-pulse analysis. An acute cal recordings from putative parvocellular neurons in PVN brain
20-min glucocorticoid application directly to brain slices had no slices taken after 3 weeks of a chronic variable stress treatment,
rapid effect (<20 min) on GABAergic synaptic currents (Di et al., which leads to sustained elevated glucocorticoids (Herman et al.,
2003), but resulted in a similar presynaptic suppression of GABA 1995), revealed a decrease in GABAergic inhibitory synaptic inputs
inputs to PVN parvocellular neurons 15 h later (Verkuyl et al., to parvocellular neurons without an accompanying reduction in
2005), suggesting that these presynaptic glucocorticoid actions are the probability of release of GABA, suggesting a reduced num-
transcriptional and occur directly in the hypothalamus. ber of GABAergic synapses (Joels et al., 2004; Verkuyl et al., 2004).
GABAergic synaptic plasticity may also play a role in the HPA A confocal immunouorescence study, however, failed to detect
response to hypoglycemic stress via signaling through neuropep- a reduction in the density of GABAergic synaptic boutons on
tide Y (NPY). There is a high density of NPY projections that CRH neurons in the medial parvocellular PVN, although these
terminate in the medial parvocellular PVN (Cowley et al., 1999), measurements were taken following a shorter exposure (1 week)
many of which originate in the arcuate nucleus, a hypothalamic to the chronic variable stress treatment (Flak et al., 2009). The
region that provides access to the brain of circulating nutritional acute glucocorticoid-induced reduction in GABA release proba-
signals (Wang et al., 2004). Both insulin-induced hypoglycemia bility and the chronic glucocorticoid-induced decrease in GABA
(Tuchelt et al., 2000) and intracerebroventricular injection of NPY synapses, in conjunction with the adrenalectomy-induced increase
(Dimitrov et al., 2007) activate the HPA axis and cause an increase in GABAergic synapses, suggest that there is an inverse causal
in the circulating glucocorticoid level. In vitro electrophysiological relationship between glucocorticoid levels and the efcacy of
studies reported that NPY caused a decrease in the probabil- GABAergic synaptic inhibition of CRH cell activity. Both acute
ity of GABA release onto putative parvocellular PVN neurons and chronic stress, therefore, appear to downregulate GABAergic
via actions at multiple presynaptic NPY receptors (Cowley et al., synaptic transmission in PVN parvocellular neurons, albeit in dif-
1999). The role of HPA axis activation in response to hypo- ferent ways. The progression from acute stress to chronic stress
glycemia may be related to the fact that glucocorticoids increase plasticity in the functional inhibitory synaptic regulation of par-
glucose bioavailability by shifting metabolic processes towards vocellular PVN neurons appears generally to involve a transition
catabolism. from a modulation of GABAergic synaptic strength to a structural
modication of afferent GABA inputs.
Chronic stress plasticity of GABA inputs While, to our knowledge, an analysis of the changes in the den-
Chronic stress and sustained changes in systemic glucocorticoid sity of GABAA receptors in the PVN following chronic variable
levels also lead to long-term shifts in the GABAergic synaptic stress is still lacking, molecular studies on chronic stress-induced
innervation of the PVN parvocellular neurons. Adrenalectomy, changes in GABAA receptor expression in the PVN have been
for example, which eliminates the main endogenous source of performed, and have produced somewhat contradictory ndings.
circulating glucocorticoids, leads to an increase in the number An in situ hybridization study reported a decrease in the expres-
of GABAergic synapses and in the density of GABA receptors sion of the GABAA receptor 1 and 2 subunits (Cullinan and
on CRH neurons, suggesting that the loss of circulating gluco- Wolfe, 2000). Another study using a single-cell RNA amplication
corticoids induces a proliferation of afferent GABAergic synaptic approach reported no change in the receptor subunit expres-
inputs to CRH neurons. Thus, quantitative electron microscopic sion, but an elevated expression of the 5 subunit and decreased
analysis of immunocytochemically labeled GABAergic synaptic expression of the subunit in putative parvocellular PVN neurons
proles revealed a 55% increase in the number of GABAer- following chronic variable stress (Verkuyl et al., 2004). However,
gic synapses on CRH cells in the PVN of adrenalectomized rats the absence of a change in -subunit expression in the latter study
(Miklos and Kovacs, 2002). An in vitro electrophysiological study may have been a consequence of the small sample size, as the

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 4

fncel-06-00024 2012/5/9 20:17 page 4 #4


Levy and Tasker Synaptic regulation of the HPA axis

investigators themselves concede. While the regulation of the 5 (mGluRs) in the PVN. One in situ hybridization study that used
and GABAA receptor subunits may suggest a stress-induced riboprobes reported that mRNAs for the NMDA receptor sub-
change in extrasynaptic GABAA receptor signaling, the decrease units GluN1, GluN2A, and GluN2B are abundantly expressed
in 1 and 2 subunits expression should lead to the downregu- in the medial parvocellular PVN, whereas there is signicantly
lation of functional synaptic GABAA receptors and/or a change less expression of the GluN2C and GluN2D subunits (Herman
in synaptic GABAA receptor efcacy. Corresponding changes in et al., 2000). This study also reported substantial expression in the
postsynaptic GABAA receptor sensitivity have not been reported medial parvocellular PVN of the kainate receptor subunits GluK2
to date. and GluK5 and of the AMPA receptor GluA1 subunit. Another
in situ hybridization study that used oligonucleotide probes to test
GLUTAMATE INPUTS for the colocalization of ionotropic glutamate receptor subunits
Double immunohistochemical labeling for the vesicular glutamate with CRH found mRNAs for the AMPA receptor subunits GluA1
transporter VGlut2 and for CRH has shown that PVN CRH neu- and GluA2 to be present in 46 and 21% of CRH cells, respectively,
rons are densely innervated by glutamatergic bers (Wittmann and mRNA for the kainate receptor subunit GluK2 to be present in
et al., 2005). Electrophysiological experiments have determined 31% of CRH cells (Aubry et al., 1996). This study also found that
that functional local circuit glutamatergic innervation of puta- 70% of CRH cells expressed the NMDA receptor subunit GluN1
tive parvocellular neurons originates in areas adjacent to the PVN, mRNA, but no colocalization with either GluN2A or GluN2B sub-
largely overlapping sources of peri-PVN GABAergic input, but at units was detected. The difference in GluN2 expression between
a much lower density (Boudaba et al., 1997; Tasker et al., 1998). these two studies may be due to the higher sensitivity of riboprobes
There is also evidence that glutamatergic input to PVN neurons relative to oligonucleotide probes. Thus, it appears that kainate,
originates within the PVN itself (Figure 2). First, CRH-, oxytocin-, AMPA, and NMDA receptors all play a role in mediating gluta-
and VP-expressing neurons in the PVN have all been shown matergic input to CRH cells, with the AMPA receptor subunit
to co-express VGlut2 mRNA (Hrabovszky and Liposits, 2008). GluA1 predominating over the GluA2 subunit, and the NMDA
In vitro brain slice electrophysiological recordings from putative receptor subunits GluN2A and GluN2B predominating over the
magnocellular neurons of the PVN revealed a norepinephrine- GluN2C and GluN2D subunits.
evoked stimulation of local glutamate circuits within the PVN In addition to ionotropic glutamate receptors, mGluRs play
that was spike-dependent, suggesting the presence of glutamater- a role in the regulation of CRH cell activity (Durand et al.,
gic neurons in the PVN that mediate intra-PVN signaling (Daftary 2008). Double-label immunocytochemistry revealed the pres-
et al., 1998). Furthermore, intra-PVN glutamatergic circuits were ence of mGluR1a in a signicant number of CRH cells (Kocsis
also revealed in an autoradiographic study in which sources of et al., 1998). Intracerebroventricular injections of selective group I
glutamatergic input to the PVN were identied using the radi- and group III mGluR agonists were found to elicit an increase
olabeled retrograde marker [3 H]D-aspartate, which selectively in circulating glucocorticoids (Lang and Ajmal, 1995; Johnson
labels glutamate neurons (Csaki et al., 2000). In addition to the et al., 2001). Unexpectedly, intracerebroventricular injections of
peri-PVN and intra-PVN sources of glutamatergic inputs to the antagonists of the group I mGluRs were also found to trigger
PVN, signicant glutamatergic afferents to the PVN have also the activation of the HPA axis (Bradbury et al., 2003). Based
been identied emanating from the ventromedial hypothalamic on previous ndings of a pre- and postsynaptic modulation of
nucleus, the posterior hypothalamic nucleus, the dorsomedial magnocellular neuroendocrine cells by group I and III mGluRs
hypothalamic nucleus, the anterior hypothalamic nucleus, the lat- (Schrader and Tasker, 1997; Tasker et al., 1998), a similar model
eral hypothalamic nucleus, the paraventricular thalamic nucleus, for the regulation of CRH cell activity by group I and group III
and the medial nucleus of the amygdala (Csaki et al., 2000; mGluRs was proposed (Johnson et al., 2001). According to this
Ulrich-Lai et al., 2011). model, group III mGluRs disinhibit CRH cell activity by suppress-
Microinjection of glutamate into the PVN caused a depletion of ing the release of GABA onto CRH cells, and group I mGluRs
CRH in the median eminence and activation of the HPA axis (Feld- inhibit CRH cells through stimulation of afferent GABAergic neu-
man and Weidenfeld, 1997), and microinjection of an ionotropic rons, while postsynaptic group I mGluRs stimulate CRH cells
glutamate receptor antagonist into the PVN attenuated the HPA directly.
hormonal response to acute restraint stress (Ziegler and Herman,
2000). The inconsistent activation of CRH neurons and the HPA STRESS PLASTICITY OF GLUTAMATE INPUTS
axis by direct PVN glutamate microinjection reported in another Acute stress plasticity of glutamate inputs
study (Cole and Sawchenko, 2002) is likely due to the desensiti- Glutamatergic synaptic inputs to CRH cells are suppressed by
zation of the ionotropic glutamate receptors on the PVN neurons rapid glucocorticoid actions that appear to be involved in the
and/or stimulation of opposing inhibitory inputs via the activation glucocorticoid-mediated fast negative feedback of the HPA axis.
of local circuit neurons upstream from the PVN neurons. Double- Thus, corticosterone and dexamethasone cause a rapid (within
label immunohistochemical assays, for example, have shown that 35 min) decrease in the frequency of miniature excitatory post-
glutamate microinjection into the PVN increases Fos induction synaptic currents (mEPSCs) in each of the major neuroendocrine
in peri-PVN GABAergic neurons, which may project to the CRH cell types of the PVN, including in CRH cells, recorded in hypotha-
cells and oppose the direct excitatory actions of glutamate. lamic slices (Di et al., 2003). This rapid glucocorticoid effect is
Molecular studies have reported the expression of multiple mediated by a non-genomic mechanism via the activation of a
subtypes of ionotropic and metabotropic glutamate receptors membrane-associated receptor. Importantly, this glucocorticoid

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 5

fncel-06-00024 2012/5/9 20:17 page 5 #5


Levy and Tasker Synaptic regulation of the HPA axis

effect was blocked by antagonists and mimicked and occluded by N -methyl-D-aspartate receptor expression in the medial par-
agonists of the cannabinoid type 1 receptor (CB1 R), suggesting the vocellular PVN has also been reported to be modulated by a
involvement of the endocannabinoid system. We recently corrob- single exposure to a stress stimulus. An in situ hybridization study
orated this with the nding that the rapid glucocorticoid effect is found that acute immobilization stress caused a 35% increase
absent in CB1 R knockout mice (Nahar et al., submitted). This was in the expression of mRNA for the GluN1 subunit of the NMDA
supported by the nding that glucocorticoid rapidly (<10 min) receptor in several brain regions, including in the medial par-
triggers the synthesis of the major endocannabinoids anandamide vocellular PVN (Bartanusz et al., 1995). The increased GluN1
(AEA) and 2-arachidonoylglycerol (2-AG) in slices of the PVN expression suggests the possibility that acute stress may cause an
and immediate surround (Malcher-Lopes et al., 2006). Endo- overall increase in NMDA receptor function in the medial par-
cannabinoids signal in a retrograde fashion (i.e., from postsynaptic vocellular PVN, since functional NMDA receptors are comprised
membrane to presynaptic membrane) and cause the inhibition of of a requisite GluN1 subunit (Cull-Candy et al., 2001). However,
presynaptic neurotransmitter release (Freund et al., 2003). Consis- a brain slice electrophysiological study demonstrated depressed
tent with the endocannabinoid acting as a retrograde messenger NMDA receptor function in individual PVN parvocellular neu-
to inhibit glutamate release at glutamate synapses, the glucocor- rons following a 30-min acute immobilization stress (Kuzmiski
ticoid effect was prevented by blockade of G protein and protein et al., 2010). Paradoxically, the depression of NMDA receptor cur-
kinase activity and Ca2+ signaling specically in the postsynaptic rents in the parvocellular neurons following acute stress placed
cell (Malcher-Lopes et al., 2006; Harris and Tasker, 2011). the glutamate synapses in a permissive state that allowed them to
Interestingly, the glucocorticoid-induced rapid suppression of undergo activity-dependent short-term potentiation in response
glutamate release in the PVN in vitro is not reversible in brain slice to high frequency stimulation. Thus, the NMDA receptor mRNA
electrophysiological studies for over an hour after dexamethasone expression and the functionality of the receptor do not appear to
administration (Di et al., 2003), which indicates that glucocor- be regulated in parallel by stress, which suggests that there may be
ticoids trigger a form of endocannabinoid-mediated long-term changes in the receptor protein expression or trafcking that may
depression of synaptic excitation; long-term depression is recog- account for this discrepancy.
nized as a widespread form of synaptic plasticity in the brain that
reduces the efcacy of synaptic interactions (Heifets and Castillo, Chronic stress plasticity of glutamate inputs
2009). The physiological signicance of the extended duration In addition to inducing an apparent functional downregulation
of the glucocorticoid-induced suppression of excitatory inputs to in the inhibitory synaptic inputs to PVN parvocellular neuroen-
CRH cells remains unknown; however, we have preliminary evi- docrine cells (Verkuyl et al., 2004), the chronic variable stress
dence for the desensitization to the rapid glucocorticoid-induced model of stress plasticity also gives rise to signicant changes in the
suppression of glutamate release in brain slices from animals that excitatory synaptic innervation of PVN parvocellular neurons. A
had been subjected to an acute 30-min restraint stress prior to sac- double-labeling immunohistochemical study found that chronic
rice (Jiang and Tasker, unpublished observation). This effect is variable stress signicantly increased the density of synaptophysin-
likely due to the long-term depression of glutamatergic synap- expressing synaptic boutons and VGlut2-expressing glutamater-
tic inputs by a tonic activation of presynaptic CB1 receptors gic bers in the medial parvocellular PVN, and increased the
via glucocorticoid-induced retrograde endocannabinoid release, number of glutamatergic boutons directly contacting CRH-
since a CB1 receptor-mediated inhibitory tone was observed in immunoreactive somata and dendrites (Flak et al., 2009). This
glutamate inputs to parvocellular neurons from acutely stressed suggests that chronic stress causes an increase in the glutamatergic
rats, but not from unstressed rats. A downregulation of rapid synaptic innervation of the CRH neurons, which, along with the
glucocorticoid-induced endocannabinoid suppression of synaptic reduced GABAergic innervation of medial parvocellular neurons
excitation was also reported in putative parvocellular neuroen- (Verkuyl et al., 2004), should lead to an increase in the excitability
docrine cells of juvenile rats subjected to a repeated immobi- of these neurons following chronic stress exposure. Preliminary
lization stress, although the mechanism proposed was a chronic electrophysiological ndings from our laboratory suggest that the
stress-induced desensitization of the cannabinoid signaling system increased density of immunolabeled glutamate synapses on the
(Wamsteeker et al., 2010), rather than the increased endocannabi- CRH neurons gives rise to an increase in the functional excitatory
noid inhibitory tone that we nd after single exposure to an acute synaptic inputs to these cells (Franco et al., 2007).
stress or stress level of glucocorticoid. It remains to be deter- Glutamate receptor expression in the PVN has also been
mined how long the glucocorticoid-induced long-term depression reported to be modied by chronic stress exposure. An in situ
of synaptic excitation lasts, but it may be in place to provide the hybridization study found that 2 weeks of chronic variable stress
time necessary for the peptidergic stores in the somata and axon caused a decrease in the mRNA expression of the NMDA recep-
terminals to replenish after a rapid and robust secretion of CRH tor subunit GluN2B in the medial parvocellular PVN, but found
and VP into the portal circulation. Also, for those mechanisms no changes in GluN1 or GluN2A subunit expression (Ziegler
that are sensitive to rates-of-change in glucocorticoid concen- et al., 2005). This effect differed from the glutamate receptor plas-
tration (Jacobson and Sapolsky, 1993), long-term depression of ticity induced by acute immobilization stress, which caused an
the synaptic excitation of CRH neurons may reduce CRH release increase in the expression of GluN1 reported in the aforemen-
to allow for clearance of circulating glucocorticoids in order to tioned study (Bartanusz et al., 1995). The fact that the decrease in
ensure the sensitivity of glucocorticoid targets to the secretion of GluN2B induction caused by chronic stress was not accompanied
a subsequent bolus. by a change in GluN1 expression suggests that the downregulated

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 6

fncel-06-00024 2012/5/9 20:17 page 6 #6


Levy and Tasker Synaptic regulation of the HPA axis

GluN2B does not reect a decrease in the number of functional psychological stress are driven by regions in the forebrain (Li et al.,
NMDA receptors, since the GluN1 subunit is an essential com- 1996). In support of this division, the glucocorticoid response to
ponent of functional NMDA receptors (Cull-Candy et al., 2001). immune stress caused by intraperitoneal injection of interleukin-
The decrease in NMDA receptor mRNA expression with chronic 1 was found to be attenuated by depletion of noradrenergic
stress is surprising given the observation of an increase in the den- inputs to the PVN, whereas no such effect of the depleted inputs
sity of glutamate synapses with the same chronic stress paradigm was observed on the HPA responses to restraint stress (Chuluyan
(Flak et al., 2009). This suggests either that NMDA receptor et al., 1992) or footshock stress (Li et al., 1996). However, later nd-
mRNA and protein expression are differentially modulated, or ings would suggest that ascending noradrenergic signaling plays a
that there is less of a contribution of NMDA receptors relative to role in driving HPA responses to both systemic and psycholog-
AMPA receptors at glutamate synapses on CRH neurons follow- ical stress (Pacak et al., 1998; Dayas et al., 2001). More recently,
ing chronic variable stress. Follow-up electrophysiological studies Herman et al. (2003) presented an alternative model regarding the
will be necessary to distinguish between these possibilities. Inter- role of the brainstem in HPA responses to stress. According to
estingly, the stress-induced downregulation of GluN2B mRNA is this model, the extent to which the NTS promotes HPA activity
apparently not mediated by glucocorticoids, as neither adrenalec- results from an interaction between ascending sensory inputs to
tomy nor high-dose glucocorticoid administration produced any the NTS and descending inputs to the NTS from the forebrain.
change in GluN2B mRNA expression in the medial parvocellular It was proposed that the ascending input to the NTS transmits
PVN (Ziegler et al., 2005). Thus, this plasticity of NMDA recep- information regarding the current homeostatic state, whereas
tor expression following chronic variable stress may result from descending input to the NTS transmits information regarding an
repeated activation of glutamatergic receptors with the chronic anticipated homeostatic state. As a result of this convergence of
activation of stress circuits. signaling in the NTS, the HPA response to an anticipated home-
Interestingly, glutamatergic synaptic plasticity resulting from ostatic state would depend on the current homeostatic state, so
the neonatal environment might be involved in lifelong changes that inhibitory signaling from the forebrain can be overridden by
in HPA responsivity. Pups that are handled daily during the rst current somatic, visceral, or humoral disturbances; alternatively,
10 days of life have been found to exhibit a suppressed HPA excitatory signaling from the periphery to the medial parvocellular
response to a novel stressor as adults (Liu et al., 1997). It is pos- PVN could be modulated by descending contextual information
sible that this effect of handling is mediated by an increase in (Herman et al., 2003).
maternal attention, as handled pups receive more frequent bouts Electrophysiological analyses have revealed mixed effects of
of maternal care in the form of licking, grooming, and arched- norepinephrine on putative parvocellular neuroendocrine cells
back nursing. One study using an early-life handling paradigm of the PVN; some cells are excited and other cells are inhibited
found that handled pups have lower VGlut2 protein content in the by norepinephrine (Yang et al., 2007). The excitatory actions of
PVN, fewer VGlut2-immunoreactive synaptic boutons contacting norepinephrine on CRH cells appear largely to be mediated by
CRH cells, and fewer asymmetrical, putative excitatory, synapses modulation of glutamatergic synaptic inputs. Thus, the surge
on CRH cells (Korosi et al., 2010). Electrophysiological record- in circulating ACTH and glucocorticoids resulting from electri-
ings in putative parvocellular PVN neurons from these animals cal stimulation of the ventral noradrenergic bundle was found
found that the frequency of miniature EPSCs also was decreased, to be inhibited by intra-PVN microinjection of ionotropic gluta-
suggesting a reduced excitatory synaptic input to the CRH cells. mate receptor antagonists in a dose-dependent manner (Feldman
The reduced glutamatergic input in this study was found exclu- and Weidenfeld, 2004). An electrophysiological study found that
sively in the pups and not following maturation to adulthood, about 36% of putative parvocellular neurons responded to nore-
but the handled animals exhibited life-long reductions in PVN pinephrine with an increase in glutamatergic input, while another
CRH expression, which suggests that reductions in glutamater- 14% responded with a hyperpolarization (Daftary et al., 2000).
gic input early in life may be involved in a cascade of events that Post-recording biocytin labeling revealed that the cells that had
leads to a life-long reduction in CRH expression and HPA stress exhibited a norepinephrine-induced hyperpolarization tended to
responsivity. be located close to the third ventricle in a region known to con-
tain dopamine and/or somatostatin parvocellular neurons, so it
NOREPINEPHRINE INPUTS is possible that none of the hyperpolarized neurons were CRH
Noradrenergic input to the medial parvocellular PVN originates cells. The norepinephrine-induced increase in glutamatergic activ-
in the brainstem, particularly in the nucleus of the solitary tract ity was mediated by 1 -adrenoceptors and was spike-dependent.
(NTS; Herman et al., 2003). Norepinephrine has long been known Perhaps the most surprising result from our electrophysiologi-
to play a prominent role in the regulation of the HPA axis and cal studies is the absence of putative parvocellular neurons that
has been shown to be generally excitatory with regard to modu- are excited by norepinephrine directly (Daftary et al., 2000). Elec-
lation of CRH neuron activity (Plotsky et al., 1987). Accordingly, tron microscopy has revealed the presence of adrenergic boutons
microinjection of norepinephrine directly into the PVN increases forming synaptic specializations with CRH neurons (Liposits et al.,
CRH mRNA expression and elevates circulating ACTH in a dose- 1986) and dual in situ hybridization has demonstrated that the
dependent manner (Itoi et al., 1994). There has been debate, 1b -adrenoceptor is expressed by nearly all the CRH neurons of
however, regarding the role of the brainstem in driving HPA the PVN (Day et al., 1999). However, we have found no evidence,
responses to stress. It has been proposed that the brainstem and there have been no electrophysiological reports, of nore-
drives HPA responses to systemic stress, whereas HPA responses to pinephrine directly activating an inward current or depolarization

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 7

fncel-06-00024 2012/5/9 20:17 page 7 #7


Levy and Tasker Synaptic regulation of the HPA axis

in parvocellular neuroendocrine cells. As noradrenergic recep- adrenalectomy (Day et al., 1999). Microdialysis analyses revealed
tors are metabotropic, noradrenergic receptors on CRH cells may that adrenalectomy increases the synthesis, release, and turnover
initiate signal transduction pathways without altering membrane of norepinephrine in the PVN (Pacak et al., 1993), whereas 1 week
conductance. On the other hand, postsynaptic noradrenergic of hypercortisolemia, induced by administration of exogenous
receptors may stimulate the synthesis of a retrograde messenger glucocorticoid, reduced the synthesis, release, and turnover of
that could trigger the activation of upstream local glutamatergic norepinephrine in the PVN (Pacak et al., 1995). This last nding
circuits. We have preliminary evidence for just such a retrograde is surprising inasmuch as 1 week of chronic variable stress has
mechanism, in which a messenger is released from CRH neurons in been found to increase both the density of noradrenergic synaptic
the PVN and stimulates a spike-dependent increase in glutamater- boutons in the PVN and the number of noradrenergic boutons
gic inputs to the CRH neurons (Jiang and Tasker, unpublished abutting CRH-immunoreactive somata and dendrites (Flak et al.,
observation; Figure 2). 2009). Additionally, 1 week of repeated intermittent cold stress was
In addition to regulating glutamate release, norepinephrine reported to enhance HPA responsivity to an acute immobilization
modulates GABA release onto parvocellular neurons in the PVN stress via an increase in 1 -adrenoceptor signaling within the PVN
(Figure 2). An electrophysiological study found that about 58% of (Ma and Morilak, 2005).
putative parvocellular neurons responded to norepinephrine with In addition to the effects of adrenalectomy on norepinephrine
an increase in the frequency of spontaneous inhibitory postsynap- release and adrenoceptor expression in the PVN, electrophysiolog-
tic currents (IPSCs) that was mediated by 1 -adrenoceptors and ical recordings revealed that adrenalectomy results in an increased
was spike-dependent, suggestive of a locus of action upstream fraction of putative parvocellular neuroendocrine cells that are
of the GABAergic axon (Han et al., 2002). In another 33% of excited by norepinephrine and a decreased fraction inhibited by
the neurons tested, norepinephrine reduced the IPSC frequency, norepinephrine (Yang et al., 2007). Additionally, it was found that
and this reduction was mediated by 2 -adrenoceptors and was the fraction of cells in which norepinephrine reduced GABA input
spike-independent, suggesting a presynaptic terminal locus of was doubled following adrenalectomy, whereas there was an 80%
action (Han et al., 2002). In another study that specically tar- decrease in the fraction of cells in which norepinephrine enhanced
geted parvocellular neuroendocrine cells with electrophysiological GABA input (Yang et al., 2008).
markers and retrograde staining from the median eminence, nore- Although norepinephrine is thought to be generally excitatory
pinephrine suppressed GABAergic input in 46% of the cells tested, with regard to CRH release, there is a body of evidence that sug-
and this was increased to 100% following blockade of either 1 - gests that norepinephrine suppresses CRH neuron activity in the
adrenoceptors or spike generation (Yang et al., 2008). Consistent absence of glucocorticoids; thus noradrenergic regulation of CRH
with 2 -adrenoceptors located on GABAergic boutons inhibiting cells may be more complex than previously thought. While nore-
GABA release onto CRH cells, binding assays have reported sub- pinephrine normally enhances CRH secretion from organotypic
stantial binding of radiolabeled clonidine, an 2 -adrenoceptor hypothalamic cultures, it was found to reduce basal secretion of
agonist, in the CRH cell region of the medial parvocellular CRH from cultures maintained in a glucocorticoid-free medium
PVN (Cummings and Seybold, 1988). Moreover, electrophys- (Szafarczyk et al., 1995). Also, while the 2 -adrenoceptor agonist
iological analyses indicate that the 2 -adrenoceptor-mediated clonidine normally promotes CRH cell activity (Calogero et al.,
decrease in GABA input is reective of a reduced probability 1988), it was found to reduce basal CRH release from organ-
of GABA release (Yang et al., 2008). In keeping with its sup- otypic hypothalamic cultures maintained in a glucocorticoid-free
pressive effect on neurotransmitter release, the 2 -adrenoceptor medium (Feuvrier et al., 1998). If noradrenergic signaling from
is commonly found to suppress norepinephrine release as an the brainstem does in fact play a more prominent role in driv-
autoreceptor (Raiteri et al., 1992). In support of a stimula- ing HPA responses to systemic stress than to psychological stress,
tory effect of the 2 -adrenoceptor on the HPA axis, clonidine adrenalectomized rats might be expected to respond differently to
has been reported to enhance CRH secretion from organ- certain stressors than do intact rats.
otypic hypothalamic cultures (Calogero et al., 1988), and intrac-
erebroventricular (Szafarczyk et al., 1990) and intraperitoneal PERSPECTIVES AND OUTLOOK
(Shimizu, 1984) injections of clonidine in vivo induce a surge in Plasticity of the neural circuitry regulating the HPA axis may
ACTH release. constitute an etiological link between stress exposure and the
subsequent development of HPA dysregulation and associated
PLASTICITY OF NOREPINEPHRINE INPUTS pathologies. Research into dysregulation of the HPA axis has
Soon after the discovery that norepinephrine regulates HPA activ- focused mainly on hypersecretion of glucocorticoids, which is
ity, it was found that ascending noradrenergic signaling plays a a condition that has been associated with depressive illness.
role in the glucocorticoid-induced suppression of the HPA axis, Autopsies of depressed patients have reported higher than nor-
since depletion of noradrenergic inputs via knife cuts to the mal levels of CRH and VP mRNA in the PVN (Raadsheer et al.,
medulla attenuated the increases in hypothalamic CRH expres- 1994, 1995), which is suggestive of elevated pre-mortem CRH
sion and CRH release caused by adrenalectomy (Sawchenko, cell activity. It is believed that diminished glucocorticoid-induced
1988). Adrenalectomy was also found to reduce binding of radi- suppression of the HPA axis is an underlying factor in the HPA
olabeled clonidine in the CRH-expressing region of the medial hyperactivity found in depressive illness (Gillespie and Nemeroff,
parvocellular PVN (Cummings and Seybold, 1988), while 1B - 2005). Consistent with this, depressed patients exhibit an atten-
adrenoceptor expression in the PVN was upregulated following uated suppression of glucocorticoid secretion when given the

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 8

fncel-06-00024 2012/5/9 20:17 page 8 #8


Levy and Tasker Synaptic regulation of the HPA axis

dexamethasone suppression test (i.e., the suppression of HPA in the onset of each of these disorders (Hatcher and House,
activity by dexamethasone administration; Juruena et al., 2006). 2003; Gupta and Silman, 2004). There is also evidence that HPA
It has been suggested that this glucocorticoid resistance could hypoactivity is involved in some cases of post-traumatic stress dis-
result from a reduced expression of the glucocorticoid receptor order (PTSD; Yehuda, 2006) and depression (Morphy et al., 1985;
(GR; Modell et al., 1997; Boyle et al., 2005), an idea supported by Penninx et al., 2007; Ahrens et al., 2008). Sexually abused girls
reports of reduced GR expression in post-mortem brain tissue of (ages 57 years) have exhibited reduced salivary glucocorticoid
depressed patients (Webster et al., 2002). A similar downregula- levels within months following an abusive incident (King et al.,
tion of GR is seen in the rat brain following chronic stress (Kitraki 2001). Moreover, adult women who were victims of childhood
et al., 1999). It is possible that a stress-induced downregulation of sexual abuse have exhibited enhanced suppression of glucocor-
GR serves as a protective mechanism to limit allostatic load during ticoid release in response to the dexamethasone suppression test
periods of elevated glucocorticoid levels. Indeed, there is evidence (Stein et al., 1997). Lastly, in rats, stress can result in HPA hypoac-
that GR mediates the hippocampal deterioration that results from tivity under certain circumstances. One study using female rats
prolonged glucocorticoid exposure (Packan and Sapolsky, 1990). found that exposure to intense footshock, followed by 3 weekly
Stress could then result in hypercortisolemia by downregulating reminders of the stimulus, resulted in lower glucocorticoid lev-
GR expression, thereby diminishing glucocorticoid-induced feed- els than those recorded previous to the footshock (Louvart et al.,
back inhibition of the HPA axis. However, we propose that a 2005). Reminders of the footshock involved placing the rat in
modied central drive underlies, at least in part, the link between a compartment adjacent to the compartment where the foot-
exposure to stress and the subsequent development of HPA abnor- shock had previously occurred, in what was intended as a rodent
malities. To be clear, we refer to the central drive as it pertains to model of PTSD. Another study found that rats subjected to chronic
the neural circuitry, namely the glutamatergic, GABAergic and variable stress, followed by several days recovery from the stress,
noradrenergic circuitry that regulates the CRH neuronal activity. exhibit attenuated pituitaryadrenal responses to acute novel envi-
There is evidence that enhanced central drive of the HPA axis ronment and restraint stresses (Ostrander et al., 2006). It should
contributes to the hypercortisolemia associated with depressive be noted that there are some mechanisms through which glu-
illness. Specically, dexamethasone-resistant depressed patients cocorticoids could actually enhance HPA activity. For example,
exhibit an enhanced ACTH response to metyrapone, a glucocor- glucocorticoids seem to attenuate GABAergic restraint on CRH
ticoid synthesis inhibitor (Fava et al., 1984; Ur et al., 1992). This cells, as described above; thus, in certain cell types, downregulated
observation is consistent with a hyperactive drive of the HPA axis GR expression in response to stress could promote subsequent
that is, in fact, suppressed by glucocorticoids. This glucocorticoid- HPA hypoactivity. However, there is evidence that stress can, at
independent overdrive of the HPA axis may reect neural circuitry times, result in upregulated GR. GR number was found to be larger
that favors CRH cell hyperactivity. Interestingly, an enhanced in lymphocytes of combat veterans than in those of non-veterans
ACTH response to metyrapone in depressed patients has been (Yehuda et al., 1995). Also, increased GR binding capacity has been
correlated with a reduced efcacy of the selective serotonin re- reported in the rat hippocampus following repeated inescapable
uptake inhibitor uoxetine for the relief of depressive symptoms footshock stress (van Dijken et al., 1993). Moreover, the enhanced
(Young et al., 2004), which suggests that resetting to a normal level response to the dexamethasone suppression test in adult victims
of central drive to the CRH neurons may prove an effective ther- of childhood sexual abuse (Stein et al., 1997) is suggestive of an
apy in cases of depression that are resistant to serotonin-based increased sensitivity to glucocorticoids, at least at the anterior
antidepressants. pituitary (Cole et al., 2000). Upregulated GR expression could be
Stress- and glucocorticoid-induced plasticity of neural cir- a protective response to hypocortisolemia, as some degree of GR
cuitry regulating the HPA axis may constitute a means through activity may be critical for survival (Cole et al., 1995), and the
which exposure to stress could play a role in a spectrum of initial hypocortisolemia could result from stress-induced synap-
HPA abnormalities, including aberrant HPA circadian rhythms, tic plasticity that promotes CRH cell suppression. Indeed, GR
abnormalities in HPA responsivity to stress, and basal HPA dys- expression generally increases following adrenalectomy (Olpe and
regulation. We hypothesize that this synaptic plasticity constitutes McEwen, 1976; Svec et al., 1989; Isenovic et al., 2006).
an etiological link between exposure to stress and a range of affec- With respect to future investigations of neural regulation of the
tive and stress-related disorders. We frame this hypothesis on the HPA axis, recent advances in circuit tracing technology have seen
basis of three premises: (1) exposure to stress increases the likeli- the development of a Cre-dependent retrogradely transported
hood of developing subsequent HPA dysregulation and associated marker derived from the rabies virus that will allow investiga-
pathologies, (2) rodent models have demonstrated that the synap- tors to trace the precise sources of innervation to CRH cells (Wall
tic mechanisms that regulate the HPA axis are highly plastic, and et al., 2010). This tool will inevitably improve our understanding
that synaptic plasticity is induced by both glucocorticoids and of how the brain integrates pertinent information into an HPA
stress, and (3) stress exposure and associated pathologies have, response. Also, one investigative team recently reported that peri-
at times, been linked to hypoactivity of the HPA axis, and this natal stress, caused by an active construction site adjacent to the
phenomenon cannot be explained by diminished glucocorticoid- rodent vivarium, resulted in hypercortisolemia that persisted even
induced suppression of the HPA axis. With regard to this last after the construction had ended (ORegan et al., 2010), which
premise, bromyalgia (Tanriverdi et al., 2007) and chronic fatigue underscores the sensitivity of HPA regulatory mechanisms to envi-
syndrome (Van Houdenhove et al., 2009) have both been linked to ronmental inuences (at least perinatally), but also serves as a
HPA hypoactivity, and stressful life events have been implicated warning to investigators of the importance of documenting the

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 9

fncel-06-00024 2012/5/9 20:17 page 9 #9


Levy and Tasker Synaptic regulation of the HPA axis

living conditions under which their subjects are kept. Lastly, it evidence that stress can result in persistent changes in HPA reg-
is important that assays of morphological plasticity and changes ulation. Rodent models of the past several years have provided
in ionotropic receptor expression are accompanied by electro- signicant insight into the etiology of stress-induced aberrations
physiological recordings, as electrophysiology provides invaluable in HPA activity, but our understanding is still in its infancy. Studies
information regarding functional manifestations of synaptic activ- of synaptic transmission will remain invaluable to this provocative
ity. Dysregulation of the HPA axis may be a causal factor in a range and promising line of research, as we uncover future targets for
of affective and physiological disorders, and there is increasing pharmacological intervention.

REFERENCES and behavior. Proc. Natl. Acad. Sci. gene expression in the paraventricu- neurons in the rat hypothalamic par-
Abelson, J. L., Khan, S., Liberzon, I., and U.S.A. 102, 473478. lar nucleus of the hypothalamus. J. aventricular nucleus via intranuclear
Young, E. A. (2007). HPA axis activ- Bradbury, M. J., Giracello, D. R., Neurosci. 22, 959969. glutamatergic circuits. J. Neurosci. 18,
ity in patients with panic disorder: Chapman, D. F., Holtz, G., Cole, T. J., Blendy, J. A., Monaghan, 1061910628.
review and synthesis of four studies. Schaffhauser, H., Rao, S. P., Varney, A. P., Krieglstein, K., Schmid, W., Daftary, S. S., Boudaba, C., and Tasker,
Depress. Anxiety 24, 6676. M. A., and Anderson, J. J. (2003). Aguzzi, A., Fantuzzi, G., Hummler, E., J. G. (2000). Noradrenergic regu-
Ahrens, T., Deuschle, M., Krumm, Metabotropic glutamate receptor 5 Unsicker, K., and Schutz, G. (1995). lation of parvocellular neurons in
B., Van Der Pompe, G., Den Boer, antagonist-induced stimulation of Targeted disruption of the glucocor- the rat hypothalamic paraventricular
J. A., and Lederbogen, F. (2008). hypothalamicpituitaryadrenal axis ticoid receptor gene blocks adren- nucleus. Neuroscience 96, 743751.
Pituitaryadrenal and sympathetic activity: interaction with serotoner- ergic chromafn cell development Day, H. E., Campeau, S., Watson, S. J.
nervous system responses to stress gic systems. Neuropharmacology 44, and severely retards lung maturation. Jr., and Akil, H. (1999). Expression
in women remitted from recurrent 562572. Genes Dev. 9, 16081621. of alpha(1b) adrenoceptor mRNA
major depression. Psychosom. Med. Brawman-Mintzer, O., Monnier, J., Cowley, M. A., Pronchuk, N., Fan, W., in corticotropin-releasing hormone-
70, 461467. Wolitzky, K. B., and Falsetti, S. A. Dinulescu, D. M., Colmers, W. F., containing cells of the rat hypotha-
Alon, T., Zhou, L., Perez, C. A., (2005). Patients with generalized and Cone, R. D. (1999). Integra- lamus and its regulation by corticos-
Gareld, A. S., Friedman, J. M., anxiety disorder and a history of tion of NPY, AGRP, and melanocortin terone. J. Neurosci. 19, 1009810106.
and Heisler, L. K. (2009). Trans- trauma: somatic symptom endorse- signals in the hypothalamic paraven- Dayas, C. V., Buller, K. M., and Day,
genic mice expressing green uo- ment. J. Psychiatr. Pract. 11, 212215. tricular nucleus: evidence of a cellular T. A. (2001). Medullary neurones
rescent protein under the control Calogero, A. E., Gallucci, W. T., basis for the adipostat. Neuron 24, regulate hypothalamic corticotropin-
of the corticotropin-releasing hor- Chrousos, G. P., and Gold, P. W. 155163. releasing factor cell responses to an
mone promoter. Endocrinology 150, (1988). Catecholamine effects upon Csaki, A., Kocsis, K., Halasz, B., and Kiss, emotional stressor. Neuroscience 105,
56265632. rat hypothalamic corticotropin- J. (2000). Localization of glutamater- 707719.
Aubry, J. M., Bartanusz, V., Pagliusi, releasing hormone secretion in vitro. gic/aspartatergic neurons projecting Di, S., Malcher-Lopes, R., Halmos, K. C.,
S., Schulz, P., and Kiss, J. Z. (1996). J. Clin. Invest. 82, 839846. to the hypothalamic paraventricular and Tasker, J. G. (2003). Nongenomic
Expression of ionotropic glutamate Choi, D. C., Furay, A. R., Evanson, nucleus studied by retrograde trans- glucocorticoid inhibition via endo-
receptor subunit mRNAs by paraven- N. K., Ostrander, M. M., Ulrich-Lai, port of [3H]D-aspartate autoradiog- cannabinoid release in the hypotha-
tricular corticotropin-releasing fac- Y. M., and Herman, J. P. (2007). raphy. Neuroscience 101, 637655. lamus: a fast feedback mechanism. J.
tor (CRF) neurons. Neurosci. Lett. Bed nucleus of the stria terminalis Cull-Candy, S., Brickley, S., and Far- Neurosci. 23, 48504857.
205, 9598. subregions differentially regulate rant, M. (2001). NMDA receptor Dimitrov, E. L., Dejoseph, M. R.,
Bali, B., and Kovacs, K. J. (2003). hypothalamicpituitaryadrenal axis subunits: diversity, development and Browneld, M. S., and Urban, J. H.
GABAergic control of neuropeptide activity: implications for the integra- disease. Curr. Opin. Neurobiol. 11, (2007). Involvement of neuropep-
gene expression in parvocellular neu- tion of limbic inputs. J. Neurosci. 27, 327335. tide Y Y1 receptors in the regulation
rons of the hypothalamic paraven- 20252034. Cullinan, W. E. (2000). GABA(A) of neuroendocrine corticotropin-
tricular nucleus. Eur. J. Neurosci. 18, Choi, D. C., Furay, A. R., Evanson, N. K., receptor subunit expression within releasing hormone neuronal activity.
15181526. Ulrich-Lai, Y. M., Nguyen, M. M., hypophysiotropic CRH neurons: a Endocrinology 148, 36663673.
Bartanusz, V., Aubry, J. M., Pagliusi, S., Ostrander, M. M., and Herman, J. P. dual hybridization histochemical Durand, D., Pampillo, M., Caruso,
Jezova, D., Baf, J., and Kiss, J. Z. (2008). The role of the posterior study. J. Comp. Neurol. 419, 344351. C., and Lasaga, M. (2008). Role
(1995). Stress-induced changes in medial bed nucleus of the stria termi- Cullinan, W. E., and Wolfe, T. J. of metabotropic glutamate receptors
messenger RNA levels of N -methyl- nalis in modulating hypothalamic (2000). Chronic stress regulates levels in the control of neuroendocrine
D-aspartate and AMPA receptor sub- pituitaryadrenocortical axis respon- of mRNA transcripts encoding beta function. Neuropharmacology 55,
units in selected regions of the siveness to acute and chronic stress. subunits of the GABA(A) receptor in 577583.
rat hippocampus and hypothalamus. Psychoneuroendocrinology 33, 659 the rat stress axis. Brain Res. 887, Fava, G. A., Carson, S. W., Perini, G. I.,
Neuroscience 66, 247252. 669. 118124. Morphy, M. A., Molnar, G., and Jusko,
Boudaba, C., Schrader, L. A., and Tasker, Chuluyan, H. E., Saphier, D., Rohn, Cummings, S., and Seybold, V. W. J. (1984). The metyrapone test in
J. G. (1997). Physiological evidence W. M., and Dunn, A. J. (1992). (1988). Relationship of alpha-1- affective disorders and schizophrenia.
for local excitatory synaptic circuits in Noradrenergic innervation of the and alpha-2-adrenergic-binding sites J. Affect. Disord. 6, 241247.
the rat hypothalamus. J. Neurophys- hypothalamus participates in adre- to regions of the paraventricular Feldman, S., and Weidenfeld, J. (1997).
iol. 77, 33963400. nocortical responses to interleukin-1. nucleus of the hypothalamus con- Hypothalamic mechanisms medi-
Boudaba, C., Szabo, K., and Tasker, Neuroendocrinology 56, 106111. taining corticotropin-releasing factor ating glutamate effects on the
J. G. (1996). Physiological map- Cole, M. A., Kim, P. J., Kalman, B. A., and vasopressin neurons. Neuroen- hypothalamopituitaryadrenocort-
ping of local inhibitory inputs and Spencer, R. L. (2000). Dex- docrinology 47, 523532. ical axis. J. Neural Transm. 104,
to the hypothalamic paraventricular amethasone suppression of corticos- Daban, C., Vieta, E., Mackin, P., and 633642.
nucleus. J. Neurosci. 16, 71517160. teroid secretion: evaluation of the Young, A. H. (2005). Hypothalamic Feldman, S., and Weidenfeld, J.
Boyle, M. P., Brewer, J. A., Funatsu, M., site of action by receptor measures pituitaryadrenal axis and bipolar (2004). Involvement of endoge-
Wozniak, D. F., Tsien, J. Z., Izumi, and functional studies. Psychoneu- disorder. Psychiatr. Clin. North Am. neous glutamate in the stimulatory
Y., and Muglia, L. J. (2005). Acquired roendocrinology 25, 151167. 28, 469480. effect of norepinephrine and sero-
decit of forebrain glucocorticoid Cole, R. L., and Sawchenko, P. E. (2002). Daftary, S. S., Boudaba, C., Szabo, tonin on the hypothalamopituitary
receptor produces depression-like Neurotransmitter regulation of cel- K., and Tasker, J. G. (1998). Nora- adrenocortical axis. Neuroendocrinol-
changes in adrenal axis regulation lular activation and neuropeptide drenergic excitation of magnocellular ogy 79, 4353.

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 10

fncel-06-00024 2012/5/9 20:17 page 10 #10


Levy and Tasker Synaptic regulation of the HPA axis

Feuvrier, E., Aubert, M., Mausset, A. Noradrenaline excites and inhibits Jacobson, L., and Sapolsky, R. (1993). Kuzmiski, J. B., Marty, V., Baimoukha-
L., Alonso, G., Gaillet, S., Malaval, F., GABAergic transmission in parvo- Augmented ACTH responses to stress metova, D. V., and Bains, J. S. (2010).
and Szafarczyk, A. (1998). Glucocor- cellular neurons of rat hypothalamic in adrenalectomized rats replaced Stress-induced priming of glutamate
ticoids provoke a shift from alpha2- paraventricular nucleus. J. Neuro- with constant, physiological levels synapses unmasks associative short-
to alpha1-adrenoreceptor activities in physiol. 87, 22872296. of corticosterone are partially nor- term plasticity. Nat. Neurosci. 13,
cultured hypothalamic slices lead- Harris, C. C., and Tasker, J. G. (2011). malized by acute increases in cor- 12571264.
ing to opposite noradrenaline effect A Calcium Requirement For Rapid ticosterone. Neuroendocrinology 58, Lang, C. H., and Ajmal, M.
on corticotropin-releasing hormone Glucocorticoid Dependent Modula- 420429. (1995). Metabolic, hormonal, and
release. J. Neurochem. 70, 11991209. tion Of Glutamate Release In The Joels, M., Karst, H., Alfarez, D., Heine, hemodynamic changes induced by
Flak, J. N., Ostrander, M. M., Tasker, Supraoptic Nucleus. Program No. V. M., Qin, Y., Van Riel, E., Verkuyl, metabotropic excitatory amino acid
J. G., and Herman, J. P. (2009). 501.01/VV32. Washington, DC: Neu- M., Lucassen, P. J., and Krugers, H. J. agonist (1S,3R)-ACPD. Am. J. Phys-
Chronic stress-induced neurotrans- roscience Meeting Planner, Society (2004). Effects of chronic stress on iol. 268, R1026R1033.
mitter plasticity in the PVN. J. Comp. for Neuroscience. Available at: http:// structure and cell function in rat hip- Lenze, E. J., Mantella, R. C., Shi, P.,
Neurol. 517, 156165. www.sfn.org/am2011/pdf/prelim/ pocampus and hypothalamus. Stress Goate, A. M., Nowotny, P., Butters,
Franco, A. J., Zsombok, A., and Tasker, MON_Poster_PM_v2.pdf 7, 221231. M. A., Andreescu, C., Thomp-
J. G. (2007). Rapid Glucocorticoid- Hatcher, S., and House, A. (2003). Johnson, M. P., Kelly, G., and Cham- son, P. A., and Rollman, B. L.
Induced, Endocannabinoid-Mediated Life events, difculties and dilemmas berlain, M. (2001). Changes in rat (2011). Elevated cortisol in older
Inhibition of Synaptic Excitation in the in the onset of chronic fatigue syn- serum corticosterone after treatment adults with generalized anxiety dis-
Hypothalamus Maintained Following drome: a casecontrol study. Psychol. with metabotropic glutamate recep- order is reduced by treatment: a
Chronic Variable Stress. Program No. Med. 33, 11851192. tor agonists or antagonists. J. Neu- placebo-controlled evaluation of esc-
298.25/TT27. Washington, DC: Neu- Heifets, B. D., and Castillo, P. E. roendocrinol. 13, 670677. italopram. Am. J. Geriatr. Psychiatry
roscience Meeting Planner, Society (2009). Endocannabinoid signaling Juruena, M. F., Cleare, A. J., Papadopou- 19, 482490.
for Neuroscience. Available at: http:// and long-term synaptic plasticity. los, A. S., Poon, L., Lightman, S., Li, H. Y., Ericsson, A., and Sawchenko,
www.sfn.org/index.aspx?pagename= Annu. Rev. Physiol. 71, 283306. and Pariante, C. M. (2006). Differ- P. E. (1996). Distinct mechanisms
abstracts_ampublications Herman, J. P., Adams, D., and Pre- ent responses to dexamethasone and underlie activation of hypothalamic
Freund, T. F., Katona, I., and Piomelli, D. witt, C. (1995). Regulatory changes prednisolone in the same depressed neurosecretory neurons and their
(2003). Role of endogenous cannabi- in neuroendocrine stress-integrative patients. Psychopharmacology (Berl.) medullary catecholaminergic affer-
noids in synaptic signaling. Physiol. circuitry produced by a variable stress 189, 225235. ents in categorically different stress
Rev. 83, 10171066. paradigm. Neuroendocrinology 61, Kendler, K. S., Kessler, R. C., Walters, paradigms. Proc. Natl. Acad. Sci.
Galanopoulou, A. S., and Moshe, S. L. 180190. E. E., Maclean, C., Neale, M. C., U.S.A. 93, 23592364.
(2003). Role of sex hormones in Herman, J. P., Eyigor, O., Ziegler, D. R., Heath, A. C., and Eaves, L. J. (1995). Liposits, Z., Phelix, C., and Paull, W.
the sexually dimorphic expression of and Jennes, L. (2000). Expression of Stressful life events, genetic liability, K. (1986). Adrenergic innervation of
KCC2 in rat substantia nigra. Exp. ionotropic glutamate receptor sub- and onset of an episode of major corticotropin releasing factor (CRF)-
Neurol. 184, 10031009. unit mRNAs in the hypothalamic depression in women. Am. J. Psychi- synthesizing neurons in the hypotha-
Garno, J. L., Goldberg, J. F., Ramirez, paraventricular nucleus of the rat. J. atry 152, 833842. lamic paraventricular nucleus of
P. M., and Ritzler, B. A. (2005). Comp. Neurol. 422, 352362. Kim, J. S., Kim, W. B., Kim, Y. B., Lee, the rat. A combined light and
Impact of childhood abuse on the Herman, J. P., Figueiredo, H., Mueller, Y., Kim, Y. S., Shen, F. Y., Lee, S. W., electron microscopic immunocyto-
clinical course of bipolar disorder. Br. N. K., Ulrich-Lai, Y., Ostrander, Park, D., Choi, H. J., Hur, J., Park, J. J., chemical study. Histochemistry 84,
J. Psychiatry 186, 121125. M. M., Choi, D. C., and Cullinan, Han, H. C., Colwell, C. S., Cho, Y. W., 201205.
Gillespie, C. F., and Nemeroff, W. E. (2003). Central mechanisms and Kim, Y. I. (2011). Chronic hyper- Liu, D., Diorio, J., Tannenbaum, B.,
C. B. (2005). Hypercortisolemia of stress integration: hierarchical osmotic stress converts GABAergic Caldji, C., Francis, D., Freedman, A.,
and depression. Psychosom. Med. circuitry controlling hypothalamo inhibition into excitation in vaso- Sharma, S., Pearson, D., Plotsky, P.
67(Suppl. 1), S26S28. pituitaryadrenocortical responsive- pressin and oxytocin neurons in the M., and Meaney, M. J. (1997). Mater-
Goodwin, R. D., Fergusson, D. M., and ness. Front. Neuroendocrinol. 24, rat. J. Neurosci. 31, 1331213322. nal care, hippocampal glucocorti-
Horwood, L. J. (2005). Childhood 151180. King, J. A., Mandansky, D., King, S., coid receptors, and hypothalamic
abuse and familial violence and the Hewitt, S. A., Wamsteeker, J. I., Kurz, Fletcher, K. E., and Brewer, J. (2001). pituitaryadrenal responses to stress.
risk of panic attacks and panic dis- E. U., and Bains, J. S. (2009). Alte- Early sexual abuse and low corti- Science 277, 16591662.
order in young adulthood. Psychol. red chloride homeostasis removes sol. Psychiatry Clin. Neurosci. 55, Louvart, H., Maccari, S., Ducrocq, F.,
Med. 35, 881890. synaptic inhibitory constraint of 7174. Thomas, P., and Darnaudery, M.
Gupta, A., and Silman, A. J. (2004). the stress axis. Nat. Neurosci. 12, Kitraki, E., Karandrea, D., and Kit- (2005). Long-term behavioural alter-
Psychological stress and bromyalgia: 438443. tas, C. (1999). Long-lasting effects ations in female rats after a single
a review of the evidence suggesting Hrabovszky, E., and Liposits, Z. (2008). of stress on glucocorticoid recep- intense footshock followed by sit-
a neuroendocrine link. Arthritis Res. Novel aspects of glutamatergic sig- tor gene expression in the rat brain. uational reminders. Psychoneuroen-
Ther. 6, 98106. nalling in the neuroendocrine system. Neuroendocrinology 69, 331338. docrinology 30, 316324.
Gustafsson, P. E., Gustafsson, P. A., J. Neuroendocrinol. 20, 743751. Kocsis, K., Kiss, J., Gorcs, T., and Halasz, Luther, J. A., Daftary, S. S., Boudaba,
Ivarsson, T., and Nelson, N. (2008). Isenovic, E. R., Radojcic, M., Zakula, Z., B. (1998). Metabotropic glutamate C., Gould, G. C., Halmos, K. C., and
Diurnal cortisol levels and cortisol Koricanac, G., and Ribarac-Stepic, N. receptor in vasopressin, CRF and VIP Tasker, J. G. (2002). Neurosecretory
response in youths with obsessive- (2006). Effect of acute adrenalectomy hypothalamic neurones. Neuroreport and non-neurosecretory parvocellu-
compulsive disorder. Neuropsychobi- on rat liver glucocorticoid receptor. 9, 40294033. lar neurones of the hypothalamic
ology 57, 1421. Arch. Biol. Sci. 58, 153159. Korosi, A., Shanabrough, M., Mcclel- paraventricular nucleus express dis-
Haam, J., Popescu, I. R., Morton, L. A., Itoi, K., Suda, T., Tozawa, F., Dobashi, land, S., Liu, Z. W., Borok, tinct electrophysiological properties.
Halmos, K. C., Teruyama, R., Ueta, I., Ohmori, N., Sakai, Y., Abe, K., and E., Gao, X. B., Horvath, T. L., J. Neuroendocrinol. 14, 929932.
U., and Tasker, J. G. (2012). GABA Demura, H. (1994). Microinjection and Baram, T. Z. (2010). Early- Luther, J. A., and Tasker, J. G.
is excitatory in adult vasopressinergic of norepinephrine into the paraven- life experience reduces excitation to (2000). Voltage-gated currents distin-
neuroendocrine cells. J. Neurosci. 32, tricular nucleus of the hypothalamus stress-responsive hypothalamic neu- guish parvocellular from magnocel-
572582. stimulates corticotropin-releasing rons and reprograms the expression lular neurones in the rat hypothala-
Han, S. K., Chong, W., Li, L. H., Lee, I. factor gene expression in conscious of corticotropin-releasing hormone. mic paraventricular nucleus. J. Phys-
S., Murase, K., and Ryu, P. D. (2002). rats. Endocrinology 135, 21772182. J. Neurosci. 30, 703713. iol. 523(Pt 1), 193209.

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 11

fncel-06-00024 2012/5/9 20:17 page 11 #11


Levy and Tasker Synaptic regulation of the HPA axis

Ma, S., and Morilak, D. A. (2005). the paraventricular nucleus during 2-autoreceptors in human brain cor- I. (1990). [Implication of alpha-2
Chronic intermittent cold stress sen- immobilization stress. Endocrinology tex. Br. J. Pharmacol. 107, 11461151. adrenergic post-synaptic receptors in
sitises the hypothalamicpituitary 133, 14041410. Roland, B. L., and Sawchenko, P. the central catecholaminergic stim-
adrenal response to a novel acute Pacak, K., Palkovits, M., Kvetnansky, R., E. (1993). Local origins of some ulation of the corticotropic axis in
stress by enhancing noradrenergic Matern, P., Hart, C., Kopin, I. J., GABAergic projections to the par- rats]. C R Acad. Sci. III. 311,
inuence in the rat paraventricu- and Goldstein, D. S. (1995). Cate- aventricular and supraoptic nuclei of 8188.
lar nucleus. J. Neuroendocrinol. 17, cholaminergic inhibition by hyper- the hypothalamus in the rat. J. Comp. Tanriverdi, F., Karaca, Z., Unluhizarci,
761769. cortisolemia in the paraventricular Neurol. 332, 123143. K., and Kelestimur, F. (2007). The
Majewska, M. D., Bisserbe, J. C., and nucleus of conscious rats. Endocrinol- Sarkhel, S., Praharaj, S. K., and Sinha, hypothalamopituitaryadrenal axis
Eskay, R. L. (1985). Glucocorticoids ogy 136, 48144819. V. K. (2011). Role of life events in in chronic fatigue syndrome and
are modulators of GABAA receptors Pacak, K., Palkovits, M., Yadid, G., obsessive compulsive disorder. Isr. J. bromyalgia syndrome. Stress 10,
in brain. Brain Res. 339, 178182. Kvetnansky, R., Kopin, I. J., and Gold- Psychiatry Relat. Sci. 48, 182185. 1325.
Malcher-Lopes, R., Di, S., Marcheselli, stein, D. S. (1998). Heterogeneous Sawchenko, P. E. (1988). Effects of Tasker, J. G., Boudaba, C., and
V. S., Weng, F. J., Stuart, C. T., neurochemical responses to different catecholamine-depleting medullary Schrader, L. A. (1998). Local glu-
Bazan, N. G., and Tasker, J. G. (2006). stressors: a test of Selyes doctrine of knife cuts on corticotropin-releasing tamatergic and GABAergic synaptic
Opposing crosstalk between leptin nonspecicity. Am. J. Physiol. 275, factor and vasopressin immunoreac- circuits and metabotropic glutamate
and glucocorticoids rapidly modu- R1247R1255. tivity in the hypothalamus of normal receptors in the hypothalamic par-
lates synaptic excitation via endo- Packan, D. R., and Sapolsky, R. M. and steroid-manipulated rats. Neu- aventricular and supraoptic nuclei.
cannabinoid release. J. Neurosci. 26, (1990). Glucocorticoid endanger- roendocrinology 48, 459470. Adv. Exp. Med. Biol. 449, 117121.
66436650. ment of the hippocampus: tissue, Schrader, L. A., and Tasker, J. G. Tasker, J. G., and Dudek, F. E.
Miklos, I. H., and Kovacs, K. J. (2002). steroid and receptor specicity. Neu- (1997). Presynaptic modulation by (1991). Electrophysiological proper-
GABAergic innervation of corti- roendocrinology 51, 613618. metabotropic glutamate receptors of ties of neurones in the region of the
cotropin-releasing hormone (CRH)- Penninx, B. W., Beekman, A. T., excitatory and inhibitory synaptic paraventricular nucleus in slices of
secreting parvocellular neurons and Bandinelli, S., Corsi, A. M., Brem- inputs to hypothalamic magnocel- rat hypothalamus. J. Physiol. 434,
its plasticity as demonstrated by mer, M., Hoogendijk, W. J., Guralnik, lular neurons. J. Neurophysiol. 77, 271293.
quantitative immunoelectron micro- J. M., and Ferrucci, L. (2007). Late- 527536. Tuchelt, H., Dekker, K., Bahr, V., and
scopy. Neuroscience 113, 581592. life depressive symptoms are asso- Shimizu, K. (1984). Effect of alpha 1- Oelkers, W. (2000). Dose-response
Modell, S., Yassouridis, A., Huber, J., and ciated with both hyperactivity and and alpha 2-adrenoceptor agonists relationship between plasma ACTH
Holsboer, F. (1997). Corticosteroid hypoactivity of the hypothalamo and antagonists on ACTH secretion and serum cortisol in the insulin-
receptor function is decreased in pituitaryadrenal axis. Am. J. Geriatr. in intact and in hypothalamic deaf- hypoglycaemia test in 25 healthy sub-
depressed patients. Neuroendocrinol- Psychiatry 15, 522529. ferentated rats. Jpn. J. Pharmacol. 36, jects and 109 patients with pituitary
ogy 65, 216222. Plotsky, P. M., Otto, S., and Sutton, S. 2333. disease. Clin. Endocrinol. (Oxf.) 53,
Morphy, M. A., Fava, G. A., Perini, (1987). Neurotransmitter modula- Simmons, D. M., and Swanson, L. W. 301307.
G. I., and Molnar, G. (1985). The tion of corticotropin releasing fac- (2009). Comparison of the spatial Ulrich-Lai, Y. M., Jones, K. R., Ziegler,
metyrapone test in depressed males. tor secretion into the hypophysial- distribution of seven types of neu- D. R., Cullinan, W. E., and Her-
Prog. Neuropsychopharmacol. Biol. portal circulation. Life Sci. 41, roendocrine neurons in the rat par- man, J. P. (2011). Forebrain ori-
Psychiatry 9, 187191. 13111317. aventricular nucleus: toward a global gins of glutamatergic innervation
Nahar, J., Haam, J., Glatzer, N. R., Hal- Raadsheer, F. C., Hoogendijk, W. J., 3D model. J. Comp. Neurol. 516, to the rat paraventricular nucleus
mos, K. C., Muglia, L. J., Dohanich, Stam, F. C., Tilders, F. J., and Swaab, 423441. of the hypothalamus: differential
G. P., and Tasker, J. G. (2012). Rapid D. F. (1994). Increased numbers Stein, M. B., Yehuda, R., Koverola, C., inputs to the anterior versus poste-
glucocorticoid actions in hypothala- of corticotropin-releasing hormone and Hanna, C. (1997). Enhanced dex- rior subregions. J. Comp. Neurol. 519,
mic neuroendocrine cells are depen- expressing neurons in the hypotha- amethasone suppression of plasma 13011319.
dent on the glucocorticoid receptor. lamic paraventricular nucleus of cortisol in adult women traumatized Ur, E., Dinan, T. G., Okeane, V.,
Endocrinology (submitted). depressed patients. Neuroendocrinol- by childhood sexual abuse. Biol. Psy- Clare, A. W., Mcloughlin, L., Rees,
ORegan, D., Kenyon, C. J., Seckl, J. R., ogy 60, 436444. chiatry 42, 680686. L. H., Turner, T. H., Grossman, A.,
and Holmes, M. C. (2010). Environ- Raadsheer, F. C., Van Heerikhuize, Stern, J. E. (2001). Electrophysio- and Besser, G. M. (1992). Effect of
mental disturbance confounds pre- J. J., Lucassen, P. J., Hoogendijk, logical and morphological proper- metyrapone on the pituitaryadrenal
natal glucocorticoid programming W. J., Tilders, F. J., and Swaab, ties of pre-autonomic neurones in axis in depression: relation to dexam-
experiments in Wistar rats. Lab. D. F. (1995). Corticotropin-releasing the rat hypothalamic paraventricular ethasone suppressor status. Neuroen-
Anim. 44, 199205. hormone mRNA levels in the par- nucleus. J. Physiol. 537, 161177. docrinology 56, 533538.
Olpe, H. R., and McEwen, B. S. (1976). aventricular nucleus of patients with Svec, F., Gordon, S., and Tate, D. (1989). van Dijken, H. H., De Goeij, D. C.,
Glucocorticoid binding to receptor- Alzheimers disease and depression. Glucocorticoid receptor regulation: Sutanto, W., Mos, J., De Kloet,
like proteins in rat brain and pitu- Am. J. Psychiatry 152, 13721376. the effects of adrenalectomy, exoge- E. R., and Tilders, F. J. (1993).
itary: ontogenetic and experimentally Radley, J. J., Gosselink, K. L., and nous glucocorticoid, and stress on Short inescapable stress produces
induced changes. Brain Res. 105, Sawchenko, P. E. (2009). A discrete hepatic receptor number in male and long-lasting changes in the brain
121128. GABAergic relay mediates medial female mice. Biochem. Med. Metab. pituitaryadrenal axis of adult male
Ostrander, M. M., Ulrich-Lai, Y. M., prefrontal cortical inhibition of the Biol. 41, 224233. rats. Neuroendocrinology 58, 5764.
Choi, D. C., Richtand, N. M., and neuroendocrine stress response. J. Szafarczyk, A., Feuvrier, E., Siaud, P., Van Houdenhove, B., Van Den Eede,
Herman, J. P. (2006). Hypoactiv- Neurosci. 29, 73307340. Rondouin, G., Lacoste, M., Gaillet, F., and Luyten, P. (2009). Does
ity of the hypothalamopituitary Radley, J. J., and Sawchenko, P. E. (2011). S., Malaval, F., and Assenmacher, I. hypothalamicpituitaryadrenal axis
adrenocortical axis during recov- A common substrate for prefrontal (1995). Removal of adrenal steroids hypofunction in chronic fatigue syn-
ery from chronic variable stress. and hippocampal inhibition of the from the medium reverses the drome reect a crash in the
Endocrinology 147, 20082017. neuroendocrine stress response. J. stimulating effect of catecholamines stress system? Med. Hypotheses 72,
Pacak, K., Kvetnansky, R., Palkovits, Neurosci. 31, 96839695. on corticotropin-releasing hormone 701705.
M., Fukuhara, K., Yadid, G., Raiteri, M., Bonanno, G., Maura, neurons in organotypic cultures. Verkuyl, J. M., Hemby, S. E., and Joels,
Kopin, I. J., and Goldstein, D. S. G., Pende, M., Andrioli, G. C., Neuroendocrinology 61, 517524. M. (2004). Chronic stress attenuates
(1993). Adrenalectomy augments in and Ruelle, A. (1992). Subclassi- Szafarczyk, A., Gaillet, S., Barbanel, GABAergic inhibition and alters gene
vivo release of norepinephrine in cation of release-regulating alpha G., Malaval, F., and Assenmacher, expression of parvocellular neurons

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 12

fncel-06-00024 2012/5/9 20:17 page 12 #12


Levy and Tasker Synaptic regulation of the HPA axis

in rat hypothalamus. Eur. J. Neurosci. nucleus by glucose and feeding- Yehuda, R. (2006). Advances in under- Ziegler, D. R., and Herman, J. P.
20, 16651673. relevant peptides. Diabetes 53, standing neuroendocrine alterations (2002). Neurocircuitry of stress inte-
Verkuyl, J. M., and Joels, M. (2003). 19591965. in PTSD and their therapeutic impli- gration: anatomical pathways reg-
Effect of adrenalectomy on minia- Webster, M. J., Knable, M. B., Ogrady, cations. Ann. N.Y. Acad. Sci. 1071, ulating the hypothalamopituitary
ture inhibitory postsynaptic currents J., Orthmann, J., and Weickert, C. S. 137166. adrenocortical axis of the rat. Integr.
in the paraventricular nucleus of the (2002). Regional specicity of brain Yehuda, R., Boisoneau, D., Lowy, M. Comp. Biol. 42, 541551.
hypothalamus. J. Neurophysiol. 89, glucocorticoid receptor mRNA alter- T., and Giller, E. L. Jr. (1995).
237245. ations in subjects with schizophrenia Doseresponse changes in plasma
Verkuyl, J. M., Karst, H., and Joels, M. and mood disorders. Mol. Psychiatry cortisol and lymphocyte glucocor-
(2005). GABAergic transmission in 7, 985994; 924. ticoid receptors following dexam- Conflict of Interest Statement: The
the rat paraventricular nucleus of the Wittmann, G., Lechan, R. M., ethasone administration in combat authors declare that the research was
hypothalamus is suppressed by corti- Liposits, Z., and Fekete, C. (2005). veterans with and without posttrau- conducted in the absence of any com-
costerone and stress. Eur. J. Neurosci. Glutamatergic innervation of corti- matic stress disorder. Arch. Gen. mercial or nancial relationships that
21, 113121. cotropin-releasing hormone- and Psychiatry 52, 583593. could be construed as a potential con-
Wall, N. R., Wickersham, I. R., Cetin, A., thyrotropin-releasing hormone-syn- Young, E. A., Altemus, M., Lopez, J. F., ict of interest.
De La Parra, M., and Callaway, E. M. thesizing neurons in the hypothala- Kocsis, J. H., Schatzberg, A. F., Debat-
(2010). Monosynaptic circuit trac- mic paraventricular nucleus of the tista, C., and Zubieta, J. K. (2004). Received: 12 March 2012; accepted: 26
ing in vivo through Cre-dependent rat. Brain Res. 1039, 5362. HPA axis activation in major depres- April 2012; published online: 11 May
targeting and complementation of Yang, J. H., Li, L. H., Lee, S., Jo, I. H., Lee, sion and response to uoxetine: a 2012.
modied rabies virus. Proc. Natl. S. Y., and Ryu, P. D. (2007). Effects pilot study. Psychoneuroendocrinol- Citation: Levy BH and Tasker JG (2012)
Acad. Sci. U.S.A. 107, 21848 of adrenalectomy on the excitability ogy 29, 11981204. Synaptic regulation of the hypothalamic
21853. of neurosecretory parvocellular neu- Ziegler, D. R., Cullinan, W. E., and pituitaryadrenal axis and its modula-
Wamsteeker, J. I., Kuzmiski, J. B., rones in the hypothalamic paraven- Herman, J. P. (2005). Organization tion by glucocorticoids and stress. Front.
and Bains, J. S. (2010). Repeated tricular nucleus. J. Neuroendocrinol. and regulation of paraventricular Cell. Neurosci. 6:24. doi: 10.3389/fncel.
stress impairs endocannabinoid sig- 19, 293301. nucleus glutamate signaling systems: 2012.00024
naling in the paraventricular nucleus Yang, J. H., Li, L. H., Shin, S. Y., N -methyl-D-aspartate receptors. J. Copyright 2012 Levy and Tasker. This
of the hypothalamus. J. Neurosci. 30, Lee, S., Lee, S. Y., Han, S. K., Comp. Neurol. 484, 4356. is an open-access article distributed under
1118811196. and Ryu, P. D. (2008). Adrenalec- Ziegler, D. R., and Herman, J. P. (2000). the terms of the Creative Commons
Wang, R., Liu, X., Hentges, S. T., Dunn- tomy potentiates noradrenergic sup- Local integration of glutamate signal- Attribution Non Commercial License,
Meynell, A. A., Levin, B. E., Wang, pression of GABAergic transmission ing in the hypothalamic paraventric- which permits non-commercial use, dis-
W., and Routh, V. H. (2004). The in parvocellular neurosecretory neu- ular region: regulation of glucocor- tribution, and reproduction in other
regulation of glucose-excited neu- rons of hypothalamic paraventricular ticoid stress responses. Endocrinology forums, provided the original authors and
rons in the hypothalamic arcuate nucleus. J. Neurophysiol. 99, 514523. 141, 48014804. source are credited.

Frontiers in Cellular Neuroscience www.frontiersin.org May 2012 | Volume 6 | Article 24 | 13

fncel-06-00024 2012/5/9 20:17 page 13 #13

You might also like