You are on page 1of 22

126 Vermetten et al.

DEPRESSION AND ANXIETY 15:126–147 (2002)

Review Article
CIRCUITS AND SYSTEMS IN STRESS.
I. PRECLINICAL STUDIES
Eric Vermetten, M.D.,1,4 and J. Douglas Bremner, M.D.1–4

This paper reviews the preclinical literature related to the effects of stress on
neurobiological and neuroendocrine systems. Preclinical studies of stress provide
a comprehensive model for understanding neurobiological alterations in post-
traumatic stress disorder (PTSD). The pathophysiology of stress reflects long-
standing changes in biological stress response systems and in systems involved in
stress responsivity, learning, and memory. The neural circuitry involved in-
cludes systems mediating hypothalamic-pituitary-adrenal (HPA) axis, norepi-
nephrine (locus coeruleus), and benzodiazepine, serotonergic, dopaminergic,
neuropeptide, and central amino acid systems. These systems interact with brain
structures involved in memory, including hippocampus, amygdala, and prefron-
tal cortex. Stress responses are of vital importance in living organisms; however
excessive and/or repeated stress can lead to long-lasting alterations in these cir-
cuits and systems involved in stress responsiveness. Intensity and duration of the
stressor, and timing of the stressor in life, have strong impact in this
respect. Depression and Anxiety 15:126–147, 2002. © 2002 Wiley-Liss, Inc.

Key words: stress; animals; neurobiology; circuits; neuroendocrinology;


rceptors; hippocampus; amygdala; learning; memory; brain; PTSD

INTRODUCTION response, which include the hypothalamic-pituitary-


adrenal (HPA) axis and the locus coeruleus (LC). Of
O ver the past few years, our understanding of the key importance in these major systems are mediating
brain, neuroendocrine circuits, and involvement of trans- hormones and neurotransmitters, glucocorticoids,
mitter systems that are linked to the stress response has
increased substantially. Accumulated evidence has dem- 1
onstrated that stress results in long-standing changes in Department of Psychiatry and Behavioral Sciences, Emory
biological stress response systems that underlie many of University School of Medicine, Atlanta, Georgia
2
Department of Radiology, Emory University School of Medi-
the symptoms of post-traumatic stress disorder (PTSD)
cine, Atlanta, Georgia
and other stress-related disorders [see also Friedman et 3
Emory Center for Positron Emission Tomography, Emory
al., 1995; Charney et al., 1998a,b; Bremner et al., 1999a; University School of Medicine, Atlanta, Georgia
Charney and Bremner, 1999; Lopez et al., 1999; New- 4
Atlanta VAMC, Decatur, Georgia
port and Nemeroff, 2000; McEwen, 2000; Bremner and
Vermetten, 2001; Vermetten et al., 2002]. Eric Vermetten is now at the Department Psychiatry, University
While the blueprint for development of the brain is Medical Center/Utrecht Military Hospital, Utrecht, The Netherlands.
most likely specified by the genome, fine details of the
central nervous system are shaped by experience and Correspondence to: Dr. Eric Vermetten, Emory Clinical Neuro-
formed in interaction with the environment. This in- sciences Research Unit, Emory University School of Medicine/
teractive process shapes an organism’s sensitivity and Atlanta VAMC, 1256 Briarcliff Rd NE, Atlanta GA 30306.
responsivity to the environment and programs the E-mail: evermet@emory.edu
stress response system. Central aspects of the stress
Received for publication 13 April 2001; Accepted 17 September
response system involve perception of a stressor or 2001
fear-producing stimulus, processing, and transduction
of this information into neurohormonal, neurobio- DOI: 10.1002/da.10016
logical, and behavioral responses. This review focuses Published online in Wiley InterScience (www.interscience.wiley.
on neural circuits and systems involved in the stress com).

© 2002 WILEY-LISS, INC.


Review Article: Circuits and Systems in PTSD, Preclinical Data 127

norepinephrine, and benzodiazepine, serotonin, dopa- repeated stressors, such as electric (inescapable) foot-
minergic, neuropeptide, and central amino acid systems, shock or being forced to swim in cold water [e.g., van
as well as brain memory systems, such as hippocampus, Dijken et al., 1993; Marti et al., 2001]. Other fre-
amygdala, and prefrontal cortex. A knowledge of the ef- quently used models use a social stress or social defeat
fects of stress on neurobiology provides a basis for un- model [Henry, 1992; Koolhaas et al., 1997]. The stres-
derstanding the pathophysiology of PTSD. This can be sors used here can be predator exposure or varying
used to piece together a comprehensive model for how housing characteristics. The immediate and long-term
stress can affect neurobiological systems in humans and effects of defeat have been studied in a variety of re-
to formulate hypotheses for testing in patients with sponse patterns, e.g., neurobiological and behavioral
PTSD. This paper will briefly summarize preclinical characteristics of mobilization of aggression vs. avoid-
findings in animals related to brain circuits and systems ance, as well as environmental effects like environmen-
affected by stress. Studies of the neurobiology of PTSD tal enrichment vs. poor housing [Ruis et al., 1999; de
in clinical populations, as well as future avenues for the Jong et al., 2000]. Early handling has also been used as
development of psychopharmacological agents, are re- a model of stress (see below). In this model, pups are
viewed elsewhere [Vermetten and Bremner, 2002]. deprived for varying lengths of time of maternal care
(maternal deprivation) or are placed in a situation
where there is enriched maternal care (after depriva-
MODELS OF STRESS IN tion) [Meany et al., 1988; van Oers et al., 1998; Plotsky
and Meaney, 1993; Ladd et al., 1996]. By combining
ANIMAL RESEARCH various models developmental effects of stress respon-
The use of animal models is well established in the sivity have been studied [e.g., Oitzl et al., 2000]. In
study of biological bases in psychiatric disorders. De- addition, pharmacological challenges and effects of
spite traditional difficulties in accepting animal models stress-reducing agents have been tested in models that
for psychopathology that stem from the argument that have been well described and standardized.
there is no evidence for concluding that what occurs in The acute biobehavioral manifestations of stress in
the brain of the animal is equivalent to what occurs in animals are similar to symptoms of human anxiety and
the brain of a human, these models have become an in- PTSD, including trembling, hyperarousal, restlessness,
valuable tool in the analysis of the multitude of causes, increased gastrointestinal motility, increased perspira-
genetic, environmental, or pharmacological, that can tion, changes in food intake, increased defecation, sleep
bring about symptoms homologous to those of patients disturbances, deficits in memory and attention, or
with a specific disorder. Animal models can also allow avoidance of novel stimuli such as an open field [Weiss
the study of the mechanisms of specific behaviors and and Simson, 1985; Lechin et al., 1996]. The bio-
their pathophysiology, and can aid in developing and behavioral parameters parallel to some extent the main
predicting therapeutic responses to pharmacologic symptoms of PTSD in humans and can provide a frame-
agents. In these respects, animal models of stress are work for understanding the main features of the psycho-
useful for understanding the basic neurobiology of stress biological model of PTSD (fear conditioning, failure/
and for generating hypotheses about the neurobiology inhibition of extinction of fear, and stress sensitization)
of PTSD, which subsequently can be tested in popula- as described by Charney et al. [1993] (Table 1).
tions of PTSD patients [Yehuda and Antelman, 1993; It is important to understand the limitations of the
Pynoos and Ritzmann, 1996; Liberzon et al., 1997; application of animal models to PTSD. No single ani-
Rasmusson and Charney, 1997]. There is a long history mal model will address the myriad facets of PTSD or
of animal models for the study of depression [see factors that lead to a PTSD-like syndrome in humans.
McKinney and Bunney, 1969; Redei et al., 2001] and Animal models typically involve a single type of stres-
anxiety [see Treit, 1985]. Many of these animal models sor, for example, exposure to electric shock, whereas
that were developed as indirect models of depression stressors in humans are diverse and multi-factorial.
(e.g., escape behavior after footshock) can well be There are also multiple factors that influence the im-
viewed as direct models of PTSD since exposure to ex- pact of the stressor on the individual, including devel-
treme stress (typically uncontrollable) is required for the opmental epoch, coping and psychological defenses,
diagnosis of PTSD. In addition, behavior seen in ro- social support, the meaning of the event to the indi-
dents exposed to stress in many ways parallels behaviors vidual, prior exposure to stressors, and psychiatric
seen in humans with a history with PTSD [Nijenhuis et comorbidity [e.g., Bremner and Narayan, 1998, Benot-
al., 1998]. Decreased exploration in open field can be sch et al., 2000; Brady et al., 2000; Bremner and
seen as avoidance, decreased grooming and play activity Vermetten, 2001]. In addition, it should be noted that
can be seen as being cut off, and decreased responding most animal models are based on the rat and that the
for rewards and self-stimulation, analgesia, and anesthe- human brain contains a much greater complexity, spe-
sia as emotional numbing. cifically the development. Psychological factors also
One of the most popular models of animal research have to be taken into account [Weiss, 1972], which in-
that has been used is that of inescapable or restraint clude factors such as controllability and predictability
stress. In this model, animals are exposed to single or of the stressor, which can account for a differentiated
128 Vermetten et al.

TABLE 1. Relation between stress in different animal models of stress, their acute and long-term alterations, and the
parallels with symptom clusters in PTSD in humans*

Animal model Acute symptoms Long-term biobehavioral Symptoms cluster Psychobiological


of stress of stress manifestation in PTSD model of PTSD
Inescapable shock Trembling Contextual fear Increased arousal Stress sensitization
Unpredictable shock Restlessness Fear to explicitly conditioned Re-experiencing Fear conditiong
Social defeat Rapid heart rate aversive stimuli Numbing Failure of extinction/
Chronic restraint Increased perspiration Stress-induced analgesia Difficulties with attention inhibition of fear
Cold water swim Increased gastrointestinal Impairment in active avoidance and concentration
Forced swim motility Enhanced passive avoidance Avoidance of stimuli
Predator exposure Urge to urinate Freezing previously associated
Shuttlebox escape Muscle tension Learned helplessness with the trauma
Maternal deprivation Arousal Behavioral despair
Postnatal handling Shortness of breath Fear potentiated startle
Elevated T-maze
*The symptom clusters are in turn reflected in the psychobiological model of PTSD in which stress sensitization, fear conditioning, and failure of extinction/
inhibition of fear are the core concepts that capture the symptomatology. Although the stressors can have different dimensional characteristics, similar pathways
may lead to the same long-term effects on stress responsivity and subsequent biobehavioral manifestations.

response that is not always reflected in animal models. been postulated. Rearing effects have been well de-
Discrete differences in neurobiological parameters scribed as important factors that influence brain and
were described in an experiment that used inescapable behavior in mice [e.g., Greenough et al., 1973]. In ro-
stress vs. escapable stress [Shors et al., 1989]. The dents, the development of behavioral and endocrine re-
same differentiated response has been described for sponse to stress in the offspring has been shown to be
predictability of the stressor [Shors et al., 1990]. influenced by variations in maternal care, such as ma-
ternal separation, postnatal handling, or repetitive pain
[Anand et al., 1999; Francis et al., 1999; Hennessy et al.,
EARLY PROGRAMMING OF 1999; Liu et al., 2000]. These can be beneficial or
adversive according to the model used. They support
STRESS RESPONSIVITY the notion that early stress can have an effect on the set
Stress responsivity is the result of complex interac- point of neuroendocrine responses that last until adult
tions between genetic susceptibility and environmen- life. Recent research in neonatal rat pups has shown
tal stress. The challenge is to identify modifiers of the that rearing effects in the early postnatal period (post-
regulation and to design strategies to modify underly- natal handling) can also attenuate the reactivity of
ing pathways. The availability of genomic databases stress-related neuronal circuitries in the adult rat brain
for many species is uncovering networks of conserved [Abraham et al., 2000] and thus has important effects
stress-related genes within given physiological path- on the development of behavioral and endocrine re-
ways. A genetic-based model of stress responsivity can sponses to stress. For example, daily handling within
be envisaged based on the disruption of integrated ge- the first few weeks of life (picking up rat pups and then
nomic circuits that control morphogenesis of brain returning them to their mother) resulted in increased
structures, cell survival, and stress regulation. Recent hippocampal type II glucocorticoid receptor binding
work has begun to identify the various transcription that persisted throughout life. This was associated with
factors that are associated with brief or intermediate increased feedback sensitivity to glucocorticoids and re-
duration of a single or repeated stress. These studies duced glucocorticoid-mediated hippocampal damage in
suggest that dynamic interplay is involved in convert- later life [Meaney et al., 1988, 1989]. As adults, the off-
ing the transient increases in the rate of transcription spring of mothers that exhibited more licking and
into prolonged (potentially adaptive or maladaptive) grooming of pups during the first 10 days of life
changes in gene expression [Sabban and Kvetnansky, showed reduced plasma ACTH and corticosterone
2001]. Recently an animal model of congenital learned responses to acute stress, increased hippocampal gluco-
helpless (cLH) has been used to study the effects of corticoid receptor mRNA expression, enhanced gluco-
genetic disposition as a risk factor for the development corticoid feedback sensitivity, and decreased levels of
of PTSD-like behaviors. This showed physiologic hypothalamic CRF mRNA [Liu et al., 1997, 2000].
symptoms of analgesia, cognitive deficits, and hypo- These effects appear to be due to a type of “stress in-
responsivity of the HPA axis, similar to those observed oculation” from the mothers repeated licking of the
in human subjects with PTSD [King et al., 2001]. handled pups. Taken together, postnatal handling can
In addition to a genetic contribution to the regula- dampen HPA responsivity to stress, while if neonates
tion of stress responsivity, a preeminent role of early were subjected to a protracted separation from the
life events in the pathogenesis of these disorders has mother, the adult response to a stressor was exagger-
Review Article: Circuits and Systems in PTSD, Preclinical Data 129

ated, e.g., showing persistently elevated cerebrospinal trolled by hypothalamic secretion of CRF into pituitary
fluid (CSF) concentrations of CRF [Coplan et al., 1996] portal blood. However, arginine vasopressin (AVP),
and other stress-related long-term alterations [Plotsky which is co-localized in the PVN, can modulate the ac-
and Meaney, 1993; Anisman et al., 1998; Oitzl et al., tions of CRF [Sawchenko et al., 1992]. Both neuropep-
2000]. These and other findings in the effects of early tides are capable of potentiating each others’ action
life stresses suggest that early in the postnatal period and create a system in which hypothalamic control of
there is a naturally occurring brain plasticity in key neu- the pituitary-adrenocortical system has a remarkable
ral systems that may “program” an organism’s biologi- degree of redundancy [Makara, 1992]. Even though a
cal response to stressful stimuli. In addition, recent dual ACTH-secreting pathway in anterior pituitary
work on gene expression has begun to identify tran- cells has been described, which may be controlled
scription processes that are associated with the dura- separately by AVP and CRF, corticotrope cells require
tion of a single or repeated stress. These studies CRF to maintain their capacity to secrete ACTH
suggest that dynamic interplay is involved in convert- [Antoni, 1993]. When CRF is released from the hypo-
ing the transient increases in the rate of transcription thalamus, this causes release of ACTH from the ante-
into prolonged (potentially adaptive or maladaptive) rior pituitary, which stimulates release of cortisol (the
changes in gene expression [Sabban and Kvetnansky, major stress hormone) from the adrenals. This axis is
2001]. The effects may culminate in adult life or at turned “on” by stress and is regulated negatively by
senescence in amplification of individual differences in glucocorticoids in a negative feedback loop (as well as
stress responsivity. regulatory feedback with the noradrenergic system,
which is discussed in more detail below).
CRF is an amino acid-containing peptide that plays
CIRCUITS AND SYSTEMS OF a fundamental role in integrating stress-related re-
STRESS REGULATION sponses throughout the neuro-immuno-endocrine axis
[reviewed in Steckler and Holsboer, 1999]. CRF has
IN ADULT LIFE been hypothesized to have direct behavioral effects in
The corticotropin releasing factor (CRF)/HPA-axis the brain that lead to increased arousal, alertness, in-
and the autonomic nervous system are the important creased attention, and increased readiness, which to-
stress response systems that serve to maintain homeo- gether resembles anxiety-like behavior [Behan et al.,
stasis during exposure to stress. Other important 1996]. CRF is released from the PVN of the hypo-
neurohormones and transmitter systems that are re- thalamus; it activates anterior pituitary corticotropes,
viewed here are the dopaminergic system, serotonin, which results in a release of ACTH in the blood-
endogenous benzodiazepine, the central amino acids stream. In addition to the PVN of the hypothalamus,
glutamate and γ-aminobutyric acid (GABA), and neu- binding sites for CRF are located in multiple central
ropeptides like endogenous opioids, neurotensin, so- brain areas involved in fear and the stress response
matostatin, cholecystokinin (CCK), and neuropeptide (e.g., locus coeruleus) and also in the periphery
Y (NPY). The CRF/HPA-axis is mediated by the re- (spleen and direct actions on immunocytes) [De Souza
lease of CRF (sometimes referred to as “hormone”) by et al., 1991]. There are two CRF receptors, CRF1 and
paraventricular nucleus (PVN) of the hypothalamus. CRF2. The latter has two splice variants, CRF2α and
The autonomic nervous system is regulated by the LC CRF2β [Chalmers et al., 1996]. Both receptor types are
and its projecting noradrenergic brain neurons. Both distinct in their localization in the brain. CRF1 recep-
systems are key players in the stress responsivity and tors are most abundant in neocortical, cerebellar, hy-
are functionally interrelated. They both have a differ- pothalamic, pituitary, and sensory relay structures,
entiated anatomical and functional response to differ- hippocampus and amygdala; CRF2 receptors are gen-
ent kinds of stressors, e.g., the hypothalamic PVN erally localized to specific subcortical structures, most
neuron appears to be affected by multiple sources, in- notably lateral septal nuclei, choroid plexus, olfactory
cluding brainstem aminergic and peptidergic afferents; bulb, amygdaloid nuclei, and various hypothalamic areas.
blood-borne information, indirect input from limbic CRF has a higher affinity for CRF1 than CRF2. With-
system, and associated regions, including the prefrontal in the pituitary, CRF1 expression predominates over
cortex, hippocampus, and amygdala; and local-circuit CRF2 expression, suggesting that CRF1 receptors may
interactions [Herman et al., 1996]. The neurohor- mediate CRF-induced changes in ACTH release.
mones and neurotransmitters involved in stress regula- Central injection of CRF potently stimulates the re-
tion are summarized in Table 2. lease of ACTH from the pituitary [Adamec et al.,
1998], resulting in physiological and behavioral re-
CRF/HPA-AXIS sponses, which are adaptive during stress and which
Hypothalamus - CRF. The PVN is the site of the are considered to be characteristic of anxiety re-
majority of CRF-containing neurons in the hypothala- sponses. These include increased motor activity, loco-
mus and is an important site in effecting cardiovascular motion and grooming in an open field environment,
and neuroendocrine responses to stress. Adreno corti- and decrease in punished responding and time spent
cotropin hormone (ACTH) secretion is primarily con- on the open arm of an elevated T-maze. When in-
130
TABLE 2. Overview of CNS neurotransmitter and neurohormonal systems involved in stress regulation

Brain regions Neurobiological


Agent Receptor involved CNS actions effects of stress Biobehavioral symptoms Remarks
HPA-axis
CRF (AVP) CRF1 NEOCOR, CEREB, Release of ACTH in bloodstream; ACUTE/CHRONIC: Metabolic activation, arousal, Receptor block shows
HIP, AMYG, LC, stimulates NE systems via LC increased release alertness, readiness to res- elimination of ACTH
THAL,PAG pond, increased startle, response; affinity of CRF
CRF2α SUBCOR, septal retention of inhibitory for CRF1>CRF2
CRF2β nuclei, choroid avoidance response, anxiety
plexus, olf. bulb,
AMYG, HYPO
ACTH ACTH-R ACUTE/CHRONIC:
dependent of model:
increased release,
sensitized release
Glucocorticoids/
Cortisol Type I HIP and brain Regulation of energy utilization; ACUTE: Hyperarousal; fight-flight;
inhibition of growth, and increased release coping with threat,
Type II Throughout the reproductive systems; CHRONIC: classic stress effects: hyper-

Vermetten et al.
brain containment of inflammatory dependent of model: tension, diabetes, gastric
response increase or decrease ulceration, immunosupression,
osteoporosis, muscle atrophy,
in CNS: hippocampal atrophy
Locus coeruleus
NE α adrenergic LC, COR, SUBCOR, Drives up glucose metabolism ACUTE: Anxiety, fear, hypervigilance, α2-antagonist (yohimbine)
receptor HIP, HYPO, in brain regions; stimulates CRF increased turnover; hyperarousal, irritability, blocks presynaptic
(and AMYG,THAL, systems increased responsive- attention, encoding of autoreceptor; stress
subtypes) bed n. stria ness of LC neurons memories sensitization; condi-
terminalis, NA, to repeated stress tioned fear
β adrenergic desc. proj. thor. CHRONIC:
receptor spinal cord depletion
(and
subtypes)
Dopamine
DA D1 receptor PFC, NA Control of locomotion, cog- ACUTE/CHRONIC: Vigilance, paranoia?, planning, Preferential increase in PFC
D2 receptor nition, affect and neuro- increased release, monitoring functions in stress; modulation of
endocrine secretion; turnover response by NMDA and
increase the gain of neuronal opiate receptor blockade,
information processing or preadmin with Bzs
Serotonin
Serotonin 5HT1 (and mPFC, NA, AMYG, Regulation of neuroendocrine ACUTE: Anxiety, vigilance control of Learned helplessness;
subtypes) HYPO, LC, HIP function: stimulatory at PVN, increased turnover mood; perception-learning; reversibility by SSRI;
5HT2 (and and stimulatory at adrenal CHRONIC: sleep induction; regulation preadmin of TCA Bz or
subtypes) level (?) depletion; changes in of food intake; control of SSRI is preventative against
receptor density aggression decreases in serotonin
(continued)
TABLE 2. (Continued).

Brain regions Neurobiological


Agent Receptor involved CNS actions effects of stress Biobehavioral symptoms Remarks
Endogenous Bzs
Endogenous Bz receptors Throughout the ACUTE/CHRONIC: Alterations in memory; Functional coupling with
Bzs brain, in particu- decreased Bmax anxiety and fear GABA receptor; prevention
lar HIP and COR by preadmin with Bzs, or
flumazenil
Peptides
Endogenous µ MB-PAG, medulla, Decreased firing of LC; ACUTE/CHRONIC: Analgesia; numbing; Receptor antagonist naltrexone
δ n raphe, HIP, COR inhibition of HPG axis increased release encoding of memories blocks SIA
κ decrease density of
υ-receptors
NT NT receptor HYPO, lat septum, Endogenous modulator of ACUTE/CHRONIC: NT prevents gastric ulcer
AMYG, HIPP various functions (e.g., increased release formation in chronically
attenuation of DA-induced stressed animals
inhibition)
Somatostatin SRIF receptor HYPO, AMYG, HIP, Inhibitor of HG secretion ACUTE/CHRONIC: Changes in sleep pattern,
COR, median increased release changes in food intake;
preoptic area, NA changes in locomotor
activity; changes in memory
CCK4/CCK8 CCKA COR, AMYG, HIPP, Modulates activity of opoids ACUTE/CHRONIC: Anxiogenic effects, alterations Receptor block has anxiolytic
CCKB1 PAG, s nigra, raphe and DA; stimulatory increased release in attention and memory effect, including alterations
CCKB2 nuclei (CCKB pre- effect on HIP in attention and memory
dominates in the (CCKB1 effect)
brain)
NPY NPY-1 Basal ganglia, NA, Modulation of fear, ACUTE/CHRONIC: Attenuated sensitivity to
NPY-2 Caudate Putamen, spatial memory increased release stress; impairment of
AMYG, HYPO, acquisition spatial memory acquisition
COR
Central amino acids
Glutamate MPFC, HIP, NA Regulation of emotion and ACUTE: Effect of stress-induced
mood increased release increase is blocked by
CHRONIC: adrenalectomy
attenuation of
Review Article: Circuits and Systems in PTSD, Preclinical Data

increased release
GABA GABA(A) MPFC, HIP, and Regulation of emotion and ACUTE/CHRONIC: Effect of stress-induced
GABA(B) other brain mood increased turnover increase is blocked by
regions increased level of adrenalectomy; functional
neurosteroids coupling with Bz-receptor
modulation of DA neurons
by GABA-ergic system
HIP, Hippocampus; HYPO, hypothalamus; PITU, pituitary; LC, locus coeruleus; (NEO)COR, cerebral cortex; AMYG, amygdala; PFC, prefrontal cortex; NA, nucleus accumbens; MB, midbrain; THAL,
thalamus; PAG, periaquaeductal gray. Other abbreviations are explained in the text.
131
132 Vermetten et al.

jected into the amygdala, CRF causes an increase in of glucocorticoid activity will occur following chronic
the magnitude of the startle response and significantly stress [Yehuda et al., 1991].
improved retention of the inhibitory avoidance re- The HPA-axis has important functional interactions
sponse, a measure of learning and memory [Dunn and with the norepinephrine system that facilitate a so-
Berridge, 1990]. Blockade of CRF can eliminate the phisticated range of responses to stress. An enhance-
ACTH response to various stress conditions. Mice de- ment of the catecholaminergic system has stimulatory
ficient for CRF2 are found to be hypersensitive for effects on ACTH secretion. Subsequently, glucocorti-
stress and display anxiety-like behavior [Bale et al., coids inhibit stress-induced activation of catechola-
2000]. These findings suggest that CRF receptor an- mine synthesis in the PVN [Kvetansky et al., 1993;
tagonists may be useful for the treatment of disease Pacak et al., 1993]. Data obtained from adrenalecto-
states where CRF is elevated. mized rats, with or without glucocorticoid replace-
Pituitary - ACTH. Corticotropin or ACTH is a ment, and from hypercortisolemic rats suggest that
peptide that acts at the adrenal cortex to release glu- glucocorticoids feedback to inhibit CRH release in the
cocorticoids (cortisol and corticosterone), the primary PVN, via attenuation of noradrenergic activation
peripheral stress hormones. In normal circumstances, [Pacak et al., 1995]. CRF, on the other hand, increases
the rate of secretion is maintained in narrow limits; activity of the LC [Valentino and Foote, 1988; Valen-
however, conditions of physical and emotional stress tino and Wehby, 1988] and CRF injected into the LC
result in release of CRF as well as ACTH and intensifies anxiety-related responses [Butler et al.,
glucocorticoids. The ACTH receptor (ACTH-R) is 1990]. Forced swim stress selectively altered regula-
mainly expressed in the adrenocortical cells. Gluco- tion of LC sensitivity to CRF 24 hr after the swim
corticoids as well as cortisol, exerting powerful in- stress, showing that stress functionally alters CRF re-
hibitory influences on the secretion of ACT, effect ceptors that have an impact on LC activity [Curtis et
their actions intracellularly, mainly via glucocorticoid al., 1999]. These findings support the notion of stress-
receptors (GRs). The mechanism responsible for induced regulation of LC sensitivity to CRF and that
transient stress-induced hyperadrenocorticism and CRF serves as an excitatory neurotransmitter in the LC
feedback resistance may involve a downregulation of in the initiation of stress responses, which may repre-
glucocorticoid receptors [Sapolsky and Plotsky, 1990]. sent the pathway for the behavioral effects of CRF.
High glucocorticoid levels (such as those elicited by Adrenal gland - Cortisol. Adrenocortical produc-
chronic stress) decrease the number of hippocampal tion of corticosteroids is regulated by pituitary release
GRs, resulting in increased corticosterone secretion of ACTH. Exposure to acute stress results in an in-
and feedback resistance. Following stress termina- crease in glucocorticoids, which is mediated by CRF
tion, when glucocorticoid levels decrease, a physi- and ACTH [McEwen et al., 1986]. Glucocorticoid se-
ological downregulation takes place where receptor cretion is tightly regulated by negative feedback on
numbers are increased and feedback sensitivity is CRF and ACTH [McEwen, 1979]. The adrenal gland
normalized [Sapolsky et al., 1984a,b]. combines essential components of the autonomic ner-
The effects of chronic stress on ACTH and corti- vous system and the HPA axis in close contact. The
costerone secretion vary, depending on the experi- chromaffin cells of the adrenal medulla and the steroid-
mental paradigm. It has been reported that when rats producing cells of the adrenal cortex are extensively in-
were placed in a stressful environment for 24 hr with termingled and functionally interrelated. Cortisol is
intermittent additional exposure to inescapable foot- converted from cholesterol in different steps. Binding
shock, levels of plasma corticosterone were elevated sites for glucocorticoids include the type I (corticoster-
during the first days in the stressful environment but one or mineralocorticoid) receptor and type II (gluco-
returned to normal levels later. Levels of plasma corticoid) receptor, with different affinities for cortisol
ACTH were similar in stressed and control rats and the other glucocorticoids (such as dexamethasone).
throughout this time [Kant et al., 1987]. Other inves- The highest number of bindings sites for type I recep-
tigations have revealed increases of plasma corticoster- tor is in the hippocampus [Reul and de Koet, 1986],
one concentrations after exposure to acute inescapable while type II is found ubiquitously throughout the
shock. Plasma corticosterone concentrations were el- brain. The type II receptor becomes more critically in-
evated irrespective of whether the animals received volved as plasma levels rise.
single or repeated sessions of inescapable footshock Cortisol stimulates increased gluconeogenesis to
[Irwin et al., 1986]. There is also evidence that the ex- regulate energy utilization in the acute situation.
perience of prior stress results in sensitized corticos- When stress lasts, it starts to exert a role in inhibition
terone responses to a subsequent stress exposure. of growth processes, inhibition of reproductive sys-
Animals exposed to footshocks 10 days before testing tems, and containment of inflammatory responses
exhibited a higher plasma corticosterone response to [Baxter and Rousseau, 1979]. More recently a function
testing than did control animals that were not prior of cortisol role in rewarding processes has been hy-
exposed to footshocks [Dallman and Jones, 1973; pothesized, in which cortisol counteracts the aversive
Caggiula et al., 1989]. It is not exactly known which effects of external aggression and also allows a better
factors determine whether adaptation or sensitization coping with threatening situations [Piazza and Le-
Review Article: Circuits and Systems in PTSD, Preclinical Data 133

Moal, 1997]. Some studies have shown a decrease in 1980; Nisenbaum et al., 1991; Tanaka et al., 2000].
glucocorticoid levels following exposure to chronic This sensitized NE release in LC and hippocampus
stress, while others have shown an increase [see has been demonstrated to be dose-dependent in-
Bremner, 2001]. Animals with a history of prior expo- creased with subsequent stressors [Page and Aber-
sure to stress respond to subsequent stressors with a crombie, 1999], illustrating the mechanism of stress
delay in the return of glucocorticoids to baseline fol- sensitization. This mechanism refers to a stressor-in-
lowing exposure to the stressor [reviewed in Nemeroff duced increase in behavioral, physiologic, and bio-
and Schatzberg, 1988]. chemical responding to subsequent stressors of the
same or lesser magnitude. Drugs that decrease the
LOCUS COERULEUS/NOREPINEPHRINE function of the LC by interacting with inhibitory opi-
SYSTEM ate, benzodiazepine, and a2-adrenergic receptors on the
Accumulated evidence suggests a relationship be- LC decrease fearful behavior and antagonize the effects
tween alterations in noradrenergic brain systems and of electrical stimulation of the LC [Charney et al.,
stress [reviewed in Bremner et al., 1996a,b; Tanaka et 1990]. Cocaine inhibits spontaneous LC neuron activ-
al., 2000]. The majority of noradrenergic cell bodies ity, which could be reversed by the specific a2-adrener-
are located in the brainstem, in the dorsal pontine teg- gic antagonist piperoxone, and pretreatment with
mentum, with a dense network of axons that extend another a2-adrenergic antagonist, yohimbine, also at-
throughout the cerebral cortex and to multiple corti- tenuated the inhibitory effect of cocaine on the LC
cal and subcortical areas, including hippocampus, [Redmond and Huang, 1979; Pitts and Marwah, 1987].
amygdala, thalamus and hypothalamus, bed nucleus of The LC/NE and CRF/HPA-axis system mutually
stria terminalis, and nucleus accumbens, as well as de- stimulate each other and respond similarly to messen-
scending projections that synapse at the level of the gers in the neurochemical environment. What both
thoracic spinal cord [Foote et al., 1983]. The locus co- systems have in common is that they are inhibited by
eruleus/norepinephrine (LC/NE) network serves glo- opiates and γ-aminobutyric acidergic (GABA) agents,
bally as a generalized warning system and helps and excited by serotonergic and cholinergic agents,
determine whether, under threat, an individual turns and both systems are inhibited by glucocorticoids
attention towards external sensory stimuli or to inter- [Michelson et al., 1995]. They are different in their
nal vegetative states [Nakamura and Sakaguchi, 1990]. temporal response patterns: the LC/NE system is fast
The LC utilizes mainly NE as their neurotransmit- and depletes earlier than the CRF/HPA-axis that starts
ter, accounting for most of the noradrenergic input to after some minutes but lasts longer.
the limbic system [van Dongen, 1981]. Ascending NE
pathways project to the PVN and exert excitatory ef-
fects on HPA-axis. LC also uses peptides like neu- DOPAMINERGIC SYSTEM
ropeptide Y (NPY) and galanin, thereby showing In the central nervous system (CNS), dopamine is
heterogeneity of transmitter content. The neuroana- predominantly released from the ventral tegmental
tomical formation of the LC/NE system makes it well area and is involved in the control of locomotion, cog-
suited to rapidly and globally modulate brain function nition, affect, and neuroendocrine secretion [Smythe,
in response to changes in the environment. Electrical 1977; Bunny et al., 1980; Jaber et al., 1996]. Dopam-
stimulation of the LC in the cat produces a behavioral ine (DA) also has been assigned the capacity to in-
response similar to naturally occurring fear. The lim- crease the gain of neuronal information processing
bic and cortical regions innervated by the LC are [Haracz et al., 2000]. Molecular biology and pharma-
those thought to be involved in the elaboration of cology studies have revealed existence of at least five
adaptive responses to stress, such as an arched back, dopamine receptor subtypes, namely, D1, D2, D3,
piloerection, flattened ears, increased heart rate and D4, and D5. The widespread biological actions of DA
blood pressure, and mydriasis [Foote et al., 1983; are mediated by two major classes of receptor, the
Abercrombie and Jacobs, 1987a,b]. Decreasing func- D(1) class [D(1) and D(5)] and the D(2) class [D(2),
tion of the LC (interacting with inhibitory opiates, D(3), and D(4)] [LaHoste et al., 2000]. Three major
benzodiazepines, and a2-receptors on LC) results in dopaminergic neuronal systems have been described,
decreases in fearful behavior. Acute stressors such as a which include nigrostriatal (projecting from substantia
cat seeing a dog or another aggressive cat result in an nigra to striatum), mesolimbic (projection from mid-
acute increase in firing of neurons in the LC [Levine brain to nucleus accumbens), and mesocortical/meso-
et al., 1990], the hippocampus, and medial prefrontal prefrontal (projection from midbrain to prefrontal
cortex [Finlay et al., 1995]. If the cat would have been cortex) systems. DA innervation of the medial pre-
chronically stressed, it would demonstrate an en- frontal cortex (mPFC) appears to be particularly vul-
hanced NE release [Nisenbaum et al., 1991]. Repeti- nerable to even mild and brief stress. Preclinical
tive stress in the form of re-exposure to a stressor after studies are in support of the fact that both acute and
a period of time without stress results in an increased chronic stress may have a negative impact on the nor-
turnover and release of NE in the cortex, hippocam- mal function of the DA system. Sufficiently low inten-
pus, amygdala, hypothalamus, and LC [Cassens et al., sity stress (such as that associated with conditioned
134 Vermetten et al.

fear) or brief exposure to stress increases dopamine re- brain areas like hippocampus. A critical range of DA
lease and metabolism in the prefrontal cortex in the turnover is necessary for keeping this “working
absence of overt changes in other mesotelencephalic memory system” active and ready for optimal cogni-
DA regions [Deutch et al., 1985; Deutch and Roth, tive functioning [Horger and Roth, 1996], a situation
1990; Thierry et al., 1998]. Low intensity electric that is impaired in situations extreme or chronic stress
footshock increases in vivo tyrosine hydroxylase and [Arnsten, 2000]. In summary, the DA system is impor-
dopamine turnover in the mPFC, but not the nucleus tant for general emotional responses, selective infor-
accumbens or striatum [Roth et al., 1988]. Daily treat- mation processing, for hedonic impact or reward
ments of inescapable footshocks to rats results in an in- learning, and, in a broader sense, for reactivity to per-
crease level of dopamine metabolites, DOPAC, in the turbation in environmental conditions, which are es-
prefrontal cortex, and HVA in the nucleus accumbens sential functions in the ability (or failure) to cope with
[Kalivas and Duffy, 1989]. Stress can enhance DA re- the external world [reviewed in Pani et al., 2000].
lease and metabolism in other areas that receive DA
innervation, provided that greater intensity or longer SEROTONIN
duration stress is used [Roth et al., 1988; Deutch and Serotonin is involved in a wide variety of processes:
Roth, 1990]. Thus, the mPFC DA innervation is anxiety, arousal, vigilance, aggression, mood, impul-
preferentially activated by stress compared with sivity, and regulation of food-intake. The role of sero-
mesolimbic and nigrostriatal systems, whereas the tonin in stress responsivity is best understood as
mesolimbic dopamine innervation appears to be more stressor specific. Anatomical as well as functional evi-
sensitive to stress than the striatal dopamine innerva- dence is supportive of a regulatory role for serotonin
tion [Deutch et al., 1985; Abercrombie et al., 1989; on stress-induced HPA activity [Phelix et al., 1992;
Mantz et al., 1989; Deutch and Roth, 1990]. The fact Dinan, 1996]. At the same time however, serotonin is
that the dopaminergic system after stress responds, able to increase corticosterone secretion at the adrenal
preferentially, in the mPFC, is thought to serve as a level [Bagdy et al., 1989]. Animals exposed to a variety
protection against positive psychotic symptoms, since of stressors including footshock, tail shock, tail pinch,
the increased DA activity in the mPFC suppresses lim- and restraint stress have all been shown to produce an
bic DA transmission. Mesoprefrontal DA neurons lack increase in serotonin turnover in the medial prefrontal
or have decreased densities of specific autoreceptors af- cortex [Petty and Sherman, 1983; Adell et al., 1988;
fecting autoregulatory capabilities, which is thought to Pei et al., 1990; Inoue et al., 1994], nucleus ac-
contribute to the fact that mesoprefrontal DA neurons cumbens, amygdala, and lateral hypothalamus, and LC
exhibit increased rates of burst firing and DA turnover [Kaehler et al., 2000] with preferential release during
relative to other midbrain dopaminergic projections. conditioned fear in mPFC [Inoue et al., 1994]. A more
Stress-induced increases in mesoprefrontal cortical widespread activation after more severe stress is
dopamine release are susceptible to modulation by sev- thought to relate to behavioral changes that reflect an
eral neurotransmitter systems, including GABA, N- augmentation of fear; chronic electric shock produc-
methyl-D-aspartate (NMDA), serotonin, excitatory ing “learned helplessness” behavioral deficits were as-
amino acid, substance P, opiate, and noradrenergic sys- sociated with reduced in-vivo release of serotonin in
tems. Any or all of these factors can contribute to the frontal cortex [Petty et al., 1992], probably reflecting a
selective activation of mesoprefrontal DA neurons fol- situation where synthesis is not able to keep pace with
lowing exposure to low intensity stressors. Blockade of demand. After inescapable stress, 5-HT2A receptor
these neurotransmitter systems prevents stress-induced density was decreased in the hypothalamus in helpless
activation of the cortical dopamine system [Kalivas and rats as compared to control rats that were stressed but
Abhold, 1987]. Despite empirical problems, intracra- not helpless. While there were no changes in density
nial “self-stimulation” of dopaminergic systems has in 5-HT1A receptors in any brain region, decreases
been used as a model for anhedonia, or decreased plea- were found for 5-HT2A receptor density in hippocam-
sure to engage in activities, suggesting that numbing, pus and amygdala, related to stress but not related to
decreased interest, or being cut off may be related to helplessness. In mPFC, the serotonin transport sites
alterations in dopaminergic systems [Salamone et al., showed decreased density in helpless rats as compared
1997]. Serotonin is thought to play an important role to controls but not to nonhelpless rats [Wu et al.,
in lack of stimulation of dopamine release in the 1999]. Chronic restraint, however, has shown to result
nucleus accumbens, thus leading to anhedonia and lack in a decrease in 5HT1A binding in the hippocampus
of motivation, and therefore may be a target for modu- [Mendelson and McEwen, 1991; Watanabe et al.,
lation by antidepressant drugs [Zangen et al., 2001]. 1993]. Animals exposed to social stress also had an in-
Long-term treatment with serotonergic agents (imi- crease in binding of 5-HT1A receptors in hippocampus
pramine and mirtazapine) has shown to inhibit the ef- and dentate gyrus, and a decrease in 5-HT2 binding in
fect of footshock on cortical dopamine output in rats parietal cortex [McKittrick et al., 1995]. Preclinical
[Dazzi et al., 2001]. studies have provided evidence that the capability to
The prefrontal cortex has been suggested to play a increased serotonin metabolism during exposure to in-
role in “working memory” in conjunction with other escapable stress prevents helplessness [Ronan et al.,
Review Article: Circuits and Systems in PTSD, Preclinical Data 135

2000]. Serotonin antagonists produce behavioral defi- to be specific to uncontrollable stress, as opposed to
cits resembling those seen following inescapable controllable stress, and are prevented by pre-adminis-
shock. Drugs that enhance serotonin neurotransmis- tration of benzodiazepines [Drugan et al., 1984]. In
sion (SSRI) are effective in reversing the learned help- addition, animals exposed to inescapable stress exhibit
lessness [Sherman and Petty, 1980; Martin et al., decreases in binding of the Bz-receptor antagonist
1990]. Pre-administration of benzodiazepines or tricy- flumazenil [Drugan et al., 1989], which are associated
clic antidepressants has been described to prevent with deficits in learning, and decreased depolariza-
stress-induced decreases in serotonin and the acquisi- tion-induced release of GABA relative to controls
tion of behavioral deficits [Petty et al., 1992], while [Petty and Sherman, 1981]. A decrease in Bz-receptor
injection of serotonin (5HT) into the frontal cortex binding (Bmax) has been demonstrated in the so-called
after stress exposure reverses behavioral deficits [Sher- Maudsley genetically fearful strain of rat in compari-
man and Petty, 1982]. Administration of 5-HT1A ago- son to non-fearful rats in several brain structures in-
nists such as buspirone resulted in a reversal of cluding the hippocampus [Robertson et al., 1978].
stress-induced behavioral deficits [Drugan et al., 1989;
Przegalinski, et al., 1995]. A natural increase in the NEUROPEPTIDES
level of 5-HIAA in the lateral septum seemed protec- Exposure to stress has marked effects on the activity
tive from adverse behavioral consequences of inescap- of a number of other CNS neuropeptides systems.
able stress [Ronan et al., 2000]. The neuropeptides that are considered to mediate the
response to stress, based upon preclinical studies, are
ENDOGENOUS BENZODIAZEPINES CRF (see above), endogenous opioid peptides, neuro-
Endogenous benzodiazepines (Bz) also play an im- tensin, somatostatin, cholecystokinin (CCK), neuro-
portant role in the stress response and anxiety [re- peptide Y, and others, such as substance P, vasopressin,
viewed in Guidotti et al., 1990]. Bz-receptors are oxytocin, vasointestinal polypeptide (VIP), and thy-
present throughout the brain with the highest concen- rotropin-releasing hormone (TRH) [Stout et al.,
tration in cortical gray matter. Benzodiazepines poten- 1995]. Neuropeptides account for neurotransmission
tiate and prolong the synaptic actions of the inhibitory at a very large percentage of synapses in the brain.
neurotransmitter GABA. Central Bz-receptors and Since many neuropeptides are hypothalamic pituitary
GABA receptors are part of the same macromolecular hormones, directly controlling the secretion of ante-
complex. These receptors have distinct binding sites, rior pituitary hormones, they can function both as hor-
although they are functionally coupled and regulate mones in the hypothalamic-hypophysial portal system
each other in an allosteric manner. Administration of and as neurotransmitters in CNS. There is a different
inverse agonists of Bz-receptors, such as β-carboline- response of specific neuropeptides circuits to distinct
3-carboxylic acid ethyl ester (β-CCE), result in behav- stressors.
ioral and biological effects similar to those seen in Endogenous opioids. All of the endogenous
anxiety and stress, including increases in heart rate, opioids have a role in stress responsiveness. The opioid
blood pressure, plasma cortisol, and catecholamines peptides comprise three sets of peptide classes, β-en-
[Braestrup et al., 1982]. These effects are blocked by dorphin, enkephalins, and dynorphins [Olson et al.,
administration of Bz [Ninan et al., 1982] or pretreat- 1985]. The β-endorphins are secreted by the pituitary
ment with the benzodiazepine antagonist flumazenil gland and reach all tissues present in the body by diffu-
(formerly designated Ro-15-1788) [Drugan et al., sion. There are three major opioid receptor types: µ, δ,
1985]. Animals exposed to acute inescapable stress in and κ [Loh and Smith, 1990]. Naloxone or naltrexone
the form of cold swim or footshock develop a decrease is an example of a rather selective µ-receptor antago-
in Bz-receptor binding (Bmax) in frontal cortex, with nist. The design of more selective antagonists for each
mixed results for cerebral cortex, hippocampus, and of the opioid receptors has enabled more accurate dis-
hypothalamus, and no change in occipital cortex, stria- section of the role of different opioid systems. The
tum, midbrain, thalamus, cerebellum, and pons [Lippa neural circuitry of the opioid system is very complex
et al., 1978; Skerritt et al., 1981; Medina et al., and is thought to produce its action through the mid-
1983a,b; Drugan et al., 1986; Weizman et al., 1990]. brain periaquaeductal gray (PAG), projecting to the
Chronic stress in the form of footshock or cold swim medulla, including the nucleus raphe magnus, which
resulted in decreases in Bz-receptor binding in cere- sends out noradrenergic and serotonergic projections
bral cortex, frontal cortex, hippocampus, and hypo- to the spinal cord [see review Vaccarino et al., 1999]. A
thalamus, with mixed results for cerebellum, midbrain, variety of stressful stimuli have been documented to
and striatum, and no changes in occipital cortex or produce diminished responsiveness to pain or stress-
pons [Braestrup et al., 1979; Drugan et al., 1986, induced analgesia (SIA) [Terman et al., 1984]. Stress is
1989; Weizman et al., 1990; Lehmann et al., 1999]. associated with an increase in endogenous opiate re-
Decreases in Bz-receptor binding (Bmax) are associated lease [Madden et al., 1977; Grau et al., 1981] with de-
with alterations in memory manifested by deficits in creased density of µ-opiate receptors [Stuckey et al.,
maze-escape behaviors [Weizman et al., 1989; Drugan 1989], which may mediate the analgesia associated with
et al., 1989]. Changes in Bz-receptor function appear stress. The reversal of some forms of SIA by naloxone
136 Vermetten et al.

has suggested that opioids play an important role in this sized in the gastrointestinal tract and exerts its effects
response. The opioids have another role in the stress there as well as widespread in the brain. It has only
responsivity by their role to inhibit the hypothalamic- recently been suggested as a neural substrate for hu-
pituitary-gonadal (HPG) axis. Exercise or psychological man anxiety. CCK exists in the CNS both as an oc-
stress in women has been described to result in amenor- tapeptide (CCK-8) and as a tetrapeptide (CCK-4).
rhea or cause other dysregulations in fertility [Magiakou The octapeptide form CCK-8 in particular in the ce-
et al., 1997; Dobson and Smith, 2000]. β-endorphins rebral cortex, the amygdala, and the hippocampus.
[Norman and Smith, 1992] and CRF antagonists [Rivier The CCK receptors are also found in the midbrain in-
et al., 1986] have demonstrated to be able to block cluding the periaqueductal gray, substantia nigra, and
stress-induced inhibition of HPG-axis function and thus raphe nuclei. CCK is co-localized with opioid pep-
are both considered to be mediators of HPG-axis sup- tides and dopamine and modulates the activity of these
pression. CRF-opioid interactions are common in many neurotransmitter systems. Two receptor subtypes have
stress responses. been cloned; a CCKA and CCKB receptor. CCKB pre-
Neurotensin. Neurotensin (NT) also has a pri- dominates in the brain. Preclinical data suggest that
mary role as a neurotransmitter in CNS. NT and its agonists of CCKB produce anxiogenic-like effects,
receptor are distributed in hypothalamus, septum, while CCK B antagonists induce anxiolytic-like re-
amygdala, and hippocampus, and the receptors are sponses in several models of anxiety [Fekete et al.,
proximal to the cell bodies of origin of the classical 1984; Van Megen et al., 1994; Cohen et al., 1999].
neurotransmitters [Jennes et al., 1982]. The modulatory CCKB is also involved in attention and memory, in de-
effect of NT, that is, the attenuation of DA-induced in- pendence of the dopaminergic system. More recently,
hibition, has been most extensively examined [Shi and two different CCKB bindings sites have been hypoth-
Bunney, 1992]. A role for NT in stress is suggested by esized, CCKB1 and CCK B2, of which in particular
the protective effects of centrally administered NT on CCKB1 appears to be responsible for the effects of anxi-
restraint-stress-induced gastric ulcers in rats [Nemeroff ety [Dauge and Lena, 1998]. Iontophoretic administra-
et al., 1982], by its role as endogenous modulator of tion of CCK has depolarizing effects on pyramidal
body temperature or nociceptive sensory information neurons, suggesting that it may serve as an excitatory
[Kalivas et al., 1982] and by its role in the acute increase neurotransmitter [Bradwejn and DeMontigny 1984,
of the activity of mesolimbic dopamine neurons, the se- 1985]. Both CCK-4 and CCK-8 have stimulatory ef-
cretion of ACTH, and an increase in serum corticoster- fects on action potentials in the dentate gyrus of the
oids after central administration [Gudelsky et al., 1989]. hippocampus. Activation of hippocampal neurons to
This leads to another role for NT in the CNS response CCK is suppressed by low-dose benzodiazepines, more
to stress, which comes from the functional and ana- attributable to their role as anxiolytic drug than as seda-
tomical association between NT- and DA-containing tive drug [Bouthillier and DeMontigny, 1988].
neurons in the brain. Some studies have found support Neuropeptide Y. Neuropeptide Y (NPY) is one of
for increases in concentration of NT in patterned de- the most abundant neuropeptides in the brain. It is
pendence of mesotelencephalic dopaminergic neurons present in brainstem nuclei, nucleus accumbens,
by conditioned stress experiments [Deutch et al., 1987; amygdala, hypothalamus, and cerebral cortex [Adrian
Kilts et al., 1992]. et al., 1983]. Two receptor subtypes have been de-
Somatostatin. Somatostatin (somatotropin re- scribed, the Y1 and Y2 receptor [Wahlestedt et al.,
lease-inhibiting factor, SRIF) is the major inhibitor of 1990]. It is suggested to act as an endogenous anxio-
growth hormone (GH) secretion. It is found widely lytic by diminishing the effects of other peptides as
distributed in the brain with high concentrations in CRF. Direct injection of NPY in the amygdala has an
PVN of the hypothalamus, amygdala, hippocampus, anxiolytic effect [Heilig et al., 1993] and is protective
cerebral cortex, median preoptic area, and nucleus against restraint stress-induced gastric ulceration in
accumbens [Krisch, 1981]. Behavioral effects of cen- rats [Penner et al., 1993]. NPY-transgenic rats provide
trally administered SRIF include delayed extinction of a good model to examine the role of endogenous NPY
avoidant behavior, changes in sleep pattern, food in- in control of stress- and anxiety-related behaviors.
take, locomotor activity, and memory processes [Vec- Transgenic rats showed a markedly attenuated sensi-
sei et al., 1984]. SRIF appears to be released in tivity to behavioral consequences of stress; they were
response to CRF and increases in response to stress insensitive to the normal anxiogenic-like effect of re-
immobilization [Benyassi et al., 1993a]. Chronic daily straint stress and displayed absent fear suppression of
immobilization stress has resulted in an increased behavior in a punished drinking test. In these rats a
basal, as well as stress-induced SRIF release and de- selective impairment of spatial memory acquisition
creased growth hormone release; prolonged increase was found in conjunction with a decreased NPY-Y1
in SRIF has been reported to counter an increase in binding within the hippocampus, thus providing novel
GRF and suppresses GH secretion [Armario et al., evidence for a role of NPY in learning and memory
1993; Benyassi et al., 1993b]. [Thorsell et al., 2000].
Cholecystokinin. Cholecystokinin (CCK) is a Other neuropeptides. Other neuropeptides and
potent anxiogenic gastrinlike neuropeptide synthe- their role in stress responsiveness have been less stud-
Review Article: Circuits and Systems in PTSD, Preclinical Data 137

ied. However, some have important indirect contri- CENTRAL AMINO ACIDS
butions to the stress responsiveness. The neurokinin Of the central amino acids, glutamate and GABA
substance P (SP) has been previously shown to in- are the most important ones in stress research.
hibit basal HPA-axis activity [Donnerer et al., 1991]. Glutamate is the main representative of excitatory
The SP receptor is highly expressed in areas of the amino acids; GABA the prototypical inhibitory
brain that are implicated in anxiety and stress but amino acid. Both have a role in the organism’s re-
also in other areas such as the nucleus accumbens, sponse to stress and are believed to influence emotive
which mediate the motivational properties. Recent states in the organism.
data show that endogenous SP does not inhibit the Glutamate. Glutamate has a role as a neurotransmit-
initial magnitude of the HPA axis response to re- ter acting via several types of receptor, including the N-
straint stress but does act through neurokinin-1 re- methyl-D-aspartate (NMDA) receptor, non-NMDA
ceptors at a central level to reduce the duration of inotropic receptor subtypes, and glutamate receptors. It
the response to stress, suggesting that SP may be an is involved in long-term synaptic connectivity by initi-
important central agent controlling the transition be- ating long-term potentiating (LPT) and depression
tween acute and chronic stress [Jessop et al., 2000]. (LTD) and produces long-lasting changes in synaptic
Its role as a regulatory peptide has also been de-
structure and function, neuronal migration, and neu-
scribed in other studies involving SIA where it
ronal viability. Exposure to stress has been shown to
modulates sensitivity to pain [De Felipe et al., 1998]
increase the outflow of glutamate in the prefrontal
and acts as a mediator in stress induced immune ac-
cortex and hippocampus [Pepeu and Blandina, 1998].
tions [Fehder et al., 1997].
Adrenalectomy attenuated the stress-induced outflow
Vasopressin (VP) is released from the nerve termi-
of glutamate, which was abolished by glucocorticoid
nals in the pituitary and has a major role in fluid and
replacement, showing that the outflow is mediated by
electrolyte balance in response to dehydration. It also
glucocorticoids [Moghaddam et al., 1994]. Blockade of
contributes to HPA-axis regulation, potentiating the NMDA receptors can acutely increase glutamate re-
effect of CRF on ACTH release. It is also suggested lease by reducing excitation of inhibitory GABA neu-
to have an effect on the firing rate of hippocampal rons [Moghaddam et al., 1997]. Since NMDA receptors
neurons. have a unique role in modulating the mesoprefrontal
Systemic effects of oxytocin (OT) include milk let- DA-ergic response to stress experiments with NMDA
down and uterine contractility at the time of parturi- receptor antagonists have been performed to block the
tion. It also stimulates prolactin secretion and, like mesoprefrontal DA-ergic response to conditioned fear
VP, it potentiates CRF-stimulated ACTH release [Goldstein et al., 1994]. This is a potential target for
[Gibbs, 1986]. There is considerable evidence that blocking effects of stress and has been used in many
CRF and, to a lesser extent, VP and OT are not only studies [see review Zigmond et al., 1998].
involved in the regulation of ACTH secretion in re-
Excessive glutamate release, in situations of severe
sponse to stress, but are also major regulators of stress
stress, and subsequent glutamatergic neuronal stimu-
responsiveness.
Vasoactive intestinal peptide (VIP) is a peptide first lation increase the production of reactive oxygen spe-
isolated from the intestine and is distributed over the cies, which, through the process of oxidative stress,
entire body but primarily in the nervous system. It is induces excitotoxicity and neuronal damage [Coyle
important in a variety of extra-intestinal actions, and Puttfarcken 1993; Savolainen et al., 1995; Nico-
which are typical of the body’s reaction to stress, such tera et al., 1997]. This process could interfere with neu-
as lipolysis, glycogenolysis, and modulation of anterior ronal and glial proliferation and has been thought to
pituitary hormone secretion. It is released in response contribute to hippocampal neural toxicity and hippoc-
to stress and has demonstrated to have preventive ef- ampal neuronal necrosis. Protection by glutamate-re-
fects on gastric ulcer formation in a cold-restraint ceptor antagonists does not necessarily imply inhibition
stress model [Tuncel et al., 1998]. of these excitotoxic abnormalities but may render the
Thyrotropin releasing hormone (TRH) and thyroid cell more resistant to other deleterious mechanisms
stimulating hormone (TSH) have been less used in (e.g., mitochondrial injury and oxidative stress) [Ob-
animal models of stress, probably because the com- renovitch, 1999].
plexity of the TSH response to stressors indicates that GABA. Stress has been shown to increase GABA
this hormone is not an adequate index of the stress ex- turnover in frontal cortex [Otero Losada, 1988]. Two
perienced by the animals [Armario and Jolin, 1989]. receptors types have been identified, GABA(A) and
More neuropeptides are being found, such as Pro- GABA(B), of which GABA(A) is most prevalent in the
lactin (PRL)-releasing peptide (PRP), which is a re- brain and extensively studied. Alterations in GABA re-
cently discovered hypothalamic peptide [Watanobe et ceptors have been observed in response to various
al., 2000]. Stress also has important effects on the im- stressors. This is accompanied by increased levels of
mune system that are not reviewed here in detail [for neurosteroids in the cortex, acting as a potent allos-
more detail, see accompanying paper: Vermetten and teric modulator of the GABA(A) receptor that could
Bremner, 2002]. serve as a possible coping to the stress. Like with
138 Vermetten et al.

glutamate, the effect stress of induced GABA increase attenuation of the distracting properties of irrelevant
is prevented by adrenalectomy [Schwartz et al., 1987]. stimuli following stimulation of noradrenergic activity
Mesoprefrontal DA neurons are uniquely modulated [Coull, 1994]. Release of norepinephrine in projection
by GABA-ergic systems, and since they are lacking areas from the LC is thought to modulate neuronal
regulating autoreceptors, they rely heavily on the activity and gene transcription and therefore the lay-
GABA-ergic influence. ing down of memory traces. Epinephrine and norepi-
A functional role has been suggested for neuro- nephrine can therefore be considered as endogenous
steroids and GABA(A) receptors in the modulation of memory-modulators in this respect since they are re-
emotional behavior and mood. Animals isolated for 30 leased during arousing and stressful circumstances.
days immediately after weaning have exhibited an Mizoguchi et al. [2000] examined whether chronically
anxiety-like behavioral profile with reduced brain stressed (4 weeks) and recovered (10 days) rats show a
GABA(A) receptor function [Serra et al., 2000]. The working memory impairment caused by reduced DA
GABA(A) receptor has been used as a target for a transmission in the PFC, demonstrating that the stress
number of anxiolytic drugs. impaired spatial working memory as evaluated by T-
maze task and induced a marked reduction of DA
transmission in the PFC. Another important role for
SYSTEMS INVOLVED IN the medial prefrontal cortex is described in relation to
LEARNING AND MEMORY the neural basis of extinction [Morgan, 1993; Falls and
Davis, 1995]. This is discussed the accompanying pa-
Stress has effects on memory processes that are
per [Vermetten and Bremner, 2002].
regulated by neurohormonal modulation of the laying
down of memory traces. Animal studies have provided
HIPPOCAMPUS
evidence for the various ways in which exposure to
stressful experiences influences learning and memory The hippocampus, which is particularly vulnerable
formation in animals [for review, see De Wied and to stress, has a unique role in mediating new learning
Croiset, 1991]. Apart from their principal role in and memory (explicit recall), emotional memory re-
stress regulations, neurotransmitters can contribute to lated to a stressor, and modulation of neurobiological
memory formation in their role as neuromodulators systems, such as the HPA axis. Lesions of the hippoc-
and in gene transcription. In addition, neurohormones ampus have been shown to block the acquisition of
may modulate the strength of connections between conditioned fear responding to the context (i.e., the
neurons, thus influencing the mechanism by which box) of a stressor (i.e., electric footshock), suggesting
memory traces are formed and stored [reviewed in that the hippocampus plays an important role in
Martin et al., 2000]. We will briefly discuss the neuro- memory for the emotional context of a stressor [Ross
biological correlates of the effects of stress on memory et al., 1984; Phillips and LeDoux, 1992; Kim and
in animal studies. Application to human learning and Fanselow, 1992].
memory systems will be discussed elsewhere [Vermet- Recent data has demonstrated a biochemical se-
ten and Bremner, 2002]. quence of events in the rat hippocampus that is neces-
sary for memory formation of the inhibitory avoidance
MEMORY AND NORADRENERGIC behavior [Izquierdo and Medina, 1997]. These events
FUNCTION involve regulation early after training by neurohor-
Increased level of norepinephrine released during monal processes related to alertness, anxiety, and stress
stress can modulate memory formation through action (involving GABA-ergic, cholinergic, and noradrenergic
on the brain. A number of studies have shown that synapses and by connections with amygdala and possi-
norepinephrine modulates aspects of memory: acquisi- bly the medial septum) and several hours after training
tion of new information [Cole and Robbins, 1992], the by pathways related to mood and affect (involving
attentional component of memory storage [Brozoski dopamine D1, β-adrenergic, and 5HT1A receptors).
et al., 1979], and working memory [Arntsten and This activity is subsequently transmitted to other areas,
Goldman-Rakic, 1985]. Studies with humans and pri- including the source of the dopaminergic, noradrener-
mates have provided support for the view that norad- gic, and serotonergic pathways, and involves participa-
renaline is important in cognitive functions associated tion of entorhinal and posterior parietal cortex to
with the frontal lobes, particularly the prevention of complete memory consolidation. Emotionally arousing
distractibility by irrelevant stimuli. The a2-receptors situations can modulate this process of memory con-
of the prefrontal cortex appear to be of particular im- solidation [Cahill and McGaugh, 1996].
portance in this respect. Pharmacological stimulation A number of studies have shown that stressors, such
with the a2-adrenoreceptor agent yohimbine in aged as footshock or forced swim, result in deficits in new
monkeys that had a degenerated LC, with depletion of learning as measured by paradigms as maze-escape be-
noradrenaline in the prefrontal cortex, has shown to haviors in the rat [reviewed in Bremner et al., 1993,
reverse deficits in performance on a delayed response 1995]. Studies in a variety of animal species [Aus der
task. It is hypothesized that this could be explained by Muhlen and Ockenfels, 1969; Sapolsky et al., 1988]
Review Article: Circuits and Systems in PTSD, Preclinical Data 139

suggest that direct glucocorticoid exposure results in terations in hippocampal morphology [Nibuya et al.,
decreased dendritic branching [Woolley et al., 1990] 1995, 1996; Smith et al., 1995; Schaaf et al., 2000].
and a loss of neurons [Uno et al., 1990], which are ste- Following termination of stress and of corticosterone
roid- and tissue-specific [Sapolsky, 1985; Packan and treatment in rats, signs of structural reorganization in
Sapolsky, 1990]. Monkeys who died spontaneously hippocampal neurons and synapses have been re-
following exposure to severe stress were found on au- ported, correlating with performance on learning and
topsy to have multiple gastric ulcers, consistent with memory tests [Sousa et al., 2000].
exposure to chronic stress, and hyperplastic adrenal
cortices, consistent with sustained glucocorticoid re- AMYGDALA
lease. High levels of glucocorticoids released during The amygdala has long been implicated in condi-
stress in these monkeys were also associated with dam- tioned fear [Davis, 1992] and has a role in influencing
age to the CA3 region of the hippocampus [Uno et al., memory-storage processes in other brain regions, such
1989; Sapolsky et al., 1990; Sapolsky, 1996], associated as the hippocampus, striatum, and neocortex [re-
with direct exposure of glucocorticoids to the hippoc- viewed in Cahill and McGaugh, 1998]. Conditioned
ampus [Sapolsky et al., 1988]. Damage to this region fear, in which pairing of a neutral (“conditioned”)
of the hippocampus has been associated with deficits stimulus to a fear-inducing (“unconditioned”) stimulus
in memory [Arbel et al., 1994; Luine et al., 1994; results in fear responses to the neutral (“conditioned”)
McEwen et al., 1992; Sapolsky, 2000]. Prenatal expo- stimulus alone, has been used as a probe of amygdala
sure to elevated levels of glucocorticoids has also function [LeDoux et al., 1990; Davis, 1992]. Lesions
shown to result in hippocampal damage [Uno et al., of the central nucleus of the amygdala have been
1990]. Corticosterone treatment can also induce these shown to completely block fear conditioning [Hitch-
neuropathological alterations [Woolley et al., 1990]. cock and Davis, 1986; Hitchcock et al., 1989], while
Long-term subcutaneous implants of glucocorticoids, electrical stimulation of the central nucleus increases
which mimic the chronic stress situation, were shown acoustic startle [Rosen and Davis, 1988].
to result in deficits in new learning and memory for The central nucleus of the amygdala projects to a
maze-escape behaviors. Moreover, the magnitude of variety of brain structures via the stria terminalis and
deficits in new learning of T-maze escape behaviors the ventral amygdalofugal pathway. One pathway is
was correlated with the number of damaged cells in from the central nucleus to the brainstem startle reflex
the CA3 region of the hippocampus [Arbel et al., circuit (nucleus reticularis pontis caudalis) [Davis,
1994]. Applying chronic psychosocial stress as well as 1992]. Pathways from the amygdala to the lateral hy-
long-term cortisol treatment for 15 weeks to tree pothalamus effect peripheral sympathetic responses to
shrews impaired hippocampus-mediated memory with stress [Iwata et al., 1986]. Early findings have been re-
a trend towards a reduction of the hippocampal vol- ported of electrical stimulation of the amygdala in
ume [Ohl et al., 2000]. These studies provided evi- cats, which resulted in peripheral signs of autonomic
dence in an animal model that the hippocampus is hyperactivity and fear-related behaviors seen in the
affected by stress and stress hormones and that the wild when the animal is being attacked or is attacking,
stress-induced damage to the hippocampus, which can including alerting, chewing, salivation, piloerection,
be reflected in its volume, is associated with deficits in turning, facial twitching, arching of the back, hissing,
memory function. and snarling, associated with an increase in catechola-
Recently, neurons within the hippocampus (dendate mine turnover [Hilton and Zbrozna, 1963]. Electrical
gyrus) were found to be unique within the brain in stimulation of the amygdala in human subjects re-
showing the capacity to regenerate themselves [Gould sulted in signs and symptoms of fear and anxiety in-
et al., 1998]. Studies in animals have demonstrated cluding an increase in heart rate and blood pressure,
several agents (e.g., phenytoin, tianeptine, dihydro- increased muscle tension, subjective sensations of fear
epiandosterone, and fluoxetine) with potentially ben- or anxiety [Chapman et al., 1954], and increases in pe-
eficial effects on the reversibility of the glucocorticoid
ripheral catecholamines [Gunne and Reis, 1963].
mediated hippocampal toxicity [Watanabe et al., 1992;
These findings demonstrate that the amygdala plays an
Duman et al., 1997]. These in-vitro studies report on
neurogenesis in the hippocampus through a regulation important role in conditioned fear and emotional re-
of brain-derived neurotrophic factor (BDNF) and sponses, as well as modulating peripheral stress re-
cAMP by SSRI, with long-term effects on brain func- sponses. There are also important connections between
tion [Duman et al., 1999]. Only chronic administra- cortical association areas, thalamus and amygdala that
tion of these drugs could induce the changes in are important in shaping the emotional valence of the
morphology [Nibuya et al., 1996]. cognitive response to stressful stimuli. In addition to
Emerging studies have found evidence for the idea thalamo-cortico-amygdala connections, there are direct
that other factors besides glucocorticoids, such as pathways from thalamus to amygdala, which could ac-
BDNF, trkB mRNA, and Nerve Growth Factor (NGF), count for fear responses below the level of conscious
which have a regulatory effect on neuronal morphol- awareness [Romanski and LeDoux, 1992].
ogy and proliferation, may mediate stress-induced al- Several neurotransmitter systems are involved in
140 Vermetten et al.

function of the amygdala. The mesencephalic dopam- from animal stress studies, between animals and be-
inergic system provides a rich innervation to the tween studies, it is important to know about the rearing
amygdala [Fallon and Ciofi, 1992]. Specific activation conditions.
of the amygdaloid dopaminergic system has been re- The work in animal models of stress has shown that
ported to occur in response to conditioned fear-arous- coordinated functional interactions between the HPA-
ing stimuli [Coco et al., 1992], suggesting that dopamine axis and LC/NE system are critical in promoting
release in the amygdala on the D1 receptor may con- adaptive responses to stress, anxiety, or fear. These
tribute to the acquisition and/or expression of Pavlov- two systems may differentially be employed by differ-
ian conditioned fear [Guarraci et al., 1999]. Preclinical ent classes of stressors, e.g., acute vs. chronic; with or
studies in rats have also pointed to a role for β-adreno- without prior exposure; escapable or inescapable stress;
receptors in the basolateral nucleus of the amygdala stressors that involve interpretation, and stressors that
(BLA) in mediating acute effects of epinephrine and glu- do not involve interpretation of the stressful stimulus.
cocorticoids on memory consolidation [McGaugh et al., These stressors all excite different neural circuits, e.g.,
1996; Roozendaal, 2000]. This system mediates the in- severe physiologic stress has shown to trigger brainstem
fluence of other neuromodulatory systems such as opioid systems that project directly to the PVN, but stressors
peptidergic and GABA-ergic systems on memory storage that involve interpretation may require interaction with
[Ferry et al., 1999]. Lesions of the BLA or infusions of β- homeostatic information prior to promoting an HPA
adrenoceptor antagonists into the BLA appear to block response and reach the PVN by way of multisynaptic
the memory-modulatory effects of systemic injections of limbic pathways. They also differ in their mobilization
glucocorticoids. When shortly after the training GR of a multiplicity of neurotransmitters and neuropep-
agonists are administered directly into the BLA an en- tides, allowing for a differential pattern of release in re-
hanced memory consolidation is seen, whereas a GR an- sponse to a wide range of conditions, with involvement
tagonist impair this process. Lesions of the BLA or of different brain structures.
inactivation of β-adrenoceptors within the BLA also Chronic stress can cause structural as well as func-
block the memory-modulatory effects of intrahippo- tional changes in the brain, involving the suppression
campal administration of a GR agonist or antagonist, in- of ongoing neurogenesis in the dental gyrus of the
dicating that the BLA interacts with the hippocampus in hippocampus, which is reflected in long-term changes
mediating glucocorticoid-induced modulatory influences in change brain structure (neuronal loss and loss of
on memory consolidation [Roozendaal, 2000]. dendritic branching) and function (impairments in
memory function).
The review is not conclusive. It has highlighted the
CONCLUDING REMARKS main findings in the key systems in preclinical studies
Stress leads to long-term dysregulation of neuro- in stress to lay the foundation for a working model for
biological systems. Repeated stressors result in a po- human studies in PTSD. In addition it provides
tentiated release of transmitters with exposure to knowledge to better understand the effects of stress on
subsequent stressors (stress sensitization). These neu- the brain and delineates where pharmacological devel-
rochemical changes are accompanied by behavioral opments might be possible. Studies of the neurobiol-
changes (fear conditioning and learned helplessness) ogy of PTSD in clinical populations are reviewed
that correlate with specific biological changes, are elsewhere [Vermetten and Bremner, 2002]. Future
similar to symptoms of human anxiety, and have an animal studies will be needed to further understand
impact on areas of the brain that regulate processes of when stress responsivity is adaptive and when it is
extinction (extinction of fear) [Weis and Simson, maladaptive. These issues will help to further under-
1985; Charney et al., 1993; Morgan et al., 1993; stand the nature of stress responsivity and prove the
Lechin et al., 1996]. basis for understanding findings in clinical research of
While the brain is the starting point for stress stress related disorders.
responsivity, the brain has also shown to be a key target
organ for stress-related changes, e.g., through gluco-
corticoids, which contribute to its set point of function- REFERENCES
ing, with subsequent adult HPA-axis activity, behavior, Abercrombie ED, Jacobs BL. 1987a. Single-unit response of norad-
and cognition. As discussed, the set point of the HPA renergic neurons in the locus coeruleus of freely moving cats. I.
responsivity to stress appears to be programmed early Acutely presented stressful and nonstressful stimuli. J Neurosci
in life [Shanks et al., 2000], potentially affecting devel- 7:2837–2843.
oping organ systems, and permanently altering struc- Abercrombie ED, Jacobs BL. 1987b. Single-unit response of nora-
drenergic neurons in the locus coeruleus of freely moving cats.
ture and function throughout life [Welberg and Seckl, II. Adaptation to chronically presented stressful stimuli. J
2001]. Key targets for this programming include GR Neurosci 7:2844–2848.
gene expression and the CRF system. Animals’ prior Abercrombie ED, Keefe KA, DiFrischia DS, Zigmond MJ. 1989.
experiences to the stress experiment can therefore have Differential effect of stress on in vivo dopamine release in stria-
important implications for the interpretation of various tum, nucleus accumbens, and medial frontal cortex. J Neuro-
data obtained in stress research. To compare results chem 52:1655–1658.
Review Article: Circuits and Systems in PTSD, Preclinical Data 141

Abraham IM, Kovacs KJ. 2000. Postnatal handling alters the activa- matostatin in the acute immobilization stress-induced GH de-
tion of stress-related neuronal circuitries. Eur J Neurosci crease in rat. Life Sci 52:361–370.
12:3003–3014. Benyassi A, Roussel JP, Rougeot C, Gavalda A, Astier H, Arancibia
Adamec R, Kent P, Anisman H, Shallow T, Merali Z. 1998. Neural S. 1993b. Chronic stress affects in vivo hypothalamic somatosta-
plasticity, neuropeptides and anxiety in animals: implications for tin release but not in vitro GH responsiveness to somatostatin in
understanding and treating affective disorder following traumatic rats. Neurosci Lett 159:166–170.
stress in humans. Neurosci Biobehav Rev 23:301–318. Bouthillier A, De Montigny C. 1988. Long-term benzodiazepine
Adell A, Garcia-Marquez C, Armario A, Gelpi E. 1988. Chronic treatment reduces neuronal responsiveness to cholecystokinin:
stress increases serotonin and noradrenaline in rat brain and sen- an electrophysiological study in the rat. Eur J Pharmacol
sitizes their responses to a further acute stress. J Neurochem 151:135–138.
50:1678–1681. Bradwejn J, De Montigny C. 1984. Benzodiazepines antagonize
Adrian TE, Allen JM, Bloom SR, Ghatei MA, Rossor MN, Roberts cholecystokinin-induced activation of rat hippocampal neurones.
GW, Crow TJ, Tatemoto K, Polak JM. 1983. Neuropeptide Y Nature 312:363–364.
distribution in human brain. Nature 306:584–686. Bradwejn J, De Montigny, C. 1985. Effects of PK 8165, a partial
Anand KJ, Coskun V, Thrivikraman KV, Nemeroff CB, Plotsky benzodiazepine receptor agonist, on cholecystokinin-induced ac-
PM. 1999. Long-term behavioral effects of repetitive pain in tivation of hippocampal pyramidal neurons: a microionto-
neonatal rat pups. Physiol Behav 66:627–637. phoretic study in the rat. Eur J Pharmacol 112:415–418.
Anisman H, Zaharia MD, Meaney MJ, Merali Z. 1998. Do early- Brady KT, Killeen TK, Brewerton T, Lucerini S. 2000a. Co-
life events permanently alter behavioral and hormonal responses morbidity of psychiatric disorders and posttraumatic stress disor-
to stressors? Int J Dev Neurosci 16:149–164. der. J Clin Psychiatry 61 Suppl 7:22–32.
Antoni FA. 1993. Vasopressinergic control of pituitary adreno- Braestrup C, Nielsen M, Nielsen EB, Lyon M. 1979. Benzodiaz-
corticotropin secretion comes of age. Front Neuroendocrinol epine receptors in the brain as affected by different experimental
14:76–122. stresses: the changes are small and not undirectional. Psychop-
Arbel I, Kadar T, Silbermann M, Levy A. 1994. The effects of harmacology (Berlin). 65:273–277.
long-term corticosterone administration on hippocampal mor- Braestrup C, Schmiechen R, Neef G, Nielsen M, Petersen, EN.
phology and cognitive performance of middle-aged rats. Brain 1982. Interaction of convulsive ligands with benzodiazepine re-
Res 19:227–235. ceptors. Science 216:1241–1243.
Armario A, Jolin T. 1989. Influence of intensity and duration of Bremner JD, Scott TM, Delaney RC, Southwick SM, Mason JW,
exposure to various stressors on serum TSH and GH levels in Johnson DR, Innis RB, McCarthy G, Charney DS. 1993. Defi-
adult male rats. Life Sci 44:215–221. cits in short-term memory in posttraumatic stress disorder. Am J
Armario A, Marti O, Gavalda A, Giralt M, Jolin T. 1993. Effects of Psychiatry 150:1015–1019.
chronic immobilization stress on GH and TSH secretion in the Bremner JD, Krystal JH, Southwick SM, Charney DS. 1995. Func-
rat: response to hypothalamic regulatory factors. Psychoneuro- tional neuroanatomical correlates of the effects of stress on
endocrinology 18:405–413. memory. J Trauma Stress 8:527–554.
Arnsten AF, Goldman-Rakic PS. 1985. Catecholamines and cogni- Bremner JD, Krystal JH, Southwick SM, Charney DS. 1996a. No-
tive decline in aged nonhuman primates. Ann NY Acad Sci radrenergic mechanisms in stress and anxiety. I. Preclinical stud-
444:218–234. ies. Synapse 23:28–38.
Arnsten AF. 2000. Stress impairs prefrontal cortical function in rats Bremner JD, Krystal JH, Southwick SM, Charney DS. 1996b. No-
and monkeys: role of dopamine D1 and norepinephrine alpha-1 radrenergic mechanisms in stress and anxiety. II. Clinical studies.
receptor mechanisms. Prog Brain Res 126:183–192. Synapse 23:39–51.
Aus der Muhlen K, Ockenfels H. 1969. Morphologische Verander- Bremner JD, Narayan M. 1998. The effects of stress on memory and
ungen im Diencephalon und Telencephalon nach Störungen des the hippocampus throughout the life cycle: implications for child-
Regelkreises Adenohypophyse-Nebennierenrinde. III. Ergeb- hood development and aging. Dev Psychopathol 10:871–885.
nisse beim Meerschweinchen nach Verabreichung von Cortison Bremner JD, Southwick SM, Charney DS. 1999a. The neurobiol-
und Hydrocortison. Z Zellforsch 93:126–138. ogy of posttraumatic stress disorder: An integration of animal
Bagdy G, Calogero, AE, Murphy DL, Szemeredi K. 1989. Seroto- and human research. In: Saigh PA, Bremner JD, editors. Post-
nin agonists cause parallel activation of the sympathoadreno- traumatic stress disorder: a comprehensive text. New York: Allyn
medullary system and the hypothalamo-pituitary- adrenocortical and Bacon. p 103–143.
axis in conscious rats. Endocrinology 125:2664–2669. Bremner JD. 2001. Hypotheses and controversies related to effects
Bale TL, Contarino A, Smith GW, Chan R, Gold LH, Sawchenko of stress on the hippocampus: an argument for stress-induced
PE, Koob GF, Vale WW, Lee KF. 2000. Mice deficient for corti- damage to the hippocampus in patients with posttraumatic stress
cotropin-releasing hormone receptor-2 display anxiety-like be- disorder. Hippocampus 11:75–81.
haviour and are hypersensitive to stress. Nat Genet 24:410–414. Bremner JD, Vermetten E. 2001. Stress and development: behav-
Baxter JD, Rousseau GG. 1979.Glucocorticoid hormone action: an ioral and biological consequences. Dev Psychopathol 13:473–489
overview. Monogr Endocrinol 12:1–24. Brozoski T J, Brown RM, Rosvold HE, Goldman PS. 1979. Cogni-
Behan DP, Grigoriadis DE, Lovenberg T, Chalmers D, Heinrichs tive deficit caused by regional depletion of dopamine in prefron-
S, Liaw C, De Souza EB. 1996. Neurobiology of corticotropin tal cortex of rhesus monkey. Science 205:929–932.
releasing factor (CRF) receptors and CRF- binding protein: im- Bunney, BS, Grace AA, Hommer DW. 1980. Changing concepts of
plications for the treatment of CNS disorders. Mol Psychiatry nigral dopamine system function within the basal ganglia: rel-
1:265–277. evance to extrapyramidal disorders. J Neural Transm Suppl
Benotsch EG, Brailey K, Vasterling JJ, Uddo M, Constans JI, 16:17–23.
Sutker PB. 2000. War zone stress, personal and environmental Butler PD, Weiss JM, Stout JC, Nemeroff CB. 1990. Corticotro-
resources, and PTSD symptoms in Gulf War veterans: a longitu- pin-releasing factor produces fear-enhancing and behavioral acti-
dinal perspective. J Abnorm Psychol 109:205–213. vating effects following infusion into the locus coeruleus. J
Benyassi A, Gavalda A, Armario A, Arancibia S. 1993a. Role of so- Neurosci 10:176–183.
142 Vermetten et al.

Caggiula AR, Antelman SM, Aul E, Knopf S, Edwards DJ. 1989. Davis M. 1992. The role of the amygdala in fear-potentiated
Prior stress attenuates the analgesic response but sensitizes the startle: implications for animal models of anxiety. Trends
corticosterone and cortical dopamine responses to stress 10 days Pharmacol Sci 13:35–41.
later. Psychopharmacology (Berlin) 99:233–237. Dazzi L, Spiga F, Pira L, Ladu S, Vacca G, Rivano A, Jentsch JD,
Cahill L, McGaugh JL. 1996. Modulation of memory storage. Curr Biggio G. 2001. Inhibition of stress- or anxiogenic-drug-induced
Opin Neurobiol 6:237–242. increases in dopamine release in the rat prefrontal cortex by
Cahill L, McGaugh JL. 1998. Mechanisms of emotional arousal long-term treatment with antidepressant drugs. J Neurochem
and lasting declarative memory. Trends Neurosci 21:294–299. 76:1212–1220.
Cassens G, Roffman M, Kuruc A, Schildkraut JJ. 1980. Alterations De Felipe C, Herrero JF, O’Brien JA, Palmer JA, Doyle CA, Smith
in brain norepinephrine metabolism induced by environmental AJ, Laird JM. Belmonte C, Cervero F, Hunt SP. 1998. Altered
stimuli previously paired with inescapable shock. Science 209: nociception, analgesia and aggression in mice lacking the recep-
1138–1140. tor for substance P. Nature 392:394–397.
Chalmers DT, Lovenberg TW, Grigoriadis DE, Behan DP, De Souza de Jong IC, Prelle IT, van de Burgwal JA, Lambooij E, Korte SM,
EB. 1996. Corticotrophin-releasing factor receptors: from molecu- Blokhuis HJ, Koolhaas JM. 2000. Effects of environmental en-
lar biology to drug design. Trends Pharmacol Sci 17:166–172. richment on behavioral responses to novelty, learning, and
Chapman WP, Schoeder HR, Gruyer G, Brazier MAB, Fager C, memory, and the circadian rhythm in cortisol in growing pigs.
Poppen JL, Solomon HC, Yakolev PI. 1954. Physiological evi- Physiol Behav 68:571–578.
dence concerning the importance of the amygdaliod nuclear re- De Souza EB, Grigoriadis DE, Webster EL. 1991. Role of brain,
gion in the integration of circulating functions and emotion in pituitary and spleen corticotropin-releasing factor receptors in
man. Science 129:949–950. the stress response. Methods Achiev Exp Pathol 14:23–44.
Charney DS, Woods SW, Price LH, Goodman WK, Glazer WM, Deutch AY, Tam SY, Roth RH. 1985. Footshock and conditioned
Heninger GR. 1990. Noradrenergic dysregualtion in panic dis- stress increase 3,4-dihydroxyphenylacetic acid (DOPAC) in the
order. In: Ballenger JC, editor. Neurobiology of panic disorders. ventral tegmental area but not substantia nigra. Brain Res
New York: Alan R. Liss. p 91–105. 333:143–146.
Charney DS, Deutch AY, Krystal JH, Southwick SM, Davis M. Deutch AY, Bean AJ, Bissette G, Nemeroff CB, Robbins RJ, Roth
1993. Psychobiologic mechanisms of posttraumatic stress disor- RH. 1987. Stress-induced alterations in neurotensin, somatosta-
der. Arch Gen Psychiatry 50:295–305. tin and corticotropin-releasing factor in mesotelencephalic
Charney DS, Grillon CC, Bremner JD. 1998a. The neurobiologi- dopamine system regions. Brain Res 417:350–354.
cal basis of anxiety and fear: Circuits, mechanisms, and neuro- Deutch AY, Roth RH. 1990. The determinants of stress-induced
chemical interactions (Part I). Neuroscientist 4:35–44. activation of the prefrontal cortical dopamine system. Prog Brain
Charney DS, Grillon CC, Bremner JD. 1998b. The neurobiologi- Res 85:367–402.
cal basis of anxiety and fear: Circuits, mechanisms, and neuro- Dinan TG. 1996. Serotonin and the regulation of hypothalamic-
chemical interactions (Part II). Neuroscientist 4:122–132. pituitary-adrenal axis function. Life Sci 58:1683–1694.
Charney DS, Bremner JD. 1999. Psychobiology of posttraumatic Dobson H, Smith RF. 2000. What is stress, and how does it affect
stress disorder. In: Bunney S, Nestler E, Charney DS, editors. reproduction? Anim Reprod Sci 60–61:743–752.
Neurobiology of psychiatric disorders. New York: Oxford Press. Donnerer J, Amann R, Skofitsch G, Lembeck F. 1991. Substance P
p 494–517. afferents regulate ACTH-corticosterone release. Ann NY Acad
Coco ML, Kuhn CM, Ely TD, Kilts CD. 1992. Selective activation Sci 632:296–303.
of mesoamygdaloid dopamine neurons by conditioned stress: at- Drugan RC, Ryan SM, Minor TR, Maier SF. 1984. Librium pre-
tenuation by diazepam. Brain Res 590:39–47 vents the analgesia and shuttlebox escape deficit typically ob-
Cohen H, Kaplan Z, Kotler M. 1999. CCK-antagonists in a rat served following inescapable shock. Pharmacol Biochem Behav
exposed to acute stress: implication for anxiety associated with 21:749–754.
post-traumatic stress disorder. Depress Anxiety 10:8–17. Drugan RC, Maier SF, Skolnick P, Paul SM, Crawley JN. 1985. An
Cole BJ, Robbins TW. 1992. Forebrain norepinephrine: role in anxiogenic benzodiazepine receptor ligand induces learned help-
controlled information processing in the rat. Neuropsycho- lessness. Eur J Pharmacol 31:453–457.
pharmacology 7:129–142. Drugan RC, Basile AS, Crawley JN, Paul SM, Skolnick P. 1986.
Coplan JD, Andrews MW, Rosenblum LA, Owens MJ, Friedman Inescapable shock reduces Ro 5-4864 binding to “peripheral-
S, Gorman JM, Nemeroff CB. 1996. Persistent elevations of type” benzodiazepine receptors in the rat. Pharmacol Biochem
cerebrospinal fluid concentrations of corticotropin-releasing fac- Behav 24:1673–1677.
tor in adult nonhuman primates exposed to early-life stressors: Drugan RC, Morrow AL, Weizman R, Weizman A, Deutsch SI,
implications for the pathophysiology of mood and anxiety disor- Crawley JN, Paul SM. 1989. Stress-induced behavioral depres-
ders. Proc Natl Acad Sci USA 93:1619–1623. sion in the rat is associated with a decrease in GABA receptor-
Coull JT. 1994. Pharmacological manipulations of the a2-noradren- mediated chloride ion flux and brain benzodiazepine receptor
ergic system: effects on cognition. Drugs Aging 5:116–126. occupancy. Brain Res 487:45–51.
Coyle JT, Puttfarcken P. 1993. Oxidative stress, glutamate, and Duman RS, Heninger GR, Nestler EJ. 1997. A molecular and cel-
neurodegenerative disorders. Science 262:689–695. lular theory of depression. Arch Gen Psychiatry 54:597–606.
Curtis AL, Pavcovich LA, Valentino, RJ. 1999. Long-term regula- Duman RS, Malberg J, Thome J. 1999. Neural plasticity to stress
tion of locus ceruleus sensitivity to corticotropin- releasing fac- and antidepressant treatment. Biol Psychiatry 46:1181–1191.
tor by swim stress. J Pharmacol Exp Ther 289:1211–1219. Dunn AJ, Berridge CW. 1990. Physiological and behavioral re-
Dallman MF, Jones MT. 1973. Corticosteroid feedback control of sponses to corticotropin-releasing factor administration: is CRF a
ACTH secretion: effect of stress-induced corticosterone secre- mediator of anxiety or stress response? Brain Res Rev 15:71–100.
tion on subsequent stress responses in the rat. Endocrinology Fallon JH, Ciofi P. 1992. Distribution of monoamines within the
92:1367–1375. amygdala. In: Aggleton JP, editor. The amygdala: neurobiologi-
Dauge V, Lena I. 1998. CCK in anxiety and cognitive processes. cal aspects of emotion, memory and mental dysfunction. New
Neurosci Biobehav Rev 22:815–825. York: Wiley. p 97–114.
Review Article: Circuits and Systems in PTSD, Preclinical Data 143

Falls WA, Davis M. 1995. Behavioral and physiological analysis of Hennessy MB, Davis HN, McCrea AE, Harvey AT, Williams MT.
fear inhibition. In: Neurobiological and clinical consequences of 1999. Short- and long-term consequences of corticotropin-releas-
stress: from normal adaptation to PTSD. Friedman MJ, Charney ing factor in early development. Ann NY Acad Sci 897:76–91.
DS, Deutch AY, editors. New York: Raven Press. p 177–202. Henry JP. 1992. Biological basis of the stress response. Integr
Fehder WP, Sachs J, Uvaydova M, Douglas SD. 1997. Substance P Physiol Behav Sci 27:66–83.
as an immune modulator of anxiety. Neuroimmunomodulation Herman JP, Prewitt CM, Cullinan WE. 1996. Neuronal circuit
4:42–48. regulation of the hypothalamo-pituitary-adrenocortical stress
Fekete M, Lengyel A, Hegedus B, Penke B, Zarandy M, Toth G, axis. Crit Rev Neurobiol 10:371–394.
Telegdy G. 1984. Further analysis of the effects of cholecystoki- Hilton SM, Zbrozyna AW. 1963. Amygdaloid region for defense
nin octapeptides on avoidance behaviour in rats. Eur J Pharma- reactions and it efferent pathway to the brain stem. J Physiol
col 98:79–91. 165:160–173.
Ferry B, Roozendaal B, McGaugh JL. 1999. Role of norepineph- Hitchcock J, Davis M. 1986. Lesions of the amygdala, but not of
rine in mediating stress hormone regulation of long-term the cerebellum or red nucleus, block conditioned fear as mea-
memory storage: a critical involvement of the amygdala. Biol sured with the potentiated startle paradigm. Behav Neurosci
Psychiatry 46:1140–1152. 100:11–22.
Finlay JM, Zigmond MJ, Abercrombie ED. 1995. Increased Hitchcock JM, Sananes CB, Davis M. 1989. Sensitization of the
dopamine and norepinephrine release in medial prefrontal cortex startle reflex by footshock: blockade by lesions of the central
induced by acute and chronic stress: effects of diazepam. Neuro- nucleus of the amygdala or its efferent pathway to the brainstem.
science 64:619–628. Behav Neurosci 103:509–518.
Foote SL, Bloom FE, Aston-Jones G. 1983. Nucleus locus coer- Horger BA, Roth RH. 1996. The role of mesoprefrontal dopamine
uleus: new evidence of anatomical and physiological specificity. neurons in stress. Crit Rev Neurobiol 10:395–418.
Physiol Rev 63:844–914. Inoue T, Tsuchiya K, Koyama T. 1994. Regional changes in
Francis D, Diorio J, Liu D, Meaney MJ. 1999. Nongenomic trans- dopamine and serotonin activation with various intensity of
mission across generations of maternal behavior and stress re- physical and psychological stress in the rat brain. Pharmacol
sponses in the rat. Science 286:1155–1158. Biochem Behav 49:911–920.
Friedman MJ, Charney DS, Deutch AY. 1995. Neurobiological and Irwin J, Ahluwalia P, Anisman H. 1986. Sensitization of norepi-
clinical consequences of stress: from normal adaptation to nephrine activity following acute and chronic footshock. Brain
PTSD. New York: Raven Press. Res 379:98–103.
Gibbs DM. 1986. Stress-specific modulation of ACTH secretion Iwata J, LeDoux JE, Meeley MP, Arneric S, Reis DJ. 1986. Intrinsic
by oxytocin. Neuroendocrinology 42:456–458. neurons in the amygdaloid field projected to by the medial gen-
Goldstein LE, Rasmusson AM, Bunney BS, Roth RH. 1994. The iculate body mediate emotional responses conditioned to acous-
NMDA glycine site antagonist (+)-HA-966 selectively regulates tic stimuli. Brain Res 383:195–214.
conditioned stress-induced metabolic activation of the meso- Izquierdo I, Medina JH. 1997. Memory formation: the sequence of
prefrontal cortical dopamine but not serotonin systems: a behav- biochemical events in the hippocampus and its connection to activ-
ioral, neuroendocrine, and neurochemical study in the rat. J ity in other brain structures. Neurobiol Learn Mem 68:285–316.
Neurosci 14:4937–4950. Jaber M, Robinson SW, Missale C, Caron MG. 1996. Dopamine re-
Gould E, Tanapat P, McEwen BS, Flugge G, Fuchs E. 1998. Prolif- ceptors and brain function. Neuropharmacology 35:1503–1519.
eration of granule cell precursors in the dentate gyrus of adult Jennes L, Stumpf WE, Kalivas PW. 1982. Neurotensin: topo-
monkeys is diminished by stress. Proc Natl Acad Sci USA graphical distribution in rat brain by immunohistochemistry. J
95:3168–3171. Comp Neurol 210:211–224.
Grau JW, Hyson RL, Maier SF, Madden J, Barchas JD. 1981. Jessop DS, Renshaw D, Larsen PJ, Chowdrey HS, Harbuz MS.
Long-term stress-induced analgesia and activation of the opiate 2000. Substance P is involved in terminating the hypothalamo-
system. Science 213:1409–1411. pituitary-adrenal axis response to acute stress through centrally
Greenough WT, Volkmar FR, Juraska JM. 1973. Effects of rearing located neurokinin-1 receptors. Stress 3:209–220.
complexity on dendritic branching in frontolateral and temporal Kaehler ST, Singewald N, Sinner C, Thurnher C, Philippu A.
cortex of the rat. Exp Neurol 41:371–378. 2000. Conditioned fear and inescapable shock modify the release
Guarraci FA, Frohardt RJ, Kapp BS. 1999. Amygdaloid D1 dopam- of serotonin in the locus coeruleus. Brain Res 859:249–254.
ine receptor involvement in Pavlovian fear conditioning. Brain Kalivas PW, Jennes L, Nemeroff CB, Prange AJ. 1982. Neuro-
Res 827:28–40. tensin: topographical distribution of brain sites involved in hypo-
Gudelsky GA, Berry SA, Meltzer HY. 1989. Neurotensin activates thermia and antinociception. J Comp Neurol 210:225–238.
tuberoinfundibular dopamine neurons and increases serum corticos- Kalivas PW, Abhold R. 1987. Enkephalin release in to the ventral
terone concentrations in the rat. Neuroendocrinology 49:604–609. tegmental area in response to stress: modulation of mesocortical
Guidotti A, Baraldi M, Leon A, Costa E. 1990. Benzodiazepines: a dopamine. Biol Psychiatry 24:339–348.
tool to explore the biochemical and neuro-physiological basis of Kalivas PW, Duffy P. 1989. Similar effects of daily cocaine and
anxiety. FASEB J 39:1039–1042. stress on mesocorticolimbic dopamine neurotransmission in the
Gunne LM, Reis DJ. 1963. Changes in brain catecholamines asso- rat. Biol Psychiatry 25:913–928.
ciated with electrical stimulation of amygdaloid nucleus. Life Sci Kant GJ, Leu JR, Anderson SM, Mougey EH. 1987. Effects of
11:804–809. chronic stress on plasma corticosterone, ACTH and prolactin.
Haracz JL, Tschanz JT, Wang Z, Griffith KE, Rebec GV. 1998. Physiol Behav 40:775–779.
Amphetamine effects on striatal neurons: implications for models Kilts CD, Coco M, Ely TD, Bissette G, Nemeroff CB. 1992. Dif-
of dopamine function. Neurosci Biobehav Rev 22:613–622. ferential effects of conditioned and unconditioned stress on the
Heilig M, McLeod S, Brot M, Heinrichs SC, Menzaghi F, Koob neurotensin content of dopamine cell body groups of the ventral
GF, Britton KT. 1993. Anxiolytic-like action of neuropeptide Y: mesencephalon. Ann NY Acad Sci 668:266–276.
mediation by Y1 receptors in amygdala, and dissociation from Kim JJ, Fanselow MS. 1992. Modality-specific retrograde amnesia
food intake effects. Neuropsychopharmacology 8:357–363. of fear. Science 256:675–677.
144 Vermetten et al.

King JA, Abend S, Edwards E. 2001. Genetic predisposition and dopamine neurons: selective activation of the mesocortical sys-
the development of posttraumatic stress disorder in an animal tem. Brain Res 476:377–381.
model. Biol Psychiatry 50:231–237. Marti O, Garcia A, Velles A, Harbuz MS, Armario A. 2001. Evi-
Koolhaas, JM, De Boer SF, De Rutter AJ, Meerlo P, Sgoifo A. dence that a single exposure to aversive stimuli triggers long-
1997. Social stress in rats and mice. Acta Physiol Scand Suppl. lasting effects in the hypothalamus-pituitary-adrenal axis that
640:69–72. consolidate with time. Eur J Neurosci 13:129–136.
Krisch B. 1981. Somatostatin-immunoreactive fiber projections Martin P, Soubrie P, Puech AJ. 1990. Reversal of helpless behavior
into the brain stem and the spinal cord of the rat. Cell Tissue by serotonin uptake blockers in rats. Psychopharmacology (Ber-
Res 217:531–552. lin) 101:403–407.
Kvetnansky R, Fukuhara K, Pacak K, Cizza G, Goldstein DS, Martin SJ, Grimwood PD, Morris RG. 2000. Synaptic plasticity
Kopin IJ. 1993. Endogenous glucocorticoids restrain catechola- and memory: an evaluation of the hypothesis. Annu Rev Neuro-
mine synthesis and release at rest and during immobilization sci 23:649–711.
stress in rats. Endocrinology 133:1411–1419. McEwen BS. 1979. Influences of adrenocortical hormones on pitu-
Ladd CO, Owens MJ, Nemeroff CB. 1996. Persistent changes in itary and brain function. Monogr Endocrinol 12:467–492.
corticotropin-releasing factor neuronal systems induced by ma- McEwen BS, de Kloet E, Rostene W. 1986. Adrenal steroid recep-
ternal deprivation. Endocrinology 137:1212–1218. tors and actions in the central nervous system. Physiol Rev
LaHoste GJ, Henry BL, Marshall JF. 2000. Dopamine D1 recep- 66:1121–1189.
tors synergize with D2, but not D3 or D4, receptors in the stria- McEwen BS, Gould EA, Sakai RR. 1992. The vulnerability of the
tum without the involvement of action potentials. J Neurosci hippocampus to protective and destructive effects of glucocorti-
20:6666–6671. coids in relation to stress. Br J Psychiatry Suppl. 15:18–23
Lechin F, Van der Dijs B, Benaim M. 1996. Stress versus depres- McEwen BS. 2000. The neurobiology of stress: from serendipity to
sion. Prog Neuropsychopharmacol Biol Psychiatry 20:899–950. clinical relevance. Brain Res 886:172–189.
LeDoux JE, Cicchetti P, Xagoraris A, Romanski LM. 1990. The McGaugh JL, Cahill L, Roozendaal B. 1996. Involvement of the
lateral amygdaloid nucleus: sensory interface of the amygdala in amygdala in memory storage: interaction with other brain sys-
fear conditioning. J Neurosci 10:1062–1069. tems. Proc Natl Acad Sci USA 93:13508–13514.
Lehmann J, Weizman R, Pryce CR, Leschiner S, Allmann I, McKinney WT, Bunney WE. Animal model of depression. 1969. I.
Feldon J, Gavish M. 1999. Peripheral benzodiazepine receptors Review of evidence: implications for research. Arch Gen Psy-
in cerebral cortex, but not in internal organs, are increased fol- chiatry 21:240–248.
lowing inescapable stress and subsequent avoidance/escape McKittrick CR, Blanchard DC, Blanchard RJ, McEwen BS, Sakai
shuttle-box testing. Brain Res 851:141–147. RR. 1995. Serotonin receptor binding in a colony model of
Levine ES, Litto WJ, Jacobs BL. 1990. Activity of cat locus coer- chronic social stress. Biol Psychiatry 37:383–393.
uleus noradrenergic neurons during the defense reaction. Brain
Meaney MJ, Aitken DH, van Berkel C, Bhatnagar S, Sapolsky RM.
Res 531:189–195.
1988. Effect of neonatal handling on age-related impairments as-
Liberzon I, Krstov M, Young EA. 1997. Stress-restress: effects on
sociated with the hippocampus. Science 239:766–8.
ACTH and fast feedback. Psychoneuroendocrinology 22:443–453.
Meaney MJ, Aitken DH, Viau V, Sharma S, Sarrieau A. 1989.
Lippa AS, Klepner CA, Yunger L, Sano MC, Smith WV, Beer B.
1978. Relationship between benzodiazepine receptors and experi- Neonatal handling alters adrenocortical negative feedback sensi-
mental anxiety in rats. Pharmacol Biochem Behav 9:853–856. tivity and hippocampal type II glucocorticoid receptor binding in
Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, Freedman A, the rat. Neuroendocrinology 50:597–604.
Sharma S, Pearson D, Plotsky PM, Meaney MJ. 1997. Maternal Medina JH, Novas ML, Wolfman CN, Levi de Stein M, De
care, hippocampal glucocorticoid receptors, and hypothalamic- Robertis E. 1983a. Benzodiazepine receptors in rat cerebral cor-
pituitary-adrenal responses to stress. Science 277:1659–1662. tex and hippocampus undergo rapid and reversible changes after
Liu D, Caldji C, Sharma S, Plotsky PM, Meaney MJ. 2000. Influence acute stress. Neuroscience 9:331–335.
of neonatal rearing conditions on stress-induced adrenocorticotro- Medina JH, Novas ML, De Robertis E. 1983b. Changes in benzo-
pin responses and norepinepherine release in the hypothalamic diazepine receptors by acute stress: different effect of chronic di-
paraventricular nucleus. J Neuroendocrinol 12:5–12. azepam or RO 15-1788 treatment. Eur J Pharmacol 96:181–185.
Loh HH, Smith AP. 1990. Molecular characterization of opioid re- Mendelson SD, McEwen BS. 1991. Autoradiographic analyses of
ceptors. Annu Rev Pharmacol Toxicol 30:123–147. the effects of restraint-induced stress on 5-HT1A, 5-HT1C and 5-
Lopez JF, Akil H, Watson SJ. 1999. Neural circuits mediating HT2 receptors in the dorsal hippocampus of male and female
stress. Biol Psychiatry 46:1461–1471. rats. Neuroendocrinology 54:454–461.
Luine VN. 1994. Steroid hormone influences on spatial memory. Michelson D, Licino J, Gold PW. 1995. Mediation of the stress
Ann NY Acad Sci 743:201–211. response by the hypothalamic-pituitary-adrenal axis. In: neuro-
Madden J, Akil H, Patrick RL, Barchas JD. 1977. Stress-induced biological and clinical consequences of stress: from normal adap-
parallel changes in central opioid levels and pain responsiveness tation to PTSD. Friedman MJ, Charney DS, Deutch AY, editors.
in the rat. Nature 265:358–360. New York: Raven Press. p 225–238.
Magiakou MA, Mastorakos G, Webster E, Chrousos GP. 1997. Mizoguchi K, Yuzurihara M, Ishige A, Sasaki H, Chui DH, Tabira
The hypothalamic-pituitary-adrenal axis and the female repro- T. 2000. Chronic stress induces impairment of spatial working
ductive system. Ann NY Acad Sci 816:42–56. memory because of prefrontal dopaminergic dysfunction. J
Makara GB. 1992. The relative importance of hypothalamic neu- Neurosci 20:1568–1574.
rons containing corticotropin-releasing factor or vasopressin in Moghaddam, B, Bolinao ML, Stein-Behrens B, Sapolsky R. 1994.
the regulation of adrenocorticotropic hormone secretion. Ciba Glucocorticoids mediate the stress-induced extracellular accu-
Found Symp 168:43–51. mulation of glutamate. Brain Res 655:251–254.
Mantz J, Thierry AM, Glowinski J. 1989. Effect of noxious tail Moghaddam B, Adams B, Verma A, Daly D. 1997. Activation of
pinch on the discharge rate of mesocortical and mesolimbic glutamatergic neurotransmission by ketamine: a novel step in the
Review Article: Circuits and Systems in PTSD, Preclinical Data 145

pathway from NMDA receptor blockade to dopaminergic and nucleus and pituitary-adrenocortical and sympathoadrenal activity:
cognitive disruptions associated with the prefrontal cortex. J in vivo microdialysis studies. Front Neuroendocrinol 16:89–150.
Neurosci 17:2912–2127. Packan DR, Sapolsky RM. 1990. Glucocorticoid endangerment of
Morgan MA, Romanski LM, LeDoux JE. 1993. Extinction of emo- the hippocampus: tissue, steroid and receptor specificity. Neu-
tional learning: contribution of medial prefrontal cortex. Neuro- roendocrinology 51:613–618.
sci Lett 163:109–113. Page ME, Abercrombie ED. 1999. Discrete local application of
Nakamura S, Sakaguchi T. 1990. Development and plasticity of the corticotropin-releasing factor increases locus coeruleus discharge
locus coeruleus: a review of recent physiological and pharmaco- and extracellular norepinephrine in rat hippocampus. Synapse
logical experimentation. Prog Neurobiol 34:505–526. 33:304–313.
Nemeroff CB, Hernandez DE, Orlando RC, Prange AJ. 1982. Pani L, Porcella A, Gessa GL. 2000. The role of stress in the
Cytoprotective effect of centrally administered neurotensin on pathophysiology of the dopaminergic system. Mol Psychiatry
stress-induced gastric ulcers. Am J Physiol 242:342–346. 5:14–21.
Nemeroff CB, Schatzberg AF. 1988. The hypothalamic-pituitary- Pei Q, Zetterstrom T, Fillenz M. 1990. Tail pinch-induced
adrenal axis: physiology, pathophysiology, and psychiatric impli- changes in the turnover and release of dopamine and 5-hydrox-
cations. New York: Raven Press. ytryptamine in different brain regions of the rat. Neuroscience
Newport DJ, Nemeroff CB. 2000. Neurobiology of posttraumatic 35:133–138.
stress disorder. Curr Opin Neurobiol 10:211–218. Penner SB, Smyth DD, Glavin GB. 1993. Effects of neuropeptide
Nibuya M, Morinobu S, Duman RS. 1995. Regulation of BDNF Y on experimental gastric lesion formation and gastric secretion
and trkB mRNA in rat brain by chronic electroconvulsive seizure in the rat. J Pharmacol Exp Ther 266:339–343.
and antidepressant drug treatments. J Neurosci 15:7539–7547. Pepeu G, Blandina P. 1998. The acetylcholine, GABA, glutamate
Nibuya M, Nestler EJ, Duman RS. 1996. Chronic antidepressant triangle in the rat forebrain. J Physiol Paris 92:351–355.
administration increases the expression of cAMP response ele- Petty F, Sherman AD. 1981. GABAergic modulation of learned
ment binding protein (CREB) in rat hippocampus. J Neurosci helplessness. Pharmacol Biochem Behav 15:567–570.
16:2365–2372. Petty F, Sherman AD. 1983. Learned helplessness induction de-
Nicotera P, Ankarcrona M, Bonfoco E, Orrenius S, Lipton SA. creases in vivo cortical serotonin release. Pharmacol Biochem
1997. Neuronal necrosis and apoptosis: two distinct events in- Behav 18:649–650.
duced by exposure to glutamate or oxidative stress. Adv Neurol Petty F, Kramer G, Wilson L. 1992. Prevention of learned help-
72:95–101. lessness: in vivo correlation with cortical serotonin. Pharmacol
Nijenhuis ER, Vanderlinden J, Spinhoven P. 1998. Animal defen- Biochem Behav 43:361–367.
sive reactions as a model for trauma-induced dissociative reac- Phelix CF, Liposits Z, Paull WK. 1992. Serotonin-CRF interaction
tions. J Trauma Stress 11:243–260. in the bed nucleus of the stria terminalis: a light microscopic
Ninan PT, Insel TM, Cohen RM, Cook JM, Skolnick P, Paul SM. double-label immunocytochemical analysis. Brain Res Bull
1982. Benzodiazepine receptor-mediated experimental ‘anxiety’ 28:943–948.
in primates. Science 218:1332–1334. Phillips RG, LeDoux JE. 1992. Differential contribution of
Nisenbaum LK, Zigmond MJ, Sved AF, Abercrombie ED. 1991. amygdala and hippocampus to cued and contextual fear condi-
Prior exposure to chronic stress results in enhanced synthesis tioning. Behav Neurosci 106:274–285.
and release of hippocampal norepinephrine in response to a Piazza PV, Le Moal M. 1997. Glucocorticoids as a biological sub-
novel stressor. J Neurosci 11:1478–1484. strate of reward: physiological and pathophysiological implica-
Norman RL, Smith CJ. 1992. Restraint inhibits luteinizing hor- tions. Brain Res Brain Res Rev 25:359–372.
mone and testosterone secretion in intact male rhesus macaques: Pitts DK, Marwah J. 1987. Electrophysiological actions of cocaine
effects of concurrent naloxone administration. Neuroendocrinol- on noradrenergic neurons in rat locus ceruleus. J Pharmacol Exp
ogy 55:405–415. Ther 240:345–351.
Obrenovitch TP. 1999. High extracellular glutamate and neuronal Plotsky PM, Meaney MJ. 1993. Early postnatal experience alters
death in neurological disorders: cause, contribution or conse- hypothalamic corticotropin-releasing factor (CRF) mRNA, me-
quence? Ann NY Acad Sci 890:273–286. dian eminence CRF content and stress-induced release in adult
Ohl F, Michaelis T, Vollmann-Honsdorf GK, Kirschbaum C, rats. Brain Res Mol Brain Res 18:195–200.
Fuchs E. 2000. Effect of chronic psychosocial stress and long- Przegalinski E, Moryl E, Papp M. 1995. The effect of 5-HT1A re-
term cortisol treatment on hippocampus-mediated memory and ceptor ligands in a chronic mild stress model of depression. Neu-
hippocampal volume: a pilot-study in tree shrews. Psycho- ropharmacology 34:1305–1310.
neuroendocrinology 25:357–363. Pynoos RS, Ritzmann RF, Steinberg AM, Goenjian A, Prisecaru I.
Oitzl, MS, Workel JO, Fluttert M, Frosch F, De Kloet ER. 2000. 1996. A behavioral animal model of posttraumatic stress disorder
Maternal deprivation affects behavior from youth to senescence: featuring repeated exposure to situational reminders. Biol Psy-
amplification of individual differences in spatial learning and chiatry 39:129–134.
memory in senescent Brown Norway rats. Eur J Neurosci Rasmusson AM, Charney DS. 1997. Animal models of relevance to
12:3771–3780. PTSD. Ann NY Acad Sci 821:332–351.
Olson GA, Olson RD, Kastin AJ. 1985. Endogenous opiates: 1984. Redei EE, Ahmadiyeh N, Baum AE, Sasso DA, Slone JL, Solberg
Peptides 6:769–791. LC, Will CC, Volenec A. 2001. Novel animal models of affec-
Otero Losada ME. 1988. Changes in central GABAergic function tive disorders. Semin Clin Neuropsychiatry 6:43–67.
following acute and repeated stress. Br J Pharmacol 93:483–490. Redmond DE, Huang YH. 1979. Current concepts. II. New evi-
Pacak K, Kvetnansky R, Palkovits M, Fukuhara K, Yadid G, Kopin dence for a locus coeruleus-norepinephrine connection with
IJ, Goldstein DS. 1993. Adrenalectomy augments in vivo release anxiety. Life Sci 25:2149–2162.
of norepinephrine in the paraventricular nucleus during immobi- Reul JM, de Kloet ER. 1986. Anatomical resolution of two types of
lization stress. Endocrinology 133:1404–1410. corticosterone receptor sites in rat brain with in vitro autorad-
Pacak K, Palkovits M, Kopin IJ, Goldstein DS. 1995. Stress-induced iography and computerized image analysis. J Steroid Biochem
norepinephrine release in the hypothalamic paraventricular 24:269–272.
146 Vermetten et al.

Rivier C, Rivier J, Vale W. 1986. Stress-induced inhibition of re- fects on BDNF expression in the hippocampus: Implications for
productive functions: role of endogenous corticotropin-releasing memory formation. Stress 3:201–208.
factor. Science 231:607–609. Schwartz RD, Wess MJ, Labarca R, Skolnick P, Paul SM. 1987.
Robertson HA, Martin IL, Candy JM. 1978. Differences in benzo- Acute stress enhances the activity of the GABA receptor-gated
diazepine receptor binding in Maudsley reactive and Maudsley chloride ion channel in brain. Brain Res 411:151–155.
non-reactive rats. Eur J Pharmacol 50:455–457. Serra M, Pisu MG, Littera M, Papi G, Sanna E, Tuveri F, Usala L,
Romanski LM, LeDoux JE. 1992. Equipotentiality of thalamo- Purdy RH, Biggio G. 2000. Social isolation-induced decreases in
amygdala and thalamo-cortico-amygdala circuits in auditory fear both the abundance of neuroactive steroids and GABA(A) recep-
conditioning. J Neurosci 12:4501–4509. tor function in rat brain 75:732–740.
Ronan PJ, Steciuk M, Kramer GL, Kram M, Petty F. 2000. In- Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram
creased septal 5-HIAA efflux in rats that do not develop CD, Lightman SL. 2000. Early-life exposure to endotoxin alters
learned helplessness after inescapable stress. J Neurosci Res 61: hypothalamic-pituitary-adrenal function and predisposition to
101–106. inflammation. Proc Natl Acad Sci USA 97:5645–5650.
Roozendaal B. 2000. 1999 Curt P. Richter award. Glucocorticoids Sherman AD, Petty F. 1980. Neurochemical basis of the action of
and the regulation of memory consolidation. Psychoneuroendo- antidepressants on learned helplessness. Behav Neural Biol
crinology 25:213–238. 30:119–134.
Rosen JB, Davis M. 1988. Enhancement of acoustic startle by elec- Sherman AD, Petty F. 1982. Additivity of neurochemical changes
trical stimulation of the amygdala. Behav Neurosci 102:195–202, in learned helplessness and imipramine. Behav Neural Biol
324. 35:344–353.
Ross RT, Orr WB, Holland PC, Berger TW. 1984. Hippo- Shi WX, Bunney BS. 1992. Actions of neurotensin: a review of the
campectomy disrupts acquisition and retention of learned condi- electrophysiological studies. Ann NY Acad Sci 668:129–145.
tional responding. Behav Neurosci 98:211–225. Shors TJ, Seib TB, Levine S, Thompson RF. 1989. Inescapable
Roth RH, Tam SY, Ida Y, Yang JX, Deutch AY. 1988. Stress and versus escapable shock modulates long-term potentiation in the
the mesocorticolimbic dopamine systems. Ann NY Acad Sci rat hippocampus. Science 244:224–226.
537:138–147. Shors TJ, Foy MR, Levine S, Thompson RF. 1990. Unpredictable
Ruis MA, te Brake JH, Buwalda B, De Boer SF, Meerlo P, Korte and uncontrollable stress impairs neuronal plasticity in the rat
SM, Blokhuis HJ, Koolhaas JM. 1999. Housing familiar male hippocampus. Brain Res Bull 24:663–667.
wildtype rats together reduces the long-term adverse behavioral Skerritt JH, Trisdikoon P, Johnston GA. 1981. Increased GABA
and physiological effects of social defeat. Psychoneuroendo- binding in mouse brain following acute swim stress. Brain Res
crinology 24:285–300. 215:398–403.
Sabban EL, Kvetnansky R. 2001. Stress-triggered activation of Smith MA, Makino S, Kvetnansky R, Post RM. 1995. Effects of
gene expression in catecholaminergic systems: dynamics of tran- stress on neurotrophic factor expression in the rat brain. Ann NY
scriptional events. Trends Neurosci 24:91–98. Acad Sci 771:234–239.
Salamone JD, Cousins MS, Snyder BJ. 1997. Behavioral functions Smythe GA. 1977. The role of serotonin and dopamine in hypotha-
of nucleus accumbens dopamine: empirical and conceptual prob- lamic-pituitary function. Clin Endocrinol 7:325–341.
lems with the anhedonia hypothesis. Neurosci Biobehav Rev Sousa N, Lukoyanov NV, Madeira MD, Almeida OF, Paula-Barbosa
21:341–359. MM. 2000. Reorganization of the morphology of hippocampal
Sapolsky RM, Krey LC, McEwen BS. 1984a. Stress down-regulates neurites and synapses after stress-induced damage correlates with
corticosterone receptors in a site-specific manner in the brain. behavioral improvement. Neuroscience 97:253–266.
Endocrinology 114:287–292. Steckler T, Holsboer F. 1999. Corticotropin-releasing hormone re-
Sapolsky RM, Krey LC, McEwen BS. 1984b. Glucocorticoid-sensi- ceptor subtypes and emotion. Biol Psychiatry 46:1480–1508.
tive hippocampal neurons are involved in terminating the adreno- Stout SC, Kilts CD, Nemeroff CB. 1995. Neuropeptides and
cortical stress response. Proc Natl Acad Sci USA 81:6174–6177. Stress: Preclinical Findings and Implication for Pathophysiology.
Sapolsky RM. 1985. A mechanism for glucocorticoid toxicity in the In: Neurobiological and clinical consequences of stress: from
hippocampus: increased neuronal vulnerability to metabolic in- normal adaptation to PTSD. Friedman MJ, Charney DS,
sults. J Neurosci 5:1228–1232. Deutch AY, editors. New York: Raven Press. p 103–123.
Sapolsky RM, Packan DR, Vale WW. 1988. Glucocorticoid toxic- Stuckey J, Marra S, Minor T, Insel TR. 1989. Changes in µ-opiate
receptors following inescapable shock. Brain Res 476:167–169.
ity in the hippocampus: in vitro demonstration. Brain Res
Tanaka M, Yoshida M, Emoto H, Ishii H. 2000. Noradrenaline
453:367–371.
systems in the hypothalamus, amygdala and locus coeruleus are
Sapolsky RM, Plotsky PM. 1990. Hypercortisolism and its possible
involved in the provocation of anxiety: basic studies. Eur J
neural bases. Biol Psychiatry 27:937–952.
Pharmacol 405:397–406.
Sapolsky RM, Uno H, Rebert CS, Finch CE. 1990. Hippocampal Terman GW, Shavit Y, Lewis JW, Cannon JT, Liebeskind JC.
damage associated with prolonged glucocorticoid exposure in 1984. Intrinsic mechanisms of pain inhibition: activation by
primates. J Neurosci 10:2897–2902. stress. Science 226:1270–1277.
Sapolsky RM. 1996. Why stress is bad for your brain. Science Thierry AM, Pirot S, Gioanni Y, Glowinski J. 1998. Dopamine
273:749–750. function in the prefrontal cortex. Adv Pharmacol 42:717–720.
Sapolsky RM. 2000. Glucocorticoids and hippocampal atrophy in Thorsell A, Michalkiewicz M, Dumont Y, Quirion R, Caberlotto L,
neuropsychiatric disorders. Arch Gen Psychiatry 57:925–935. Rimondini R, Mathe AA, Heilig M. 2000. Behavioral insensitivity
Savolainen KM, Loikkanen J, Naarala J. 1995. Amplification of to restraint stress, absent fear suppression of behavior and impaired
glutamate-induced oxidative stress. Toxicol Lett 82–83:399–405. spatial learning in transgenic rats with hippocampal neuropeptide Y
Sawchenko PE, Imaki T, Vale W. 1992. Co-localization of neuro- overexpression. Proc Natl Acad Sci USA 97:12852–12857.
active substances in the endocrine hypothalamus. Ciba Found Treit D. 1985. Animal models for the study of anti-anxiety agents:
Symp 168:16–30. a review. Neurosci Biobehav Rev 9:203–222.
Schaaf MJ, De Kloet ER, Vreugdenhil E. 2000. Corticosterone ef- Tuncel N, Erkasap N, Sahinturk V, Ak DD, Tuncel M. 1998. The
Review Article: Circuits and Systems in PTSD, Preclinical Data 147

protective effect of vasoactive intestinal peptide (VIP) on stress- 1992. Phenytoin prevents stress- and corticosterone-induced at-
induced gastric ulceration in rats. Ann NY Acad Sci 865:309–322. rophy of CA3 pyramidal neurons. Hippocampus 2:431–435.
Uno H, Tarara R, Else JG, Suleman MA, Sapolsky RM. 1989. Hip- Watanabe Y, Sakai RR, McEwen BS, Mendelson S. 1993. Stress
pocampal damage associated with prolonged and fatal stress in and antidepressant effects on hippocampal and cortical 5-HT1A
primates. J Neurosci 9:1705–1711. and 5-HT2 receptors and transport sites for serotonin. Brain Res
Uno H, Lohmiller L, Thieme C, Kemnitz JW, Engle MJ, Roecker 615:87–94.
EB, Farrell PM. 1990. Brain damage induced by prenatal expo- Watanobe H, Schiloth HB, Wikberg JE, Suda T. 2000. Evaluation
sure to dexamethasone in fetal rhesus macaques. I. Hippocam- of the role for prolactin-releasing peptide in prolactin secretion
pus. Brain Res Dev Brain Res 53:157–167. induced by ether stress and suckling in the rat: comparison with
Vaccarino AL, Olson GA, Olson RD, Kastin AJ. 1999. Endogenous vasoactive intestinal peptide. Brain Res 865:91–96.
opiates: 1998. Peptides 20:1527–1574. Weiss JM. 1972. Psychological factors in stress and disease. Sci Am.
Valentino RJ, Foote SL. 1988. Corticotropin-releasing hormone in- 226:104–113.
creases tonic but not sensory-evoked activity of noradrenergic locus Weiss JM, Simson PG. 1985. Neurochemical basis of stress-in-
coeruleus neurons in unanesthetized rats. J Neurosci 8:1016–1025. duced depression. Psychopharmacol Bull 21:447–457.
Valentino RJ, Wehby RG. 1988. Corticotropin-releasing factor: Weizman R, Weizman A, Kook KA, Vocci F, Deutsch SI, Paul SM.
evidence for a neurotransmitter role in the locus coeruleus dur- 1989. Repeated swim stress alters brain benzodiazepine receptors
ing hemodynamic stress. Neuroendocrinology 48:674–677. measured in vivo. J Pharmacol Exp Ther 249:701–707.
Van Dongen PA. 1981. The central noradrenergic transmission and Weizman A, Weizman R, Kook KA, Vocci F, Deutsch SI, Paul SM.
the locus coeruleus: a review of the data, and their implications 1990. Adrenalectomy prevents the stress-induced decrease in
for neurotransmission and neuromodulation. Prog Neurobiol vivo. Brain Res 519:347–350.
16:117–143. Welberg LA, Seckl JR. 2001. Review article: prenatal stress gluco-
Van Dijken HH, de Goeij DC, Sutanto W, Mos J, de Kloet ER, corticoids and the programming of the brain. J Neuroendocrinol
Tilders FJ. 1993. Short inescapable stress produces long-lasting 13:113–128.
changes in the brain- pituitary-adrenal axis of adult male rats. de Wied D, Croiset G. 1991. Stress modulation of learning and
Neuroendocrinology 58:57–64. memory processes. Methods Achiev Exp Pathol 15:167–199.
Van Megen HJ, Westenberg HG, den Boer JA, Haigh JR, Traub Woolley CS, Gould E, McEwen BS. 1990. Exposure to excess glu-
M. 1994. Pentagastrin induced panic attacks: enhanced sensitiv- cocorticoids alters dendritic morphology of adult hippocampal
ity in panic disorder patients. Psychopharmacology (Berlin) pyramidal neurons. Brain Res 531:225–231.
114:449–455. Wu J, Kramer GL, Kram M, Steciuk M, Crawford IL, Petty F.
van Oers HJ, de Kloet ER, Levine S. 1998. Early vs. late maternal 1999. Serotonin and learned helplessness: a regional study of 5-
deprivation differentially alters the endocrine and hypothalamic HT1A, 5-HT2A receptors and the serotonin transport site in rat
responses to stress. Brain Res Dev Brain Res 111:245–252. brain. J Psychiatr Res 33:17–22.
Vecsei L, Kiraly C, Bollok I, Nagy A, Varga J, Penke B, Telegdy G. Yehuda R, Giller EL, Southwick SM, Lowy MT, Mason JW. 1991.
1984. Comparative studies with somatostatin and cysteamine in Hypothalamic-pituitary-adrenal dysfunction in posttraumatic
different behavioral tests on rats. Pharmacol Biochem Behav stress disorder. Biol Psychiatry 30:1031–1048.
21:833–837. Yehuda R, Antelman SM. 1993. Criteria for rationally evaluating
Vermetten E, Charney DS, Bremner DS. 2002. PTSD. In Current animal models of posttraumatic stress disorder. Biol Psychiatry
Concepts in Psychiatry. Helmchen H, Henn FA, Lauter H, Sar- 33:479–486.
torius N, editors. Springer Verlag, Berlin-Heidelberg (in press). Zangen A, Nakash R, Overstreet DH, Yadid G. 2001. Association
Vermetten E, Bremner JD. Circuits and systems in Stress: II. Ap- between depressive behavior and absence of serotonin-dopamine
plications to Neurobiology and Treatment in PTSD. Depress interaction in the nucleus accumbens. Psychopharmacology
Anxiety (in press). (Berlin) 155:434–439.
Wahlestedt C, Grundemar L, Hakanson R, Heilig M, Shen GH, Zigmond MJ, Castro SL, Keefe KA, Abercrombie ED, Sved AF.
Zukowska-Grojec Z, Reis DJ. 1990. Neuropeptide Y receptor 1998. Role of excitatory amino acids in the regulation of
subtypes, Y1 and Y2. Ann NY Acad Sci 611:7–26. dopamine synthesis and release in the neostriatum. Amino Ac-
Watanabe Y, Gould E, Cameron HA, Daniels DC, McEwen BS. ids 14:57–62.

You might also like