You are on page 1of 18

Stress, September 2011; 14(5): 503–519

q Informa Healthcare USA, Inc.


ISSN 1025-3890 print/ISSN 1607-8888 online
DOI: 10.3109/10253890.2011.596864

Stress modulation of cognitive and affective processes

SERGE CAMPEAU1, ISRAEL LIBERZON2, DAVID MORILAK3, & KERRY RESSLER4


1
Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA, 2Department of
Psychiatry and Psychology, University of Michigan, Ann Arbor, MI, USA, 3Department of Pharmacology, University of Texas
Health Science Center, San Antonio, TX, USA, and 4Department of Psychiatry and Behavioral Sciences, Emory University,
Atlanta, GA, USA

(Received 20 October 2010; revised 18 April 2011; accepted 1 June 2011)

Abstract
This review summarizes the major discussion points of a symposium on stress modulation of cognitive and affective processes,
which was held during the 2010 workshop on the neurobiology of stress (Boulder, CO, USA). The four discussants addressed
a number of specific cognitive and affective factors that are modulated by exposure to acute or repeated stress. Dr David
Morilak discussed the effects of various repeated stress situations on cognitive flexibility, as assessed with a rodent model of
attentional set-shifting task, and how performance on slightly different aspects of this test is modulated by different prefrontal
regions through monoaminergic neurotransmission. Dr Serge Campeau summarized the findings of several studies exploring
a number of factors and brain regions that regulate habituation of various autonomic and neuroendocrine responses to
repeated audiogenic stress exposures. Dr Kerry Ressler discussed a body of work exploring the modulation and extinction of
fear memories in rodents and humans, especially focusing on the role of key neurotransmitter systems including excitatory
amino acids and brain-derived neurotrophic factor. Dr Israel Liberzon presented recent results on human decision-making
processes in response to exogenous glucocorticoid hormone administration. Overall, these discussions are casting a wider
framework on the cognitive/affective processes that are distinctly regulated by the experience of stress and some of the brain
regions and neurotransmitter systems associated with these effects.

Keywords: Cognitive flexibility, prefrontal cortex, decision making, habituation, extinction, fear conditioning

Introduction of cognitive and affective processes, often long after


the precipitating episode(s). Improved knowledge of
This review provides a synopsis of the major discus-
the exact cognitive/affective processes influenced
sion points presented during the session on ‘Stress
by various stressful experiences is already leading to
modulation of cognitive and affective processes’ that
a refinement of understanding of the brain circuits
took place as part of the 2010 workshop on the
and local neural mechanisms affected, and is guiding
neurobiology of stress held in Boulder, CO, USA, on new or enhanced treatment avenues for stress-related
15 – 17 June 2010. Drs David Morilak and Serge disorders. In addition, it is becoming increasingly clear
Campeau focused on different aspects of the effects of that acute or chronic stress situations simultaneously
repeated stress exposures on specific cognitive func- initiate a number of changes across several cogniti-
tions and habituation in rats, while Dr Kerry Ressler ve/affective functions, which are likely to depend on
focused on the modulation of rat and human aversive the specific stress situations, and unfold with varying
memories, and Dr Israel Liberzon presented recent temporal characteristics. The following synopsis
results on human decision-making processes in generally describes each presentation and the major
response to exogenous glucocorticoid hormone points that were raised in several of the presentations
administration. As the title implied, the emphasis of on the modulation of cognitive and affective processes
this session was on how stress modulates a number by acute or repeated stress exposures.

Correspondence: S. Campeau, Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
Tel: 1-303-492-5693. Fax: 1-303-492-2967. E-mail: serge.campeau@colorado.edu
504 S. Campeau et al.

Chronic stress and monoaminergic modulatory influence distracting or irrelevant dimension. Then, once this is
in prefrontal cortex functions (David Morilak, PhD, mastered, the first odor may become negative and the
University of Texas Health Science Center, previously negative odor becomes the positive cue,
San Antonio, TX) a stimulus reversal (R). Then, all new stimuli may be
introduced in a new acquisition task, also called an
The association of stress with a number of mood
intra-dimensional (ID) set-shift because an entirely
disorders, including, but not limited to, post-
new contingency must be learnt, but the salient
traumatic stress disorder, clinical depression, and
dimension is the same (in this example, odor). After
other anxiety disorders is well established (Kendler
several tasks in which the salient dimension remains
et al. 1999; Chrousos 2000; Tafet and Bernardini
constant, rats develop what is called a cognitive set, or
2003). In turn, frontal cortical dysfunctions have been
a ‘rule about the rules’, in which they have learnt
repeatedly reported in patients afflicted with such
which stimulus dimension will be informative when
stress-related psychiatric disorders [see Drevets et al. the rules are changed. Then, the extra-dimensional
(1997), Bremner (2002), Anisman and Matheson (ED) set-shift task is a new acquisition in which the
(2005) for reviews]. Many examples of these cognitive salient dimension is also changed, forcing the animals
dysfunctions have been observed with the Wisconsin to shift their cognitive set, and abandon the previously
Card Sorting task, used extensively to test cognitive learnt higher-order executive rule, a very challenging
flexibility and executive functions in human subjects test of cognitive flexibility that depends on the func-
(Merriam et al. 1999; Fossati et al. 2002). Indeed, tional integrity of the medial prefrontal cortex
deficits of cognitive flexibility leading to emotionally (mPFC; Birrell and Brown 2000; Lapiz and Morilak
biased cognitive processing have been suggested to 2006). By proceeding through this series of increas-
underlie the mood symptoms of depression (see Beck ingly difficult discriminations, from simple to
2005). Given the extensive association of the compound discriminations, reversals, ID and ED
prefrontal cortex with the expression of behavioral set-shifts, the AST allows one to study mechanisms
and cognitive flexibility and executive functions on underlying distinct aspects of attention, cognitive
one hand (Ragozzino et al. 1999; Dalley et al. 2004; flexibility, and executive function in rats.
Ghahremani et al. 2010), and the abnormal prefrontal Using this test (see Figure 1), 2 weeks of chronic
cortical activity noted in stress-related clinical intermittent cold (CIC) stress induced a selective
disorders on the other hand (with a preponderance impairment in one form of cognitive flexibility, reversal
indicating hypoactivity; Austin et al. 2001; Davidson learning, without affecting ED cognitive set-shifting
et al. 2002; Rogers et al. 2004), an important question (Lapiz-Bluhm et al. 2009; Lapiz-Bluhm and Morilak
is how stress, and particularly chronic stress, might 2010). In contrast to ED set-shifting, reversal learning
modify or predispose organisms to dysfunctional is associated more with the functional integrity of the
prefrontal cortical activity, and how this dysregulation orbitofrontal cortex (Schoenbaum et al. 2002;
might be remedied. McAlonan and Brown 2003; Kim and Ragozzino
There is indeed accumulating evidence that chronic 2005). Thus, this chronic physiologic stressor
stress in laboratory animals has a negative impact on appeared to impact the orbitofrontal subregion of
cognitive flexibility, the process by which feedback prefrontal cortex to a greater extent than it did the
from the environment is used to modify previously mPFC. In contrast, also shown in Figure 1, a 2-week
learnt or prepotent behavioral patterns. A test to assess regimen of chronic unpredictable stress (CUS),
various aspects of cognitive flexibility has been which is a robust psychological stressor, more reliably
developed specifically for rodents, the attentional disrupted ED cognitive set-shifting (Bondi et al. 2008,
set-shifting test (AST; Birrell and Brown 2000). In the 2010), similar to the effect reported after chronic
AST, rats are trained to dig for a food reward in small intermittent restraint (Liston et al. 2006). Thus, chronic
pots filled with material such as sawdust and styrofoam stressors with a substantive psychological component
beads and scented with fragrant oils. Thus, the pots affect cognitive set-shifting, and are thus likely to
are identified by cues along two different stimulus involve subregions of mPFC (prelimbic and infralimbic
dimensions (digging medium and odor), and the rats cortices) which have been more specifically linked to this
must learn which of these dimensions signals reward process (Ragozzino et al. 1999; Birrell and Brown 2000;
and which is irrelevant. After successfully learning a Chudasama and Robbins 2003; Lapiz et al. 2007).
given contingency, the rules are changed, similar to An integral aspect of the above series of studies
several human neuropsychological tests of cognitive examining the detrimental impact of chronic stress on
flexibility, such as the Wisconsin Card Sort Test. cognitive flexibility was based on the fact that
For instance, in the first discrimination, one odor monoaminergic neurotransmission in prefrontal cor-
(e.g. lemon) may signal the location of the reward and tical regions has been linked both to stress and to
the other odor (e.g. mint) is explicitly not associated modulation of several cognitive functions (Devauges
with the reward. Thus, odor is the salient dimension, and Sara 1990; Cole and Robbins 1992; Page and
lemon is the positive cue, and digging medium is the Lucki 2002; Ramos and Arnsten 2007). In addition,
Stress modulation of cognitive and affective processes 505

Figure 1. Left panel: CUS produced a significant deficit in cognitive flexibility on the AST, manifested as a significantly higher number of
trials required to reach criterion on the reversal tasks (R1, R3) and the ED cognitive set-shift task; þp , 0.05 compared with non-stressed
controls on the same stage. White bars: unstressed controls; Gray bars: chronically stressed rats; data expressed as mean ^ SEM
(n ¼ 14/group). Reproduced with permission from Bondi et al. (2008). Right panel: In contrast, CIC stress produced a selective deficit in
reversal learning on the AST. Stressed rats required significantly more trials to criterion on the R1 reversal task compared with non-stressed
controls (*p , 0.01 compared with control rats on the same stage; þp , 0.05 compared with other tasks for the same group. White bars:
unstressed controls; Gray bars: chronically stressed rats (n ¼ 12/group). Reproduced with permission from Lapiz-Bluhm et al. (2009).

these neurotransmitter systems remain the leading acute elevations of norepinephrine by an a2-adrener-
targets of therapeutic interventions in treating stress- gic autoreceptor antagonist (atipamezole) can facili-
related mood disorders (Schatzberg and Schildkraut tate ED set-shifting (Lapiz and Morilak 2006), a
1995; Ressler and Nemeroff 2000; Morilak and Frazer facilitation that was observed even after a deficit had
2004). In view of the above results demonstrating been induced by CUS treatment (Bondi et al. 2010).
subtle but different cognitive functional alterations Together, the above studies suggest that with
with different chronic stress modalities, the question different stressor modalities, different modulatory
of putative neurotransmitter specificity could also be neurotransmitter systems can have distinguishable
addressed. Of interest, Morilak and collaborators effects on specific cognitive functions in different
showed that serotonergic depletion with PCPA sub-regions of prefrontal cortex. It is also becoming
(4-chloro-DL -phenylalanine) selectively disrupted clear that these same prefrontal cortical regions
reversal learning in rats to levels comparable to that and their associated cognitive functions can be
obtained following chronic cold stress, and acute differentially altered by chronic stress. Monoamine
treatment with the serotonin specific reuptake blocker neurotransmission can facilitate cognitive flexibility
citalopram blocked the chronic cold stress-induced even in the presence of stress-induced prefrontal
reversal learning deficit (Lapiz-Bluhm et al. 2009). cortical dysfunction, providing one specific mechan-
This effect was specific to the serotonin system because ism via which stress-related cognitive dysfunctions can
desipramine treatment (a norepinephrine reuptake be improved by pharmacotherapy, but not necessarily
blocker) did not reduce the chronic cold stress- cured. This is perhaps consistent with recent findings
induced reversal deficit (Lapiz-Bluhm and Morilak that antidepressant treatment in depressed patients
2010), supporting prior findings of serotonin-specific can rapidly ameliorate some symptoms, including
modulation of reversal learning in the orbitofrontal cognitive dysfunction, while producing no immediate
cortex (Clarke et al. 2005, 2007). On the other hand, effects on ratings of mood or anxiety (Harmer et al.
chronic treatments with either the norepinephrine 2009). Although these modulatory systems are likely
reuptake blocker desipramine (see Figure 2) or the to be both activated and altered by chronic stress
selective serotonin reuptake blocker escitalopram pre- exposure impacting cognitive flexibility, it would
vented the deficit in ED cognitive set-shifting induced appear that the disruption of monoaminergic neuro-
by the psychogenic stressor, CUS (Bondi et al. 2008, transmission itself does not represent the primary
2010). Furthermore, at least for the noradrenergic pathology underlying the cognitive dysfunction that is
system, activation of post-synaptic a1-adrenergic associated with stress-related psychiatric disorders.
receptors at the level of the prefrontal cortex during Given the increasing refinements of our under-
the cognitive task is necessary for the effects of chronic standing of the functional cognitive modulation by
desipramine in blocking the CUS-induced deficit in stress and monoaminergic transmission, the work of
cognitive set-shifting (Bondi et al. 2010). Moreover, Morilak and collaborators has recently shifted to a
506 S. Campeau et al.

Figure 2. The beneficial effects of chronic treatment with the NE reuptake blocker and antidepressant drug, desipramine (DMI,
7.5 mg/kg day, given by osmotic minipump), against the detrimental cognitive effects of CUS are mediated by a1-adrenergic receptors in
mPFC. CUS induced a significant set-shifting deficit on the ED task compared with unstressed controls (*p , 0.001), which was prevented by
chronic DMI treatment (þp , 0.01 compared with vehicle-treated, chronically stressed rats). Bilateral microinjections of the a1-adrenergic
receptor antagonist benoxathian (2.0 nM per side), given into mPFC immediately prior to testing on the ED cognitive set-shifting task,
significantly blocked the protective effect of chronic DMI treatment against the detrimental effects of CUS (#p , 0.024 compared with CUS-
exposed rats that were DMI-treated and vehicle-microinjected). All data expressed as mean ^ SEM, n ¼ 5–8/group. Reproduced with
permission from Bondi et al. (2010).

similarly refined analysis in animals undergoing early effects induced by repeated or chronic stress
life stress, as one possible factor responsible for later exposures that are known to either increase or
vulnerability to stress-induced cognitive dysfunctions decrease the unconditioned or prepotent responses
in adulthood. Additional tasks, including the extinc- elicited directly by stressful situations. An example is
tion of conditioned fear, are being assessed as possible the reduction of behavioral, autonomic, and neuro-
cognitive functions also modulated by stress, and endocrine responses to the repeated intermittent
particularly early life stress. A promising new line of exposures to the same stressful situation (homotypic
research under investigation by this group, which has stress), a phenomenon defined as stress habituation
to date been little explored, is the possibility that (Marti and Armario 1998). Reduction of stress
cognitive training of rodents on tasks such as the responses through prior experience with the same
attentional set-shifting paradigm at some critical stressor has been studied for many years [see Grissom
developmental stages might protect against the and Bhatnagar (2008), for a review]. However, with
negative effects of later chronic stress. The mechan- few exceptions (Cole et al. 2000; Bhatnagar et al.
isms whereby such protection might be conferred 2002; Jaferi et al. 2003), the brain regions and neuro-
could provide novel targets for behavioral or pharma- plastic mechanisms subserving this process have
cotherapy, before or after chronic stress. remained surprisingly elusive. This presentation
discussed the results of several studies aimed at further
defining the putative processes and brain regions
Learning to live with stress: Modulation of stress responses associated with stress habituation.
by prior stress (Serge Campeau, PhD, University of For many years, loud noise (audiogenic stress) has
Colorado, Boulder, CO) been employed as an effective stimulus to activate
The previous section introduced some of the effects a number of responses traditionally associated with
of stress chronicity on specific cognitive responses. stressor exposure, including the neuroendocrine
Although CUS was reported to modulate cognitive hypothalamo – pituitary – adrenocortical (HPA) axis
functions, the same situations (cold or restraint) [as indexed by the release of glucocorticoids and
experienced repeatedly were also found to alter adrenocorticotropin hormone (ACTH; Henkin and
cognitive functions, albeit with some differences as Knigge 1963; Borrell et al. 1980; Segal et al. 1989;
to the specific cognitive processes and neurotransmit- Campeau and Watson 1997)], the autonomic system
ter systems involved. There are a number of additional [as indexed by peripheral catecholamine release, heart
Stress modulation of cognitive and affective processes 507

rate, blood pressure, or core body temperature A Plasma CORT Response - 30 min Noise
measurements (De Boer et al. 1989; Overton et al.

micrograms/dl +/– SEM)


Plasma CORT (mean
1991; Gamallo et al. 1992; Saha et al. 1996; Bao et al. 35 *
30 No Noise
1999; Masini et al. 2008)], and various behavioral
25 Acute
responses [measured with locomotor activity, or 20 Spaced
inhibition of feeding and drinking (Irwin et al. 1989; 15 Massed
Segal et al. 1989; Britton et al. 1992; Campeau and 10
Watson 1997; Masini et al. 2008)]. Importantly, most 5
of the above responses to acute loud noise exposure 0
EXP6 TEST
are significantly and robustly reduced upon repeated
Exposure
exposures to the same loud noise (Armario et al. 1984;
De Boer et al. 1988; van Raaij et al. 1997; Campeau B Plasma ACTH Response- 30 min Noise
et al. 2002; Masini et al. 2008). At the intensities and

Plasma ACTH (mean


350
durations employed, habituation to repeated loud 300

pg/ml +/– SEM)


noise exposures is not a simple process of progressive 250
sensory hearing loss because loud noise habituated 200
rats display no deficits in acoustic thresholds, as 150
100
measured with auditory-evoked brainstem potentials,
50
the acoustic startle reflex, or prepulse inhibition or 0
facilitation of the acoustic startle reflex (Campeau EXP 6 TEST
et al. 2002; Masini et al. 2008). Exposure
One of the factors, which has received very limited
attention, was the question of whether interstimulus Figure 3. Mean (^SEM) plasma levels of corticosterone (A) and
ACTH (B) immediately following control quiet exposure to the
interval between stressor presentations was an experimental chambers (No noise, Acute, n’s ¼ 4), or the 6th
important variable that might modulate the develop- (EXP6) loud noise (95 dB) exposure, with either 1-h (massed,
ment of habituated responses to repeated stress n ¼ 4) or 24-h (spaced, n ¼ 4) interstressor intervals. Note the
exposures. This question appeared to be unresolved similar and habituated responses in the two different interstressor
with regard to endocrine and autonomic responses to interval groups after six loud noise exposures. Each graph also
displays mean (^SEM) plasma levels of corticosterone (A) and
repeated stressful exposures, even if it had led to an ACTH (B) 48 h later (TEST) following an additional control
important distinction between short- and long-term chamber exposure (no noise, n ¼ 8), acute loud noise (n ¼ 8), or the
behavioral response habituation and their associated 7th loud noise (30 min, 95 dB) exposure in the spaced (n ¼ 8), and
neural mechanisms (Hinde 1954; Davis 1970; Carew massed (n ¼ 8) groups. Note that the 1-h interstressor interval
et al. 1972; Menzel et al. 2001; Rose and Rankin massed group displays significant recovery of corticosterone and
ACTH levels compared to their levels on the 6th exposure, and
2001). Indeed, this phenomenon might be related to compared to the 24-h interval group. *Indicates a significant
the distributed/spaced and massed training/practice in difference between massed and spaced groups ( p , 0.05).
more traditional learning paradigms, in which longer Reproduced with permission from Masini et al. (2008).
interstimulus intervals (distributed/spaced) are corre-
lated with better retention or performance on memory
tasks or tests with increasing (longer-term) retention feedback effects of corticosterone in the 60-min
intervals (Cepeda et al. 2006). Using loud noise stress, interstressor interval groups (Masini et al. 2008).
it was determined that 24 h (1 day) intervals between These results strongly suggested that there exist
six repeated exposures led to reliable habituation of distinct neural processes responsible for short- and
ACTH and corticosterone release in rats, compared to long-term habituation to stress, similar to the effects of
the initial acute noise exposure (Masini et al. 2008). distributed vs. massed practice effects in various other
Importantly, the same six 30-min noise exposures learning conditions (Cepeda et al. 2006). The exact
given 60 min apart on the same day led to a very neural bases of these distinct processes are currently
similar reduction of HPA axis activity on the last unknown.
exposure (short-term test), comparable to the 24 h Another question relating to stress habituation is the
interstressor condition described above (see Figure 3). extent to which this process is associative. Indeed, an
However, 2 days after the last (6th) stressor exposure influential associative learning model accounting for
(longer-term test), an additional acute noise exposure habituation to repeated stimulus exposures argues that
revealed that habituation was moderately reversed, contextual cues become associated with the repeated
but only in the 60-min interstressor interval stimulus, such that exposure to the contextual cues
group. Furthermore, the same pattern of results was alone gradually primes the retrieval from the memory
obtained in different rats instrumented for measure- of the repeatedly experienced stimulus (Wagner 1978,
ments of heart rate, core body temperature, and 1979, 1981). Importantly, for this associative model,
locomotor activity, indicating that the above neuroen- the primed memory of a stimulus is suggested to
dorine results were not simply due to short-term inhibit the elicitation of prepotent responses normally
508 S. Campeau et al.

triggered by exposure to the stimulus, thereby


providing the basis for the reduction of responses
defined as habituation. Some of the main predictions
of this habituation model generally have been
supported, especially through contextual manipula-
tions modifying habituation of behavioral responses in
predicted ways (Jordan et al. 2000). With regard to the
habituation of neuroendocrine responses, some find-
ings have suggested that a change in contextual cues
following the acquisition of habituation can restore
habituated neuroendocrine responses (Dobrakovova
et al. 1993; Grissom et al. 2007). In these studies,
however, the situations employed to modify contex-
tual cues were confounded with their familiarity, so it
was unclear whether the observed response recovery
was a more general response to novelty rather than a
reflection of the associative role of contextual cues.
In a recent series of studies (Nyhuis et al. 2010),
traditional contextual manipulations known to modu-
late habituation of behavioral responses were tested
while measuring HPA axis responses to repeated
loud noise exposures. An initial study demonstrated
that habituated ACTH and corticosterone release
display very modest, if any, spontaneous recovery Figure 4. (A) Mean plasma corticosterone responses (^1 SEM)
up to 4 weeks after seven daily repeated loud noise over the course of seven consecutive loud noise exposures (30 min,
exposures. A classical extinction procedure involving 95 dB), and during an additional exposure 48 h later when rats were
exposure to the contextual cues daily for 14 days after tested in the same context in which they were habituated (A/A,
habituation training, in the absence of the loud noise, C/C), or in a different context (A/C, C/A). Corticosterone values did
not significantly differ between the groups on Days 1, 3, 7 or the test
provided no evidence of ACTH or corticosterone loud noise exposure, whether rats were tested in the same context or
response recovery on a post-extinction loud noise test a different context, as long as rats had been equally familiarized in
day. Importantly, it was shown that modifying the both contexts (n’s ¼ 8; ACTH results were similar, but are not
contextual cues can restore habituated ACTH and shown here). (B) Mean plasma corticosterone responses (^1 SEM)
corticosterone responses, but only when the cues are on an acute loud noise exposure (30 min, 95 dB) in contexts A or C,
in rats preexposed to the same (12-S) or a different test context
novel, as shown in Figure 4; if the context was made (12-D) for 12 consecutive days. The graph shows that only 12
familiar prior to exposure to the loud noise exposures, preexposure days (30 min daily exposures) to the same context in
no response recovery was observed (Nyhuis et al. which they were tested (12-S) reduced loud noise-induced
2010). The lack of contextual control over habituation corticosterone release compared to acute responses without any
to repeated loud noise exposure was not due to a lack preexposure (Acute), or preexposure in a different context (12-D).
*, A significant difference from Control group; #, a significant
of context discrimination, as an additional experiment difference from both the Acute and 12-D groups (Tukey’s
demonstrated that 11 daily preexposures to contextual HSD; p’s , 0.05). Reproduced with permission from Nyhuis
cues specifically reduced acute loud noise-induced et al. (2010).
ACTH and corticosterone responses (Figure 4).
This also provided one of the first demonstrations
that prior experience with contextual stimuli signifi- to stress under some conditions. It is conceivable that
cantly modifies acute stress responses, akin to the the very characteristics of some of the stress situations
latent inhibition effect described for Pavlovian fear often employed in stress studies (continuous and
conditioning (Lubow and Moore 1959). A recent relatively long-lasting exposures to loud noise,
study also employing repeated restraint stress failed to restraining/immobilizing situations, etc.) simply over-
demonstrate a link between contextual manipulations ride the need to rely on static contextual cues, which
and neuroendocrine response habituation (Rabasa
become redundant when the stress stimuli themselves
et al. 2011). These results highlight the importance of
predict the situation, similar to the overshadowing
controlling for overall experimental familiarity, given
the known role of environmental novelty in inducing effect reported in Pavlovian fear conditioning
stress responses (Bassett et al. 1973; Hennessy et al. paradigms (Kamin 1969). Additional studies will
1977; Pfister 1979). Although these studies do not be essential to answer questions regarding context-
provide support for an associative role of contextual dependent vs. context-independent forms of
stimuli in stress habituation, they do not rule out the habituation to stress and whether these are supported
possibility that such stimuli may influence habituation by distinct neural mechanisms.
Stress modulation of cognitive and affective processes 509

An analysis of the putative brain regions associated

Mean Plasma Corticosterone


with HPA axis habituation to repeated loud noise 30
exposures was begun using a systematic approach 25 MGN ACSF

(ug/dl +SEM)
capitalizing on the well-defined sensory pathways 20 MGN Muscimol
mediating responsiveness to acoustic stimuli. An 15
initial study of regional brain activation, as measured
10 *
by induction of the immediate-early gene c-fos mRNA,
indicated very slight, if any, reduction in lower 5
brainstem auditory nuclei such as the cochlear nuclei, 0
the nuclei of the trapezoid bodies, superior olivary Training – – +
complex, or lateral lemniscus, or various inferior
Testing – + +
colliculus nuclei (Campeau et al. 2002). However, a
broad regional reduction in c-fos mRNA induction in Training & Testing Noise Conditions
habituated compared to acutely noise-exposed rats
Figure 5. Mean plasma corticosterone levels (mg/dl þ 1 SEM)
was observed, including auditory thalamic and measured immediately following 30 min loud noise (95 dB,
cortical levels. This pattern of c-fos mRNA induction testing þ ) or quiet chamber (testing -) exposure on the 48 h test, in
was similar to that obtained following complete rats previously injected in the auditory thalamus (medial geniculate
bilateral auditory thalamic lesions, which specifically nucleus; MGN) with ACSF (gray bars) or muscimol (black bars) and
exposed to loud noise (training þ ) or quiet chambers (training -).
blocked acute loud-noise-induced HPA axis responses
Note the significant reduction of corticosterone release in the rats
(Campeau et al. 1997). These findings strongly previously treated with intra-MGN ACSF and noise, and tested again
suggest that the neuroplasticity associated with HPA with noise, compared to the complete block of this reduction in the
axis habituation to loud noise takes place at auditory rats receiving intra-MGN muscimol. *Statistically different from
thalamic levels or some of its targets (Campeau and muscimol-injected group repeatedly exposed to loud noise,
p ¼ 0.003. Reproduced with permission from Day et al. (2009).
Watson 2000). To more precisely test the hypothesis
that the auditory thalamus is a necessary component
of the circuit mediating habituation to repeated loud pinpoint specific mechanisms associated with this
noise exposures, bilateral reversible inactivation of the important function. The existence of interstressor
auditory thalamus using muscimol was performed interval and other temporal effects (Jordan et al. 2000)
daily during the first two of three repeated noise suggests the possibility of multiple neural mechan-
exposures (Day et al. 2009). This reversible inacti- isms, but conclusive evidence is yet to be obtained.
vation of the auditory thalamus, unlike the permanent
lesions of the prior study, allowed a final test of the
Modulation and extinction of aversive memories:
repeatedly presented loud noise with a functional
from rodents to humans (Kerry Ressler, M.D., PhD,
thalamus. As shown in Figure 5, this study revealed
Emory University, Atlanta, GA)
the necessity of this region for HPA axis habituation to
repeated loud noise exposures. A common theme of discussion thus far has been that
Of special significance, preliminary results obtained stress is robustly associated with a number of mood
following large temporal (auditory) cortex lesions disorders. From the multiple symptom clusters
either before or after HPA axis habituation to repeated associated with different mood and anxiety disorders,
loud noise exposures produced no disruption of acute it is now recognized that enhanced fear or an inability
or habituated ACTH and corticosterone responses to to suppress fear responses is common across several
loud noises (C.V. Masini and S. Campeau, unpub- disorders (Kessler et al. 1995; Breslau et al. 2000;
lished observations). Additional targets of the auditory Lissek et al. 2005). Although these observations often
thalamus, including the amygdaloid complex, and blur clear distinctions, and stoke debates, about the
some hypothalamic regions need to be tested to existence of separate clinical categories, a trend
establish their putative role, or that of the auditory gaining momentum is to focus on specific symptoms
thalamus itself, in HPA axis habituation to repeated as putative biomarkers in the hope that such fine
loud noise exposures. grained analysis could discriminate between currently
Overall, the above studies are continuing to more closely related disorders. Phenotypic differences
precisely define the conditions under which stress associated with fear responses in various psychiatric
habituation takes place, and are honing in on some of populations have been at the center of several recent
the brain regions mediating these response reductions. studies, indicating that fear processing represents a
Some pressing questions revolve around defining target function discriminating between some mood
more precisely the multiple processes required for the disorders, and offering new preclinically driven
development of habituation to stressful stimuli, strategies in their treatments. An example of this
including the possibility that redundant neural emerging trend is the recent discovery that the
mechanisms are responsible for habituation to acoustic startle or its potentiation by fear-related
repeated stress, which might explain the difficulty to stimuli is enhanced in various mood and anxiety
510 S. Campeau et al.

disorder populations (Butler et al. 1990; Grillon et al. previously, PTSD patients had no HPA hormone
1993, 1994, 1998a,b; Morgan et al. 1995; Grillon and differences at baseline, but had reliably more
Morgan 1999; Lissek et al. 2010). Notably, the use of suppressed cortisol levels than non-PTSD trauma-
more precise discrimination tasks indicates that tized controls following dexamethasone (Jovanovic
patients with post-traumatic stress disorder (PTSD) et al. 2010b). Compared with the non-PTSD
or panic disorder fail to suppress fear to safety signals. traumatized controls, the PTSD patients again
Figure 6 demonstrates that control subjects and displayed an inability to suppress fear on the safe
subjects with major depression show much more trials, and this was significantly correlated with both
physiological fear to the danger cue (AX þ ) than they cortisol and ACTH at baseline and following
do to safety cues (BX 2 and AB). In contrast, those dexamethasone treatment. In addition, the lack of
with PTSD are unable to perform this emotional differences at baseline between diagnostic groups but
discrimination task (see Figure 6), an impairment that the correlation with physiological fear suggests that
is not observed in major depression or generalized fear physiology may be an intermediate phenotype
that is more closely related to HPA axis functioning
anxiety (Lissek et al. 2009, 2010; Jovanovic et al.
than is the diagnosis of PTSD per se.
2010a,b). An additional possible phenotypic discri-
The high degree of correlations observed between
minator between PTSD and panic disorder may
HPA axis and fear reactivity abnormalities may
involve the normal conscious and appropriate expect-
suggest that this link results from a common
ancy of danger and safety signals by PTSD patients
underlying neural dysfunction in this disorder.
(Jovanovic et al. 2009), which appears disrupted in Indeed, recent genetic and environmental studies of
panic disorder patients (Lissek et al. 2009). PTSD-related risk factors indicate that the strong
Modulation of the HPA axis is also a hallmark of association of child and adult trauma with PTSD
several mood disorders, and one of the distinguishing (Bremner et al. 1993; Widom 1999; Heim and
characteristics of trauma-related disorders, including Nemeroff 2001; Lang et al. 2006; Stovall-McClough
PTSD, is the exaggerated suppression of HPA- and Cloitre 2006) interacts significantly with specific
induced hormone release in the dexamethasone allelic genotypes of the FKBP5 protein (see Figure 7),
suppression test (Yehuda et al. 2002, 2004a,b; de a modulator of glucocorticoid receptor sensitivity
Kloet et al. 2007). To determine if HPA axis and fear (Binder et al. 2008; Yehuda et al. 2009; Xie et al.
abnormalities interact in PTSD, as described above, 2010). These studies are therefore beginning to offer
traumatized patients with and without PTSD were rational explanations for the large individual variations
given a fear-potentiated startle test under control or observed with regard to trauma-related disorders,
dexamethasone suppression conditions. As reported with vulnerability determined by a combination of
developmental/environmental (social support, trau-
200
matic events, etc.) and genetic factors (FKBP5 and
AX+ BX– AB other recently noted genes), together with improving
180
Percent Fear-Potentiated Startle

predictive power for individual risk, and a chance to


160
develop better intervention approaches to mitigate the
140 effects of existing susceptibilities.
120 ** Besides abnormalities in suppressing fear to safety
**
100 ** signals, deficits in the extinction of fear responses to
*
80 fear stimuli or situations, even when no longer
60
associated with aversive outcomes, are frequently
reported in PTSD populations (Rothbaum and Davis
40
2003; Bremner et al. 2005; Gillespie and Ressler
20
2005; Wessa and Flor 2007). Most recently, we have
0 further examined this phenomenon in a large
NO DX PTSD ONLY MDD ONLY
group (N ¼ 127) of all-traumatized individuals with
Figure 6. Different fear responses across traumatized subjects with and without PTSD (Norrholm et al. 2011). During
PTSD vs. depression (MDD). Fear-potentiated startle on AX þ fear extinction, the PTSD group showed elevated fear-
(danger), BX 2 (safety), and AB (transfer) trials across diagnostic potentiated startle responses to the previously
groups. The Y-axis represents average percent startle potentiation
for each trial type. These data show that control and depressed
reinforced CS þ during the early and middle stages
groups show good discrimination with the highest level of startle to of extinction. During the acquisition and extinction
the danger cue relative to the safety and transfer tasks. In contrast, phases, PTSD participants with higher levels of re-
those with PTSD have higher fear-potentiated startle to fear and experiencing symptoms exhibited greater potentiated
safety cues compared to others, suggesting that they are unable to startle responses to the CS þ compared to PTSD
discriminate these conditioned cues. *, within-subject trial type
effect, p , 0.05; **, within-subject trial type effect, p , 0.01.
participants with lower re-experiencing symptoms.
Reproduced and adapted with permission from Jovanovic et al. These results suggest that PTSD is associated with
(2010a). enhanced fear learning and diminished fear extinction,
Stress modulation of cognitive and affective processes 511

rs3800373 rs9296158
A 40 B 40
AA
AA AG
35 AC 35 GG
CC
PTSD Symptom Score (PSS)

30 30

25 25

PSS Score
20 20

15 15

10 10

5 5

0 0
No childhood 1 type of childhood 2 types of childhood No childhood 1 type of childhood 2 types of childhood
abuse abuse abuse abuse abuse abuse

Figure 7. Gene £ environment interactions predict risk for adult PTSD. Interaction of FKBP5 polymorphisms rs3800373 (A) or rs9296158
(B) with level of physical, emotional, or sexual child abuse history (x-axis) predicts level of PTSD symptoms (PTSD symptom scale, y-axis).
Reproduced and adapted with permission from Binder et al. (2008).

in part due to a greater ‘fear load’ to extinguish after bdnf SNP, an intriguing remediation possibility would
conditioned fear is acquired. be to ‘rescue’ these deficits by providing TrkB
Several animal studies have focused on neural stimulation directly. This possibility is currently
mechanisms mediating fear extinction in the pre- being tested using a recently discovered TrkB receptor
frontal cortex, amygdaloid complex, and hippocampal agonist that crosses the blood –brain barrier, 7-8-
formation (Milad et al. 2006; Chang et al. 2009), dihydroxyflavone (Jang et al. 2010), and has been
regions that are also of primary interest in mood shown to rescue the deficits induced in a region-
disorders, including PTSD (Liberzon and Sripada specific BDNF knock-out mice (Choi et al. 2010).
2008). Initial work provided evidence for the role of Recent demonstrations in enhancing fear extinction
the brain-derived neurotrophic factor (BDNF) system (see Figure 8), and restoration of extinction-like beha-
in the acquisition (Rattiner et al. 2004), and extinction viors in an animal model of stress-induced extinction
of fear memories (Chhatwal et al. 2006). For instance, deficits (Andero et al. 2011), could quickly translate to
injection of a lentiviral vector coding a truncated copy human trials similarly investigating the possibility of
of the relatively BDNF-specific tyrosine kinase enhancing fear extinction in PTSD, and other mood
receptor B (TrkB) neurotrophin receptor in the disorders. It is important to note that any cognitive
amygdala significantly retards extinction of fear- enhancer strategy to enhance fear extinction would
potentiated startle in rats (Chhatwal et al. 2006). need to also carefully take into account the possible
Additional preclinical studies also suggest a role of the effects on non-emotional memories as well. Only
hippocampal BDNF system in fear extinction, but not giving a TrkB agonist acutely in a limited fashion, at
its acquisition (Heldt et al. 2007). Recent results in the time of extinction-based exposure therapy, and not
humans generally support the animal studies whereby in a chronic, or ongoing fashion, might be one way to
a polymorphism of the human BDNF gene (Val66Met circumvent these potential concerns.
single nucleotide polymorphism—SNP) confers, The strategy of enhancing specific functions given
through reduced BDNF signaling, a retarded fear the deficits observed in patient populations such as
extinction phenotype in human heterozygote and PTSD is gaining support from a different, but similarly
homozygote Met carriers, as it does in knock-in mice preclinically driven approach, surrounding the role of
with analogous genotypes (Soliman et al. 2010). The excitatory N-methyl-D -aspartate (NMDA) receptor-
potential role of BDNF in mood disorders is further mediated neurotransmission in the acquisition
supported by associations with anxiogenic phenotypes (Miserendino et al. 1990; Kim et al. 1991) and
in normal and anxious/depressed patients (Lau et al. extinction (Falls et al. 1992) of learned fear. Whereas
2010; Montag et al. 2010), leading to hyperresponsive interference of NMDA receptor functions systemically
hippocampal and amygdala functional magnetic or directly at the level of the amygdaloid complex is
resonance imaging (fMRI) responses to aversive known to produce fear learning and extinction deficits,
stimulation. Given this association with a specific enhancement of their functions with the NMDA
512 S. Campeau et al.

IMO - Vehicle
cures rather than symptom control. Answers to
80 remaining questions that could potentially move the
IMO - 7,8-DHF
Freezing (% Time Spent)
70 field forward is the extent to which the two inhibitory
*** ***
60 *** functions previously discussed, stimulus safety dis-
50 ** crimination and extinction of aversive memories/
40 responses, are related to each other and mediated by
30
unique or multiple underlying neural processes. This
is a clear case in which the emerging clinical outcomes
20
suggest specific questions that could now be addressed
10
with animal research. An additional interesting
0 question concerns the extent to which habituation of
No Stimuli 1-6 7-12 13-18 19-24 25-30
prepotent responses to aversive stimuli is similar to
Conditioned stimuli
extinction of learned aversive memories, given the
Blocks of 6 trials phenomenological similarity of these two forms of
aversive response reduction. The emerging field of
Figure 8. Effect of a BDNF agonist molecule, 7,8-DHF on
extinction of fear in an animal model of PTSD. 7,8-DHF decreased
genetics and environmental interactions certainly
freezing in the within-session extinction trials in mice with a prior indicates a complex overall picture of the antecedent
history of immobilization. Previously, we have shown that prior conditions leading to mood and anxiety disorders, but
immobilization led to deficits in extinction of fear as a model of how these interactions modify specific cognitive and
PTSD. Freezing (average of 6 trials within each bin) is shown during emotional functions still has to be clearly defined.
within-extinction session, 1 h after 7,8-DHF or vehicle.
(***p , 0.001, **p , 0.01) Results are expressed as mean þ SD.
Reproduced and adapted with permission from Andero et al. (2011). Cortisol effects on human decision making (Israel
Liberzon, M.D., University of Michigan and Veterans
receptor partial agonist D -cycloserine facilitates fear Administration Medical Center, Ann Arbor, MI)
extinction rates (Walker et al. 2002). Interestingly, in As discussed in Dr Morilak’s and Ressler’s sections,
a BDNF Val66Met polymorphic animal model stress, frontal dysfunctions, and mood disorders are
displaying clear signs of extinction deficits following frequently associated together. An area of active
aversion learning, systemic D -cycloserine was effective research is related to the way humans select among
in normalizing extinction in this model (Yu et al. multiple alternative responses, especially under
2009). These animal studies are serving as the basis ambiguous options, which falls in the general category
for a treatment approach in which exposure therapy, of decision making. Theories based on the concept
a method of choice clinically employed to reduce or of utility, a measure of human value, defined by
minimize fear reactions to specific situations, could be considering the probabilities of reward and punish-
augmented by adjunct pharmacotherapy (D -cycloser- ment magnitudes associated with each option, have
ine) during exposure to the fear-eliciting situations generally been employed to explain decision making.
under controlled conditions (Davis et al. 2006). However, it was soon noted that people transform
Instead of focusing on reducing fear reactions directly probabilities and values of options in complex
with anxiolytics, this strategy focuses on improving the nonlinear manners, and that these transformations
learning and retention of ‘safety’ conditions, which are different for rewards or punishments, as described
would subsequently reduce fear reactions in those in a series of observations now embodied in the
situations. Several clinical trials have reported ‘cumulative prospect theory’ of decision making
statistically significant improvements in subjective (Tversky and Kahneman 1981, 1992). In addition,
and physiological measures following D -cycloserine to the observed overweighting of small probabilities
augmented exposure therapy in phobia (Ressler et al. and underweighting of large probabilities, people are
2004), social anxiety disorder (Hofmann et al. 2006; much more sensitive to losses than gains, which has
Guastella et al. 2008), and obsessive-compulsive led to the identification and mathematical formulation
disorder (Kushner et al. 2007; Wilhelm et al. 2008). of a loss aversion parameter in the prospect theory.
Current trials are underway to determine if D - Thus, decision making, as behaviorally defined in
cycloserine-augmented exposure to virtual reality- the cumulative prospect theory, is a final outcome of
based presentations of Iraq war cues will be effective in multiple processes that contribute to value estimate
reducing some of the PTSD symptoms from returning and probability assessment that are described by three
Iraqi war veterans (Gerardi et al. 2008). specific functions: loss aversion, reward discrimin-
These studies reveal the importance of focusing on ability, and a decision-weighting function.
precise neural functions in the overall approach to In recent years, a number of functional neuroima-
detail dysfunctions associated with specific mood and ging studies have attempted to identify specific regions
anxiety disorders. The goal of these rationally based distinctly associated with overall value representation
novel approaches is to provide a more direct path to in the brain and specifically in performing the distinct
Stress modulation of cognitive and affective processes 513

functions described above in people subjected to Britton et al. 2005; Phan et al. 2006; Urry et al. 2006).
uncertainty and potential loss during decision making. However, these original studies did not clearly
For instance, in a study designed to identify brain distinguish the possibility that the correlated neural
regions associated with behavioral parameters of loss activities directly influenced HPA axis responses, or
aversion, the surprising finding was that structures that the ensuing cortisol elevations from the activation
generally associated with aversive responses (amyg- of the HPA axis could have modified the observed
dala, insular cortex) were not identified (Tom et al. neural activity, due to the lengthy test sessions
2007). Instead, the same regions being selectively employed. Some of these limitations were addressed
activated during gain conditions (ventro-mPFC and in recent studies using more temporally restricted
portions of the dorsal and ventral striatum) displayed sampling methods of both the neural cerebral
decreased activity under loss conditions. Studies blood flow and the neuroendocrine release, indicating
focusing on the decision weighting function (prob- that the dorsal mPFC, the rostral anterior cingulate
ability estimate) have correlated nonlinear neural cortex, and the insular cortex were closely associated
activity associated with the probability assessment to with ACTH levels during various stress-inducing
the anterior cingulate cortex (Paulus and Frank 2006), conditions (Liberzon et al. 2007). Taking advantage of
the dorsolateral prefrontal cortex (Tobler et al. 2008), the finding that some patients show a lack of ACTH
and the ventral striatum (Hsu et al. 2009). In parallel, release (non-responders) to stress-induced HPA axis
decision-making tasks in rodents are also providing a activation (in combat-related PTSD patients as well
rich pattern of behavioral results (Simon et al. 2009), as combat-experienced non-PTSD subjects), it was
together with corresponding set of neural substrates recently reported (see Figure 9) that although the
mediating choices in these animals (St Onge and insular cortex activity increased in both ACTH
Floresco 2010). These initial general findings are responders and non-responders, mPFC activity was
indeed intriguing, but the detailed neurocircuitry increased only in non-responders (King et al. 2009),
involved in the specific components of decision which was associated with deactivation in the
making remains to be further elucidated. amygdala, hippocampus, and temporal pole area.
Activity in many of the same regions implicated in These results suggest that insular activity is associated
decision making has been recently reported to with HPA axis activation, while mPFC activity
correlate closely with the elaboration of specific stress dampens downstream brain activity including the
responses, including activation of the HPA axis, in HPA axis, as suggested from an increasing number
humans. A number of studies have linked increased of animal studies (Diorio et al. 1993; Crane et al.
activity in the amygdala, insular, dorsal and ventral 2003; Radley et al. 2006). The effects of glucocorti-
prefrontal, and anterior cingulate cortices with HPA coids alone (hydrocortisone injections) prior to a
axis activation (Shin et al. 2001; Tchiteya et al. 2003; psychological challenge have recently been found to

Figure 9. (A1, A2) Regions of greater activation in ‘ACTH responders’ vs. ‘non-responders’ (ACTH responders . nonresponders of
regional cerebral blood flow (rCBF) responses to trauma script (trauma script . neutral script contrast), (A1) insula and (A2) temporal pole.
p , 0.005 uncorrected). (B) Regions of greater activation in ‘ACTH non-responders’ vs. ‘responders’ (ACTH nonresponders . responders
rCBF responses to trauma script. Rostral anterior cingulate. All combat-exposed subjects (n ¼ 13 responders, n ¼ 12 ACTH nonresponders).
Trauma scripts were participant-specific emotionally arousing autobiographical script-driven imagery. Reproduced and adapted with
permission from King et al. (2009). (C, D), Areas of covariation with postscan plasma ACTH responses during 10 emotion–induction scans
in combat-PTSD patients and combat-exposed healthy comparison subjects. Regions of covariation in the rostral anterior cingulate (C1),
anterior insula (C2) in PTSD patients, and dorsal mPFC (Brodmann’s area 9) in healthy combat-exposed controls. Covarying voxels
( p , 0.005, uncorrected) are projected onto a canonical Montreal Neurological Institute brain. Reproduced and adapted with permission
from Liberzon et al. (2007).
514 S. Campeau et al.

moderate amygdaloid activity, which was again related The fact that stress influences multiple cognitive/
to mPFC activity (Henckens et al. 2010). affective functions simultaneously raises some
Given the overlap in regional activations so far important questions. For instance, are all the
noted in response to psychological stress and decision functions impacted by stress the result of a unique
making in humans, an emerging question concerns neural modification at a regional level or do they occur
the way in which stress modifies decision making. across different regions? In addition, are these stress-
Although a number of studies have reported signifi- related changes occurring via the same underlying
cant neuropsychological effects of stress on decision mechanism or via independent mechanisms and
making (Garvey and Klein 1993; Klein 1996; Preston modifications which are associated with different
et al. 2007; Starcke et al. 2008), the neural correlates functions? The development of increasingly sophisti-
of this impaired function have not been explored. cated cognitive tests across multiple functions would
Studies are currently underway, employing the provide answers to these questions, and in turn
cumulative prospect theory of decision making to suggest appropriate treatment options. The continu-
determine how glucocorticoids and stress modify ing interplay between basic, preclinical, and clinical
the activity of the neurocircuitry so far associated research associated with stress and stress-related
with different aspects of decision making under risk disorders is also of special importance. For instance,
and uncertainty (Paulus and Frank 2006; Tom et al. the non-invasive neuroimaging and neuropsychologi-
2007; Tobler et al. 2008; Hsu et al. 2009). Preliminary cal studies in patient populations under specific
results suggest that hydrocortisone administration cognitive demands are providing increasingly specific
modulates neural activity associated with loss aversion regional targets that are so far well recapitulated in
and reward processing in insular and extended animal studies. On the other hand, the increased
amygdala regions accordingly, but the signal associ- mechanistic approach afforded by animal research
ated with probability processing of the dorsolateral serves as a hypothesis testing ground to determine the
prefrontal cortex might be blunted. These findings, role of specific neurotransmitter systems/molecular
while preliminary, suggest that a novel role for stress changes associated with the functional phenotypes.
hormones is to alter behavior via modification of An additional advantage of the interdependency of
the decision-making process through feedback, in a clinical and preclinical research based on specific
manner adaptive to stressful environment. cognitive functions is the determination of the validity
The above human studies investigating the psycho- of specific animal models of disease or dysfunction,
logical and neural decision-making processes are thus which should, at the very least, display similar loss of
beginning to identify specific aspects of this complex functions compared to the human clinical condition.
function which are coupled to relatively localized brain These approaches are still in their infancy, but hold
loci. These findings, combined with the continuing promises to improved understanding of stress-related
interest in uncovering the influences of stress, and disorders, and novel rational approaches to their
hormones released by stress, during decision making treatments.
are helping to further define the specific effects that
stress and stress hormones exert on critical com- Declaration of interest: The research reported by
ponents of decision making. Exactly how these Dr Morilak was supported by the National Institutes
functions might be altered in stress-related disorders, of Health (NIH) grants MH053851, MH072672,
such as PTSD, is only beginning to be investigated and MH083699. Dr Morilak also receives financial
(Sailer et al. 2008). support and provides consultant expertise to Forest
Laboratories, Inc. (New York, NY), which is
unrelated to the research reported herein. Dr Ressler
is a cofounder of Extinction Pharmaceuticals for
Conclusion
NMDA-based therapeutic augmentation of psycho-
Analyses of the influence of acute and chronic stress therapy. The research reported by Dr Ressler was
on increasingly basic and well-defined cognitive/affec- provided by NIH grants MH071537 and DA019624,
tive functions are allowing a more precise description and the Burroughs Wellcome Fund. Drs Liberzon
of the brain regions, and in some instances, the and Campeau report no conflicts of interest, and the
associated neurotransmitter systems, mediating these research discussed in this chapter was supported by
functions. Simultaneously, these studies provide the NIH grants MH075999 (IL) and MH077152 (SC),
basis to determine whether and where in the brain respectively.
stress-related disorders modify these functions. How
different functions may be permanently modified
by stress, and more importantly, how these modifi- References
cations could be reversed back to relative normalcy Andero R, Heldt SA, Ye K, Liu X, Armario A, Ressler KJ. 2011.
with targeted treatments is obviously a matter of Effect of 7,8-dihydroxyflavone, a small-molecule TrkB agonist,
intensive investigations. on emotional learning. Am J Psychiatry 168:163–172.
Stress modulation of cognitive and affective processes 515

Anisman H, Matheson K. 2005. Stress, depression, and anhedonia: in a subgroup of Vietnam veterans with combat-related PTSD.
Caveats concerning animal models. Neurosci Biobehav Rev 29: Am J Psychiatry 147:1308– 1312.
525– 546. Campeau S, Akil H, Watson SJ. 1997. Lesions of the medial
Armario A, Castellanos JM, Balasch J. 1984. Adaptation of anterior geniculate nuclei specifically block corticosterone release and
pituitary hormones to chronic noise stress in male rats. Behav induction of c-fos mRNA in the forebrain associated with loud
Neural Biol 41:71–76. noise stress in rats. J Neurosci 17:5979 –5992.
Austin MP, Mitchell P, Goodwin GM. 2001. Cognitive deficits in Campeau S, Dolan D, Akil H, Watson SJ, Jr. 2002. c-fos mRNA
depression: Possible implications for functional neuropathology. induction in acute and chronic audiogenic stress: Possible role of
Br J Psychiatry 178:200 –206. the orbitofrontal cortex in habituation. Stress 5:121–130.
Bao G, Metreveli N, Fletcher EC. 1999. Acute and chronic Campeau S, Watson SJ. 1997. Neuroendocrine and behavioral
blood pressure response to recurrent acoustic arousal in rats. responses and brain pattern of c-fos induction associated with
Am J Hypertens 12:504–510. audiogenic stress. J Neuroendocrinol 9:577–588.
Bassett JR, Cairncross KD, King MG. 1973. Parameters of novelty, Campeau S, Watson SJ. 2000. Connections of some auditory-
shock predictability and response contingency in corticosterone responsive posterior thalamic nuclei putatively involved in
release in the rat. Physiol Behav 10:901–907. activation of the hypothalamo–pituitary–adrenocortical axis
Beck AT. 2005. The current state of cognitive therapy: A 40-year in response to audiogenic stress in rats: An anterograde
retrospective. Arch Gen Psychiatry 62:953–959. and retrograde tract tracing study combined with Fos
Bhatnagar S, Huber R, Nowak N, Trotter P. 2002. Lesions of the expression. J Comp Neurol 423:474–491.
posterior paraventricular thalamus block habituation of hypo- Carew TJ, Pinsker HM, Kandel ER. 1972. Long-term habituation
thalamic – pituitary –adrenal responses to repeated restraint. of a defensive withdrawal reflex in Aplysia. Science 175:
J Neuroendocrinol 14:403–410. 451 –454.
Binder EB, Bradley RG, Liu W, Epstein MP, Deveau TC, Mercer Cepeda NJ, Pashler H, Vul E, Wixted JT, Rohrer D. 2006.
KB, Tang Y, Gillespie CF, Heim CM, Nemeroff CB, Schwartz Distributed practice in verbal recall tasks: A review and
AC, Cubells JF, Ressler KJ. 2008. Association of FKBP5 quantitative synthesis. Psychol Bull 132:354–380.
polymorphisms and childhood abuse with risk of posttraumatic Chang CH, Knapska E, Orsini CA, Rabinak CA, Zimmerman JM,
stress disorder symptoms in adults. J Am Med Assoc 299: Maren S. 2009. Fear extinction in rodents. Curr Protoc
1291–1305. Neurosci 47:8.23.1 –8.23.17.
Birrell JM, Brown VJ. 2000. Medial frontal cortex mediates
Chhatwal JP, Stanek-Rattiner L, Davis M, Ressler KJ. 2006.
perceptual attentional set shifting in the rat. J Neurosci 20:
Amygdala BDNF signaling is required for consolidation but not
4320–4324.
encoding of extinction. Nat Neurosci 9:870–872.
Bondi CO, Jett JD, Morilak DA. 2010. Beneficial effects of
Choi DC, Maguschak KA, Ye K, Jang SW, Myers KM, Ressler KJ.
desipramine on cognitive function of chronically stressed rats are
2010. Prelimbic cortical BDNF is required for memory of
mediated by alpha1-adrenergic receptors in medial prefrontal
learned fear but not extinction or innate fear. Proc Natl Acad
cortex. Prog Neuropsychopharmacol Biol Psychiatry 34:
Sci USA 107:2675–2680.
913– 923.
Chrousos GP. 2000. The role of stress and the hypothalamic–
Bondi CO, Rodriguez G, Gould GG, Frazer A, Morilak DA. 2008.
pituitary–adrenal axis in the pathogenesis of the metabolic
Chronic unpredictable stress induces a cognitive deficit and
syndrome: Neuro-endocrine and target tissue-related causes.
anxiety-like behavior in rats that is prevented by chronic
Int J Obes Relat Metab Disord 24:S50–S55.
antidepressant drug treatment. Neuropsychopharmacology 33:
Chudasama Y, Robbins TW. 2003. Dissociable contributions of the
320– 331.
orbitofrontal and infralimbic cortex to pavlovian autoshaping
Borrell J, Torrellas A, Guaza C, Borrell S. 1980. Sound stimulation
and discrimination reversal learning: Further evidence for the
and its effects on the pituitary-adrenocortical function and brain
functional heterogeneity of the rodent frontal cortex. J Neurosci
catecholamines in rats. Neuroendocrinology 31:53–59.
Bremner JD. 2002. Neuroimaging studies in post-traumatic stress 23:8771–8780.
disorder. Curr Psychiatry Rep 4:254–263. Clarke HF, Walker SC, Crofts HS, Dalley JW, Robbins TW, Roberts
Bremner JD, Southwick SM, Johnson DR, Yehuda R, Charney DS. AC. 2005. Prefrontal serotonin depletion affects reversal
1993. Childhood physical abuse and combat-related posttrau- learning but not attentional set shifting. J Neurosci 25:532–538.
matic stress disorder in Vietnam veterans. Am J Psychiatry 150: Clarke HF, Walker SC, Dalley JW, Robbins TW, Roberts AC. 2007.
235– 239. Cognitive inflexibility after prefrontal serotonin depletion is
Bremner JD, Vermetten E, Schmahl C, Vaccarino V, Vythilingam behaviorally and neurochemically specific. Cereb Cortex 17:
M, Afzal N, Grillon C, Charney DS. 2005. Positron emission 18–27.
tomographic imaging of neural correlates of a fear acquisition Cole BJ, Robbins TW. 1992. Forebrain norepinephrine: Role in
and extinction paradigm in women with childhood sexual-abuse- controlled information processing in the rat. Neuropsychophar-
related post-traumatic stress disorder. Psychol Med 35: macology 7:129–142.
791– 806. Cole MA, Kalman BA, Pace TW, Topczewski F, Lowrey MJ,
Breslau N, Davis GC, Peterson EL, Schultz LR. 2000. A second Spencer RL. 2000. Selective blockade of the mineralocorticoid
look at comorbidity in victims of trauma: The posttraumatic receptor impairs hypothalamic – pituitary – adrenal axis
stress disorder-major depression connection. Biol Psychiatry 48: expression of habituation. J Neuroendocrinol 12:1034 –1042.
902– 909. Crane JW, Ebner K, Day TA. 2003. Medial prefrontal cortex
Britton JC, Phan KL, Taylor SF, Fig LM, Liberzon I. 2005. suppression of the hypothalamicpituitaryadrenal axis response
Corticolimbic blood flow in posttraumatic stress disorder during to a physical stressor, systemic delivery of interleukin-1. Eur J
script-driven imagery. Biol Psychiatry 57:832–840. Neurosci 17:1473 –1481.
Britton KT, Segal DS, Kuczenski R, Hauger R. 1992. Dissociation Dalley JW, Cardinal RN, Robbins TW. 2004. Prefrontal executive
between in vivo hippocampal norepinephrine response and and cognitive functions in rodents: Neural and neurochemical
behavioral/neuroendocrine responses to noise stress in rats. substrates. Neurosci Biobehav Rev 28:771–784.
Brain Res 574:125–130. Davidson RJ, Pizzagalli D, Nitschke JB, Putnam K. 2002. Depres-
Butler RW, Braff DL, Rausch JL, Jenkins MA, Sprock J, Geyer MA. sion: Perspectives from affective neuroscience. Annu Rev
1990. Physiological evidence of exaggerated startle response Psychol 53:545–574.
516 S. Campeau et al.

Davis M. 1970. Effects of interstimulus interval length and Grillon C, Morgan CA, III, Davis M, Southwick SM. 1998b. Effect
variability on startle-response habituation in the rat. of darkness on acoustic startle in Vietnam veterans with PTSD.
J Comp Physiol Psychol 72:177–192. Am J Psychiatry 155:812–817.
Davis M, Ressler K, Rothbaum BO, Richardson R. 2006. Effects of Grissom N, Bhatnagar S. 2008. Habituation to repeated stress:
D -cycloserine on extinction: Translation from preclinical to Get used to it. Neurobiol Learn Mem 92:215– 224.
clinical work. Biol Psychiatry 60:369–375. Grissom N, Iyer V, Vining C, Bhatnagar S. 2007. The physical
Day HE, Masini CV, Campeau S. 2009. Reversible inactivation of context of previous stress exposure modifies hypothalamic–
the auditory thalamus disrupts HPA axis habituation to repeated pituitary–adrenal responses to a subsequent homotypic stress.
loud noise stress exposures. Brain Res 1276:123–130. Horm Behav 51:95– 103.
De Boer SF, Slangen JL, van der Gugten J. 1988. Adaptation of Guastella AJ, Richardson R, Lovibond PF, Rapee RM, Gaston JE,
plasma catecholamine and corticosterone responses to short- Mitchell P, Dadds MR. 2008. A randomized controlled trial of
D -cycloserine enhancement of exposure therapy for social
term repeated noise stress in rats. Physiol Behav 44:273–280.
De Boer SF, Van der Gugten J, Slangen JL. 1989. Plasma anxiety disorder. Biol Psychiatry 63:544–549.
catecholamine and corticosterone responses to predictable and Harmer CJ, O’Sullivan U, Favaron E, Massey-Chase R, Ayres R,
Reinecke A, Goodwin GM, Cowen PJ. 2009. Effect of acute
unpredictable noise stress in rats. Physiol Behav 45:789–795.
antidepressant administration on negative affective bias in
de Kloet CS, Vermetten E, Heijnen CJ, Geuze E, Lentjes EGWM,
depressed patients. Am J Psychiatry 166:1178–1184.
Westenberg HGM. 2007. Enhanced cortisol suppression in
Heim C, Nemeroff CB. 2001. The role of childhood trauma in the
response to dexamethasone administration in traumatized
neurobiology of mood and anxiety disorders: Preclinical and
veterans with and without posttraumatic stress disorder.
clinical studies. Biol Psychiatry 49:1023–1039.
Psychoneuroendocrinology 32:215–226.
Heldt SA, Stanek L, Chhatwal JP, Ressler KJ. 2007. Hippocampus-
Devauges V, Sara SJ. 1990. Activation of the noradrenergic system
specific deletion of BDNF in adult mice impairs spatial memory
facilitates an attentional shift in the rat. Behav Brain Res 39: and extinction of aversive memories. Mol Psychiatry 12:
19–28. 656 –670.
Diorio D, Viau V, Meaney MJ. 1993. The role of the medial Henckens MJAG, van Wingen GA, Joels M, Fernandez G. 2010.
prefrontal cortex (cingulate gyrus) in the regulation of Time-dependent effects of corticosteroids on human amygdala
hypothalamic–pituitary– adrenal responses to stress. J Neurosci processing. J Neurosci 30:12725–12732.
13:3839– 3847. Henkin RI, Knigge KM. 1963. Effect of sound on the
Dobrakovova M, Kvetnansky R, Oprsalova Z, Jezova D. 1993. hypothalamic – pituitary – adrenal axis. Am J Physiol 204:
Specificity of the effect of repeated handling on sympathetic- 910 –914.
adrenomedullary and pituitary-adrenocortical activity in rats. Hennessy JW, Levin R, Levine S. 1977. Influence of experiential
Psychoneuroendocrinology 18:163–174. factors and gonadal hormones on pituitary-adrenal response of
Drevets WC, Price JL, Simpson JR, Jr, Todd RD, Reich T, the mouse to novelty and electric shock. J Comp Physiol Psychol
Vannier M, Raichle ME. 1997. Subgenual prefrontal cortex 91:770–777.
abnormalities in mood disorders. Nature 386:824 –827. Hinde RA. 1954. Factors governing the changes in strength of a
Falls WA, Miserendino MJD, Davis M. 1992. Extinction of fear- partially inborn response, as shown by the mobbing behaviour of
potentiated startle: Blockade by infusion of an NMDA the chaffinch (Fringilla coelebs). I. The nature of the response,
antagonist into the amygdala. J Neurosci 12:854–863. and an examination of its course. Proc R Soc Lond B Biol Sci
Fossati P, Ergis AM, Allilaire JF. 2002. Executive functioning in 142:306–331.
unipolar depression: A review. Encephale 28:97–107. Hofmann SG, Meuret AE, Smits JA, Simon NM, Pollack MH,
Gamallo A, Alario P, Gonzalez-Abad MJ, Villanua MA. 1992. Eisenmenger K, Shiekh M, Otto MW. 2006. Augmentation of
Acute noise stress, ACTH administration, and blood pressure exposure therapy with D -cycloserine for social anxiety disorder.
alteration. Physiol Behav 51:1201 –1205. Arch Gen Psychiatry 63:298–304.
Garvey BJ, Klein K. 1993. Releationship of life stress and body Hsu M, Krajbich I, Zhao C, Camerer CF. 2009. Neural response
consciousness to hypervigilant decision making. J Personality to reward anticipation under risk is nonlinear in probabilities.
Soc Psychol 64:267–273. J Neurosci 29:2231–2237.
Gerardi M, Rothbaum BO, Ressler K, Heekin M, Rizzo A. 2008. Irwin MR, Segal DS, Hauger RL, Smith TL. 1989. Individual
Virtual reality exposure therapy using a virtual Iraq: Case report. behavioral and neuroendocrine differences in responsiveness to
audiogenic stress. Pharmacol Biochem Behav 32:913–917.
J Trauma Stress 21:209–213.
Jaferi A, Nowak N, Bhatnagar S. 2003. Negative feedback functions
Ghahremani DG, Monterosso J, Jentsch JD, Bilder RM, Poldrack
in chronically stressed rats: Role of the posterior paraventricular
RA. 2010. Neural components underlying behavioral flexibility
thalamus. Physiol Behav 78:365–373.
in human reversal learning. Cereb Cortex 20:1843–1852.
Jang SW, Liu X, Yepes M, Shepherd KR, Miller GW, Liu Y, Wilson
Gillespie CF, Ressler KJ. 2005. Emotional learning and glutamate:
WD, Xiao G, Blanchi B, Sun YE, Ye K. 2010. A selective TrkB
Translational perspectives. CNS Spectr 10:831–839.
agonist with potent neurotrophic activities by 7,8-dihydroxy-
Grillon C, Ameli R, Foot M, Davis M. 1993. Fear-potentiated
flavone. Proc Natl Acad Sci USA 107:2687–2692.
startle: Relationship to the level of state/trait anxiety in healthy Jordan WP, Strasser HC, McHale L. 2000. Contextual control of
subjects. Biol Psychiatry 33:566–574. long-term habituation in rats. J Exp Psychol Anim Behav Process
Grillon C, Ameli R, Goddard A, Woods SW, Davis M. 1994. 26:323–339.
Baseline and fear-potentiated startle in panic disorder patients. Jovanovic T, Norrholm SD, Blanding NQ, Davis M, Duncan E,
Biol Psychiatry 35:431–439. Bradley B, Ressler KJ. 2010a. Impaired fear inhibition is a
Grillon C, Morgan CA, III. 1999. Fear-potentiated startle biomarker of PTSD but not depression. Depress Anxiety 27:
conditioning to explicit and contextual cues in Gulf War 244 –251.
veterans with posttraumatic stress disorder. J Abnorm Psychol Jovanovic T, Norrholm SD, Blanding NQ, Phifer JE, Weiss T, Davis
108:134– 142. M, Duncan E, Bradley B, Ressler K. 2010b. Fear potentiation is
Grillon C, Morgan CA, III, Davis M, Southwick SM. 1998a. Effects associated with hypothalamic–pituitary–adrenal axis function in
of experimental context and explicit threat cues on acoustic PTSD. Psychoneuroendocrinology 35:846–857.
startle in Vietnam veterans with posttraumatic stress disorder. Jovanovic T, Norrholm SD, Fennell JE, Keyes M, Fiallos AM,
Biol Psychiatry 44:1027–1036. Myers KM, Davis M, Duncan EJ. 2009. Posttraumatic stress
Stress modulation of cognitive and affective processes 517

disorder may be associated with impaired fear inhibition: Lissek S, Rabin SJ, McDowell DJ, Dvir S, Bradford DE, Geraci M,
Relation to symptom severity. Psychiatry Res 167:151–160. Pine DS, Grillon C. 2009. Impaired discriminative fear-
Kamin LJ. 1969. Selective association and conditioning. conditioning resulting from elevated fear responding to learned
In: Mackintosh NJ, Honig WK, editors. Fundamental issues in safety cues among individuals with panic disorder. Behav Res
associative learning. Halifax, Nova Scotia: Dalhousie University Ther 47:111–118.
Press. p 42–64. Liston C, Miller MM, Goldwater DS, Radley JJ, Rocher AB, Hof
Kendler KS, Karkowski LM, Prescott CA. 1999. Causal PR, Morrison JH, McEwen BS. 2006. Stress-induced alterations
relationship between stressful life events and the onset of major in prefrontal cortical dendritic morphology predict selective
depression. Am J Psychiatry 156:837–841. impairments in perceptual attentional set-shifting. J Neurosci
Kessler RC, Sonnega A, Bromet E, Hughes M, Nelson CB. 1995. 26:7870–7874.
Posttraumatic stress disorder in the National Comorbidity Lubow RE, Moore AU. 1959. Latent inhibition: The effect
Survey. Arch Gen Psychiatry 52:1048 –1060. of nonreinforced pre-exposure to the conditional stimulus.
Kim JJ, DeCola JP, Landeira-Fernandez J, Fanselow MS. 1991. J Comp Physiol Psychol 52:415–419.
N-methyl-D -aspartate receptor antagonist APV blocks acqui- Marti O, Armario A. 1998. Anterior pituitary response to stress:
sition but not expression of fear conditioning. Behav Neurosci Time-related changes and adaptation. Int J Dev Neurosci 16:
105:126– 133. 241 –260.
Kim J, Ragozzino ME. 2005. The involvement of the orbitofrontal Masini CV, Day HE, Campeau S. 2008. Long-term habituation to
cortex in learning under changing task contingencies. Neurobiol repeated loud noise is impaired by relatively short interstressor
Learn Mem 83:125–133. intervals in rats. Behav Neurosci 122:210–223.
King AP, Abelson JL, Britton JC, Phan KL, Taylor SF, Liberzon I. McAlonan K, Brown VJ. 2003. Orbital prefrontal cortex mediates
2009. Medial prefrontal cortex and right insula activity predict reversal learning and not attentional set shifting in the rat. Behav
plasma ACTH response to trauma recall. Neuroimage 47: Brain Res 146:97–103.
872– 880. Menzel R, Manz G, Greggers U. 2001. Massed and spaced learning
Klein G. 1996. The effect of acute stressors on decision-making. in honeybees: The role of CS, US, the intertrial interval,
In: Driskell JE, Salas E, editors. Stress and human performance. and the test interval. Learn Mem 8:198–208.
Mahwah, NJ: Erlbaum. p 49–88. Merriam EP, Thase ME, Haas GL, Keshavan MS, Sweeney JA.
Kushner MG, Kim SW, Donahue C, Thuras P, Adson D, Kotlyar 1999. Prefrontal cortical dysfunction in depression determined
M, McCabe J, Peterson J, Foa EB. 2007. D -Cycloserine by Wisconsin card sorting test performance. Am J Psychiatry
augmented exposure therapy for obsessive-compulsive disorder. 156:780–782.
Biol Psychiatry 62:835–838. Milad MR, Rauch SL, Pitman RK, Quirk GJ. 2006. Fear extinction
Lang AJ, Laffaye C, Satz LE, McQuaid JR, Malcarne VL, in rats: Implications for human brain imaging and anxiety
Dresselhaus TR, Stein MB. 2006. Relationships among disorders. Biol Psychol 73:61–71.
childhood maltreatment, PTSD, and health in female veterans Miserendino MJ, Sananes CB, Melia KR, Davis M. 1990. Blocking
in primary care. Child Abuse Negl 30:1281 –1292. of acquisition but not expression of conditioned fear-potentiated
Lapiz MD, Bondi CO, Morilak DA. 2007. Chronic treatment with startle by NMDA antagonists in the amygdala. Nature 345:
desipramine improves cognitive performance of rats in an 716 –718.
attentional set-shifting test. Neuropsychopharmacology 32: Montag C, Basten U, Stelzel C, Fiebach CJ, Reuter M. 2010.
1000–1010. The BDNF Val66Met polymorphism and anxiety: Support for
Lapiz MD, Morilak DA. 2006. Noradrenergic modulation of animal knock-in studies from a genetic association study in
cognitive function in rat medial prefrontal cortex as measured humans. Psychiatry Res 179:86–90.
by attentional set shifting capability. Neuroscience 137: Morgan CA, III, Grillon C, Southwick SM, Davis M, Charney DS.
1039–1049. 1995. Fear-potentiated startle in posttraumatic stress disorder.
Lapiz-Bluhm MDS, Morilak DA. 2010. A cognitive deficit induced Biol Psychiatry 38:378–385.
in rats by chronic intermittent cold stress is reversed by chronic Morilak DA, Frazer A. 2004. Antidepressants and brain mono-
antidepressant treatment. Int J Neuropsychopharmacol 13: aminergic systems: A dimensional approach to understanding
997– 1009. their behavioural effects in depression and anxiety disorders.
Lapiz-Bluhm MDS, Soto-Pina AE, Hensler JG, Morilak DA. 2009. Int J Neuropsychopharmacol 7:193–218.
Chronic intermittent cold stress and serotonin depletion induce Norrholm SD, Jovanovic T, Olin IW, Sands LA, Karapanou I,
deficits of reversal learning in an attentional set-shifting test in Bradley B, Ressler KJ. 2011. Fear extinction in traumatized
rats. Psychopharmacology (Berl) 202:329 –341. civilians with posttraumatic stress disorder: Relation to symptom
Lau JY, Goldman D, Buzas B, Hodgkinson C, Leibenluft E, severity. Biol Psychiatry 69:556– 563.
Nelson E, Sankin L, Pine DS, Ernst M. 2010. BDNF gene Nyhuis TJ, Sasse SK, Masini CV, Day HE, Campeau S. 2010.
polymorphism (Val66Met) predicts amygdala and anterior Lack of contextual modulation of habituated neuroendocrine
hippocampus responses to emotional faces in anxious and responses to repeated audiogenic stress. Behav Neurosci 124:
depressed adolescents. Neuroimage 53:952–961. 810 –820.
Liberzon I, King AP, Britton JC, Phan KL, Abelson JL, Taylor SF. Overton JM, Kregel KC, Davis-Gorman G, Seals DR, Tipton CM,
2007. Paralimbic and medial prefrontal cortical involvement in Fisher LA. 1991. Effects of exercise training on responses
neuroendocrine responses to traumatic stimuli. Am J Psychiatry to central injection of CRF and noise stress. Physiol Behav 49:
164:1250–1258. 93–98.
Liberzon I, Sripada CS. 2008. The functional neuroanatomy of Page ME, Lucki I. 2002. Effects of acute and chronic reboxetine
PTSD: A critical review. Prog Brain Res 167:151 –169. treatment on stress-induced monoamine efflux in the rat frontal
Lissek S, Powers AS, McClure EB, Phelps EA, Woldehawariat G, cortex. Neuropsychopharmacology 27:237–247.
Grillon C, Pine DS. 2005. Classical fear conditioning in the Paulus MP, Frank LR. 2006. Anterior cingulate activity modulates
anxiety disorders: A meta-analysis. Behav Res Ther 43: nonlinear decision weight function of uncertain prospects.
1391–1424. Neuroimage 30:668– 677.
Lissek S, Rabin S, Heller RE, Lukenbaugh D, Geraci M, Pine DS, Pfister HP. 1979. The glucocorticosterone response to novelty as
Grillon C. 2010. Overgeneralization of conditioned fear as a a psychological stressor. Physiol Behav 23:649–652.
pathogenic marker of panic disorder. Am J Psychiatry 167: Phan KL, Britton JC, Taylor SF, Fig LM, Liberzon I. 2006.
47–55. Corticolimbic blood flow during nontraumatic emotional
518 S. Campeau et al.

processing in posttraumatic stress disorder. Arch Gen Psychiatry Glover G, Ballon DJ, Liston C, Teslovich T, Van Kempen T,
63:184–192. Lee FS, Casey BJ. 2010. A genetic variant BDNF polymorphism
Preston SD, Buchanan TW, Stansfield RB, Bechara A. 2007. alters extinction learning in both mouse and human. Science
Effects of anticipatory stress on decision making in a gambling 327:863–866.
task. Behav Neurosci 121:257–263. St Onge JR, Floresco SB. 2010. Prefrontal cortical contribution to
Rabasa C, Delgado-Morales R, Munoz-Abellan C, Nadal R, risk-based decision making. Cereb Cortex 20:1816–1828.
Armario A. 2011. Adaptation of the hypothalamic–pituitary – Starcke K, Wolf OT, Markowitsch HJ, Brand M. 2008. Anticipatory
adrenal axis and glucose to repeated immobilization or restraint stress influences decision making under explicit risk conditions.
stress is not influenced by associative signals. Behav Brain Res Behav Neurosci 122:1352–1360.
217:232– 239. Stovall-McClough KC, Cloitre M. 2006. Unresolved attachment,
Radley JJ, Arias CM, Sawchenko PE. 2006. Regional Differen- PTSD, and dissociation in women with childhood abuse
tiation of the medial prefrontal cortex in regulating adaptive histories. J Consult Clin Psychol 74:219–228.
responses to acute emotional stress. J Neurosci 26: Tafet GE, Bernardini R. 2003. Psychoneuroendocrinological links
12967–12976. between chronic stress and depression. Prog Neuro-Psycho-
Ragozzino ME, Detrick S, Kesner RP. 1999. Involvement of the pharm Biol Psychiatry 27:893–903.
prelimbic-infralimbic areas of the rodent prefrontal cortex in Tchiteya BM, Lecours AR, Elie R, Lupien SJ. 2003. Impact of a
behavioral flexibility for place and response learning. J Neurosci unilateral brain lesion on cortisol secretion and emotional state:
19:4585– 4594. Anterior/posterior dissociation in humans. Psychoneuroendo-
Ramos BP, Arnsten AF. 2007. Adrenergic pharmacology and crinology 28:674–686.
cognition: Focus on the prefrontal cortex. Pharmacol Ther 113: Tobler PN, Christopoulos GI, O’Doherty JP, Dolan RJ, Schultz W.
523– 536. 2008. Neuronal distortions of reward probability without choice.
Rattiner LM, Davis M, French CT, Ressler KJ. 2004. Brain-derived J Neurosci 28:11703–11711.
neurotrophic factor and tyrosine kinase receptor B involvement Tom SM, Fox CR, Trepel C, Poldrack RA. 2007. The neural basis
in amygdala-dependent fear conditioning. J Neurosci 24: of loss aversion in decision-making under risk. Science 315:
4796–4806. 515 –518.
Ressler KJ, Nemeroff CB. 2000. Role of serotonergic and Tversky A, Kahneman D. 1981. The framing of decisions and the
noradrenergic systems in the pathophysiology of depression psychology of choice. Science 211:453 –458.
and anxiety disorders. Depress Anxiety 12(Suppl. 1):2–19. Tversky A, Kahneman D. 1992. Advances in prospect theory:
Ressler KJ, Rothbaum BO, Tannenbaum L, Anderson P, Graap K, Cumulative representation of uncertainty. J Risk Uncertain 5:
Zimand E, Hodges L, Davis M. 2004. Cognitive enhancers as 297 –323.
adjuncts to psychotherapy: Use of D -cycloserine in phobic Urry HL, van Reekum CM, Johnstone T, Kalin NH, Thurow ME,
individuals to facilitate extinction of fear. Arch Gen Psychiatry Schaefer HS, Jackson CA, Frye CJ, Greischar LL, Alexander
61:1136– 1144. AL, Davidson RJ. 2006. Amygdala and ventromedial prefrontal
Rogers MA, Kasai K, Koji M, Fukuda R, Iwanami A, Nakagome K, cortex are inversely coupled during regulation of negative affect
Fukuda M, Kato N. 2004. Executive and prefrontal dysfunction and predict the diurnal pattern of cortisol secretion among older
in unipolar depression: A review of neuropsychological and adults. J Neurosci 26:4415–4425.
imaging evidence. Neurosci Res 50:1–11. van Raaij MTM, Dobbe CJG, Elvers B, Timmerman A, Schenk E,
Rose JK, Rankin CH. 2001. Analyses of habituation in caenorhab- Oortgiesen M, Wiegant VM. 1997. Hormonal status and the
ditis elegans. Learn Mem 8:63– 69. neuroendocrine response to a novel heterotypic stressor
Rothbaum BO, Davis M. 2003. Applying learning principles to the involving subchronic noise exposure. Neuroendocrinology 65:
treatment of post-trauma reactions. Ann NY Acad Sci 1008: 200 –209.
112– 121. Wagner AR 1978. Expectancies and the priming of STM.
Saha S, Gandhi A, Das S, Kaur P, Singh SH. 1996. Effect of noise In: Hulse SH, editor. Cognitive processes in animal behavior.
stress on some cardiovascular parameters and audiovisual Hillsdale, NJ: Erlbaum. p 177–201.
reaction time. Indian J Physiol Pharmacol 40:35–40. Wagner AR. 1979. Habituation and memory. In: Dickinson A,
Sailer U, Robinson S, Fischmeister FP, Konig D, Oppenauer C, Boakes RA, editors. Mechanisms of learning and motivation.
Lueger-Schuster B, Moser E, Kryspin-Exner I, Bauer H. 2008. Hillsdale, NJ: Erlbaum. p 53–82.
Altered reward processing in the nucleus accumbens and mesial Wagner AR. 1981. A model of automatic memory processing in
prefrontal cortex of patients with posttraumatic stress disorder. animal behavior. In: Spear NE, Miller RR, editors. Information
Neuropsychologia 46:2836–2844. processing in animals: Memory mechanisms. Hillsdale, NJ:
Schatzberg AF, Schildkraut JJ. 1995. Recent studies on norepi- L. Erlbaum Associates. p 5–47.
nephrine systems in mood disorders. In: Bloom FE, Kupfer DJ, Walker DL, Ressler KJ, Lu KT, Davis M. 2002. Facilitation of
editors. Psychopharmacology: The fourth generation of pro- conditioned fear extinction by systemic administration or
gress. New York: Raven Press, Ltd. p 911–920. intra-amygdala infusions of D -cycloserine as assessed with
Schoenbaum G, Nugent SL, Saddoris MP, Setlow B. 2002. fear-potentiated startle in rats. J Neurosci 22:2343–2351.
Orbitofrontal lesions in rats impair reversal but not acquisition Wessa M, Flor H. 2007. Failure of extinction of fear responses in
of go, no-go odor discriminations. Neuroreport 13:885–890. posttraumatic stress disorder: Evidence from second-order
Segal DS, Kuczenski R, Swick D. 1989. Audiogenic stress response: conditioning. Am J Psychiatry 164:1684–1692.
Behavioral characteristics and underlying monoamine mechan- Widom CS. 1999. Posttraumatic stress disorder in abused and
isms. J Neural Transm 75:31–50. neglected children grown up. Am J Psychiatry 156:1223– 1229.
Shin LM, Whalen PJ, Pitman RK, Bush G, Macklin ML, Lasko NB, Wilhelm S, Buhlmann U, Tolin DF, Meunier SA, Pearlson GD,
Orr SP, McInerney SC, Rauch SL. 2001. An fMRI study of Reese HE, Cannistraro P, Jenike MA, Rauch SL. 2008.
anterior cingulate function in posttraumatic stress disorder. Biol Augmentation of behavior therapy with D -cycloserine for
Psychiatry 50:932–942. obsessive-compulsive disorder. Am J Psychiatry 165:335 –341,
Simon NW, Gilbert RJ, Mayse JD, Bizon JL, Setlow B. 2009. quiz 409.
Balancing risk and reward: A rat model of risky decision making. Xie P, Kranzler HR, Poling J, Stein MB, Anton RF, Farrer LA,
Neuropsychopharmacology 34:2208–2217. Gelernter J. 2010. Interaction of FKBP5 with childhood
Soliman F, Glatt CE, Bath KG, Levita L, Jones RM, Pattwell SS, adversity on risk for post-traumatic stress disorder. Neuropsy-
Jing D, Tottenham N, Amso D, Somerville LH, Voss HU, chopharmacology 35:1684 –1692.
Stress modulation of cognitive and affective processes 519

Yehuda R, Cai G, Golier JA, Sarapas C, Galea S, Ising M, Rein T, severity in the parent and child. Psychoneuroendocrinology 27:
Schmeidler J, Muller-Myhsok B, Holsboer F, Buxbaum JD. 171 –180.
2009. Gene expression patterns associated with posttraumatic Yehuda R, Halligan SL, Golier JA, Grossman R, Bierer LM. 2004b.
stress disorder following exposure to the World Trade Center Effects of trauma exposure on the cortisol response to
attacks. Biol Psychiatry 66:708–711. dexamethasone administration in PTSD and major depressive
Yehuda R, Golier JA, Halligan SL, Meaney M, Bierer LM. 2004a. disorder. Psychoneuroendocrinology 29:389–404.
The ACTH response to dexamethasone in PTSD. Am J Yu H, Wang Y, Pattwell S, Jing D, Liu T, Zhang Y, Bath KG, Lee FS,
Psychiatry 161:1397–1403. Chen ZY. 2009. Variant BDNF Val66Met polymorphism affects
Yehuda R, Halligan SL, Bierer LM. 2002. Cortisol levels in adult extinction of conditioned aversive memory. J Neurosci 29:
offspring of Holocaust survivors: Relation to PTSD symptom 4056–4064.
Copyright of Stress: The International Journal on the Biology of Stress is the property of Taylor & Francis Ltd
and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for individual use.

You might also like