Вы находитесь на странице: 1из 30

Frontiers in

Neuroendocrinology

Frontiers in Neuroendocrinology 24 (2003) 151180


www.elsevier.com/locate/yfrne

Central mechanisms of stress integration: hierarchical circuitry


controlling hypothalamopituitaryadrenocortical responsiveness
James P. Herman,a,b,* Helmer Figueiredo,a Nancy K. Mueller,a Yvonne Ulrich-Lai,a
Michelle M. Ostrander,a Dennis C. Choi,a and William E. Cullinanc
a
Department of Psychiatry, University of Cincinnati, Cincinnati, OH 45267-0559, USA
b
Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, Cincinnati, OH 45267-0559, USA
c
Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA

Abstract

Appropriate regulatory control of the hypothalamopituitaryadrenocortical stress axis is essential to health and survival. The
following review documents the principle extrinsic and intrinsic mechanisms responsible for regulating stress-responsive CRH
neurons of the hypothalamic paraventricular nucleus, which summate excitatory and inhibitory inputs into a net secretory signal at
the pituitary gland. Regions that directly innervate these neurons are primed to relay sensory information, including visceral af-
ferents, nociceptors and circumventricular organs, thereby promoting reactive corticosteroid responses to emergent homeostatic
challenges. Indirect inputs from the limbic-associated structures are capable of activating these same cells in the absence of frank
physiological challenges; such anticipatory signals regulate glucocorticoid release under conditions in which physical challenges
may be predicted, either by innate programs or conditioned stimuli. Importantly, anticipatory circuits are integrated with neural
pathways subserving reactive responses at multiple levels. The resultant hierarchical organization of stress-responsive neurocir-
cuitries is capable of comparing information from multiple limbic sources with internally generated and peripherally sensed in-
formation, thereby tuning the relative activity of the adrenal cortex. Imbalances among these limbic pathways and homeostatic
sensors are likely to underlie hypothalamopituitaryadrenocortical dysfunction associated with numerous disease processes.
2003 Elsevier Inc. All rights reserved.

Keywords: Corticotropin releasing hormone; Glucocorticoids; ACTH; Hypothalamus; Paraventricular nucleus; Nucleus of the solitary tract;
Hippocampus; Amygdala

1. Introduction [208,217], and in such fashion have rapid actions on


homeostatic regulation.
The hypothalamopituitaryadrenocortical (HPA) The end eects of glucocorticoid action include en-
axis plays a vital role in adaptation of the organism to ergy mobilization (glycogenolysis) in the liver, suppres-
homeostatic challenge. Activation of the HPA system sion of innate immunity in immune organs, inhibition of
culminates in secretion of glucocorticoids, which act at bone and muscle growth, potentiation of sympathetic
multiple levels to redirect bodily energy resources nervous system-mediated vasoconstriction, proteolysis
[198,210,262]. These hormones are recognized by glu- and lipolysis, suppression of reproductive function
cocorticoid receptor molecules in numerous organ sys- along the hypothalamopituitarygonadal axis, and
tems, and act by genomic mechanisms to modify behavioral depression (see [198,210]). The spectrum of
transcription of key regulatory proteins [198,210]. eects have led to the hypothesis that glucocorticoids act
Emerging evidence suggests that glucocorticoids also act to restore homeostasis following disruption [210]; for
by non-genomic mechanisms on cell signaling processes example, increasing glucose can replenish lost energy
stores; inhibiting T-cell proliferation will control the
inammatory response; and inhibiting other hormonal
*
Corresponding author. Fax: 1-513-558-9104. systems reduces expenditure of energy on processes
E-mail address: james.herman@uc.edu (J.P. Herman). unrelated to the immediate challenge. These restorative

0091-3022/$ - see front matter 2003 Elsevier Inc. All rights reserved.
doi:10.1016/j.yfrne.2003.07.001
152 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

processes are generally catabolic in nature, and naturally small population of cells (approximately 4000 in rat
if extended in time can take a powerful toll on the or- [290]) produce a number of ACTH-releasing factors.
ganism. Indeed, glucocorticoid hypersecretion is impli- Of these, corticotropin releasing hormone (CRH) is
cated as a major deleterious factor in the aging process required for normal ACTH release under both basal
[171,172,261], and is known to accompany numerous and stressed conditions [6,319] and is the dening
long-term metabolic, aective, and psychotic disease phenotypic feature of this cell type. The most impor-
states (see [111,198], for review). tant co-expressed peptide is arginine vasopressin, which
Accordingly, adequate control of glucocorticoids synergizes with CRH to enhance the gain of the
needs to be accomplished by the organism. Such ACTH response [6,187,319]. These neurons also pro-
negative feedback control is eciently exerted in duce numerous other peptides and neurotransmitters
healthy organisms, and involves both rapid and geno- [155], and may thus have their net activity orchestrated
mic actions at the pituitary and at multiple sites in the by multiple neuroactive species. Once released by the
brain (discussed below). Thus, a HPA response is corticotropes, ACTH travels through the systemic cir-
generally characterized by a temporally regulated culation and promotes on-site synthesis and secretion
surge of ACTH release followed by a shut-o signal of corticosteroids at the adrenal cortex. While ACTH
generated by glucocorticoid as well as neuronal feed- is the major modulator of corticosteroid release, adre-
back (see Fig. 1, [151]). The ACTH surge is initiated nocortical output can be modulated by neuronal inputs
by a discrete population of hypophysiotrophic neurons that adjust responsivity to ACTH [304].
in the medial parvocellular division of the paraven- The HPA axis operates in two equally important
tricular hypothalamic nucleus (PVN) [6,319]. This domains of activity. Under relatively unstressed condi-

Fig. 1. Overview of the hypothalamopituitaryadrenocortical axis, including principal classes of regulatory aerents and corticosteroid actions.
CRH neurons located within the hypothalamic paraventricular nucleus (PVN) drive pituitary corticotrophs via the portal vasculature, stimulating
the release of ACTH. ACTH, in turn, mediates the synthesis and release of corticosteroids from the adrenal glands. CRH neurons are regulated by
sensory aerents which are relayed via brainstem loci, and transmit reactive stimuli which are generally excitatory and relatively direct. Conversely,
limbic forebrain structures are hypothesized to convey anticipatory signals that involve processing within pathways proximal to the level of the
PVN, including in the peri-PVN area and several local hypothalamic regions. Integration of anticipatory circuits and neural pathways subserving
reactive responses occurs at multiple levels (not shown; see text).
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 153

tions, glucocorticoid secretion undergoes a daily sensory stimuli signaling homeostatic disruption. These
rhythm, with peak secretion occurring at the initiation responses are centrally generated in the absence of a
of the waking cycle in most vertebrate organisms [151]. physiological challenge, and represent an eort of the
Secretion during the waking phase permits circulating organism to mount a glucocorticoid response in antici-
glucocorticoids to partially occupy glucocorticoid re- pation of, rather than as a reaction to, homeostatic
ceptors [238], and is believed to be critical for optimizing disruption. These anticipatory responses are either
functional tone of numerous systems [67]. For example, generated by conditioning (memory) or by innate,
partial occupation of hippocampal glucocorticoid re- species-specic predispositions (e.g., recognition of pre-
ceptors is required for ecient performance of learning dators, recognition of danger associated with heights or
and memory tasks in rats [67,72], suggesting that open spaces) (Table 1). The reactive-anticipatory
glucocorticoids may set the tone for information pro- distinction is experience dependent; the environment
cessing in the brain. Control of this rhythmic activity is associated with a reactive stressor can be itself condi-
coordinated by inputs from the suprachiasmatic nucleus tioned, resulting in an anticipatory response when the
[67,72], the critical pacemaker of numerous bodily conditioned stimuli are next encountered.
rhythms. The mnemonic aspects of anticipatory stressors are
The second domain of HPA action, and the principal important determinants of the HPA response. As no-
topic of this review, is control of corticosteroid secretion ted above, the HPA response is energetically costly
following stress. Upon receipt of a stressful stimulus, and cannot be over-engaged without deleterious con-
dened here as a real or predicted threat to homeostasis, sequences [197]. As such, the brain can generate
the brain initiates an ACTH surge to promote adreno- memory-dependent inhibitory and excitatory traces to
cortical activation. The notion of real or predicted is control glucocorticoid responses. For example, mne-
important, as it highlights what we hypothesize are two monic circuits can diminish responsiveness to contex-
distinct realms of stress activation. A real stressor tual stimuli with repeated exposure (habituation), or
represents a genuine homeostatic challenge that is rec- activate responses to innocuous cues that are associ-
ognized by somatic, visceral or circumventricular sen- ated with an emergent threat. The wide spectrum of
sory pathways. These stressors would include such these responses is under exquisite control by limbic
things as marked changes in cardiovascular tone, re- brain regions such as the hippocampus, amygdala, and
spiratory distress, visceral or somatic pain, and blood- prefrontal cortex.
borne cytokine or chemokine factors signaling infection The remainder of this review will be devoted to de-
or inammation (see Table 1). As these represent a re- lineating critical circuits responsible for regulation of the
sponse of the body to a very real sensory stimulus, we HPA stress response, and developing a framework for
consider these responses to be reactive. However, HPA understanding the possible role of these hierarchical
activation can also occur in the absence of primary circuits in stress-related disease states.

Table 1
Stimuli triggering reactive vs. anticipatory HPA stress responses
Reactive responses Anticipatory responses
Pain Innate Programs
Visceral Predators
Somatic Unfamiliar environments/situations
Neuronal homeostatic signals Social challenges
Chemoreceptor stimulation Species-specic threats (e.g., illuminated spaces for rodents, dark spaces for humans)
Baroreceptor stimulation Memory programs
Osmoreceptor stimulation Classically conditioned stimuli
Humoral homeostatic signals Contextually conditioned stimuli
Glucose Negative reinforcement/frustration
Leptin
Insulin
Reninangiotensin
Atrial natriuretic peptide
Others
Humoral inammatory signals
IL-1
IL-6
TNF-a
Others
154 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 155

2. Direct PVN connections modality, indicating that NE release is not the sole de-
terminant of the stress response [221].
The medial parvocellular PVN is in receipt of syn- However, the direct involvement of NE/E pathways
aptic innervation from a relatively circumspect set of in PVN activation is not without some degree of con-
central nervous system structures, summarized in Fig. 2. troversy. First, pharmacological studies indicate that
In general, PVN projecting neurons are localized in re- high levels of NE may have inhibitory eects on ACTH
gions known to receive rst- or second-order inputs release mediated by b adrenergic receptors [230]. Sec-
from somatic nociceptors, visceral aerents or humoral ond, electrophysiologicial studies indicate that eects of
sensory pathways. As such, the majority of PVN-pro- NE on activity of parvocellular neurosecretory neurons
jecting neurons are positioned to evoke rapid, reexive can be blocked with tetrodotoxin or glutamate receptor
activation of the HPA axis. antagonists [60], suggesting that NE eects are mediated
by glutamatergic interneurons, rather than acting on
2.1. Nucleus of the solitary tract CRH neurons directly. Finally, a recent study suggests
that stimuli that sensitize HPA stress responses reduce
Brainstem catecholamine systems play a major role in NE/E innervation of the parvocellular PVN [138], sug-
excitation of the HPA axis. The PVN receives its major gesting that enhanced excitability is accompanied by
norepinephrine (NE) and epinephrine (E) input from the withdrawal of catecholaminergic inputs.
A2/C2 region, originating in the area of the nucleus of Tyrosine hydroxylase-c-fos colocalization studies re-
the solitary tract (NTS). Fibers from these regions veal that catecholaminergic cells represent only a subset
preferentially innervate the medial parvocellular zone of of stress-activated, PVN-projecting NTS neurons
the PVN, at the expense of magnocellular neurosecre- [31,46,66,223]. Recent evidence indicates that these non-
tory and parvocellular preautonomic neurons, which are catecholaminergic NTS neurons modulate HPA axis
innervated by the A1/C1 groups in the rostral ventro- activity. For example, glucagon-like peptide 1 (GLP-1),
lateral medulla [58,59]. General consensus indicates that which is richly expressed in medial parvocellular PVN
the NE/E input represents a major HPA excitatory terminals, is only synthesized in non-tyrosine hydroxy-
pathway, promoting CRH and ACTH release and CRH lase (i.e., non-catecholaminergic) neurons of the NTS
gene transcription through activation of a1 adrenergic and its immediate environs [177,204]. Exposure to lith-
receptors [230,231,294]. The excitatory nature of NE/E ium chloride, which produces visceral illness and an
inputs is further supported by evidence documenting attendant HPA response, activates NTS GLP-1 neurons
stress-induced NE release in the PVN, and by studies [240]. Importantly, ACTH and corticosterone responses
indicating that reactive HPA responses can be attenu- to lithium chloride can be inhibited by central admin-
ated by lesions of ascending medullary PVN inputs or istration of the GLP-1 receptor antagonist des-His1 ,
by selective lesions of catecholaminergic pathways using Glu9 -exendin-4, indicating that endogenous GLP-1 of
6-hydroxydopamine [108,183,266]. Finally, microdialy- NTS origin inhibits HPA activation by this stimulus
sis studies indicate that a variety of stressors directly [154]. In addition, ACTH release can be induced by
promote norepinephrine release in the PVN region [221]. direct injections of GLP-1 into the PVN (but not
The latter studies indicate that the relationship between amygdala) [154], consistent with direct GLP-1 actions
NE release and HPA activation varies with stimulus on PVN neurons.

b
Fig. 2. Major direct projections to a prototypical medial parvocellular PVN neuron. The PVN receives direct innervation from several extrahy-
pothalamic pathways regulating homeostatic functions, including: (1) the subfornical organ (SFO)-median preoptic nucleus (mnPOA)-organum
vasculosum of the lamine terminalis (OVLT), regulating uid and electrolyte balance; (2) norepinephrine (NE), epinephrine (E), glucagon-like
peptide 1 (GLP-1) and other neuropeptidergic neurons in the nucleus of the solitary tract (NTS) and parabrachial nucleus (PBN), subserving relay of
autonomic and immune system aerents; and (3) hypothalamic nuclei subserving autonomic/metabolic/immune/arousal signals, including the
dorsomedial hypothalamus (DMH), medial preoptic area (mPOA), lateral hypothalamic area (LHA), arcuate nucleus (ARC), peri-PVN zone,
anterior hypothalamic nucleus (AHN) and ventral premammillary nucleus (PMV), among others. Some of these projections are largely GABAergic
(open circles), some are predominantly glutamatergic (closed circles), whereas as others contain mixed populations of cells. The bed nucleus of the
stria terminalis (BST) is predominantly GABAergic, suggesting a largely inhibitory inuence on the PVN.

Fig. 7. Hierarchical limbic projections to reactive stress pathways. The medial prefrontal cortex (mPFC), central amygdaloid nucleus (CeA), ventral
subiculum (vSUB), medial amygdaloid nucleus (MeA), and lateral septum (LS) are limbic system regions known to aect HPA axis activation. None
of these regions send direct projections to the PVN; however, all project to select brainstem, hypothalamic, and BST regions that in turn innervate the
medial parvocellular PVN. Limbic structures subserving anticipatory stress responses can therefore modulate PVN activation through interactions
with reactive stress circuits. Superimpostion of limbic input onto brainstem and hypothalamic stress eectors forms a hierarchical system that uses
direct PVN projections to mediate both reexive (reactive) and voluntary (anticipatory) stress responses. Note that the projections of these limbic
regions have considerable, but incomplete overlap with one another, implying that the net impact of the stress response is dependent on the ensemble
activity of these structures. Symbols and other abbreviations are dened in the legend of Fig. 2; grayed-in details summarizes intrinsic PVN reg-
ulatory mechanisms, noted in Fig. 6.
156 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

In addition to GLP-1, non-aminergic PVN-projecting prefrontal cortex, central amygdaloid nucleus, and sev-
NTS neurons may also synthesize additional neurop- eral hypothalamic nuclei [273,308]. As such, the NTS
eptidergic species, including somatostatin, substance P, may not only serve as a relay for reexive inputs, but
and enkephalin [267,268]. Of these, substance P is known may respond to descending information as well; these
to inhibit stress-induced HPA activation [141,175,259], connections likely subserve the inuence of this cell
whereas enkephalin analogs can promote corticosterone group on anticipatory stress responses.
release [136]. However, all the above peptides are pro-
duced in other PVN-projecting regions as well, and it is 2.2. Raphe nuclei
not completely clear whether specic NTS peptidergic
populations specically modulate PVN activitation. Serotonergic systems are known to modulate HPA
The NTS appears to be a common site for integra- axis activity. The majority of studies indicate that
tion of reactive HPA responses. The NTS shows c-fos serotonin (5-HT) stimulates ACTH and corticosterone
activation following most stressors we classify as re- secretion by way of PVN 5HT2A and perhaps 5HT1A
active, including visceral illness (lithium chloride) receptors [225,305] (although the latter is somewhat
[170,240], cytokine/inammatory challenge (e.g., inter- controversial [318]). In addition, depletion of serotonin
leukin 1-b or lipopolysaccharide injection) [83,168], inhibits c-fos induction and CRH heteronuclear RNA
hypovolemia [302], hypoxia [297], and hypotension expression in the PVN following lipopolysaccharide in-
[167]). These responses are in keeping with the known jection [169]. Given this stimulatory role, it is somewhat
role of the NTS in relaying reexive or sensorial in- surprising that direct serotonin input to the PVN is
formation to the PVN and other forebrain structures limited to a small number of bers derived from the
[299]. However, the NTS is also activated by numerous dorsal and median raphe nucleus [272]. Indeed, the vast
stressors having mixed reactive and anticipatory attri- majority of serotonin positive bers are seen in the im-
butes, such as restraint, swim, and immobilization mediate PVN surround, raising the possibility for
[56,270], and can be activated during conditioning prominent interactions with GABAergic neurons occu-
paradigms [228], suggesting that the NTS also plays a pying the PVN shell (see below). Given the potential for
role in anticipatory stress integration. This hypothesis is injected pharmacological agents to spread to these local
supported by recent studies indicating that central in- interneurons, the exact locus of serotonergic involve-
jection of a GLP-1 receptor antagonist (des-His1 , Glu9 - ment in stress integration is dicult to pinpoint.
exendin-4) blocks ACTH and corticosterone release Lesion studies lend further support for a stimulatory
following elevated plus maze exposure [154]. As noted role of 5-HT in HPA axis regulation. Lesions of the
above, GLP-1 is only expressed in the NTS; thus, these raphe nuclei (either electrolytic or neurochemical (5,7-
data indicate that ascending NTS input is required for dihydroxytryptamine (DHT)) elicit decreased HPA re-
optimal elaboration of internally generated, anticipa- sponses to restraint stress [143], photic stimuli [87], local
tory HPA axis responses. PVN glutamate administration [93], and stimulation of
The role of the NTS in the reactive or reexive stress the dorsal hippocampus or central amygdaloid nucleus
pathway may be related to its position as a relay for [87,94]. The latter study demonstrates that local PVN
both visceral and somatic sensory information. For the injections of 5,7-DHT can block stimulation-induced
former, it is clear that the NTS is in receipt of ascending HPA activation, indicating that the eects of 5-HT may
cardiovascular aerents and plays a major role in the be exerted directly at the PVN and/or its surround.
baroreceptor reex [299]. The NTS also receives inputs Notably, 5-HT heavily innervates forebrain stress-
from brainstem regions integrally involved in respiration integrative structures, including the hippocampus, pre-
and from rostroventral medullary neurons controlling frontal cortex, amygdala, and hypothalamus (see [186]).
autonomic outow [299]. Notably, somatic pain stimuli Thus, in addition to direct actions at the PVN, systemic
also appear to activate PVN NE release via ascending or intracerebroventricular injections of serotonergic
NTS neurons, presumably via direct or indirect input drugs may also inuence HPA activity indirectly. For
from the spinoreticular [224]. In addition, the NTS is example, administration of 5HT2 receptor antagonist
densely innervated by the area postrema [299], which is a into the amygdala inhibits the ACTH response to photic
bloodbrain barrier decient region that is at least in stress [90], suggesting that 5-HT may also modulate
part responsible for cytokine-initiated HPA axis acti- HPA axis activity via upstream stress eectors.
vation [83]. There is also evidence for a role of the local The PVN is also in receipt of information from as-
NTS microvasculature in HPA integration, as studies cending pontine and midbrain pathways relevant to re-
indicate that vessels in this region can activate neurons exive stress integration. These include the parabrachial
through local synthesis of prostaglandins [82], as noted nucleus and periaqueductal gray, which are intimately
above. involved in autonomic function [255]. Evidence sup-
Importantly, the NTS also receives aerents from porting a role for the parabrachial nucleus in stress re-
limbic forebrain stress circuits, including the medial sponses centers primarily on several tract tracing studies
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 157

establishing widespread connections with many tha- decient subfornical organ (see [104]). The subfornical
lamic nuclei (including paraventricular thalamic nu- organ is also enriched in atrial natriuretic peptide and its
cleus), hypothalamic nuclei (including PVN), amygdala, cognate receptor (natriuretic peptide receptor-A)
bed nucleus of the stria terminalis (BST), NTS, ventro- [124,173], suggesting that other osmoregulatory peptides
lateral medulla, raphe nuclei, and periaqueductal gray may also signal via this mechanism.
[165,166,256]. The ventrolateral periaqueductal gray
sends projections to the medial parvocellular PVN [105], 2.4. Hypothalamus
consistent with a role in endocrine integration, and this
region shows c-fos induction in numerous stress para- The PVN receives massive input from numerous re-
digms [56,205,270]. While the parabrachial nucleus and gions of the hypothalamus. Dual-label tract tracing
ventrolateral periaqueductal gray relay information on studies indicate that the lions share of this input ema-
cardiovascular tone and pain perception to the PVN, the nates from GABAergic neurons [57,246]. Parvocellular
precise role of these structures in HPA integration re- CRH neurons are directly innervated by GABAergic
mains to be determined. boutons (Fig. 3) and express multiple GABA-A receptor
subunits [54], consistent with the importance of GABA
2.3. Subfornical organ/lamina terminalis system in central HPA integration. Notably, PVN injection of
the GABA-A agonist muscimol inhibits corticosterone
The PVN is in direct receipt of information on uid secretion following restraint [53], validating the physio-
and electrolyte status by way of the subfornical organ/ logical relevance of GABA in PVN stress integration.
lamina terminalis system. Signals are relayed through The PVN is richly innervated by GABAergic neurons
direct projections of the subfornical organ, median located in its immediate proximity [23,57,246]. Numer-
preoptic nucleus, and the organum vasculosum of the ous studies indicate a prominent role for these neurons
lamina terminalis to medial parvocellular PVN neurons in stress integration. For example, acute stress exposure
[17,271]. These regions are critically involved in control induces c-fos expression in the peri-PVN region [122],
of osmoregulation and drinking behavior, as lesions while GAD65 mRNA is up-regulated in these neurons
along this pathway can result in adipsia and dehydration following chronic stress exposure [24]. Importantly, the
[142]. Notably, like the area postrema, both the sub- peri-PVN region preferentially expresses select kainate-
fornical organ and organum vasculosum of the lamina preferring glutamate receptor subunits [125], and injec-
terminalis are bloodbrain barrier decient regions tion of a pan-ionotropic glutamate receptor antagonist
[215], providing another mechanism by which blood- (kynurenic acid) into this region exacerbates cortico-
borne signals may be communicated to the PVN. Ac- sterone responses to restraint [330]. In addition, micro-
cordingly, previous work indicates that angiotensinergic injection of glutamate in this region induces c-fos
neurons of the subfornical organ directly activate CRH expression in GAD neurons present in the immediate
secretion [232]. This response may itself be mediated by surround of the PVN (peri-PVN zone), but does not
direct interaction of circulating angiotensin II with an- activate hypophysiotrophic PVN neurons [50]. Thus,
giotensin II-receptive neurons in the bloodbrain barrier the peri-PVN appears highly receptive to glutamatergic

Fig. 3. Confocal microscopic images illustrating direct innervation of PVN CRH neurons by glutamate (vesicular glutamate transporter 2 immu-
noreactivity, VGlut2) (A,B) and GABA (glutamic acid decarboxylase immunoreactivity) (GAD) (C). (A) Image from a single 0.5 lm section, de-
picting VGlut2 immunoreactive boutons (green) in apparent apposition to individual CRH neurons (red). (B) Three-dimensional reconstruction of
VGlut2 bouton appositions to CRH neurons, using image stacks merged and rotated (Volocity software). Note close appositions of boutons to cell
somata and dendrites. (C) Processed three-dimensional image (Volocity software), illustrating GAD bouton appositions to a CRH neuron. The
processed image employs an algorithm that determines overlap of red- and green-channel uorescence in three-dimensional space. The indicated
boutons are those that have >10 voxel overlap with the CRH immunoreactive cell.
158 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

inputs, and aords a means whereby excitatory inputs to glutamate transporter 2 (VGlut2)-immunoreactive ter-
the peri-PVN region can elicit inhibition of the HPA minals (Figs. 3A and B). VGlut2 is a specic marker for
axis. glutamatergic cell populations, and is highly expressed in
Medial parvocellular PVN neurons are also richly a number of hypothalamic PVN-projecting regions, in-
innervated by GABAergic cell populations from the cluding neurons scattered throughout the dorsomedial
dorsomedial hypothalamic nucleus [57]. GABAergic hypothalamic nucleus (Fig. 5) [329]. In addition, local
neurons in the dorsomedial nucleus (particularly its infusion of the ionotropic glutamate receptor antagonist
ventrolateral part) show c-fos activation following swim kynurenic acid directly into the medial parvocellular PVN
stress ([55], Fig. 4A), consistent with a role in inhibition inhibits ACTH and corticosterone responses to stress,
of the stress response. Accordingly, lesions of this region consistent with an excitatory action of PVN glutamate
exacerbate HPA axis responses to stress [14,312], release on CRH secretion [330]. These results complement
whereas pharmacological stimulation of neurons in the previous studies showing that systemic injection of a
dorsomedial nucleus generates GABAergic inhibitory NMDA and AMPA/kainate receptor antagonist cocktail
post-synaptic potentials in PVN neurosecretory neurons inhibits ACTH responses to immobilization, but not
[23]. Recent data from our laboratory indicate that local footshock or ether [326]. Importantly, NMDA and kai-
infusion of kynurenic acid into this region prolongs nate receptor subunits are indeed expressed in the par-
stress-induced glucocorticoid secretion (Fig. 4B), indi- vocellular PVN [8,125], and are regulated by acute and
cating that blockade of glutamate in PVN-projecting chronic stressors [13], consistent with a role in processing
dorsomedial nucleus neurons disinhibits the HPA axis. of stressful information.
However, it should be noted that the role of the While the dorsomedial nucleus contains VGlut2
dorsomedial nucleus in HPA integration may be subre- neurons and projects to the PVN, combined tract-trac-
gion or cell-type specic; for example, data from Taskers ing/in situ hybridization data conrming PVN-project-
group indicate that this region supplies excitatory as well ing glutamate cells in the dorsomedial nucleus or other
as inhibitory input to PVN neurons [295], and glutamate hypothalamic regions has yet to be advanced. Alterna-
stimulation of neurons in the dorsal extent of the dorso- tive attempts to demonstrate this projection have been
medial nucleus promote rather than inhibit ACTH se- made, but have not allowed denitive conclusions to be
cretion [11]. Electrophysiological studies indicate that drawn. For example, light-microscopic glutamate im-
intrahypothalamic excitation of the PVN by the dorso- munohistochemistry has proved unsuitable in allowing
medial nucleus (as well as other local PVN projecting conrmation of a glutamatergic phenotype, and the
regions) involves glutamatergic neurotransmission [22]. method of retrograde transport of [3 H]D -aspartate [52]
Electron microscopic studies have demonstrated the has several limitations, including the diculty in limiting
presence of glutamate-immunoreactive synapses on par- injections to the PVN proper, as well as questions con-
vocellular PVN neurons [69], and our recent studies in- cerning its ability to specically mark glutamatergic
dicate direct innervation of CRH neurons by vesicular neuronal populations.

Fig. 4. Dorsomedial hypothalamic nucleus (DMH) involvement in stress integration. (A) Dual in situ hybridization image, showing co-localization of
GAD65 mRNA (dark cell bodies) with c-fos mRNA (white grains) in the ventromedial DMH following 30 min of swim stress. (B) Inhibition of
DMH ionotropic glutamate receptors by microinjection of kynurenic acid (Kyn) prolongs corticosterone (CORT) secretion following restraint stress,
consistent with loss of glutamate excitation to neurons inhibiting the PVN.
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 159

Fig. 5. Mixed populations of GABA and glutamate neurons are present in the dorsomedial hypothalamic nucleus (DMH) and preoptic area (POA).
In situ hybridization indicates that both GABA (glutamic acid decarboxylase 67 mRNA-expressing) and glutamate (VGlut2 mRNA-expressing)
neurons are present in the DMH (AC) and POA (DF). Note that GAD67 and VGlut2 populations are extensively intermingled in both regions;
however, dual in situ hybridization does not reveal VGlut2 associated grains over dark GAD67-positive neurons (C,F), indicating that these rep-
resent two distinct cell populations.

Notably, the dorsomedial nucleus possesses mutually high levels of androgen, estrogen receptor a, estrogen
exclusive populations of GABA and glutamate neurons receptor b, and progesterone receptors [9,114,279,282],
([329], Figs. 5AC). As such, this nucleus may supply and is a major target for gonadal steroid modulation of
both inhibitory and excitatory projections to the par- reproductive function. While it is not known whether
vocellular PVN, perhaps depending on which sets of steroid-receptive cells project to the PVN, it is clear that
neurons are activated by an incoming stimulus and/or the HPA axis is keenly sensitive to gonadal steroids.
the sequence/timing of synaptic activity. Early studies showed that female rats display HPA axis
The medial preoptic area also sends stress-activated, hyperactivity relative to males, predominantly during
GABAergic projections to the PVN [55]. Lesions of this proestrus [41,51,178], a phenomenon that was linked
region enhance stress-induced ACTH secretion [312], with variations in gonadal hormone levels [156,157].
whereas stimulation can block the excitatory eects of Indeed, administration of estrogen to ovariectomized
medial amygdalar stimulation on corticosterone release females enhances corticosterone secretion following
in anesthetized rats [88]. Notably, electrophysiological stress [41,100,311]. The increase in stress-induced corti-
studies suggest dierential involvement of preoptic area costerone release is not normalized by corticosterone-
subregions in PVN regulation; stimulation of the medial binding globulin [109], verifying that females have
portion, including the highly GABAergic medial pre- greater free corticosterone levels as well. In contrast,
optic nucleus [213], decreases PVN neuronal activity, testosterone inhibits HPA responses to stress when in-
whereas stimulation of more laterally situated cell jected systemically [310] or directly into the preoptic
groups increases ring rate [257]. As was the case for the area [312], consistent with an opposite role for andro-
dorsomedial nucleus, the lateral regions of the medial gens in PVN regulation.
preoptic area contain intermingled GABA and gluta- The importance of PVN gonadal steroid-receptive
mate neurons (Figs. 5DF). regions such as the medial preoptic area and perhaps
The medial preoptic area stands out as a potential the bed nucleus of the stria terminalis is highlighted by
interface between gonadal steroids and the HPA axis. the dearth of gonadal steroid receptor expression in the
This region, along with subdivisions of the bed nucleus medial parvocellular PVN proper. Thus, although CRH
of the stria terminalis (see below), express extremely and arginine vasopressin (AVP) mRNA levels in the
160 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

PVN are elevated in proestrus [20,283] and in estrogen- The PVN also receives input from nuclei of the
replaced OVX animals [227,249], these eects are likely posterior hypothalamus, with the densest innervation
mediated by transynaptic mechanisms, as the CRH se- supplied by the ventral premammillary nucleus. The
creting PVN neurons do not express substantial levels of function of these regions is ill-dened with respect to
estrogen receptor a or b (the latter receptor is, however, either reactive or anticipatory stress responses; how-
expressed in magnocellular oxytocin and vasopressin ever, it should be noted that the mammillary nuclei all
neurons [5,130]). Similarly, PVN androgen receptor ex- receive considerable input from limbic forebrain
pression appears to be limited to spinal cord and structures [235], and are thus in good position to relay
brainstem projecting cell groups [327], suggesting that stress-related information from these structures to the
testosterone acts transsynaptically as well [116]. Ex- PVN.
pression of progesterone receptor in the PVN is limited
to isolated cells in rat [107]. Thus, in all cases it appears 2.5. Bed nucleus of the stria terminalis
likely that centrally mediated eects of gonadal steroids
on the HPA axis are mediated predominantly by aerent Direct non-hypothalamic forebrain inputs to the
projection systems, of which the medial preoptic area parvocellular PVN are largely conned to discrete sub-
and related PVN projecting structures (BST, antero- nuclei of the bed nucleus of the stria terminalis
ventral periventricular nucleus) are prime candidates. (BST) and the neighboring parastrial nucleus [271,290].
Signals relevant to energy balance may be transmitted The densest PVN projections emanate from the intra-
by direct innervation of PVN neurons by the arcuate fascicular, transverse, and anterodorsal nuclei of Ju
nucleus and perhaps the lateral hypothalamus. Arcuate and Swanson [144], which contain predominantly
nucleus neurons are sensitive to circulating glucose, lep- GABAergic neurons [57,145]. As such, like hypotha-
tin, and insulin [322]. Neuropeptidergic systems resident lamic inputs, the majority of these BST inputs to the
in the arcuate nucleus have complementary roles in food PVN appear to be inhibitory. In addition, the PVN is
intake; neuropeptide Y and agouti related peptide also innervated by ventrolateral regions of the BST,
(AGRP) neurons promote food intake, whereas proopi- including the fusiform nucleus [76]. This pathway may
omelanocortin peptides have anorectic actions [322]. be of importance in relaying information from the cen-
Hypothalamic neuropeptide Y (NPY) and AGRP infu- tral amygdaloid nucleus to the PVN.
sions increase HPA axis activation [71,181,315] and The valence of BST action on HPA secretory activity
stimulate CRH release [71]. However, injections of appears to be location-specic. Our group has shown
a-melunocyte stimulating hormone (a-MSH) also elevate that lesions of the posterior BST, which include the in-
ACTH and corticosterone secretion, induce rapid cyclic trafascicular and transverse subnuclei, enhance expres-
AMP response element binding protein phosphorylation sion of CRH mRNA in the PVN, consistent with an
in CRH neurons [264] and enhance CRH release in vitro inhibitory role in HPA regulation [120]. In contrast,
[71]. These data indicate that the HPA axis is activated by lesions of the anterior BST, including the anterodorsal
negative as well as positive energy balance. Thus, both and fusiform subnuclei, decrease CRH mRNA expres-
signals may be interpreted as stressors by the organism. sion [120]. These results largely agree with studies of
Given the largely excitatory actions of both the NPY Gray et al. [113], which demonstrate an excitatory role
and a-MSH systems, it is somewhat surprising to note for anterolateral BST regions in conditioned cortico-
that neonatal monosodium glutamate lesions of the sterone responses. In addition, stimulation of anterior
arcuate, which destroy both systems [2,152], enhance BST structures, including PVN projecting regions in the
both basal and stress-induced corticosterone release anterodorsal and fusiform subnuclei, increase cortico-
[176,189]. These data suggest a role for the arcuate nu- sterone secretion, whereas stimulation in regions corre-
cleus in HPA inhibition, which is not in keeping with the sponding to the intrafascicular and transverse subnuclei
known role of its PVN-projecting peptidergic subpop- reduce circulating glucocorticoids [78]. Importantly, the
ulations. However, it is important to note that lesions anterolateral BST is positioned to modulate behavioral
performed early in postnatal development may trigger stress responses as well, as restraint stress and lateral
compensation by other regulatory pathways. BST stimulation produce similar changes in stress-re-
Tracing studies indicate that a sizable proportion of lated behaviors (home cage immobility, aggressive be-
PVN-projecting arcuate nucleus neurons are GABAer- havior) [42].
gic [12]. These GABAergic neurons tend to be concen- Like the medial preoptic area, the principle encap-
trated in the medial region of the nucleus, corresponding sulated, intrafascicular and transverse subnuclei ro-
with the location of NPY/AGRP neurons [12], and at bustly express gonadal steroid receptors [279,282]. Thus,
least a subpopulation of these neurons co-express eects of gonadal steroids on the HPA axis may be also
GABA and NPY [236]. However, the specic role of be modulated by receptive neurons in these regions.
GABA in arcuate nucleus control of HPA axis activa- Analysis of the respective roles of these BST regions in
tion has yet to be evaluated. HPA integration remains to be evaluated.
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 161

As several subdivisions of the BST are heavily pop- 3. Intrinsic PVN information processing
ulated with CRH neurons [145], it is important to
comment on the potential involvement of CRH neurons The PVN is well positioned to receive direct input
in regulation of the HPA axis. Several lines of evidence from blood- and CSF-borne factors (Fig. 6). This nu-
implicate CRH as a positive modulator of PVN stress cleus is endowed with a dense capillary plexus [200,307];
responses. Electron microscopic studies indicate the while there is no evidence that these vessels are fenes-
presence of axodendritic and axosomatic CRH-positive trated, it is clear that this plexus can allow ready access
contacts on PVN CRH neurons [185,281], and it is well of bloodbrain barrier permeable factors, including
established that intracerebroventricular injections of steroid hormones. Indeed, there is ample evidence that
CRH can promote immediate early gene induction glucocorticoids exert local feedback eects in the region
[133,226] and CRH gene transcription [226] in medial of the PVN, inhibiting CRH gene expression and adre-
parvocellular PVN neurons. While CRH receptor ex- nalectomy-induced ACTH hypersecretion [163,265].
pression in the PVN is minimal, CRHR1 expression is This capacity for local action suggests that enhanced
rapidly potentiated in response to either stress or CRH vascularity may play a role in direct feedback processes.
[132,191], implying a possible role in prolonged or In addition, aldosterone and gonadal steroid hormones
chronic stress conditions. The fusiform nucleus of the also pass readily through the blood-brain barrier, and as
BST is known to send a CRHergic projection to the noted above, there is evidence for androgen and estro-
PVN [76], and is a possible candidate for BST-mediated gen receptors in the PVN, though not necessarily in
excitation of the PVN. However, as was the case for medial parvocellular neurons [116,279,282]. Finally, re-
glutamate, CRH innervation of the PVN may emanate cent data indicate that brain microvessels may play a
from several regions, including several hypothalamic major role in communicating cytokine signals to the
nuclei (perifornical, dorsal hypothalamic nucleus, and CNS, through activation of prostaglandin biosynthesis
dorsal hypothalamic area), dorsal raphe or Barringtons [243]. Notably, the medial parvocellular PVN richly
nucleus [45]. In addition, it is also possible that axo- expresses prostaglandin receptors [243], suggesting a
dendritic CRHCRH contacts may be axon collaterals capacity for local integration of immune signals within
of PVN neurons [185]. the PVN proper.
Dendrites of PVN neurons may also contact the ce-
2.6. Thalamus rebrospinal uid under some conditions. For example,
studies performed in sh, amphibians, and reptiles re-
Recent evidence suggests that the PVN may receive veal that dendrites of PVN neurons contact the third
inputs from thalamic sensory nuclei as well. Notably, ventricle [313]. Such contacts are not observed in normal
neurons in auditory-responsive posterior thalamic mammalian hypothalami [313]. However, axonal pro-
nuclei, including the subparafascicular and posterior jections to the ventricular wall are noted in long-term
intralaminar regions, send direct projections to the dehydrated rats [70], suggesting that neuronal contacts
medial parvocellular PVN [36], and are likely involved with the ventricular system may be physiologically reg-
in pathways mediating activation of the HPA axis by ulated. The signicance of these ndings remains to be
audiogenic stressors [33,35]. These regions are known evaluated.
to be responsive to cardiovascular stimuli and may The intrinsic organization of the PVN is positioned to
play a role in noise-induced sympathetic activation allow considerable cross-talk among hypophysial, neu-
[180]. It remains to be determined whether other rohypophysial, and preautonomic cell populations.
sensory modalities (e.g., somatosensory/pain input via Morphological studies using Golgi impregnation or
the spinothalamic pathway [49]) can connect with intracellular lls indicate that the dendrites of PVN
the PVN through the same or parallel thalamic cir- neurons ramify freely among various PVN subnuclei
cuitry. [239,307]. Notably, previous EM studies demonstrate
The parvocellular PVN is also innervated by neurons CRH synapses on non-CRH containing PVN dendrites
of the anteromedial zona incerta [271,314], many of [185,281], consistent with anatomical interactions
which are dopaminergic [47]. Zona incerta neurons show among the various PVN cell phenotypes. In addition to
minimal stress-induced c-fos expression, but do show axosomatic interactions, there is evidence for dendritic
rapid induction of other immediate early genes (e.g., release of neurohypophysial peptides [188], indicating an
NGFI-A) following restraint or swim [56], suggesting additional avenue for intranuclear communication. Fi-
that activity of these neurons is modulated during stress nally, recent studies indicate that PVN neurons, partic-
exposure. Dopamine is known to activate the HPA axis ularly those in magnocellular subdivisions, contain
[21]; however, it is not known whether eects are exerted neuronal nitric oxide synthase and heme oxygenases 1
directly at the PVN (possibly via local zona incerta and 2 [244,245,324]. These neurons may therefore
projections) or via actions on upstream PVN projec- modulate medial parvocellular activation through re-
tions. lease of nitric oxide and carbon monoxide. Consistent
162 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

Fig. 6. Intrinsic PVN-regulatory pathways. PVN neurons may be inuenced by several processes eected within the PVN proper. First (1) the PVN
contains a dense capillary plexus, aording ready access to steroid hormones, such as glucocorticoids (GC). Second (2) PVN capillaries also syn-
thesize prostaglandin E2 , which can interact with receptors on parvocellular PVN neurons to provide a mechanism for local cytokine signaling. Third
(3) though dendrites do not extend outside the nucleus, they ramify extensively within the nucleus, aording interaction with other cell populations.
Fourth (4) neurons within the parvocellular and/or magnocellular subdivisions contain the cellular machinery to synthesize the gaseous neuro-
transmitters nitric oxide (NO) and carbon monoxide (CO), both of which aect HPA axis activation. Fifth (5) there is evidence for local lateral
innervation of parvocellular neurons by intra-PVN axons, suggesting an opportunity for synaptic interactions within and across PVN cell groups.
Finally (6) terminals in the median eminence have access to blood-borne and glial constituents that can be retrogradely transported to aect growth
and plasticity.

with this hypothesis, studies from Riviers laboratory PVN neurons have a limited capacity to sample the -
indicate that nitric oxide promotes synthesis of CRH ber-rich peri-PVN region directly. The importance of
and AVP in parvocellular neurons, suggesting an acti- this distinction cannot be overestimated, as a wealth of
vational role within the PVN. However, additional data bers from limbic forebrain structures terminate (see
suggest that nitric oxide can inhibit ACTH release to below) in this peri-PVN zone.
mild stressors, perhaps at the level of the pituitary; thus, Finally, HPA activation by medial parvocellular
the net eect on the HPA axis may depend on the locus CRH neurons may be modulated by non-neuronal fac-
of nitric oxide biosynthesis [244,245]. tors. For example, the median eminence is a blood
Morphometric analyses indicate that dendrites of brain barrien decient region, and as such may allow
PVN axons are largely contained within the PVN proper blood-borne factors access to PVN terminal arbors. In
[239,307]. While there is evidence for contacts between addition, specialized glial elements in the hypothalamus
limbic aerents and dendrites of magnocellular neurons and median eminence produce receptors for growth-as-
in the peri-PVN zone [214], the extent of extra-PVN sociated neuregulin molecules and insulin-like growth
dendritic ramications is quite limited, and has not been factor-1, which may promote neurite outgrowth and
demonstrated for CRH-containing neurons. As such, thus aect secretory potential [98,212,233].
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 163

4. Indirect paths to the PVN iting responses to anticipatory stressors, as lesion of this
region enhances responsiveness to restraint and open
Previous work reveals rich interactions between lim- eld exposure, but not to ether vapors [119,123,209].
bic brain structures and HPA activation. These struc- This region projects extensively to BST and hypotha-
tures, including regions such as the hippocampus, lamic neurons relaying information to the PVN, as well
prefrontal cortex, amygdala, septum, and midline thal- as to the peri-PVN zone [57], and as such are well-po-
amus, are critical for emotional responses and memory, sitioned to mediate hippocampal HPA inhibition.
and are thus logical candidates for modulating pitui- Overall, the observation that hippocampal damage
taryadrenal secretions with respect to previous experi- can delay termination of corticosterone release is con-
ence. As such, these structures likely play a major role in sistent with a role in glucocorticoid negative feedback
anticipatory stress responses. Importantly, all of the inhibition of the HPA axis (reviewed in [137,261]). This
above regions have very limited opportunities to interact hypothesis is supported by rich localization of gluco-
directly with medial parvocellular PVN neurons, re- corticoid receptors in the hippocampus [199,238], and by
quiring intermediary neurons to relay their inuence on lesion studies demonstrating that (1) hippocampal
ACTH secretagog release (see Fig. 7). damage can reduce the ability of dexamethasone to in-
hibit corticosterone release following ether stress [190]
4.1. Hippocampus or photic stimulation [92], and (2) mbriafornix lesions
inhibit the ability of dexamethasone to block hypoten-
Numerous studies indicate that hippocampal lesions sion-induced CRH and AVP release into hypophysial
promote basal hypersecretion of glucocorticoids portal blood [260]. Implants of glucocorticoids into the
[97,158,159]. Hippocampectomy or fornix transection ventral hippocampus atten the circadian corticosteroid
can increase basal glucocorticoid levels [97,263], pre- rhythm [284], and dorsal hippocampal implants of cor-
sumably by altering circadian rhythms [102,103,206,321]. ticosterone (but not dexamethasone) decrease adrenal-
In addition, hippocampal lesions enhance basal CRH ectomy-induced ACTH release [164]. Hippocampal
and AVP mRNA expression [119,121,127] and elevate implants of glucocorticoid and mineralocorticoid an-
basal AVP (but not CRH) in portal blood [260], con- tagonists attenuate, but do not block HPA responses to
sistent with increased ACTH secretagog capacity in PVN novel photic or acoustic stimuli [95]. However, a com-
neurons. Alterations in secretory patterns may be due to prehensive study from Dallmans group found no eect
loss of mineralocorticoid receptor signaling through this of mbriafornix lesion on glucocorticoid feedback re-
structure, which is believed to play a major role in in- sponses following hypoxia [25], and our group has been
hibiting basal HPA tone during the trough of the diurnal unable to demonstrate that ventral subiculum lesions
corticosterone rhythm [61,67]. However, it should be reverse corticosteroid feedback inhibition of an ACTH
noted that basal HPA hypersecretion following hippo- response to restraint [123]. As such, it is currently un-
campal damage is not observed in all studies clear whether hippocampal inhibition of HPA responses
[25,121,135,174]; inconsistencies among experiments to anticipatory stressors involves glucocorticoid recep-
may be related to time of day or experimental design tor activation and signaling.
issues (see Section 5). Hippocampal stimulation reduces HPA axis activity
The hippocampus is also involved in terminating in human subjects [250], inhibits the circadian rise in
HPA axis responses to stress. Several laboratories have corticosterone in freely moving animals [43], and reduces
noted that hippocampal lesions prolong corticosterone resting corticosterone levels in anesthetized prepara-
and/or ACTH release following exposure to a number of tions [79]. Likewise, dorsal hippocampal stimulation in-
stressors, including restraint [119,261], contextual con- hibits median eminence-projecting PVN neurons [258].
ditioning [148], acoustic stimulation [211], and open eld The eects of hippocampal stimulation appear to be
exposure [123]. However, hippocampal lesions are dependent on subeld, as stimulation of CA1 stimulates,
without eect on HPA axis responses to ether ([123,190]; rather than inhibits corticosterone secretion in anesthe-
however see also [321]) or hypoxia [25], indicating that tized rats [79]. However, it should be noted that much of
involvement of the hippocampus in HPA integration is the stimulation work is complicated by use of anesthesia;
stressor-specic. It is notable that stressors that are for example, studies from the Feldman group indicate
unresponsive to lesions of the hippocampus involve that dorsal hippocampal stimulation can increase corti-
stimuli that rapidly activate the reactive response costerone release in anesthetized rats [85,91], whereas
pathway, indicating they are refractory to hippocampal similar stimulation produces long-lasting inhibition of
inhibition. ACTH and corticosterone secretion following light ex-
Work from our group indicates that hippocampal posure in freely moving animals [96].
inhibition of the HPA axis is mediated by a relatively It is notable that both glucocorticoid and mineralo-
restricted set of neurons in the ventral subiculum corticoid receptors are richly expressed throughout the
[119,123]. These neurons appear responsible for inhib- hippocampus and subiculum [7,117]. Since inhibition of
164 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

the HPA axis represents a minor subset of hippocampal preoptic area are nearly all positive for VGlut1 (Fig. 8),
function, it is clear that the vast majority of hippocampal a specic phenotypic marker for corticolimbic glutamate
steroid receptors have other physiological actions. Given neurons [147,329]. When combined, the tracing data
the well-documented, inverted-U shaped relationship indicate that hippocampal inhibition of the PVN is
between glucocorticoids and memory [67,72], these re- likely mediated by a two-neuron relay, involving con-
ceptors are probably relevant to encoding the signicance tacts between glutamatergic neurons in the subiculum
of stressful stimuli viz. memory consolidation. and GABAergic neurons in the BST and hypothalamus
The impact of the hippocampus on the HPA axis may (Fig. 7).
also be mitigated by up-stream factors mediating hip- The hippocampus may also interact with the PVN by
pocampal function. For example, lesions of the septo- multi-synaptic connections. For example, the lateral
hippocampal cholinergic pathway eectively enhance septum is a major target of eerents from the entire
responses to restraint stress, suggesting that cholinergic septotemporal axis of the hippocampus [292], and in-
tone is critical for normal hippocampal inhibition of nervates numerous PVN-projecting hypothalamic nuclei
HPA stress responsiveness [115]. In addition, the dele- as well as the peri-PVN region [242]. There is also evi-
terious eects of aging on the hippocampal neurons are dence that the inhibitory eects of hippocampal stimu-
highly correlated with prolonged corticosterone re- lation on the HPA axis may be in part relayed through
sponses to restraint stress [134,261], suggesting that loss the brainstem [85,91].
of hippocampal function results in HPA axis hyper-
secretion. 4.2. Prefrontal cortex
Anatomical tracing studies reveal that the principal
hypothalamic outow of the hippocampus originates in The medial prefrontal cortex also modulates HPA
the ventral subiculum and ventral CA1 [37,57,160,203]. axis activation. The medial prefrontal cortex manifests
These neurons densely innervate the peri-PVN zone, robust c-fos induction following numerous stress mo-
but not the PVN proper [39,40,57,242], and send rich dalities (cf. [56,161,193]), and dopamine/norepinephrine
projections to numerous PVN-projecting nuclei in the release is markedly elevated in this region following
BST and hypothalamus [37,57,160,203]. As noted acute or chronic stress [101,140]. Lesion studies indicate
above, parvocellular PVN dendrites show quite limited that bilateral lesions of the anterior cingulate or pre-
ramication outside the nucleus, and thus ventral su- limbic components of the medial prefrontal cortex
biculum terminal boutons likely contact somata and enhance ACTH and corticosterone responses and PVN
processes of neurons populating the PVN surround. c-fos induction following restraint stress, but not ether
The lack of direct innervation indicates that hippo- inhalation [26,73,99]. These data are consistent with the
campal regulation of the PVN is mediated by inter- hypothesis that, like the hippocampus, dorsal subre-
vening neurons. gions of the medial prefrontal cortex selectively inhibit
Combined retrogradeanterograde tracing studies HPA axis responses to anticipatory stressors. Interest-
reveal several regions where ventral subicular eerents ingly, inhibitory eects do not generalize to the entire
contact PVN-projecting cell populations, including the structure; only lesions of ventral regions of the rat pre-
anterodorsal, intrafascicular, and transverse subnuclei frontal cortex (infralimbic cortex) reduce corticosterone
of the BST, medial preoptic area, and to a lesser extent, secretion following restraint [289]. The inuence of
the dorsomedial hypothalamic nucleus [57]. These re- medial prefrontal cortex on stress habituation may be
gions contain rich populations of GABAergic neurons lateralized; bilateral or right prefrontal cortex lesions
[24,213], and indeed the vast majority of PVN-projecting suppress corticosterone secretion, whereas unilateral,
neurons in the BST co-express the GABAergic marker left-sided lesions do not [289]. Unlike the hippocampus,
glutamic acid decarboxylase [57]. There are also sub- dorsal or ventral medial prefrontal cortex lesions have
stantial populations of GABAergic neurons in the peri- no eect on basal morning or evening ACTH and cor-
PVN region [24,213] that may serve to relay inhibitory ticosterone levels [73,99], indicating that the prefrontal
information to hypophysiotrophic PVN neurons. The cortex selectively modulates stress-induced HPA axis
implications of this synaptic arrangement is clear: rather activity.
than projecting directly to CRH neurons, subicular Like the hippocampus, the prefrontal cortex has been
neurons connect with the PVN through at least one in- implicated in negative feedback regulation of the HPA
tervening synapse. axis. Direct implants of corticosterone into the medial
Subicular aerents to PVN-projecting regions are prefrontal region decrease stress-induced ACTH and
predominantly glutamatergic. Neurochemical studies corticosterone secretion following acute or repeated re-
indicate a marked depletion of hypothalamic glutamate straint, but like hippocampal lesions, have no eect on
following mbriafornix lesions [316]. More recent work responses to ether [1,73]. These results are consistent
from our group indicates that ventral subicular neurons with stress- or modality-specic modulation of feedback
innervating PVN-projecting regions of BST and medial ecacy.
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 165

Fig. 8. Innervation of the PVN-projecting region of the bed nucleus of the stria terminalis (BST) by GABAergic and glutamatergic limbic regions.
Combined tract tracing/immunohistochemistry (Fluorogold, FG) and in situ hybridization was used to establish the neurochemical identity of medial
amygdalar (MeA) (A,B) and ventral subicular (vSUB) (C,D) neurons innervating the PVN-projecting interfascicular region of the BST. In situ
hybridization-positive neurons are indicated with arrows, unlabeled neurons with arrowheads. In the MeA, the vast majority of FG labeled neurons
(dark) contained the GABAergic marker GAD67 (grains) (A), whereas there were very few neurons expressing vesicular glutamate transporter 1
(VGT) (B), a corticolimbic glutamate marker. In contrast, the vast majority of FG-labeled neurons in the vSUB were negative for GAD67 (C), but
positive for VGT (D). The arrow in C illustrates a non-FG labeled GAD67 neuron, probably reecting a subicular GABAergic interneuron.

Anatomical studies indicate that the prefrontal cortex coeruleus, a stress-activated region that is involved in
does not innervate the PVN to any substantial degree. HPA axis integration [328]. Dopaminergic input to the
However, bers from the infralimbic cortex and to a medial prefrontal region is exquisitely stress-sensitive
lesser extent, prelimbic and anterior cingulate cortex [129], and may be sucient to alter HPA-relevant out-
innervate several predominantly GABAergic PVN- put. Finally, NTS-projecting neurons in the prefrontal
projecting regions, including the BST, perifornical nu- cortex receive direct innervation from the ventral hip-
cleus, lateral hypothalamus and dorsomedial nucleus pocampus [251], suggesting that these regions may act in
[131,277], and prominently innervate the nucleus of the concert to modulate multiple down-stream stress regu-
solitary tract [300,308]. The infralimbic and prelimbic latory circuits.
cortices also project to the amygdaloid complex and
raphe nuclei, and may thereby modulate HPA activation 4.3. Amygdala
mediated by these structures (see below) [196,277]. Fi-
nally, the medial prefrontal cortex projects heavily to the In contrast with the hippocampus and prefrontal
paraventricular thalamic nucleus [131,277] (below), a cortex, the amygdala appears to activate the HPA
structure that is intimately involved in tuning HPA axis axis. Gross stimulation of the amygdala promotes
responses to chronic stress [18]. corticosteroid biosynthesis [150,252] and secretion
Activation of the prefrontal cortex may itself be [194,195,237,296] in rats, rabbits and monkeys, and
modulated by aerent input. The medial prefrontal elicits ACTH secretion in humans [110]. Conversely,
cortex receives heavy noradrenergic input from the locus lesions of large portions of the amygdala reduce
166 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

glucocorticoid secretion following stressful stimuli, compassing groups of cells in the region of the fusiform,
including ether inhalation [158,159], leg break [3] and subcommissural and anterodorsal subnuclei [76,234].
sensory stimulation [84]. Large amygdaloid regions also The latter subgroups contain large populations of
reduce adrenalectomy-induced ACTH hypersecretion GABAergic neurons; in combination with the predom-
[4], suggestive of a role in feedback integration. inantly GABAergic phenotype of CeA projection neu-
Subsequent studies using more localized stimulation/ rons, the CeABSTPVN circuit may utilize two GABA
lesion paradigms implicate the central (CeA), medial synapses, and thus activate the PVN by disinhibition
(MeA) and basolateral (BLA) subnuclei in control of (Fig. 7).
ACTH release. While all subnuclei are implicated in The MeA also plays a role in HPA axis integration.
HPA activation, they appear to respond preferentially to Selective stimulation of the MeA increases corticoste-
dierent stimulus modalities, and therefore have distinct rone release in anesthetized rats [80] and may increase
roles in HPA integration. adrenal sensitivity to ACTH [253]. In agreement with
The central amygdaloid nucleus (CeA) is involved in these data, lesions of the medial amygdala decrease
integration of behavioral responses to fear and/or anx- ACTH and corticosterone secretion and inhibit median
iety provoking behaviors [62,179]. Accordingly, the CeA eminence CRH depletion following photic and olfactory
appears to regulate HPA axis activation in a number of stimulation [86]. However, in contrast to the CeA, le-
stress paradigms. Lesion studies indicate that selective sions of the MeA reduce c-fos activation in the PVN by
damage of the CeA decreases ACTH and corticosterone restraint [65]. Furthermore, MeA c-fos induction can be
release following immobilization stress [15], photic/ol- observed following stimuli that activate of anticipatory
factory stimuli [86] and importantly, fear conditioning pathways, including restraint [56,64], novelty [81] and
[306]. Lesions of the CeA cause depletion of CRH from fear conditioning [229], but is far less pronounced in
the median eminence under basal conditions [16], and conjunction with reactive responses to stimuli such as
block stress-induced decreases in median eminence CRH hypovolemia [302], cytokine stimulation [269] or ether
stores [86], suggesting that the CeA promotes both CRH inhalation [81].
synthesis and release. Lesions of the CeA also reduce the Like the CeA, the MeA has very few direct projections
number of c-fos positive PVN CRH neurons following to the PVN [39,112,216,234]. However, the MeA has an
interleukin-1 injection [323], consistent with an interac- extensive network of projections to PVN-projecting re-
tion between this region and cytokine-mediated activa- gions, including the intrafascicular, transverse, antero-
tion of PVN neurons. dorsal and ventral subnuclei of the BST, medial preoptic
However, other reports have failed to nd a signi- nucleus, medial preoptic area, anterior hypothalamus,
cant impact of CeA lesions on HPA axis regulation. ventral premammillary nucleus and peri-PVN zone [39].
Lesions of the CeA fail to reduce either ACTH release Combined anterograde-retrograde tracing studies indi-
or PVN c-fos induction following restraint stress cate that PVN-projecting neurons of the aforementioned
[65,234]. In combination with data showing clear in- BST subnuclei and the medial preoptic nucleus are con-
volvement of the CeA in cytokine-mediated PVN c-fos tacted by boutons labeled following posteroventral MeA
induction by the same group [323], these data have lead injections of the anterograde tracer PHA-L [234]. With
to the conclusion that the CeA is selectively involved in the possible exception of the ventral premammillary nu-
generating what we have termed reactive responses cleus, PVN-projecting neurons in these MeA-receptive
[64,118,270]. This hypothesis is supported by c-fos regions are predominantly GABAergic [55,57], thus sug-
mapping data showing preferential induction of the CeA gesting that MeAPVN relays are composed of sequential
by stressors such as hemorrhage, cytokine infusions and GABA neurons. Accordingly, combined retrograde
lithium chloride injection [269,302,325], while stimuli tracing-in situ hybridization studies reveal that majority
such as novelty, restraint, footshock or air-pu startle of MeA neurons projecting to the posterior BST express
[56,81,269,301] show a minimal CeA c-fos response. glutamic acid decarboxylase (but not VGlut1) mRNA,
Moreover, CeA c-fos responses can be conditioned by consistent with a largely GABAergic phenotype (Fig. 8).
stimuli eliciting reactive responses (e.g., lithium chlo- The stimulatory eect of the MeA on corticosterone re-
ride) [170,325], but are not induced in conjunction with lease can be blocked by lesions of the BST or medial
anticipatory stress responses (e.g., fear conditioning) preoptic area [88], supporting the importance of these
[34,229]. putative relays in HPA integration. Therefore, the MeA
The CeA has little direct interaction with the PVN, likely activates the PVN by disinhibition, using relays
limited to scattered bers present in preautonomic cell distinct from those of the CeA.
groups [112,192,234]. However, the CeA has rich con- The BLA has received considerable attention as a
nections with brainstem structures innervating the PVN, stress-regulatory structure. This region is robustly acti-
including most notably the NTS and lateral parabra- vated by anticipatory stressors, including restraint,
chial nucleus [273,308]. In addition, there is evidence for swim and footshock, but shows a substantially lower
a relatively circumspect forebrain relay in the BST, en- response to cytokine stimulation [56,270]. Stimulation of
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 167

the BLA in anesthetized rats can decrease [80] or in- The anatomy of the septal region predicts prominent
crease [89] corticosterone secretion, perhaps due to interactions between the ventrally situated septal cell
complex interactions between stimulation parameters groups (ventrolateral septal (LSv) or ventral regions of
and anesthesia. Lesions of the BLA are ineective in the rostral subdivision (LSr)) and PVN-projecting
modulating ACTH, corticosterone or CRH release fol- forebrain regions. As is the pattern for limbic stress in-
lowing photic or olfactory stimuli [86], and do not aect tegrative regions, ventrolateral septal neurons do not
corticosterone release following social interaction [201], project directly to the PVN, but innervate the peri-PVN
novelty, restraint, ether stress or cold [274]. However, it zone and numerous hypothalamic PVN relays, including
is important to note that this region may indeed play a the anterior hypothalamus, medial preoptic area and
more complex role in stress regulation; for example, the lateral hypothalamus [242]. The vast majority of neu-
BLA appears to be critical for integrating the eects of rons in this region of the septal complex express GAB-
glucocorticoids on memory consolidation [247,248], Aergic markers [241], thus suggesting an inhibitory
suggesting involvement in memory of stressful stimuli. inuence on PVN-projecting cell groups. Notably, lat-
Similarly, this region is selectively activated by novel eral septal neurons innervate PVN-projecting regions
stressors presented during a chronic stress protocol, that contain both GABAergic and glutamatergic neu-
consistent with a role in HPA axis sensitization [18]. rons [68,213,329], and thus the lateral septum is well-
Projections of the BLA are to a large extent intra- positioned to modulate either inhibition or excitation of
amygdalar. Indeed, the anterior BLA extensively in- the PVN.
nervates the CeA and MeA, suggesting that output from
this region of the amygdala are funneled through these 4.5. Thalamus
principle amygdalar output nuclei. Nonetheless, the
posterior BLA does have limited projections to the an- Several recent reports implicate limbic thalamic nu-
terodorsal BST and several medial hypothalamic nuclei clei in processes related to HPA axis regulation. The
(excluding the PVN), suggesting the capacity for this midline thalamus shows robust c-fos induction follow-
region to act via non-amygdalar circuitry as well ing psychogenic stimuli in several subregions, including
[75,293]. the paraventricular, subparafascicular, posterior intra-
laminar, anteroventral, anteromedial, paratenial, cen-
4.4. Lateral septum tromedial and rhomboid nuclei (cf. [10,18,28,56,
81,280]).
Lateral septal lesions are known to produce a con- Of the known stress-activated thalamic nuclei, the
stellation of behavioral and autonomic symptoms con- paraventricular thalamus appears to play a major role in
sistent with extreme anxiety (septal rage) [242]. integrating HPA axis responses to repeated stressors.
Accordingly, the lateral septum seems well-positioned to The paraventricular thalamus is among a handful of
modulate neuroendocrine stress responses. Indeed, nu- regions that is selectively activated by novel stressors
merous c-fos mapping studies provide circumstantial presented after a repeated stress regimen [18]. This reg-
evidence for a role of the ventrolateral septal region in imen produces a phenomenon known as facilitation,
stress regulation. Neurons in this area are robustly in- whereby the HPA axis response to the novel stressor is
duced by a variety of anticipatory stressors, such as markedly potentiated in previously stressed animals,
novelty, predator exposure and social interaction despite the presence of elevated glucocorticoid feedback.
[34,38,81,162], but show little induction following acti- Notably, lesions of the paraventricular thalamus (in
vation of the reactive stress pathway [81,270,301]. particular, its posterior subdivision) eectively block
In agreement with the c-fos mapping data, lesion chronic stress facilitation of ACTH release [18], and can
studies suggest that the septum has an inhibitory inu- indeed inhibit habituation of the HPA axis to repeated
ence on the HPA axis. Large lesions of the septum, exposure to the same stressor [19]. Overall, these data
encompassing both lateral and medial divisions, do not suggest that this region plays a prominent role in
aect basal or circadian corticosterone rhythms controlling HPA responsiveness following repeated
[74,275,320], but prolong corticosterone responses to stimulation [19].
immobilization [74]. Septal lesion animals also show The connectivity of the paraventricular thalamus
enhanced susceptibility to mild stress [27,276,303]. places it in prime position to mediate HPA responsive-
However, these early studies of septal function used ness. Combined c-fos-retrograde tracing studies indicate
large electrolytic lesions, which did extensive damage to that the paraventricular thalamus receives extensive
both the medial septum and descending mbriafornix input from numerous stress-activated regions, including
system. Thus, the results of these early studies are dif- the ventral subiculum, infralimbic/prelimbic cortex,
cult to interpret with regard to the lateral septum per BST, NTS, rostrolateral medulla, locus coeruleus, dor-
se, and rened lesion and stimulation studies of this sal raphe and parabrachial nuclei [218]. In addition, the
region are lacking. paraventricular thalamus heavily innervates several
168 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

PVN-projecting and PVN-interactive regions, including stress modality, there are some instances where very
the medial prefrontal cortex and the basolateral and similar experiments yield divergent results. We feel there
central amygdaloid nuclei [207,219]. are at least four possible explanations for such discrep-
Recent work also suggests that the anterodorsal ancies. First, the environment surrounding stress exper-
thalamic nucleus can modulate both basal and stress- iments represents an important and largely uncontrolled
induced HPA activation. Small lesions of this nucleus experimental variable. A considerable amount has been
elevate basal ACTH levels while inhibiting stress-in- learned about HPA secretory time courses, circadian
duced release [287]. This region has extensive reciprocal secretory patterns, stress sensitization, and response
connections with the subiculum [291,309], which may habituation in recent years. This knowledge emphasizes
account for its impact on HPA axis regulation. the necessity for careful control of conditions sur-
rounding stress exposure. Whereas a rigorous level of
4.6. Hypothalamus experimental control (e.g., time of day, limited housing/
procedure room access, rapid sampling, etc) is now fairly
There are several hypothalamic regions that appear standard, it is not known how tightly such control was
able to interact with the PVN through intrahypotha- exerted in earlier studies of this system. Indeed, many of
lamic relays. Prominent among these is the suprachias- the early ndings on limbic modulation of the stress re-
matic nucleus, which is the major integrator of circadian sponse occurs against a backdrop of elevated (i.e., su-
HPA rhythmicity [44]. While having minimal direct prabasal) glucocorticoids, and makes it dicult to
projections to the medial parvocellular PVN per se, the interpret the data with regard to eects on basal secre-
SCN projects heavily to GABA-rich, PVN-projecting tion vs. stress-induced release.
hypothalamic regions, including the subparaventricular Animal housing represents a second area of concern.
zone of the peri-PVN region and dorsomedial hypo- Over the last 1015 years, laboratory animal husbandry
thalamus [29,317]. The SCN inhibits circadian cortico- has improved dramatically. Even non-SPF (Specic
sterone release by way of these nuclei, apparently via Pathogen Free) rooms have high minimum standards
vasopressinergic neurons [30,146]. In addition to its for sanitization, and infections that were largely ignored
circadian inuence, the SCN has inhibitory actions on in previous decades (e.g., pinworm infections) are now
corticosterone responses to novelty stress that appears aggressively monitored, treated and eradicated. Thus, to
to involve actions at both the hypothalamus and adrenal the extent that such subchronic infections are now ob-
cortex [30]. viated, the background level of stress encountered un-
The PVN also receives transsynaptic innervation der current conditions may dier substantially from
from the ventromedial nucleus of the hypothalamus prior studies. As the existence of background stress can
(VMH). In this case, interconnectivity is likely achieved potentially sensitize or habituate HPA responses, and
through intervening neurons in the dorsomedial and both of these processes are likely modulated by limbic
lateral hypothalamic nuclei [298]. Electrolytic lesions of structures, dierences in housing may constitute a sub-
the ventromedial nucleus disrupt circadian corticoste- stantial contribution to the observed experimental dis-
rone secretion, resulting in a net elevation in secretory crepancies.
activity across the day-night period [153,288]. How- A third source of error concerns the early-life envi-
ever, colchicine inactivation of the VMH in fact ronment of the experimental subjects. Considerable data
reduces corticosterone secretion at the circadian are emerging to indicate that the quality of maternal
peak and inhibits stress-induced corticosterone secre- care can have a life-long impact on the responsiveness
tion [48]. of the HPA axis; good maternal care predisposes e-
cient HPA axis responses to stress, whereas maternal
deprivation elicits stress hyper-responsivity (cf. [32,182,
5. Synthesis 254]). Thus, substantial individual dierences may
be encountered in studies using animals from dier-
5.1. Methodological considerations in stress research: ent commercial vendors or breeding-colony environ-
sources of disagreements? ments.
A nal factor is the animal itself. There is consider-
Before generalities can be drawn concerning the vo- able evidence for strain dierences in HPA reactivity.
luminous literature on neurocircuit regulation, sources The Lewis strain exhibits substantially lower basal CRH
for some of the disagreements among studies need to mRNA levels in the PVN relative to Fischer 344 strains,
be considered. Disagreements are most pronounced and exhibits attentuated neuroendocrine responses to
among limbic-HPA studies, where conicting data exist stress [285,286]. Strains also dier in responsiveness to
regarding the eects of hippocampal and central chronic stimulation; for example, our group has noted
amygdalar lesions on ACTH secretion (see above). While that the SpragueDawley strain exhibits lower levels of
some of the conict can be resolved by consideration of chronic stress-induced corticosterone hypersecretion
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 169

than Fischer 344 animals [128] or Fisher 344 Brown guing that the road to adaptation may well have many
Norway F1 hybrids [126]. Thus, variation in response neurochemical and humoral forks.
predispositions may be present within dierent strains of Our attempt to discern a set of principles dening
rats, generating dierent baselines against which the HPA axis regulation relies heavily on the diversity of the
experience of stress is perceived. data. In this view, central stress integration relies on
hierarchical rather than dichotomous pathways. The
5.2. Principles governing neurocircuit regulation of the output of the stress response is dependent on the overall
HPA axis set of central sensorial input, which places a dierent
weight on the various central structures projecting onto
Numerous attempts have been made to distill the PVN stress integrators. This hypothesis is based on the
stress circuitry literature summarized above into a gen- following premises:
eral theory of stress integration. Perhaps the most 1. Stressors considered to be reactive invoke direct in-
durable hypotheses propose a dichotomy between neu- put pathways to the PVN, as PVN activation and glu-
rogenic/psychogenic/processive/exteroceptive responses cocorticoid responses are poorly elaborated when
and systemic/interoceptive responses [64,118,184,270]. these circuits are damaged. Induction of these sys-
This notion was rst presented by Fortier in 1951 to tems occurs the rst time the stimulus is presented,
explain why some stressors (epinephrine, cold, and and does not require input from the forebrain.
histamine) could elicit corticosteroid release when the 2. The inuence of the forebrain on the stress system is
pituitary was removed and placed in the anterior most keenly engaged following stressors that require
chamber of the eye, while others required an intact either initiation of innate anxiety or defense pro-
pituitary for elaboration (immobilization, sound) [106]. grams (e.g., presence of a predator, high places, social
The theoretical basis for these distinctions has changed conict, etc.) or conditioned responses to sensory
over the years to encompass a very dierent concept, stimuli associated with previous stressful events.
namely, that the neurogenic class of stimuli requires 3. Forebrain inuences on the PVN are polysynaptic,
some form of forebrain processing and integration prior and relay through structures that, while engaged in
to an HPA response, whereas the systemic class is cases of anticipatory stress, may also mediate reac-
elaborated by reexive pathways (spinal cord, brain- tive responses. These include regions such as the
stem, lamina terminalis continuum) [118,270]. Perhaps NTS, dorsomedial hypothalamus, preoptic area,
nowhere is this better demonstrated than in the seminal and PVN surround.
study by Sawchenkos group in 1996, demonstrating 4. Unlike the reactive pathways, forebrain regions such
that ascending medullary systems are required for PVN as the prefrontal cortex, hippocampus, and amygdala
c-fos activation by interleukin-1b, a systemic/intero- are in receipt of polysensorial and associational in-
ceptive stimulus, but do not aect c-fos induction fol- put, rather than primary sensory modalities.
lowing footshock [183]. Our own work demonstrated 5. The inuence of forebrain structures on down-stream
that lesions of the ventral subiculum impair corticoste- stress eectors varies with region and the nature of
rone responsiveness to novelty, but have no impact on the stimulus. For example, the hippocampus (ventral
responses to ether [123]. Trevor Days group has sub- subiculum) has a wealth of interactions with the BST
sequently contributed a substantial body of data and hypothalamus, but very little with the NTS; the
documenting an amygdalar dissociation of emotional prefrontal cortex has limited input to the hypothala-
from systemic stress, implicating the medial amygdala mus, but innervates the lateral BST and NTS more
in the former, and the central amygdaloid nucleus in the extensively; and so on.
latter [64]. Their work also supports a role for the All of these data agree in large part with the diverse
brainstem in both emotional and systemic stress mo- role of limbic pathways in HPA integration, while
dalities [66], and has been important in revising how we conserving the notion of direct, reactive pathways, atop
envision the overall organization of HPA integration by which the hierarchical anticipatory responses can be
the PVN. built.
Despite a common unifying theme that runs through Hierarchical organization of brain stress integration
the above lines of thought, there is a considerable body is in keeping with theories of brain evolution, whereby
of data to suggest that this dichotomy may be an increasingly complex functions are layered atop simpler
oversimplication. This is particularly evident in the reex pathways. The reex pathways operate as the de-
work of Pacak, Goldstein, Palkovits, and colleagues, fault mode, and are subsequently modied by the
who have performed numerous studies demonstrating inuence of prior experience or innate programs. The
quite unique HPA and sympathomedullary responses to circuitries underlying these internally generated neural
dierent stressors (see [220,222]). Indeed, these authors programs adjust the stress response with respect to their
use these data to revise the notion of the general ad- net actions at the PVN. The outow of the HPA axis is
aptation syndrome proposed by Selye, in essence ar- therefore a summation of integrated inputs from several
170 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

forebrain regions, including the hippocampus, pre- projections to the medial and central amygdaloid nu-
frontal cortex, amygdala, and septum. Evidence of this clei [196,277]. Recent work from our group demon-
summation may be appreciated from the fact that many strates that medial prefrontal cortex lesions inhibit
stressors produce parallel activation among numerous medial amygdalar c-fos induction following restraint
HPA-regulatory limbic regions (e.g. [56,270]), and le- stress [99], suggesting that such interconnections may
sions of dierent regions can produce similar eects on be functionally meaningful.
stress responses (for example [73,99,123]). It stands to Thus, initiation and cessation of stress responses
reason that these diverse regions will be inuenced in appears to be choreographed by sets of hierarchical
accordance with their polysensorial inputs; thus, a me- circuits converting mono- or polysensorial information
dial amygdala-loaded stimulus (e.g., responses to con- into an integrated PVN response. The simplest form is
specic aggression) is likely to exhibit a stressresponse a novel physiological challenge, which is rapidly com-
signature that is funneled primarily through the BST municated by reex pathways from surveillance sites in
and preoptic area, representing its primary terminal the periphery (nociceptors, visceral eerents) or cir-
elds. Whereas other limbic regions can participate in cumventricular organs (subfornical organ, and area
tuning the response, the heavy emphasis on the medial postrema). This requires no limbic inputs and is rela-
amygdala gives this PVN input channel priority, and the tively hard-wired. A second-level response involves in-
net HPA outow is adjusted accordingly. nate defense programs that anticipate a physiological
Preferential weighting of dierent limbic regions may challenge, in essence driving a glucocorticoid response
also be elaborated at the interface between the antici- to obviate a predicted homeostatic disruption. Due to
patory and reexive pathways. For example, our GLP-1 the necessity of comparing polysensory stimuli with
studies indicate a prominent role for the NTS in initi- these internal programs, these pathways involve limbic
ating HPA responses to the elevated plus maze [154]. outow. The valence of this outow will likely be
The elevated plus maze generates a fear response that is regulated in accordance with the relative sensory and
controlled in part by neurons of the CeA [63]. The CeA associational load to respective limbic forebrain re-
has considerable projections to NTSPVN relays, and gions, with the net limbic outow determining which
may thus be involved in generating the HPA response to PVN eector pathways are activated or inhibited. Fi-
the fear-evoking stimulus. In contrast, Sawchenkos nally, the third-level response involves learning; re-
group noted that the PVN c-fos response to footshock sponses are keyed to recollection of the signicance of
was not blocked by medullary knife cuts, suggesting this prior experience, colored itself by the glucocorticoid
pain stimulus does not require NTS input to the PVN response. These responses probably involve mnemonic
[183]. In this study animals were pre-habituated to the processing regions such as the hippocampus and me-
testing chamber for several days prior to testing; the dial prefrontal cortex. Again, the strength of the PVN
brainstem-independent response may have been gener- response will be modulated by the relative strength of
ated exclusively in limbic sites processing contextual the respective limbic signals.
information related to the test environment, such as the Many of the PVN-projecting hypothalamic and
hippocampus or perhaps the prefrontal cortex. Thus, the brainstem circuits used by limbic inputs govern basic
nature of the limbic circuits activated by specic con- homeostatic regulatory functions. Descending forebrain
stellations of polysensory input may in large part dictate inputs relayed through these circuits thus interface with
the down-stream PVN relays controlling HPA axis ac- information traversing primary somatic, visceral or hu-
tivation. moral sensory pathways. This arrangement provides a
Anatomical data also support coordination of de- signaling context with which limbic information can
scending limbic eerents distal to the PVN proper. interact; as such, the HPA response to an anticipatory
Limbic connectivity can overlap in several PVN pro- stressor will depend on the homeostatic state of the in-
jecting regions; for example, the medial amygdala, dividual. An inhibitory signal cued by contextual in-
ventral subiculum and medial prefrontal cortex all formation may be overridden by peripheral stimuli
project to posterior subnuclei of the BST and the peri- conveying energy imbalance, cardiovascular depression,
PVN region (see Fig. 7), suggesting a convergence of infection, or thirst; conversely, an excitatory signal
inputs in these areas. While it has yet to be determined originating in the periphery may be modulated by de-
whether these regions project to the same neurons, the scending contextual information. This arrangement af-
proximity of eerent targets is consistent with local fords the tone of the HPA axis to be set by both
integration within PVN-projecting cell populations. In polysynaptic forebrain activity and reexive informa-
addition, there is considerable cross-talk within limbic tion, much analogous to layered sensorymotor loops
pathways; for example, in addition to BST and septal regulating reexive and voluntary movement.
outow, the ventral subiculum also projects to the The teleological implications of hierarchical stress-
medial amygdala and medial prefrontal cortex integrative pathways are clearthe circuitry can weigh
[40,139]; likewise, the medial prefrontal cortex sends the importance of a stimulus to survival, and use the
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 171

resulting information to tune an appropriate hormonal We thank Dr. Dana Ziegler, Dr. Chantal Prewitt, Mark
response. In most organisms, the system eciently Dolgas, Garrett Bowers, and Melanie Emmert for their
modulates the HPA axis in accordance with need, as- eorts on various components of these projects. We also
suring that glucocorticoids are present only when re- acknowledge the late Bryan Bodie; his scientic and
quired. However, as is the case with all biological personal contributions are sorely missed.
processes, there is considerable individual variation in
HPA response dispositions. Recent studies reveal that
genetics, early-life experience or even trauma in adult References
life can modulate response characteristics of the HPA
axis. In rats, HPA hyperresponsiveness induced by [1] S.F. Akana, A. Chu, L. Soriano, M.F. Dallman, Corticosterone
early-life maternal separation/neglect can be correlated exerts site-specic and state-dependent eects in prefrontal
with neurochemical changes in key limbic stress circuits, cortex and amygdala on regulation of adrenocorticotropic
including reduced glucocorticoid receptor expression in hormone, insulin and fat depots, J. Neuroendocrinol. 13 (2001)
625637.
the hippocampus, elevated central amygdaloid nucleus [2] N.E. Alessi, P. Quinlan, H. Khachaturian, MSG eects on beta-
CRH expression and reduced GABA-A receptor ex- endorphin and alpha-MSH in the hypothalamus and caudal
pression in the basolateral amygdala (see [32,149,202]). medulla, Peptides 9 (1988) 689695.
This spectrum of changes implies reduced hippocampal [3] J.P. Allen, C.F. Allen, Role of the amygdaloid complexes in the
inhibition and enhanced amygdalar excitation, and to- stress-induced release of ACTH in the rat, Neuroendocrinology
15 (1974) 220230.
gether these results suggest that a coordinate disruption [4] J.P. Allen, C.F. Allen, Amygdalar participation in tonic
of hierarchical stress circuitry underlies HPA axis dys- ACTH secretion in the rat, Neuroendocrinology 19 (1975)
function. The data imply that the conguration of stress 115125.
circuitry, being subject to considerable inuence by en- [5] S.E. Alves, V. Lopez, B.S. McEwen, N.G. Weiland, Dierential
vironmental factors, is malleable even within an organ- colocalization of estrogen receptor beta (ERbeta) with oxytocin
and vasopressin in the paraventricular and supraoptic nuclei of
ism. Thus, changes in limbic integration patterns with the female rat brain: an immunocytochemical study, Proc. Natl.
experience may play a major role in HPA axis dys- Acad. Sci. USA 95 (1998) 32813286.
function. [6] F.A. Antoni, Hypothalamic control of adrenocorticotropin
The relationship between aective disease states and secretion: Advances since the discovery of 41-residue corticotro-
HPA hyperactivity may lie in the linkage between de- pin-releasing factor, Endocr. Rev. 7 (1986) 351378.
[7] J.L. Arriza, R.B. Simerly, L.W. Swanson, R.M. Evans, Neuronal
scending limbic pathways stress eectors and the PVN. mineralocorticoid receptor as a mediator of glucocorticoid
For example, major depression is associated with en- response, Neuron 1 (1988) 887900.
hanced blood ow and glucose utilization signal in the [8] J.M. Aubry, V. Bartanusz, S. Pagliusi, P. Schulz, J.Z. Kiss,
prefrontal cortex and amygdala (see [77]) and shrinkage Expression of ionotropic glutamate receptor subunit mRNAs by
of the hippocampal formation [278]. As noted above, all paraventricular corticotropin-releasing factor (CRF) neurons,
Neurosci. Lett. 205 (1996) 9598.
of these regions are implicated in HPA regulation in [9] C.J. Auger, G.J. De Vries, Progestin receptor immunoreactivity
animal studies. The characteristics of depression-in- within steroid-responsive vasopressin-immunoreactive cells in
duced HPA dysfunction, including stress hyperrespon- the male and female rat brain, J. Neuroendocrinol. 14 (2002)
siveness, disrupted circadian secretory pattern, and 561567.
glucocorticoid negative feedback resistance, can be [10] J.S. Ba, M. Palkovits, Fine topography of brain areas activated
by cold stress. A fos immunohistochemical study in rats,
replicated in some (but not all) animal lesion studies, Neuroendocrinology 72 (2000) 102113.
and are consistent with the changes in regional activity [11] T.W. Bailey, J.A. Dimicco, Chemical stimulation of the dorso-
and structure revealed by neuroimaging. Importantly, medial hypothalamus elevates plasma ACTH in conscious rats,
the HPA symptoms are paralleled by other manifesta- Am. J. Physiol. 280 (2001) R8R15.
tions of homeostatic disruptionsleep, metabolism, [12] R.A. Baker, M. Herkenham, Arcuate nucleus neurons that
project to the hypothalamic paraventricular nucleus: neuropept-
cardiovascular regulationthat are also within the ef- idergic identity and consequences of adrenalectomy on mRNA
ferent trajectory of these key limbic circuits. Thus, it levels in the rat, J. Comp. Neurol. 358 (1995) 518529.
appears that modulation of the anticipatory pathway [13] V. Bartanusz, J.M. Aubry, S. Pagliusi, D. Jezova, J. Ba, J.Z.
initiated by innate or learned response predispositions Kiss, Stress-induced changes in messenger RNA levels of N-
may form a critical neuroendocrine eector component methyl-D -aspartate and AMPA receptor subunits in selected
regions of the rat hippocampus and hypothalamus, Neuroscience
of these human disease processes. 66 (1995) 247252.
[14] S.L. Bealer, Corticosteroids and plasma restitution after hemor-
rhage and hypothalamic lesions, Am. J. Physiol. 250 (1986) R18
Acknowledgments R23.
[15] S. Beaulieu, T. DiPaolo, N. Barden, Control of ACTH secretion
by the central nucleus of the amygdala: Implication of the
This work was supported by NIH Grants MH49698 serotonergic system and its relevance to the glucocorticoid
to J.P.H., MH56577 to W.E.C., MH60819 to J.P.H. and delayed negative feedback mechanism, Neuroendocrinology 44
W.E.C., and NIH NRSA Grant MH65770 to N.K.M. (1986) 247254.
172 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

[16] S. Beaulieu, G. Pelletier, H. Vaudry, N. Barden, Inuence of the and induction of c-fos mRNA in the forebrain associated
central nucleus of the amygdala on the content of corticotropin- with audiogenic stress in rats, J. Neurosci. 17 (1997) 5979
releasing factor in the median eminence, Neuroendocrinology 49 5992.
(1989) 255261. [34] S. Campeau, W.A. Falls, W.E. Cullinan, D.L. Helmreich, M.
[17] M.L. Berk, J.A. Finkelstein, Aerent projections to the preoptic Davis, S.J. Watson, Elicitation and reduction of fear:
area and hypothalamic regions in the rat brain, Neuroscience behavioural and neuroendocrine indices and brain induction
6 (1981) 16011624. of the immediate-early gene c-fos, Neuroscience 78 (1997)
[18] S. Bhatnagar, M. Dallman, Neuroanatomical basis for facilita- 10871104.
tion of hypothalamic-pituitary- adrenal responses to a novel [35] S. Campeau, S.J. Watson, Neuroendocrine and behavioral
stressor after chronic stress, Neuroscience 84 (1998) 10251039. responses and brain pattern of c-fos induction associated with
[19] S. Bhatnagar, R. Huber, N. Nowak, P. Trotter, Lesions of the audiogenic stress, J. Neuroendocrinol. 9 (1997) 577588.
posterior paraventricular thalamus block habituation of hypo- [36] S. Campeau, S.J. Watson Jr., Connections of some auditory-
thalamic-pituitary-adrenal responses to repeated restraint, responsive posterior thalamic nuclei putatively involved in
J. Neuroendocrinol. 14 (2002) 403410. activation of the hypothalamopituitaryadrenocortical axis in
[20] H.C. Bohler Jr., R.T. Zoeller, J.C. King, B.S. Rubin, R. Weber, response to audiogenic stress in rats: an anterograde and
G.R. Merriam, Corticotropin releasing hormone mRNA is retrograde tract tracing study combined with Fos expression,
elevated on the afternoon of proestrus in the parvocellular J. Comp. Neurol. 423 (2000) 474491.
paraventricular nuclei of the female rat, Brain Res. Mol. Brain [37] N.S. Canteras, R.B. Simerly, L.W. Swanson, Connections of the
Res. 8 (1990) 259262. posterior nucleus of the amygdala, J. Comp. Neurol. 324 (1992)
[21] B. Borowsky, C.M. Kuhn, D1 and D2 dopamine receptors 143179.
stimulate hypothalamopituitaryadrenal activity in rats, Neu- [38] N.S. Canteras, R.B. Simerly, L.W. Swanson, Projections of the
ropharmacology 31 (1992) 671678. ventral premammillary nucleus, J. Comp. Neurol. 324 (1992)
[22] C. Boudaba, L.A. Schrader, J.G. Tasker, Physiological evidence 195212.
for local excitatory synaptic circuits in the rat hypothalamus, [39] N.S. Canteras, R.B. Simerly, L.W. Swanson, Organization of
J. Neurophysiol. 77 (1997) 33963400. projections from the medial nucleus of the amygdala: a PHAL
[23] C. Boudaba, K. Szabo, J.G. Tasker, Physiological mapping of study in the rat, J. Comp. Neurol. 360 (1995) 213245.
local inhibitory inputs to the hypothalamic paraventricular [40] N.S. Canteras, L.W. Swanson, Projections of the ventral
nucleus, J. Neurosci. 16 (1996) 71517160. subiculum to the amygdala, septum, and hypothalamus: a
[24] G. Bowers, W.E. Cullinan, J.P. Herman, Region-specic regu- PHAL anterograde tract-tracing study in the rat, J. Comp.
lation of glutamic acid decarboxylase (GAD) mRNA expression Neurol. 324 (1992) 180194.
in central stress circuits, J. Neurosci. 18 (1998) 59385947. [41] M.P. Carey, C.H. Deterd, J. de Koning, F. Helmerhorst, E.R. de
[25] M.J. Bradbury, A.M. Strack, M.F. Dallman, Lesions of the Kloet, The inuence of ovarian steroids on hypothalamic
hippocampal eerent pathway (mbriafornix) do not alter pituitaryadrenal regulation in the female rat, J. Endocrinol. 144
sensitivity of adrenocorticotropin to feedback inhibition by (1995) 311321.
corticosterone in rats, Neuroendocrinology 58 (1993) 396 [42] J.H. Casada, N. Dafny, Restraint and stimulation of the bed
407. nucleus of the stria terminalis produce similar stress-like behav-
[26] W.G. Brake, G. Flores, D. Francis, M.J. Meaney, L.K. iors, Brain Res. Bull. 27 (1991) 207212.
Srivastava, A. Gratton, Enhanced nucleus accumbens dopamine [43] R.L. Casady, A.N. Taylor, Eect of electrical stimulation of the
and plasma corticosterone stress responses in adult rats with hippocampus upon corticosteroid levels in the freely-behaving,
neonatal excitotoxic lesions to the medial prefrontal cortex, non-stressed rat, Neuroendocrinology 20 (1976) 6878.
Neuroscience 96 (2000) 687695. [44] C.S. Cascio, J. Shinsako, M.F. Dallman, The suprachiasmatic
[27] G.M. Brown, I.V. Uhlir, J. Seggie, A.V. Schally, A.J. Kastin, nuclei stimulate evening ACTH secretion in the rat, Brain Res.
Eect of septal lesions on plasma levels of MSH, corticosterone, 423 (1987) 173178.
GH and prolactin before and after exposure to novel environ- [45] D. Champagne, J. Beaulieu, G. Drolet, CRFergic innervation of
ment: role of MSH in the septal syndrome, Endocrinology 94 the paraventricular nucleus of the rat hypothalamus: a tract-
(1974) 593597. tracing study, J. Neuroendocrinol. 10 (1998) 119131.
[28] M. Bubser, A.Y. Deutch, Stress induces Fos expression in [46] R.K. Chan, P.E. Sawchenko, Spatially and temporally dieren-
neurons of the thalamic paraventricular nucleus that innervate tiated patterns of c-fos expression in brainstem catecholaminer-
limbic forebrain sites, Synapse 32 (1999) 1322. gic cell groups induced by cardiovascular challenges in the rat,
[29] R.M. Buijs, M. Markman, B. Nunes-Cardoso, Y.X. Hou, S. J. Comp. Neurol. 348 (1994) 433460.
Shinn, Projections of the suprachiasmatic nucleus to stress- [47] S. Cheung, J.R. Ballew, K.E. Moore, K.J. Lookingland, Con-
related areas in the rat hypothalamus: a light and electron tribution of dopamine neurons in the medial zona incerta to the
microscopic study, J. Comp. Neurol. 335 (1993) 4254. innervation of the central nucleus of the amygdala, horizontal
[30] R.M. Buijs, J. Wortel, J.J. Van Heerikhuize, A. Kalsbeek, Novel diagonal band of Broca and hypothalamic paraventricular
environment induced inhibition of corticosterone secretion: nucleus, Brain Res. 808 (1998) 174181.
physiological evidence for a suprachiasmatic nucleus mediated [48] S. Choi, L.S. Wong, C. Yamat, M.F. Dallman, Hypothalamic
neuronal hypothalamoadrenal cortex pathway, Brain Res. 758 ventromedial nuclei amplify circadian rhythms: do they contain
(1997) 229236. a food-entrained endogenous oscillator?, J. Neurosci. 18 (1998)
[31] K. Buller, Y. Xu, C. Dayas, T. Day, Dorsal and ventral 38433852.
medullary catecholamine cell groups contribute dierentially to [49] K.D. Clier, R. Burstein, G.J. Giesler Jr., Distributions of
systemic interleukin-1beta-induced hypothalamic pituitary adre- spinothalamic, spinohypothalamic, and spinotelencephalic bers
nal axis responses, Neuroendocrinology 73 (2001) 129138. revealed by anterograde transport of PHA-L in rats, J. Neurosci.
[32] C. Caldji, J. Diorio, M.J. Meaney, Variations in maternal care in 11 (1991) 852868.
infancy regulate the development of stress reactivity, Biol. [50] R.L. Cole, P.E. Sawchenko, Neurotransmitter regulation of
Psychiatr. 48 (2000) 11641174. cellular activation and neuropeptide gene expression in the
[33] S. Campeau, H. Akil, S.J. Watson, Lesions of the medial paraventricular nucleus of the hypothalamus, J. Neurosci. 22
geniculate nuclei specically block corticosterone release (2002) 959969.
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 173

[51] V. Critchlow, R.A. Liebelt, M. Bar-Sela, W. Mountcastle, H.S. [70] H.D. Dellmann, E.M. Rodriguez, P. Pena, I. Siegmund, Immu-
Lipscomb, Sex dierence in resting pituitaryadrenal function in nohistochemical investigation of the magnocellular peptidergic
the rat, Am. J. Physiol. 205 (1963) 807815. hypothalamo-neurohypophysial system of the rat chronically
[52] A. Csaki, K. Kocsis, B. Halasz, J. Kiss, Localization of stimulated by long-term administration of hypertonic saline,
glutamatergic/aspartatergic neurons projecting to the hypotha- Neuroendocrinology 47 (1988) 335342.
lamic paraventricular nucleus studied by retrograde transport of [71] W.S. Dhillo, C.J. Small, L.J. Seal, M.S. Kim, S.A. Stanley, K.G.
[3 H]D -aspartate autoradiography, Neuroscience 101 (2000) 637 Murphy, M.A. Ghatei, S.R. Bloom, The hypothalamic melano-
655. cortin system stimulates the hypothalamopituitaryadrenal axis
[53] W.E. Cullinan, Evidence for a PVN site of action of GABA in in vitro and in vivo in male rats, Neuroendocrinology 75 (2002)
the regulatory control of the rat stress axis, The Physiologist 41 209216.
(1998) 353. [72] D.M. Diamond, M.C. Bennett, M. Fleshner, G.M. Rose,
[54] W.E. Cullinan, GABA(A) receptor subunit expression within Inverted-U relationship between the level of peripheral cortico-
hypophysiotropic CRH neurons: a dual hybridization histo- sterone and the magnitude of hippocampal primed burst
chemical study, J. Comp. Neurol. 419 (2000) 344351. potentiation, Hippocampus 2 (1992) 421430.
[55] W.E. Cullinan, D.L. Helmreich, S.J. Watson, Fos expression in [73] D. Diorio, V. Viau, M.J. Meaney, The role of the medial
forebrain aerents to the hypothalamic paraventricular nucleus prefrontal cortex (cingulate gyrus) in the regulation of hypo-
following swim stress, J. Comp. Neurol. 368 (1996) 8899. thalamopituitaryadrenal responses to stress, J. Neurosci. 13
[56] W.E. Cullinan, J.P. Herman, D.F. Battaglia, H. Akil, S.J. (1993) 38393847.
Watson, Pattern and time course of immediate early gene [74] M. Dobrakovova, R. Kvetnansky, T. Torda, Changes of plasma
expression in rat brain following acute stress, Neuroscience 64 and adrenal catecholamines and corticosterone in stressed rats
(1995) 477505. with septal lesions, Physiol. Behav. 29 (1982) 4145.
[57] W.E. Cullinan, J.P. Herman, S.J. Watson, Ventral subicular [75] H.W. Dong, G.D. Petrovich, L.W. Swanson, Topography of
interaction with the hypothalamic paraventricular nucleus: projections from amygdala to bed nuclei of the stria terminalis,
evidence for a relay in the bed nucleus of the stria terminalis, Brain Res. Rev. 38 (2001) 192246.
J. Comp. Neurol. 332 (1993) 120. [76] H.W. Dong, G.D. Petrovich, A.G. Watts, L.W. Swanson, Basic
[58] E.T. Cunningham Jr., P.E. Sawchenko, Anatomical specicity of organization of projections from the oval and fusiform nuclei of
noradrenergic inputs to the paraventricular and supraoptic the bed nuclei of the stria terminalis in adult rat brain, J. Comp.
nuclei of the rat hypothalamus, J. Comp. Neurol. 274 (1988) Neurol. 436 (2001) 430455.
6076. [77] W.C. Drevets, Prefrontal cortical-amygdalar metabolism in
[59] E.T. Cunningham Jr., M.C. Bohn, P.E. Sawchenko, Organiza- major depression, Ann. N. Y. Acad. Sci. 877 (1999) 614637.
tion of adrenergic inputs to the paraventricular and supraoptic [78] J.D. Dunn, Plasma corticosterone responses to electrical stim-
nuclei of the hypothalamus in the rat, J. Comp. Neurol. 292 ulation of the bed nucleus of the stria terminalis, Brain Res. 407
(1990) 651667. (1987) 327331.
[60] S.S. Daftary, C. Boudaba, J.G. Tasker, Noradrenergic regula- [79] J.D. Dunn, S.E. Orr, Dierential plasma corticosterone re-
tion of parvocellular neurons in the rat hypothalamic paraven- sponses to hippocampal stimulation, Exp. Brain Res. 54 (1984)
tricular nucleus, Neuroscience 96 (2000) 743751. 16.
[61] M.F. Dallman, N. Levin, C.S. Cascio, S.F. Akana, L. Jacobson, [80] J.D. Dunn, J. Whitener, Plasma corticosterone responses to
R.W. Kuhn, Pharmacological evidence that the inhibition of electrical stimulation of the amygdaloid complex: cytoarchitec-
diurnal adrenocorticotropin secretion by corticosteroids is med- tonic specicity, Neuroendocrinology 42 (1986) 211217.
iated via type I corticosterone-preferring receptors, Endocrinol- [81] M.H. Emmert, J.P. Herman, Dierential forebrain c-fos mRNA
ogy 124 (1989) 28442850. induction by ether inhalation and novelty: evidence for distinc-
[62] M. Davis, The role of the amygdala in fear and anxiety, Annu. tive stress pathways, Brain Res. 845 (1999) 6067.
Rev. Neurosci. 15 (1992) 353375. [82] A. Ericsson, C. Arias, P.E. Sawchenko, Evidence for an
[63] M. Davis, Are dierent parts of the extended amygdala involved intramedullary prostaglandin-dependent mechanism in the acti-
in fear versus anxiety?, Biol. Psychiatr. 44 (1998) 12391247. vation of stress-related neuroendocrine circuitry by intravenous
[64] C.V. Dayas, K.M. Buller, J.W. Crane, Y. Xu, T.A. Day, Stressor interleukin-1, J. Neurosci. 17 (1997) 71667179.
categorization: acute physical and psychological stressors elicit [83] A. Ericsson, K.J. Kovacs, P.E. Sawchenko, A functional
distinctive recruitment patterns in the amygdala and in medullary anatomical analysis of central pathways subserving the eects
noradrenergic cell groups, Eur. J. Neurosci. 14 (2001) 11431152. of interleukin-1 on stress-related neuroendocrine neurons,
[65] C.V. Dayas, K.M. Buller, T.A. Day, Neuroendocrine responses J. Neurosci. 14. (1994) 897913.
to an emotional stressor: evidence for involvement of the medial [84] S. Feldman, N. Conforti, Amygdalectomy inhibits adrenocorti-
but not the central amygdala, Eur. J. Neurosci. 11 (1999) 2312 cal responses to somatosensory and olfactory stimulation,
2322. Neuroendocrinology 32 (1981) 330334.
[66] C.V. Dayas, K.M. Buller, T.A. Day, Medullary neurones [85] S. Feldman, N. Conforti, Eects of hypothalamic deaerenta-
regulate hypothalamic corticotropin-releasing factor cell re- tions on adrenocortical responses in the rat following hippo-
sponses to an emotional stressor, Neuroscience 105 (2001) 707 campal stimulation, Exp. Brain Res. 44 (1981) 232234.
719. [86] S. Feldman, N. Conforti, A. Itzik, J. Weidenfeld, Dierential
[67] E.R. De Kloet, E. Vreugdenhil, M.S. Oitzl, M. Joels, Brain eect of amygdaloid lesions of CRF-41, ACTH and corticoste-
corticosteroid receptor balance in health and disease, Endocrine rone responses following neural stimuli, Brain Res. 658 (1994)
Rev. 19 (1998) 269301. 2126.
[68] C. Decavel, A.N. Van Den Pol, GABA: a dominant neurotrans- [87] S. Feldman, N. Conforti, E. Melamed, Paraventricular nucleus
mitter in the hypothalamus, J. Comp. Neurol. 302 (1990) 1019 serotonin mediates neurally stimulated adrenocortical secretion,
1037. Brain Res. Bull. 18 (1987) 165168.
[69] C. Decavel, A.N. Van Den Pol, Converging GABA and [88] S. Feldman, N. Conforti, D. Saphier, The preoptic area and bed
glutamate-immunoreactive axons make synaptic contact with nucleus of the stria terminalis are involved in the eects of the
identied hypothalamic neurosecretory axons, J. Comp. Neurol. amygdala on adrenocortical secretion, Neuroscience 37 (1990)
316 (1992) 104116. 775779.
174 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

[89] S. Feldman, N. Conforti, R.A. Siegel, Adrenocortical responses rone secretions is dependent on the nature of stressors, Exp.
following limbic stimulation in rats with hypothalamic deaer- Brain Res. 87 (1991) 173180.
entations, Neuroendocrinology 35 (1982) 205211. [109] R.R. Gala, U. Westphal, Corticosteroid-binding globulin in
[90] S. Feldman, M.E. Newman, E. Gur, J. Weidenfeld, Role of the rat: studies on the sex dierence, Endocrinology 77 (1965)
serotonin in the amygdala in hypothalamopituitaryadrenocor- 841851.
tical responses, NeuroReport 9 (1998) 20072009. [110] B.B. Gallagher, H.F. Flanigin, D.W. King, W.H. Littleton, The
[91] S. Feldman, D. Saphier, N. Conforti, Hypothalamic aerent eect of electrical stimulation of medial temporal lobe structures
connections mediating adrenocortical responses that follow in epileptic patients upon ACTH, prolactin, and growth
hippocampal stimulation, Exp. Neurol. 98 (1987) 103109. hormone, Neurology 37 (1987) 299303.
[92] S. Feldman, J. Weidenfeld, The dorsal hippocampus modies the [111] P.W. Gold, G.P. Chrousos, Organization of the stress system
negative feedback eect of glucocorticoids on the adrenocortical and its dysregulation in melancholic and atypical depression:
and median eminence CRF-41 responses to photic stimulation, high vs low CRH/NE states, Mol. Psychiatr. 7 (2002) 254
Brain Res. 614 (1993) 227232. 275.
[93] S. Feldman, J. Weidenfeld, Hypothalamic mechanisms mediat- [112] T.S. Gray, M.E. Carney, D.J. Magnuson, Direct projections
ing glutamate eects on the hypothalamo pituitaryadrenocor- from the central amygdaloid nucleus to the hypothalamic
tical axis, J. Neural Transm. 104 (1997) 633642. paraventricular nucleus: possible role in stress-induced adreno-
[94] S. Feldman, J. Weidenfeld, The excitatory eects of the corticotropin release, Neuroendocrinology 50 (1989) 433446.
amygdala on hypothalamopituitaryadrenocortical responses [113] T.S. Gray, R.A. Piechowski, J.M. Yracheta, P.A. Rittenhouse,
are mediated by hypothalamic norepinephrine, serotonin, and C.L. Bethea, L.D. Van de Kar, Ibotenic acid lesions in the bed
CRF-41, Brain Res. Bull. 45 (1998) 389393. nucleus of the stria terminalis attenuate conditioned stress-
[95] S. Feldman, J. Weidenfeld, Glucocorticoid receptor antagonists induced increases in prolactin, ACTH and corticosterone,
in the hippocampus modify the negative feedback following Neuroendocrinology 57 (1993) 517524.
neural stimuli, Brain Res. 821 (1999) 3337. [114] B. Greco, E.A. Allegretto, M.J. Tetel, J.D. Blaustein, Coexpres-
[96] S. Feldman, J. Weidenfeld, Electrical stimulation of the dorsal sion of ER beta with ER alpha and progestin receptor proteins in
hippocampus caused a long lasting inhibition of ACTH and the female rat forebrain: eects of estradiol treatment, Endocri-
adrenocortical responses to photic stimuli in freely moving rats, nology 142 (2001) 51725181.
Brain Res. 911 (2001) 2226. [115] J.S. Han, J.L. Bizon, H.J. Chun, C.E. Maus, M. Gallagher,
[97] K. Fendler, G. Karmos, G. Telegdy, The eect of hippocampal Decreased glucocorticoid receptor mRNA and dysfunction of
lesion on pituitaryadrenal function, Acta Physiol. Scand. 20 HPA axis in rats after removal of the cholinergic innervation to
(1961) 293297. hippocampus, Eur. J. Neurosci. 16 (2002) 13991404.
[98] M.C. Fernandez-Galaz, E. Morschl, J.A. Chowen, I. Torres- [116] R.J. Handa, L.H. Burgess, J.E. Kerr, J.A. OKeefe, Gonadal
Aleman, F. Naftolin, L.M. Garcia-Segura, Role of astroglia and steroid hormone receptors and sex dierences in the hypothal-
insulin-like growth factor-I in gonadal hormone-dependent amopituitaryadrenal axis, Horm. Behav. 28 (1994) 464476.
synaptic plasticity, Brain Res. Bull. 44 (1997) 525531. [117] J.P. Herman, Regulation of adrenocorticosteroid receptor
[99] H.F. Figueiredo, A.E. Breustle, B.L. Bodie, C.M. Dolgas, J.P. mRNA expression in the central nervous system, Cell. Mol.
Herman, The medial prefrontal cortex dierentially regulates Neurobiol. 13 (1993) 349372.
stress-induced c-fos expression in the forebrain depending on [118] J.P. Herman, W.E. Cullinan, Neurocircuitry of stress: central
type of stressor, Eur. J. Neurosci. 18 (2003) 18. control of the hypothalamopituitaryadrenocortical axis, TINS
[100] H.F. Figueiredo, C.M. Dolgas, J.P. Herman, Stress activation of 20 (1997) 7883.
cortex and hippocampus is modulated by sex and stage of estrus, [119] J.P. Herman, W.E. Cullinan, M.I. Morano, H. Akil, S.J.
Endocrinology 143 (2002) 25342540. Watson, Contribution of the ventral subiculum to inhibitory
[101] J.M. Finlay, M.J. Zigmond, E.D. Abercrombie, Increased regulation of the hypothalamopituitaryadrenocortical axis,
dopamine and norepinephrine release in medial prefrontal cortex J. Neuroendocrinol. 7 (1995) 475482.
induced by acute and chronic stress: eects of diazepam, [120] J.P. Herman, W.E. Cullinan, S.J. Watson, Involvement of the
Neuroscience 64 (1995) 619628. bed nucleus of the stria terminalis in tonic regulation of
[102] C.T. Fischette, H.M. Edinger, A. Siegel, Temporary desynchro- paraventricular hypothalamic CRH and AVP mRNA expres-
nization among circadian rhythms with lateral fornix ablation, sion, J. Neuroendocrinol. 6 (1994) 433442.
Brain Res. 229 (1981) 85101. [121] J.P. Herman, W.E. Cullinan, E.A. Young, H. Akil, S.J. Watson,
[103] C.T. Fischette, B.R. Komisaruk, H.M. Edinger, H.H. Feder, A. Selective forebrain ber tract lesions implicate ventral hippo-
Siegel, Dierential fornix ablations and the circadian rhythmicity campal structures in tonic regulation of paraventricular nucleus
of adrenal corticosteroid secretion, Brain Res. 195 (1980) 373 CRH and AVP mRNA expression, Brain Res. 592 (1992) 228
387. 238.
[104] J.T. Fitzsimons, Angiotensin, thirst, and sodium appetite, [122] J.P. Herman, W.E. Cullinan, D.R. Ziegler, J.G. Tasker, Role of
Physiol. Rev. 78 (1998) 583686. the paraventricular nucleus microenvironment in stress integra-
[105] N.S. Floyd, K.A. Keay, C.M. Arias, P.E. Sawchenko, R. tion, Eur. J. Neurosci. 16 (2002) 381385.
Bandler, Projections from the ventrolateral periaqueductal gray [123] J.P. Herman, C.M. Dolgas, S.C. Carlson, Ventral subiculum co-
to endocrine regulatory subdivisions of the paraventricular ordinates situation-specic neuroendocrine and behavioral stress
nucleus of the hypothalamus in the rat, Neurosci. Lett. 220 responses, Neuroscience 86 (1998) 449459.
(1996) 105108. [124] J.P. Herman, C.M. Dolgas, D. Rucker, M.C. Langub Jr.,
[106] C. Fortier, Dual control of adrenocorticotropin release, Endo- Localization of natriuretic peptide-activated guanylate cyclase
crinology 49 (1951) 782788. mRNAs in the rat brain, J. Comp. Neurol. 369 (1996) 165187.
[107] K. Francis, S.L. Meddle, V.R. Bishop, J.A. Russell, Progester- [125] J.P. Herman, O. Eyigor, D.R. Ziegler, L. Jennes, Expression of
one receptor expression in the pregnant and parturient rat ionotropic glutamate receptor subunit mRNAs in the hypotha-
hypothalamus and brainstem, Brain Res. 927 (2002) 1826. lamic paraventricular nucleus of the rat, J. Comp. Neurol. 422
[108] S. Gaillet, J. Lachuer, F. Malaval, I. Assenmacher, A. (2000) 352362.
Szafarczyk, The involvement of noradrenergic ascending path- [126] J.P. Herman, B.R. Larson, D.B. Speert, A.F. Seasholtz, Hypo-
ways in the stress-induced activation of ACTH and corticoste- thalamopituitaryadrenocortical dysregulation in aging F344/
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 175

Brown-Norway F1 hybrid rats, Neurobiol. Aging 22 (2001) 323 [145] G. Ju, L.W. Swanson, R.B. Simerly, Studies on the cellular
332. architecture of the bed nucleus of the stria terminalis: II.
[127] J.P. Herman, M.K.-H. Schafer, E.A. Young, R. Thompson, J. Chemoarchitecture, J. Comp. Neurol. 280 (1989) 603621.
Douglass, H. Akil, S.J. Watson, Evidence for hippocampal [146] A. Kalsbeek, R.M. Buijs, J.J. van Heerikhuize, M. Arts, T.P. van
regulation of neuroendocrine neurons of the hypothalamo der Woude, Vasopressin-containing neurons of the suprachias-
pituitaryadrenocortical axis, J. Neurosci. 9 (1989) 30723082. matic nuclei inhibit corticosterone release, Brain Res. 580 (1992)
[128] J.P. Herman, S.J. Watson, R.L. Spencer, Defense of adrenocor- 6267.
ticosteroid receptor expression in rat hippocampus: eects of [147] T. Kaneko, F. Fujiyama, Complementary distribution of vesic-
stress and strain, Endocrinology 140 (1999) 39813991. ular glutamate transporters in the central nervous system,
[129] B.A. Horger, R.H. Roth, The role of mesoprefrontal dopamine Neurosci. Res. 42 (2002) 243250.
neurons in stress, Crit. Rev. Neurobiol. 10 (1996) 395418. [148] G.J. Kant, J.L. Meyerho, L.E. Jarrard, Biochemical indices of
[130] E. Hrabovszky, I. Kallo, T. Hajszan, P.J. Shughrue, I. Merch- reactivity and habituation in rats with hippocampal lesions,
enthaler, Z. Liposits, Expression of estrogen receptor-beta Pharmacol. Biochem. Behav. 20 (1984) 793797.
messenger ribonucleic acid in oxytocin and vasopressin neurons [149] J. Kaufman, P.M. Plotsky, C.B. Nemero, D.S. Charney, Eects
of the rat supraoptic and paraventricular nuclei, Endocrinology of early adverse experiences on brain structure and function:
139 (1998) 26002604. clinical implications, Biol. Psychiatr. 48 (2000) 778790.
[131] K.M. Hurley, H. Herbert, M.M. Moga, C.B. Saper, Eerent [150] M. Kawakami, K. Seto, E. Terasawa, K. Yoshida, T. Miyamoto,
projections of the infralimbic cortex of the rat, J. Comp. Neurol. M. Sekiguchi, Y. Hattori, Inuence of electrical stimulation and
308 (1991) 249276. lesion in limbic structure upon biosynthesis of adrenocorticoid in
[132] T. Imaki, M. Naruse, S. Harada, N. Chikada, J. Imaki, H. the rabbit, Neuroendocrinology 3 (1968) 337348.
Onodera, H. Demura, W. Vale, Corticotropin-releasing factor up- [151] M. Keller-Wood, M.F. Dallman, Corticosteroid inhibition of
regulates its own receptor mRNA in the paraventricular nucleus of ACTH secretion, Endocr. Rev. 5 (1984) 124.
the hypothalamus, Mol. Brain Res. 38 (1996) 166170. [152] L. Kerkerian, G. Pelletier, Eects of monosodium L -glutamate
[133] T. Imaki, T. Shibasaki, M. Hotta, H. Demura, Intracerebroven- administration on neuropeptide Y-containing neurons in the rat
tricular administration of corticotropin-releasing factor induces hypothalamus, Brain Res. 369 (1986) 388390.
c-fos mRNA expression in brain regions related to stress [153] B.M. King, M.F. Dallman, K.R. Esquerre, L.A. Frohman,
responses: comparison with pattern of c-fos mRNA induction Radio-frequency vs. electrolytic VMH lesions: dierential eects
after stress, Brain Res. 616 (1993) 114125. on plasma hormones, Am. J. Physiol. 254 (1988) R917R924.
[134] A.M. Issa, W. Rowe, S. Gauthier, M.J. Meaney, Hypothalamic [154] K.P. Kinzig, D.A. DAlessio, J.P. Herman, R.R. Sakai, T.P.
pituitaryadrenal activity in aged, cognitively impaired and Vahl, H.F. Figueiredo, E.K. Murphy, R.J. Seeley, CNS gluca-
cognitively unimpaired rats, J. Neurosci. 10 (1990) 32473254. gon-like peptide-1 receptors mediate endocrine and anxiety
[135] P.M. Iuvone, C. Van Hartesveldt, Locomotor activity and responses to interoceptive and psychogenic stressors, J. Neuro-
plasma corticosterone in rats with hippocampal lesions, Behav. sci. 23 (2003) 61636170.
Biol. 16 (1976) 515520. [155] J.Z. Kiss, Dynamism of chemoarchitecture in the hypothalamic
[136] S. Iyengar, H.S. Kim, P.L. Wood, Mu-, delta-, kappa- and paraventricular nucleus, Brain Res. Bull. 20 (1988) 699708.
epsilon-opioid receptor modulation of the hypothalamicpitui- [156] J.I. Kitay, Sex dierences in adrenal cortical secretion in the rat,
taryadrenocortical (HPA) axis: subchronic tolerance studies of Endocrinology 68 (1961) 818824.
endogenous opioid peptides, Brain Res. 435 (1987) 220226. [157] J.I. Kitay, M.D. Coyne, N.H. Swygert, K.E. Gaines, Eects of
[137] L. Jacobson, R.M. Sapolsky, The role of the hippocampus in gonadal hormones and ACTH on the nature and rates of
feedback regulation of the hypothalamopituitaryadrenocorti- secretion of adrenalcortical steroids by the rat, Endocrinology 89
cal axis, Endocr. Rev. 12 (1991) 118134. (1971) 565570.
[138] A.S. Jansen, E.D. Schmidt, P. Voorn, F.J. Tilders, Substance [158] K.M. Knigge, Adrenocortical response to stress in rats with
induced plasticity in noradrenergic innervation of the paraven- lesions in hippocampus and amygdala, Proc. Soc. Exp. Biol.
tricular hypothalamic nucleus, Eur. J. Neurosci. 17 (2003) 298 Med. 108 (1961) 1821.
306. [159] K.M. Knigge, M. Hays, Evidence of inhibitive role of hippo-
[139] T.M. Jay, M.P. Witter, Distribution of hippocampal CA1 and campus in neural regulation of ACTH release, Proc. Soc. Exp.
subicular eerents in the prefrontal cortex of the rat studied by Biol. Med. 114 (1963) 6769.
means of anterograde transport of Phaseolus vulgaris-leucoag- [160] C. Kohler, Subicular projections to the hypothalamus and
glutinin, J. Comp. Neurol. 313 (1991) 574586. brainstem: some novel aspects revealed in the rat by the
[140] H.P. Jedema, A.F. Sved, M.J. Zigmond, J.M. Finlay, Sensitiza- anterograde Phaseolus vulgaris leukoagglutinin (PHA-L) tracing
tion of norepinephrine release in medial prefrontal cortex: eect method, Prog. Brain Res. 83 (1990) 5969.
of dierent chronic stress protocols, Brain Res. 830 (1999) 211 [161] S. Kollack-Walker, C. Don, S.J. Watson, H. Akil, Dierential
217. expression of c-fos mRNA within neurocircuits of male hamsters
[141] D.S. Jessop, D. Renshaw, P.J. Larsen, H.S. Chowdrey, M.S. exposed to acute or chronic defeat, J. Neuroendocrinol. 11
Harbuz, Substance P is involved in terminating the hypothal- (1999) 547559.
amopituitaryadrenal axis response to acute stress through [162] S. Kollack-Walker, S.J. Watson, H. Akil, Social stress in
centrally located neurokinin-1 receptors, Stress 3 (2000) 209220. hamsters: defeat activates specic neurocircuits within the brain,
[142] A.K. Johnson, J.T. Cunningham, R.L. Thunhorst, Integrative J. Neurosci. 17 (1997) 88428855.
role of the lamina terminalis in the regulation of cardiovascular [163] K. Kovacs, J.Z. Kiss, G.B. Makara, Glucocorticoid implants
and body uid homeostasis, Clin. Exp. Pharmacol. Physiol. 23 around the hypothalamic paraventricular nucleus prevent the
(1996) 183191. increase of corticotropin-releasing factor and arginine vasopres-
[143] H. Jorgensen, U. Knigge, A. Kjaer, T. Vadsholt, J. Warberg, sin immunostaining induced by adrenalectomy, Neuroendocri-
Serotonergic involvement in stress-induced ACTH release, Brain nology 44 (1986) 229234.
Res. 811 (1998) 1020. [164] K.J. Kovacs, G.B. Makara, Corticosterone and dexamethasone
[144] G. Ju, L.W. Swanson, Studies on the cellular architecture of the act at dierent brain sites to inhibit adrenalectomy-induced
bed nucleus of the stria terminalis in the rat: I. Cytoarchitecture, adrenocorticotropin hypersecretion, Brain Res. 474 (1988) 205
J. Comp. Neurol. 587-602 (1989). 210.
176 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

[165] K.E. Krout, A.D. Loewy, Parabrachial nucleus projections to dierent stress paradigms, Proc. Natl. Acad. Sci. USA 93
midline and intralaminar thalamic nuclei of the rat, J. Comp. (1996) 23592364.
Neurol. 428 (2000) 475494. [184] H.Y. Li, P.E. Sawchenko, Hypothalamic eector neurons and
[166] T.L. Kruko, K.H. Harris, J.H. Jhamandas, Eerent projections extended circuitries activated in neurogenic stress: a compar-
from the parabrachial nucleus demonstrated with the antero- ison of footshock eects exerted acutely, chronically, and in
grade tracer Phaseolus vulgaris leucoagglutinin, Brain Res. Bull. animals with controlled glucocorticoid levels, J. Comp. Neurol.
30 (1993) 163172. 393 (1998) 244266.
[167] T.L. Kruko, D. MacTavish, K.H. Harris, J.H. Jhamandas, [185] Z. Liposits, W.K. Paull, G. Setalo, S. Vigh, Evidence for local
Changes in blood volume and pressure induce c-fos expression in corticotropin releasing factor (CRF)-immunoreactive neuronal
brainstem neurons that project to the paraventricular nucleus of circuits in the paraventricular nucleus of the rat hypothalamus.
the hypothalamus, Mol. Brain Res. 34 (1995) 99108. An electron microscopic immunohistochemical analysis, Histo-
[168] S. Lacroix, S. Rivest, Functional circuitry in the brain of chemistry 83 (1985) 516.
immune-challenged rats: partial involvement of prostaglandins, [186] C.A. Lowry, Functional subsets of serotonergic neurones:
J. Comp. Neurol. 387 (1997) 307324. implications for control of the hypothalamicpituitaryadrenal
[169] N. Laamme, E. Feuvrier, D. Richard, S. Rivest, Involvement of axis, J. Neuroendocrinol. 14 (2002) 911923.
serotonergic pathways in mediating the neuronal activity and [187] P.J. Lowry, F.E. Estivariz, G.E. Gillies, A.C. Kruseman, E.A.
genetic transcription of neuroendocrine corticotropin-releasing Linton, CRF: its regulation of ACTH and pro-opiomelanocortin
factor in the brain of systemically endotoxin-challenged rats, peptide release and its extra hypothalamic occurrence, Acta
Neuroscience 88 (1999) 223240. Endocrinol. Suppl. (Copenh.) 276 (1986) 5662.
[170] R. Lamprecht, Y. Dudai, Dierential modulation of brain [188] M. Ludwig, Dendritic release of vasopressin and oxytocin,
immediate early genes by intraperitoneal LiCl, NeuroReport 7 J. Neuroendocrinol. 10 (1998) 881895.
(1995) 289293. [189] A.M. Magarinos, F. Estivariz, M.I. Morano, A.F. de Nicola,
[171] P.W. Landeld, J.C. Eldridge, The glucocorticoid hypothesis of Regulation of the central nervous systempituitaryadrenal axis
brain aging and neurodegeneration: recent modications, Acta in rats after neonatal treatment with monosodium glutamate,
Endocrinol. 125 (1991) 5464. Neuroendocrinology 48 (1988) 105111.
[172] P.W. Landeld, J.C. Waymire, G. Lynch, Hippocampal aging [190] A.M. Magarinos, G. Somoza, A.F. DeNicola, Glucocorticoid
and adrenocorticoids: quantitative correlations, Science 202 negative feedback and glucocorticoid receptors after hippocamp-
(1978) 10981102. ectomy in rats, Horm. Metab. Res. 19 (1987) 105109.
[173] M.C. Langub Jr., R.E. Watson Jr., J.P. Herman, Distribution of [191] J.A. Mansi, S. Rivest, G. Drolet, Regulation of corticotropin-
natriuretic peptide precursor mRNAs in the rat brain, J. Comp. releasing factor type 1 (CRF1) receptor messenger ribonucleic
Neurol. 356 (1995) 183199. acid in the paraventricular nucleus of rat hypothalamus by
[174] J.P. Lanier, C. Van Hartesveldt, B. Weis, R.L. Isaacson, Eects exogenous CRF, Endocrinology 137 (1996) 46194629.
of dierential hippocampal damage upon rhythmic and stress- [192] A. Marchilhac, P. Siaud, Identication of projections from the
induced corticosterone secretion in the rat, Neuroendocrinology central nucleus of the amygdala to the paraventricular nucleus of
18 (1975) 154160. the hypothalamus which are immunoreactive for corticotrophin-
[175] P.J. Larsen, D. Jessop, H. Patel, S.L. Lightman, H.S. Chowdrey, releasing hormone in the rat, Exp. Physiol. 82 (1997) 273281.
Substance P inhibits the release of anterior pituitary adrenocor- [193] M. Martinez, P.J. Phillips, J. Herbert, Adaptation in patterns of
ticotrophin via a central mechanism involving corticotrophin- c-fos expression in the brain associated with exposure to either
releasing factor-containing neurons in the hypothalamic parav- single or repeated social stress in male rats, Eur. J. Neurosci. 10
entricular nucleus, J. Neuroendocrinol. 5 (1993) 99105. (1998) 2033.
[176] P.J. Larsen, J.D. Mikkelsen, D. Jessop, S.L. Lightman, H.S. [194] J.W. Mason, Plasma 17-hydroxycorticosteroid levels during
Chowdrey, Neonatal monosodium glutamate treatment alters electrical stimulation of the amygdaloid complex in conscious
both the activity and the sensitivity of the rat hypothalamo monkeys, Am. J. Physiol. 196 (1959) 4448.
pituitaryadrenocortical axis, J. Endocrinol. 141 (1994) 497503. [195] G.K. Matheson, B.J. Branch, A.N. Taylor, Eects of amygda-
[177] P.J. Larsen, M. Tang-Christensen, J.J. Holst, C. Orskov, loid stimulation on pituitary- -adrenal activity in conscious cats,
Distribution of glucagon-like peptide-1 and other preprogluca- Brain Res. 32 (1971) 151167.
gon-derived peptides in the rat hypothalamus and brainstem, [196] A.J. McDonald, F. Mascagni, L. Guo, Projections of the medial
Neuroscience 77 (1997) 257270. and lateral prefrontal cortices to the amygdala: a Phaseolus
[178] J.C. Le Mevel, S. Abitbol, G. Beraud, J. Maniey, Temporal vulgaris leucoagglutinin study in the rat, Neuroscience 71 (1996)
changes in plasma adrenocorticotropin concentration after 5575.
repeated neurotropic stress in male and female rats, Endocrinol- [197] B.S. McEwen, Stress, adaptation, and disease. Allostasis and
ogy 105 (1979) 812817. allostatic load, Ann. N. Y. Acad. Sci. 840 (1998) 3344.
[179] J. LeDoux, Emotional networks and motor control: a fearful [198] B.S. McEwen, E. Stellar, Stress and the individual. Mechanisms
view, Prog. Brain Res. 107 (1996) 437446. leading to disease, Arch. Intern. Med. 153 (1993) 20932101.
[180] J.E. LeDoux, C. Farb, D.A. Ruggiero, Topographic organiza- [199] B.S. McEwen, J.M. Weiss, L.S. Schwartz, Selective retention of
tion of neurons in the acoustic thalamus that project to the corticosterone by limbic structures in rat brain, Nature 220
amygdala, J. Neurosci. 10 (1990) 10431054. (1968) 911912.
[181] S.F. Leibowitz, C. Sladek, L. Spencer, D. Tempel, Neuropeptide [200] J.F. McGinty, L.Y. Koda, F.E. Bloom, A combined vascular-
Y, epinephrine and norepinephrine in the paraventricular catecholamine uorescence method reveals the relative vascular-
nucleus: stimulation of feeding and the release of corticosterone, ity of rat locus coeruleus and the paraventricular and supraoptic
vasopressin and glucose, Brain Res. Bull. 21 (1988) 905912. nuclei of the hypothalamus, Neurosci. Lett. 36 (1983) 117123.
[182] S. Levine, Primary social relationships inuence the development [201] A. McGregor, J. Herbert, Dierential eects of excitotoxic
of the hypothalamicpituitaryadrenal axis in the rat, Physiol. basolateral and corticomedial lesions of the amygdala on the
Behav. 73 (2001) 255260. behavioural and endocrine responses to either sexual or aggres-
[183] H.-Y. Li, A. Ericsson, P.E. Sawchenko, Distinct mechanisms sion-promoting stimuli in the male rat, Brain Res. 574 (1992) 920.
underlie activation of hypothalamic neurosecretory neurons and [202] M.J. Meaney, J. Diorio, D. Francis, J. Widdowson, P. LaPlante,
their medullary catecholaminergic aerents in categorically C. Caldji, S. Sharma, J.R. Seckl, P.M. Plotsky, Early environ-
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 177

mental regulation of forebrain glucocorticoid receptor gene ular nucleus and pituitaryadrenocortical and sympathoadrenal
expression: implications for adrenocortical responses to stress, activity: in vivo microdialysis studies, Front. Neuroendocrinol.
Dev. Neurosci. 18 (1996) 4972. 16 (1995) 89150.
[203] R.C. Meibach, A. Siegel, Eerent connections of the hippocam- [222] K. Pacak, M. Palkovits, G. Yadid, R. Kvetnansky, I.J. Kopin,
pal formation in the rat, Brain Res. 124 (1977) 197224. D.S. Goldstein, Heterogeneous neurochemical responses to
[204] I. Merchenthaler, M. Lane, P. Shughrue, Distribution of pre- dierent stressors: a test of Selyes doctrine of nonspecicity,
pro-glucagon and glucagon-like peptide-1 receptor messenger Am. J. Physiol. 275 (1998) R1247R1255.
RNAs in the rat central nervous system, J. Comp. Neurol. 403 [223] M. Palkovits, J.S. Ba, K. Pacak, Stress-induced Fos-like
(1999) 261280. immunoreactivity in the pons and the medulla oblongata of
[205] M. Miura, K. Takayama, J. Okada, Neuronal expression of Fos rats, Stress 1 (1997) 155168.
protein in the rat brain after baroreceptor stimulation, J. Auton. [224] M. Palkovits, J.S. Ba, K. Pacak, The role of ascending
Nerv. Syst. 50 (1994) 3143. neuronal pathways in stress-induced release of noradrenaline in
[206] G.P. Moberg, U. Scapagnini, J. deGroot, W.F. Ganong, Eect the hypothalamic paraventricular nucleus of rats, J. Neuroen-
of sectioning the fornix on diurnal uctuation in plasma docrinol. 11 (1999) 529539.
corticosterone levels in the rats, Neuroendocrinology 7 (1971) [225] L. Pan, F. Gilbert, Activation of 5-HT1A receptor subtype in the
1115. paraventricular nuclei of the hypothalamus induces CRH and
[207] M.M. Moga, R.P. Weis, R.Y. Moore, Eerent projections of the ACTH release in the rat, Neuroendocrinology 56 (1992) 797802.
paraventricular thalamic nucleus in the rat, J. Comp. Neurol. [226] D. Parkes, S. Rivest, S. Lee, C. Rivier, W. Vale, Corticotropin-
359 (1995) 221238. releasing factor activates c-fos, NGFI-B, and corticotropin-
[208] F.L. Moore, S.J. Evans, Steroid hormones use non-genomic releasing factor gene expression within the paraventricular
mechanisms to control brain functions and behaviors: a review nucleus of the rat hypothalamus, Mol. Endocrinol. 7 (1993)
of evidence, Brain Behav. Evol. 54 (1999) 4150. 13571367.
[209] N.K. Mueller, M.E. Paskitti, C.M. Dolgas, B. Bodie, J.P. [227] V.K. Patchev, S. Hayashi, C. Orikasa, O.F. Almeida, Implica-
Herman, Ventral subiculum lesions alter response to stress and tions of estrogen-dependent brain organization for gender
modulate glutamic acid decarboxylase (GAD) mRNA levels in dierences in hypothalamopituitaryadrenal regulation, FAS-
limbic stress relays, Soc. Neurosci. Abs. 27 (2001) Program No. EB. J. 9 (1995) 419423.
177.8. [228] M.A. Pezzone, W.-S. Lee, G.E. Homan, K.M. Pezzone, B.S.
[210] A. Munck, P.M. Guyre, N.J. Holbrook, Physiological functions Rabin, Activation of brainstem catecholaminergic neurons by
of glucocorticoids in stress and their relations to pharmacolog- conditioned and unconditioned aversive stimuli as revealed by
ical actions, Endocr. Rev. 5 (1984) 2544. c-Fos immunoreactivity, Brain Res. 608 (1993) 310318.
[211] K.W. Nettles, C. Pesold, M.B. Goldman, Inuence of the ventral [229] M.A. Pezzone, W.-S. Lee, G.E. Homan, B.S. Rabin, Induction
hippocampal formation on plasma vasopressin, hypothalamic of c-Fos immunoreactivity in the rat forebrain by conditioned
pituitaryadrenal axis, and behavioral responses to novel and unconditioned aversive stimuli, Brain Res. 597 (1992) 4150.
acoustic stress, Brain Res. 858 (2000) 181190. [230] P.M. Plotsky, Facilitation of immunoreactive corticotropin-
[212] S.R. Ojeda, Y.J. Ma, Glialneuronal interactions in the neuro- releasing factor secretion into the hypophysial-portal circulation
endocrine control of mammalian puberty: facilitatory eects of after activation of catecholaminergic pathways or central
gonadal steroids, J. Neurobiol. 40 (1999) 528540. norepinephrine injection, Endocrinology 121 (1987) 924934.
[213] H. Okamura, M. Abitbol, J.F. Julien, S. Dumas, A. Berod, M. [231] P.M. Plotsky, E.T. Cunningham Jr., E.P. Widmaier, Catechol-
Geard, K. Kitahama, P. Bobillier, J. Mallet, L. Wiklund, aminergic modulation of corticotropin-releasing factor and
Neurons containing messenger RNA encoding glutamate decar- adrenocorticotropin secretion, Endocr. Rev. 10 (1989) 437458.
boxylase in rat hypothalamus demonstrated by in situ hybrid- [232] P.M. Plotsky, S.W. Sutton, T.O. Bruhn, A.V. Ferguson,
ization, with special emphasis on cell groups in medial preoptic Analysis of the role of angiotensin II in mediation of adreno-
area, anterior hypothalamic area and dorsomedial hypothalamic corticotropin secretion, Endocrinology 122 (1988) 538545.
nucleus, Neuroscience 39 (1990) 675699. [233] V. Prevot, Glialneuronalendothelial interactions are involved
[214] B.J. Oldeld, A. Hou-Yu, A.J. Silverman, A combined electron in the control of GnRH secretion, J. Neuroendocrinol. 14 (2002)
microscopic HRP and immunocytochemical study of the limbic 247255.
projections to rat hypothalamic nuclei containing vasopressin [234] C.M. Prewitt, J.P. Herman, Anatomical interactions between the
and oxytocin neurons, J. Comp. Neurol. 231 (1985) 221231. central amygdaloid nucleus and the hypothalamic paraventric-
[215] B.J. Oldeld, M.J. McKinley, Circumventricular organs, in: The ular nucleus of the rat: a dual tract-tracing analysis, J. Chem.
Rat Nervous System, second ed., Academic Press, San Diego, Neuroanat. 15 (1998) 173185.
1995, pp. 391403. [235] J.L. Price, Thalamus, in: The Rat Nervous System, second ed.,
[216] B.J. Oldeld, A.J. Silverman, A light microscopic HRP study of Academic Press, Sydney, 1995, pp. 629648.
limbic projections to the vasopressin- containing nuclear groups [236] S. Pu, M.R. Jain, T.L. Horvath, S. Diano, P.S. Kalra, S.P.
of the hypothalamus, Brain Res. Bull. 14 (1985) 143157. Kalra, Interactions between neuropeptide Y and gamma-ami-
[217] M. Orchinik, T.F. Murray, F.L. Moore, A corticosteroid nobutyric acid in stimulation of feeding: a morphological and
receptor in neuronal membranes, Science 252 (1991) 18481851. pharmacological analysis, Endocrinology 140 (1999) 933940.
[218] K. Otake, K. Kin, Y. Nakamura, Fos expression in aerents to [237] E.S. Redgate, E.E. Fahringer, A comparison of the pituitary
the rat midline thalamus following immobilization stress, Neu- adrenal activity elicited by electrical stimulation of preoptic,
rosci. Res. 43 (2002) 269282. amygdaloid and hypothalamic sites in the rat brain, Neuroen-
[219] K. Otake, Y. Nakamura, Single midline thalamic neurons docrinology 12 (1973) 334343.
projecting to both the ventral striatum and the prefrontal cortex [238] J.M. Reul, E.R. deKloet, Two receptor systems for corticoste-
in the rat, Neuroscience 86 (1998) 635649. rone in rat brain: microdistribution and dierential occupation,
[220] K. Pacak, M. Palkovits, Stressor specicity of central neuroen- Endocrinology 117 (1985) 25052511.
docrine responses: implications for stress-related disorders, [239] J.H. Rho, L.W. Swanson, A morphometric analysis of function-
Endocr. Rev. 22 (2001) 502548. ally dened subpopulations of neurons in the paraventricular
[221] K. Pacak, M. Palkovits, I.J. Kopin, D.S. Goldstein, Stress- nucleus of the rat with observations on the eects of colchicine,
induced norepinephrine release in the hypothalamic paraventric- J. Neurosci. 9 (1989) 13751388.
178 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

[240] L. Rinaman, Interoceptive stress activates glucagon-like peptide- [260] R.M. Sapolsky, M.P. Armanini, S.W. Sutton, P.M. Plotsky,
1 neurons that project to the hypothalamus, Am. J. Physiol. 277 Elevation of hypophysial portal concentrations of adrenocorti-
(1999) R582R590. cotropin secretagogues after fornix transection, Endocrinology
[241] P.Y. Risold, L.W. Swanson, Chemoarchitecture of the rat lateral 125 (1989) 28812887.
septal nucleus, Brain Res. Rev. 24 (1997) 91113. [261] R.M. Sapolsky, L.C. Krey, B.S. McEwen, The neuroendocri-
[242] P.Y. Risold, L.W. Swanson, Connections of the rat lateral septal nology of stress and aging: the glucocorticoid cascade hypoth-
complex, Brain Res. Rev. 24 (1997) 115195. esis, Endocr. Rev. 7 (1986) 284301.
[243] S. Rivest, How circulating cytokines trigger the neural circuits [262] R.M. Sapolsky, L.M. Romero, A.U. Munck, How do glucocor-
that control the hypothalamicpituitaryadrenal axis, Psycho- ticoids inuence stress responses? Integrating permissive, sup-
neuroendocrinology 26 (2001) 761788. pressive, stimulatory, and preparative actions, Endocr. Rev. 21
[244] C. Rivier, Role of gaseous neurotransmitters in the hypotha- (2000) 5589.
lamicpituitaryadrenal axis, Ann. N. Y. Acad. Sci. 933 (2001) [263] R.M. Sapolsky, S. Zola-Morgan, L.R. Squire, Inhibition of
254264. glucocorticoid secretion by the hippocampal formation in the
[245] C. Rivier, Role of nitric oxide and carbon monoxide in primate, J. Neurosci. 11 (1991) 36953704.
modulating the activity of the rodent hypothalamicpituitary [264] S. Sarkar, G. Legradi, R.M. Lechan, Intracerebroventricular
adrenal axis, Front. Horm. Res. 29 (2002) 1549. administration of alpha-melanocyte stimulating hormone in-
[246] B.L. Roland, P.E. Sawchenko, Local origins of some GABAergic creases phosphorylation of CREB in TRH- and CRH-producing
projections to the paraventricular and supraoptic nuclei of the neurons of the hypothalamic paraventricular nucleus, Brain Res.
hypothalamus of the rat, J. Comp. Neurol. 332 (1993) 123143. 945 (2002) 5059.
[247] B. Roozendaal, 1999 Curt P. Richter award. Glucocorticoids [265] P.E. Sawchenko, Evidence for a local site of action for
and the regulation of memory consolidation, Psychoneuroendo- glucocorticoids in inhibiting CRF and vasopressin expression
crinology 25 (2000) 213238. in the paraventricular nucleus, Brain Res. 403 (1987) 213223.
[248] B. Roozendaal, J.L. McGaugh, Basolateral amygdala lesions [266] P.E. Sawchenko, Eects of catecholamine-depleting medullary
block the memory-enhancing eect of glucocorticoid adminis- knife cuts on corticotropin-releasing factor and vasopressin
tration in the dorsal hippocampus of rats, Eur. J. Neurosci. immunoreactivity in the hypothalamus of normal and steroid-
9 (1997) 7683. manipulated rats, Neuroendocrinology 48 (1988) 459470.
[249] B.N. Roy, R.L. Reid, D.A. Van Vugt, The eects of estrogen and [267] P.E. Sawchenko, C. Arias, J.C. Bittencourt, Inhibin beta,
progesterone on corticotropin-releasing hormone and arginine somatostatin and enkephalin immunoreactivities coexist in caudal
vasopressin messenger ribonucleic acid levels in the paraventric- medullary neurons that project to the paraventricular nucleus of
ular nucleus and supraoptic nucleus of the rhesus monkey, the hypothalamus, J. Comp. Neurol. 291 (1990) 269280.
Endocrinology 140 (1999) 21912198. [268] P.E. Sawchenko, R. Benoit, M.R. Brown, Somatostatin28-
[250] R.T. Rubin, A.J. Mandell, P.H. Crandall, Corticosteroid immunoreactive inputs to the paraventricular and supraoptic
responses to limbic stimulation in man: localization of stimula- nuclei: principal origin from non-aminergic neurons in the
tion sites, Science 153 (1966) 12121215. nucleus of the solitary tract, J. Chem. Neuroanat. 1 (1988) 8194.
[251] K.G. Ruit, E.J. Neafsey, Hippocampal input to a visceral [269] P.E. Sawchenko, E.R. Brown, R.W.K. Chan, A. Ericsson, H.-Y.
motor corticobulbar pathway: an anatomical and electrophys- Li, B.K. Roland, K.J. Kovacs, The paraventricular nucleus of
iological study in the rat, Exp. Brain Res. 82 (1990) 606616. the hypothalamus and the functional neuroanatomy of viscer-
[252] H. Saito, H. Kaba, T. Sato, A. Honmura, T. Kawakami, K. omotor responses to stress, Prog. Brain Res. 107 (1996) 201222.
Seto, H. Yamamoto, M. Kawakami, Inuence of electrical [270] P.E. Sawchenko, H.Y. Li, A. Ericsson, Circuits and mechanisms
stimulation of the limbic structure on adrenocortical steroido- governing hypothalamic responses to stress: a tale of two
genesis in hypophysectomized rats, Exp. Clin. Endocrinol. 94 paradigms, Prog. Brain Res. 122 (2000) 6178.
(1989) 387390. [271] P.E. Sawchenko, L.W. Swanson, The organization of forebrain
[253] H. Saito, T. Sato, H. Kaba, F. Okutani, G.-Z. Yu, K. Seto, F. aerents to the paraventricular and supraoptic nuclei of the rat,
Kimura, M. Kawakami, S. Roberts, Inuence of the electrical J. Comp. Neurol. 218 (1983) 121144.
stimulation of the medial amygdala on adrenocortical sensitivity [272] P.E. Sawchenko, L.W. Swanson, H.W. Steinbusch, A.A. Ver-
to adrenocorticotrophin in hypophysectomized rats, Exp. Clin. hofstad, The distribution and cells of origin of serotonergic
Endocrinol. 100 (1992) 145147. inputs to the paraventricular and supraoptic nuclei of the rat,
[254] M.M. Sanchez, C.O. Ladd, P.M. Plotsky, Early adverse expe- Brain Res. 277 (1983) 355360.
rience as a developmental risk factor for later psychopathology: [273] J.S. Schwaber, B.S. Kapp, G.A. Higgins, P.R. Rapp, Amygdala
evidence from rodent and primate models, Dev. Psychopathol. and basal forebrain direct connections with the nucleus of the
13 (2001) 419449. solitary tract and the dorsal motor nucleus, J. Neurosci. 2 (1982)
[255] C.B. Saper, Central autonomic system, in: The Rat Nervous 14241438.
System, second ed., Academic Press, Sydney, 1995, pp. 107135. [274] J. Seggie, Dierential responsivity of corticosterone and prolac-
[256] C.B. Saper, A.D. Loewy, Eerent connections of the parabra- tin to stress following lesions of the septum or amygdala:
chial nucleus in the rat, Brain Res. 197 (1980) 291317. implications for psychoNeuroendocrinol, Prog. Neuropsycho-
[257] D. Saphier, S. Feldman, Eects of stimulation of the preoptic pharmacol. Biol. Psychiatr. 11 (1987) 315324.
area on hypothalamic paraventricular nucleus unit activity and [275] J. Seggie, B. Shaw, I. Uhlir, G.M. Brown, Baseline 24-hour
corticosterone secretion in freely moving rats, Neuroendocrinol- plasma corticosterone rhythm in normal, sham-operated and
ogy 42 (1986) 167173. septally-lesioned rats, Neuroendocrinology 15 (1974) 5161.
[258] D. Saphier, S. Feldman, Eects of septal and hippocampal [276] J. Seggie, I. Uhlir, G.M. Brown, Adrenal stress responses
stimuli on paraventricular nucleus neurons, Neuroscience 20 following septal lesions in the rat, Neuroendocrinology 16 (1974)
(1987) 749755. 225236.
[259] D. Saphier, J.E. Welch, G.E. Farrar, N.Q. Nguyen, F. Aguado, [277] S.R. Sesack, A.Y. Deutch, R.H. Roth, B.S. Bunney, Topo-
T.R. Thaller, D.S. Knight, Interactions between serotonin, graphical organization of the eerent projections of the medial
thyrotropin-releasing hormone, and substance P in the CNS prefrontal cortex in the rat: an anterograde tract-tracing study
regulation of adrenocortical secretion, Psychoneuroendocrinol- with Phaseolus vulgaris leucoagglutinin, J. Comp. Neurol. 290
ogy 19 (1994) 779797. (1989) 213242.
J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180 179

[278] Y.I. Sheline, P.W. Wang, M.H. Gado, J.G. Csernansky, M.W. [297] L.J. Teppema, J.G. Veening, A. Kranenburg, A. Dahan, A.
Vannier, Hippocampal atrophy in recurrent major depression, Berkenbosch, C. Olievier, Expression of c-fos in the rat brain-
Proc. Natl. Acad. Sci. USA 93 (1996) 39083913. stem after exposure to hypoxia and to normoxic and hyperoxic
[279] P.J. Shughrue, M.V. Lane, I. Merchenthaler, Comparative hypercapnia, J. Comp. Neurol. 388 (1997) 169190.
distribution of estrogen receptor-alpha and -beta mRNA in the [298] G.J. ter Horst, P.G.M. Luiten, Phaseolus vulgaris leuco-agglu-
rat central nervous system, J. Comp. Neurol. 388 (1997) 507 tinin tracing of intrahypothalamic connections of the lateral,
525. ventromedial, dorsomedial and paraventricular hypothalamic
[280] A.L. Sica, H.E. Greenberg, S.M. Scharf, D.A. Ruggiero, nuclei in the rat, Brain Res. Bull. 18 (1987) 191203.
Chronic-intermittent hypoxia induces immediate early gene [299] G.J. ter Horst, C. Streeand, Ascending projections of the
expression in the midline thalamus and epithalamus, Brain solitary tract nuclei, in: Nucleus of the Solitary Tract, CRC
Res. 883 (2000) 224228. Press, Boca Raton, 1994, pp. 93104.
[281] A.J. Silverman, A. Hou-Yu, W.P. Chen, Corticotropin-releasing [300] R.R. Terreberry, E.J. Neafsey, The rat medial frontal cortex
factor synapses within the paraventricular nucleus of the projects directly to autonomic regions of the brainstem, Brain
hypothalamus, Neuroendocrinology 49 (1989) 291299. Res. Bull. 19 (1987) 639649.
[282] R.B. Simerly, C. Chang, M. Muramatsu, L.W. Swanson, [301] K.V. Thrivikraman, C.B. Nemero, P.M. Plotsky, Sensitivity to
Distribution of androgen and estrogen receptor mRNA-con- glucocorticoid-mediated fast-feedback regulation of the hypo-
taining cells in the rat brain: an in situ hybridization study, thalamicpituitaryadrenal axis is dependent upon stressor
J. Comp. Neurol. 294 (1990) 7695. specic neurocircuitry, Brain Res. 870 (2000) 87101.
[283] W.R. Skowsky, L. Swan, P. Smith, Eects of sex steroid [302] K.V. Thrivikraman, Y. Su, P.M. Plotsky, Patterns of Fos-
hormones on arginine vasopressin in intact and castrated male immunoreactivity in the CNS induced by repeated hemorrhage
and female rats, Endocrinology 104 (1979) 105108. in conscious rats: Correlations with pituitaryadrenal axis
[284] M.A. Slusher, Eects of cortisol implants in the brainstem and activity, Stress 2 (1997) 145158.
ventral hippocampus on diurnal corticosteroid levels, Exp. Brain [303] I. Uhlir, J. Seggie, G.M. Brown, The eect of septal lesions on
Res. 1 (1966) 184194. the threshold of adrenal stress response, Neuroendocrinology 14
[285] E.M. Sternberg, J.R. Glowa, M.A. Smith, A.E. Calogero, S.J. (1974) 351355.
Listwak, S. Aksentijevich, G.P. Chrousos, R.L. Wilder, P.W. [304] Y.M. Ulrich-Lai, W.C. Engeland, Adrenal splanchnic innerva-
Gold, Corticotropin releasing hormone related behavioral and tion modulates adrenal cortical responses to dehydration stress
neuroendocrine responses to stress in Lewis and Fischer rats, in rats, Neuroendocrinology 76 (2002) 7992.
Brain Res. 570 (1992) 5460. [305] L.D. Van de Kar, A. Javed, Y. Zhang, F. Serres, D.K. Raap,
[286] E.M. Sternberg, W.S. Young 3rd, R. Bernardini, A.E. Calogero, T.S. Gray, 5-HT2A receptors stimulate ACTH, corticosterone,
G.P. Chrousos, P.W. Gold, R.L. Wilder, A central nervous oxytocin, renin, and prolactin release and activate hypothalamic
system defect in biosynthesis of corticotropin-releasing hormone CRF and oxytocin-expressing cells, J. Neurosci. 21 (2001) 3572
is associated with susceptibility to streptococcal cell wall-induced 3579.
arthritis in Lewis rats, Proc. Natl. Acad. Sci. USA 86 (1989) [306] L.D. Van de Kar, R.A. Piechowski, P.A. Rittenhouse, T.A.
47714775. Gray, Amygdaloid lesions: dierential eect on conditioned
[287] M.M. Suarez, N.I. Perassi, Inuence of anterodorsal thalami stress and immobilization-induced increases in corticosterone
nuclei on ACTH release under basal and stressful conditions, and renin secretion, Neuroendocrinology 54 (1991) 8995.
Physiol. Behav. 62 (1997) 373377. [307] A.N. van den Pol, The magnocellular and parvocellular parav-
[288] S. Suemaru, D.N. Darlington, S.F. Akana, C.S. Cascio, M.F. entricular nucleus of rat: intrinsic organization, J. Comp.
Dallman, Ventromedial hypothalamic lesions inhibit corticoste- Neurol. 206 (1982) 317345.
roid feedback regulation of basal ACTH during the trough of the [308] D. van der Kooy, L.Y. Koda, J.F. McGinty, C.R. Gerfen, F.E.
circadian rhythm, Neuroendocrinology 61 (1995) 453463. Bloom, The organization of projections from the cortex,
[289] R.M. Sullivan, A. Gratton, Lateralized eects of medial amygdala, and hypothalamus to the nucleus of the solitary tract
prefrontal cortex lesions on neuroendocrine and autonomic in rat, J. Comp. Neurol. 224 (1984) 124.
stress responses in rats, J. Neurosci. 19 (1999) 28342840. [309] T. Van Groen, J.M. Wyss, Projections from the anterodorsal and
[290] L.W. Swanson, The hypothalamus, in: Handbook of Chemical anteroventral nucleus of the thalamus to the limbic cortex in the
Neuroanatomy, Elsevier, Amsterdam, 1987, pp. 1124. rat, J. Comp. Neurol. 358 (1995) 584604.
[291] L.W. Swanson, W.M. Cowan, An autoradiographic study of the [310] V. Viau, A. Chu, L. Soriano, M.F. Dallman, Independent and
organization of the eerent connections of the hippocampal overlapping eects of corticosterone and testosterone on corti-
formation in the rat, J. Comp. Neurol. 172 (1977) 4984. cotropin-releasing hormone and arginine vasopressin mRNA
[292] L.W. Swanson, C. K ohler, A. Bjorklund, The limbic region. I. expression in the paraventricular nucleus of the hypothalamus
The septohippocampal system, in: Handbook of Chemical and stress-induced adrenocorticotropic hormone release, J.
Neuroanatomy, Elsevier, Amsterdam, 1987, pp. 125277. Neurosci. 19 (1999) 66846693.
[293] L.W. Swanson, G.D. Petrovich, What is the amygdala?, TINS 21 [311] V. Viau, M.J. Meaney, Variations in the hypothalamicpitui-
(1998) 323331. taryadrenal response to stress during the estrous cycle in the rat,
[294] A. Szafarczyk, F. Malaval, A. Laurent, R. Gibaud, I. Assenm- Endocrinology 129 (1991) 25032511.
acher, Further evidence for a central stimulatory actions of [312] V. Viau, M.J. Meaney, The inhibitory eect of testosterone on
catecholamines on adrenocorticotropin release in the rat, Endo- hypothalamopituitaryadrenal responses to stress is mediated
crinology 121 (1987) 392883. by the medial preoptic area, J. Neurosci. 16 (1996) 18661876.
[295] J.G. Tasker, C. Boudaba, L.A. Schrader, Local glutamatergic [313] B. Vigh, I. Vigh-Teichmann, Actual problems of the cerebrospinal
and GABAergic synaptic circuits and metabotropic glutamate uid-contacting neurons, Microsc. Res. Tech. 41 (1998) 5783.
receptors in the hypothalamic paraventricular and supraoptic [314] C.K. Wagner, M.J. Eaton, K.E. Moore, K.J. Lookingland,
nuclei, Adv. Exp. Med. Biol. 449 (1998) 117121. Eerent projections from the region of the medial zona incerta
[296] A.N. Taylor, G.K. Matheson, N. Dafny, Modication of the containing A13 dopaminergic neurons: a PHA-L anterograde
responsiveness of components of the limbic-midbrain circuit by tract-tracing study in the rat, Brain Res. 677 (1995) 229237.
corticosteroids and ACTH, UCLA Forum Med. Sci. 15 (1972) [315] C. Wahlestedt, G. Skagerberg, R. Ekman, M. Heilig, F. Sundler,
6778. R. Hakanson, Neuropeptide Y (NPY) in the area of the
180 J.P. Herman et al. / Frontiers in Neuroendocrinology 24 (2003) 151180

hypothalamic paraventricular nucleus activates the pituitary- [324] K. Yamada, P. Emson, T. Hokfelt, Immunohistochemical
adrenocortical axis in the rat, Brain Res. 417 (1987) 3338. mapping of nitric oxide synthase in the rat hypothalamus and
[316] I. Walaas, F. Fonnum, Biochemical evidence for glutamate as a colocalization with neuropeptides, J. Chem. Neuroanat. 10
transmitter in hippocampal eerents to the basal forebrain and (1996) 295316.
hypothalamus in the rat brain, Neuroscience 5 (1980) 16911698. [325] T. Yamamoto, N. Sako, N. Sakai, A. Iwafune, Gustatory and
[317] A.G. Watts, L.W. Swanson, G. Sanchez-Watts, Eerent projec- visceral inputs to the amygdala of the rat: conditioned taste
tions of the suprachiasmatic nucleus: studies using anterograde aversion and induction of c-fos-like immunoreactivity, Neurosci.
transport of Phaseolus vulgaris-leucoagglutinin in the rat, Lett. 226 (1997) 127130.
J. Comp. Neurol. 258 (1987) 204229. [326] D. Zelena, G.B. Makara, D. Jezova, Simultaneous blockade of
[318] J.E. Welch, G.E. Farrar, A.J. Dunn, D. Saphier, Central5-HT1A two glutamate receptor subtypes (NMDA and AMPA) results in
receptors inhibit adrenocortical secretion, Neuroendocrinology stressor-specic inhibition of prolactin and corticotropin release,
57 (1993) 272281. Neuroendocrinology 69 (1999) 316323.
[319] M.H. Whitnall, Regulation of the hypothalamic corticotropin- [327] L. Zhou, J.D. Blaustein, G.J. De Vries, Distribution of androgen
releasing hormone neurosecretory system, Prog. Neurobiol. 40 receptor immunoreactivity in vasopressin- and oxytocin-immu-
(1993) 573629. noreactive neurons in the male rat brain, Endocrinology 134
[320] M. Wilson, V. Critchlow, Eect of septal ablation on rhythmic (1994) 26222627.
pituitaryadrenal function in the rat, Neuroendocrinology 14 [328] D.R. Ziegler, W.A. Cass, J.P. Herman, Excitatory inuence of
(1974) 333344. the locus coeruleus in hypothalamicpituitaryadrenocortical
[321] M.M. Wilson, S.E. Greer, M.A. Greer, L. Roberts, Hippocam- axis responses to stress, J. Neuroendocrinol. 11 (1999) 361
pal inhibition of pituitaryadrenocortical function in female rats, 369.
Brain Res. 197 (1980) 433441. [329] D.R. Ziegler, W.E. Cullinan, J.P. Herman, Distribution of
[322] S.C. Woods, R.J. Seeley, D. Porte Jr., M.W. Schwartz, Signals vesicular glutamate transporter mRNA in rat hypothalamus,
that regulate food intake and energy homeostasis, Science 280 J. Comp. Neurol. 448 (2002) 217229.
(1998) 13781383. [330] D.R. Ziegler, J.P. Herman, Local integration of glutamate
[323] Y. Xu, T.A. Day, K.M. Buller, The central amygdala modulates signaling in the hypothalamic paraventricular region: regulation
hypothalamicpituitaryadrenal axis responses to systemic in- of glucocorticoid stress responses, Endocrinology 141 (2000)
terleukin-1beta administration, Neuroscience 94 (1999) 175183. 48014804.

Вам также может понравиться