SlideShare a Scribd company logo
1 of 79
Every day, parents observe the growing behavioural
repertoires of their infants and young children, and
the corresponding changes in cognitive and emotional
functions. These changes are thought to relate to normal
brain development, particularly the development of the
hippocampus, the amygdala and the frontal lobes, and
the complex circuitry that connects these brain regions.
At the other end of the age spectrum, we observe changes
in cognition that accompany aging in our parents. These
changes are related to both normal and pathological
brain processes associated with aging.
Studies in animals and humans have shown that
during both early childhood and old age the brain
is particularly sensitive to stress, probably because it
undergoes such important changes during these periods.
Furthermore, research now relates exposure to early-life
stress with increased reactivity to stress and cognitive
deficits in adulthood, indicating that the effects of stress
at different periods of life interact.
Stress triggers the activation of the hypothalamus-
pituitary-adrenal (HPA) axis, culminating in the pro-
duction of glucocorticoids by the adrenals (FIG. 1).
Receptors for these steroids are expressed throughout
the brain; they can act as transcription factors and so
regulate gene expression. Thus, glucocorticoids can have
potentially long-lasting effects on the functioning of the
brain regions that regulate their release.
This Review describes the effects of stress during pre-
natal life, infancy, adolescence, adulthood and old age on
the brain, behaviour and cognition, using data from ani-
mal (BOX 1) and human studies. Here, we propose a model
that integrates the effects of stress across the lifespan,
along with future directions for stress research.
Prenatal stress
Animal studies. In animals, exposure to stress early in
life has ‘programming’ effects on the HPA axis and the
brain1. A single or repeated exposure of a pregnant
female to stress2 or to glucocorticoids3 increases mater-
nal glucocorticoid secretion. A portion of these gluco-
corticoids passes through the placenta to reach the fetus,
increasing fetal HPA axis activity and modifying brain
development4. In rats prenatal stress leads to long-term
increases in HPA axis activity 5. Controlling glucocor-
ticoid levels in stressed dams by adrenalectomy and
hormone replacement prevents these effects, indicating
that elevations in maternal glucocorticoids mediate the
prenatal programming of the HPA axis6.
Glucocorticoids are important for normal brain
maturation: they initiate terminal maturation, remodel
axons and dendrites and affect cell survival7; both sup-
pressed and elevated glucocorticoid levels impair brain
development and functioning. For example, admin-
istration of synthetic glucocorticoids to pregnant rats
delays the maturation of neurons, myelination, glia
and vasculature in the offspring, significantly altering
neuronal structure and synapse formation and inhibit-
ing neurogenesis4. Furthermore, juvenile and adult rats
exposed to prenatal stress have decreased numbers of
mineralocorticoid receptors (MRs) and glucocorticoid recep-
tors (GRs) in the hippocampus, possibly because of epi -
genetic effects on gene transcription8. The hippocampus
*Université de Montréal,
Mental Health Research
Centre, Fernand Seguin
Hôpital Louis‑ H Lafontaine,
Montreal, Quebec, H1N 3V2,
Canada.
‡Laboratory of
Neuroendocrinology, The
Rockefeller University, 1230
York Avenue, New York,
New York 10021, USA.
§Institute of Child
Development, University of
Minnesota, Minneapolis,
Minnesota 55455, USA.
||Department of Psychiatry,
Emory University, 101
Woodruff Circle, Suite 4000,
Atlanta, Georgia 30307, USA.
Correspondence to S.J.L.
e‑ mail: [email protected]
umontreal.ca
doi:10.1038/nrn2639
Published online 29 April 2009
Programming
When an environmental factor
that acts during a sensitive
developmental period affects
the structure and function of
tissues, leading to effects that
persist throughout life.
Effects of stress throughout
the lifespan on the brain,
behaviour and cognition
Sonia J. Lupien*, Bruce S. McEwen‡, Megan R. Gunnar § and
Christine Heim||
Abstract | Chronic exposure to stress hormones, whether it
occurs during the prenatal
period, infancy, childhood, adolescence, adulthood or aging, has
an impact on brain
structures involved in cognition and mental health. However,
the specific effects on the
brain, behaviour and cognition emerge as a function of the
timing and the duration of
the exposure, and some also depend on the interaction between
gene effects and previous
exposure to environmental adversity. Advances in animal and
human studies have made it
possible to synthesize these findings, and in this Revi ew a
model is developed to explain why
different disorders emerge in individuals exposed to stress at
different times in their lives.
R E V I E W S
434 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
mailto:[email protected]
mailto:[email protected]
Nature Reviews | Neuroscience
Hippocampus
Hypothalamus
Amygdala
CRH
AVP
ACTH
Glucocorticoids
Adrenal cortex
Anterior pituitary
Frontal cortex
GRs
GRs
GRs
MRs and
GRs
Mineralocorticoid receptor
A receptor that is activated by
mineralocorticoids, such as
aldosterone and deoxycorti-
costerone, as well as
glucocorticoids, such as
cortisol and cortisone. It also
responds to progestins.
Glucocorticoid receptor
A receptor that is activated by
cortisol, corticosterone and
other glucocorticoids and is
expressed in almost every cell
in the body. It regulates genes
controlling development,
metabolism and the immune
response.
inhibits HPA axis activity (FIG. 1), and a prenatal stress-
induced reduction in hippocampal MRs and GRs could
decrease this inhibition, with a resulting increase in basal
and/or stress-induced glucocorticoid secretion. In rhe-
sus monkeys, prenatal treatment with the synthetic GR
agonist dexamethasone causes a dose-dependent degen-
eration of hippocampal neurons, leading to a reduced
hippocampal volume at 20 months of age9.
Effects on other brain regions are also apparent.
Rats exposed to stress during the last week of gestation
have significantly decreased dendritic spine density in
the anterior cingulate gyrus and orbitofrontal cortex10.
Furthermore, prenatal exposure to glucocorticoids leads
to increased adult corticotropin-releasing hormone
(CRH) levels in the central nucleus of the amygdala, a
key region in the regulation of fear and anxiety11.
Exposure to prenatal stress has three major effects
on adult behaviour: learning impairments, especially
in aging rats12; enhanced sensitivity to drugs of abuse13;
and increases in anxiety- and depression-related behav-
iours14. The impaired learning is thought to result from
the effects of prenatal stress on hippocampal function15,
whereas the effects on anxiety are thought to be medi-
ated by prenatal stress-induced increases in CRH in the
amygdala11. Prenatal glucocorticoid exposure affects
the developing dopaminergic system, which is involved
in reward- or drug-seeking behaviour16, and it has been
suggested that the increased sensitivity to drugs of abuse
is related to the interaction between prenatal stress,
glucocorticoids and dopaminergic neurons16.
Human studies. In agreement with animal data, findings
from retrospective studies on children whose mothers
experienced psychological stress or adverse events or
received exogenous glucocorticoids during pregnancy
suggest that there are long-term neurodevelopmental
effects17. First, maternal stress or anxiety18, depression19
and glucocorticoid treatment during pregnancy17 have
been linked with lower birthweight or smaller size (for
gestational age) of the baby. More importantly, mater-
nal stress, depression and anxiety have been associated
with increased basal HPA axis activity in the offspring
at different ages, including 6 months20, 5 years21 and
10 years22.
Disturbances in child development (both neurologi-
cal and cognitive) and behaviour have been associated
with maternal stress23 and maternal depression dur-
ing pregnancy 24, and with fetal exposure to exogenous
gluco corticoids in early pregnancy 25. These behavioural
alterations include unsociable and inconsiderate behav-
iours, attention deficit hyperactivity disorder and sleep
disturbances as well as some psychiatric disorders,
including depressive symptoms, drug abuse and mood
and anxiety disorders. Very few studies have measured
Figure 1 | The stress system. When the brain detects a
threat, a coordinated physiological response involving
autonomic, neuroendocrine, metabolic and immune
system components is activated. A key system in the
stress response that has been extensively studied is the
hypothalamus-pituitary-adrenal (HPA) axis. Neurons in
the medial parvocellular region of the paraventricular
nucleus of the hypothalamus release corticotropin-
releasing hormone (CRH) and arginine vasopressin (AVP).
This triggers the subsequent secretion of adrenocortico-
tropic hormone (ACTH) from the pituitary gland, leading
to the production of glucocorticoids by the adrenal
cortex. In addition, the adrenal medulla releases
catecholamines (adrenaline and noradrenaline) (not
shown). The responsiveness of the HPA axis to stress is in
part determined by the ability of glucocorticoids to
regulate ACTH and CRH release by binding to two
corticosteroid receptors, the glucocorticoid receptor
(GR) and the mineralocorticoid receptor (MR). Following
activation of the system, and once the perceived stressor
has subsided, feedback loops are triggered at various
levels of the system (that is, from the adrenal gland to the
hypothalamus and other brain regions such as the
hippocampus and the frontal cortex) in order to shut the
HPA axis down and return to a set homeostatic point. By
contrast, the amygdala, which is involved in fear
processing142, activates the HPA axis in order to set in
motion the stress response that is necessary to deal with
the challenge. Not shown are the other major systems
and factors that respond to stress, including the
autonomic nervous system, the inflammatory cytokines
and the metabolic hormones. All of these are affected by
HPA activity and, in turn, affect HPA function, and they
are also implicated in the pathophysiological changes
that occur in response to chronic stress, from early
experiences into adult life.
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 435
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
changes in the brain as a function of prenatal stress in
humans. However, a recent study showed that low birth-
weight combined with lower levels of maternal care was
associated with reduced hippocampal volume in adult-
hood26. This finding is consistent with evidence that
effects of prenatal stress in humans are often moderated
by the quality of postnatal care, which in turn is consist-
ent with the protracted postnatal development of the
human brain.
Postnatal stress
Animal studies. Although in rodents the postnatal
period is relatively hyporesponsive to stress (BOX 2), one
of the most potent stressors for pups is separation from
the dam. long separation periods (3 h or more each day)
activate the pups’ HPA axis, as evidenced by increased
plasma levels of adrenocorticotropic hormone and
glucocorticoids27. Protracted maternal separation also
reduces pituitary CRH binding sites28, and low levels of
maternal care reduce GR levels in the hippocampus29.
The effects of maternal deprivation extend beyond
the HPA axis. Early prolonged maternal separation in
rats increases the density of CRH binding sites in the
prefrontal cortex, amygdala, hypothalamus, hippo-
campus and cerebellum, as measured post-infancy28. In
the hippocampus CRH mediates stress-related loss of
branches and spines30, and in the amygdala and hypotha-
lamus elevated CRH levels are associated with increased
anxiety and HPA axis activity, respectively31. Thus, the
increase in CRH-binding sites induced by maternal sep-
aration might have negative effects over time. The long-
term effects of prolonged separation depend on the age
of the pup and the duration of the deprivation, with the
effects noted above generally being greater when these
separations occur earlier in infancy and last for longer
durations32.
Although the rodent work provides a rich frame-
work for conceptualizi ng the impact of early-life stress,
the fact that the rodent brain is much less developed at
birth than the primate brain makes translation of the
findings to humans somewhat challenging (BOX 3). non-
human primates have more human-like brain matura-
tion at birth and patterns of parent–offspring relations,
and so provide an important bridge in the translation of
the rodent findings. Studies in monkeys have shown that
repeated, unpredictable separations from the mother33,
unpredictable maternal feedings34 or spontaneous mater-
nal abusive behaviour35 increases CRH concentrations
in the cerebrospinal fluid and alters the diurnal activity
of the HPA axis for months or even years after the period of
adversity: cortisol levels are lower than normal early in
the morning (around wake-up) and slightly higher than
normal later in the day, an effect that seems to reverse
over time in the absence of continued, ongoing psy-
chosocial stress35. These diurnal effects have not been
noted in rodents, but the effects on higher brain regions
seem to be comparable to the rodent findings and
include heightened fear behaviour36, exaggerated startle
responses33, hippocampal changes such as an increase in
the intensity of non-phosphorylated neurofilament pro-
tein immunoreactivity in the dentate gyrus granule cell
layer37, and atypical development of prefrontal regions
involved in emotion and behaviour control38.
Human studies. A human equivalent of the rodent
maternal separation paradigms might be studies of
children who attend full-day, out-of-home day care
centres. Studies have reported that glucocorticoid levels
rise in these children over the day, more so in toddlers
than in older preschool-aged children39,40. However, it is
important to note that the elevated glucocorticoid levels
observed are less pronounced than those observed in
rodents and monkeys exposed to maternal separation.
Moreover, although age accounts for most of the varia-
tion in the rise in glucocorticoid levels by late afternoon,
the quality of care is also important, with less supportive
care producing larger increases, especially for children
who are more emotionally negative and behaviour-
ally disorganized39. So far, there is no evidence that the
elevated glucocorticoid levels associated with being in
day care affect development; however, children who are
exposed to poor care for long hours early in develop-
ment have an increased risk of behaviour problems later
in development41.
Parent–child interactions and the psychological state
of the mother also influence the child’s HPA axis activity.
Beginning early in the first year, when the HPA system
of the infant is quite labile, sensitive parenting is associ -
ated with either smaller increases in or less prolonged
activations of the HPA axis to everyday perturbations42.
Maternal depression often interferes with sensitive and
supportive care of the infant and young child; there is
increasing evidence that offspring of depressed mothers,
Box 1 | Models to study stress in animals and humans
The hypothalamus-pituitary-adrenal axis can be activated by a
wide variety of stressors.
Some of the most potent are psychological or processive
stressors (that is, stressors that
involve higher-order sensory cognitive processing), as opposed
to physiological or
systemic stressors. Many psychological stressors are
anticipatory in nature — that is,
they are based on expectation as the result of learning and
memory (for example,
conditioned stimuli in animals and the anticipation of threats,
real or implied, in humans)
or on species-specific predispositions (for example, avoidance
of open space in
rodents or the threat of social rejection and negative social
evaluations in humans).
Animal studies allow the development of experimental protocols
in which animals
are submitted to acute and/or chronic stress and the resulting
effects on brain and
behaviour are studied. Experimental stressful ‘early-life’
manipulations in animals can
be broadly split into prenatal and postnatal manipulations.
Prenatal manipulations
involve maternal stress, exposing the mother to synthetic
glucocorticoids or maternal
nutrient restriction. Postnatal manipulations include depriving
the animal of
maternal contact, modifying maternal behaviour and exposing
the animal to synthetic
glucocorticoids. In these protocols, the cause–effects
relationship between stress and
its impact on the brain can be demonstrated. By contrast, and
because of ethical issues,
the cause–effects impact of stress on the brain cannot be studied
in humans, and most
human studies are correlational by nature. However, there are
some ‘experiments of
nature’ that can be used to inform scientists about the effects of
chronic exposure to
early adversity on brain development and of adulthood and late-
life stress effects on
the brain. Intrauterine under-growth and low birth weight are
considered indices of
prenatal stress (including malnutrition) in humans. In terms of
postnatal stress, low
socio-economic status, maltreatment and war are considered
adverse events. In adults
and older adults, studies of chronic caregivers (spouses of
patients with brain
degenerative disorders, parents of chronically sick children and
health-care
professionals) provide a human model of the impact of chronic
stress on the brain,
behaviour and cognition.
R E V I E W S
436 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
especially those who were clinically depressed in the
child’s early years, are at risk of heightened activity of
the HPA axis43 or of developing depression during ado-
lescence (controlling for maternal depression during
adolescence)44. However, it should be noted that it can
be difficult to exclude potentially confounding genetic
factors in such studies. Furthermore, preschool-aged
children of depressed mothers exhibit electroencephalo-
graphic alterations in frontal lobe activity that corre-
late with diminished empathy and other behavioural
problems45.
In contrast to findings of elevated glucocorticoid lev-
els in conditions of low parental care, studies in human
children exposed to severe deprivation (for example,
in orphanages or other institutions), neglect or abuse
report lower basal levels of glucocorticoids, similar to
what has been observed in primates39. one proposed
mechanism for the development of hypocortisolism is
downregulation of the HPA axis at the level of the pitui-
tary in response to chronic CRH drive from the hypoth-
alamus46, whereas a second possible mechanism is target
tissue hypersensitivity to glucocorticoids47. Importantly,
this hypocortisolism in humans in response to severe
stress may not be permanent: sensitive and supportive
care of fostered children normalizes their basal gluco-
corticoid levels after only 10 weeks48. Another impor-
tant finding comes from a recent study which showed
that exposure to early adversity is associated with epi -
genetic regulation of the GR receptor, as measured in the
post-mortem brains of suicide victims49.
Stress in adolescence
Animal studies. In rodents the period of adolescence has
three stages: a prepubescent or early adolescent period
from day 21 to 34, a mid-adolescent period from day
34 to 46 and a late adolescent period from day 46 to 59
(ReF. 50). In humans, adolescence is often considered
to demarcate the period of sexual maturation (that is,
starting with menarche in girls).
Although adolescence is a time of significant brain
development, particularly in the frontal lobe51, there has
been relatively little research on stress during this period
in rodents. In adolescent rodents, HPA function is char-
acterized by a prolonged activation in response to stres-
sors compared with adulthood. Moreover, prepubertal
rats have a delayed rise of glucocorticoid levels and
prolonged glucocorticoid release in response to several
types of stressors compared with adult rats52, owing to
incomplete maturation of negative-feedback systems53.
In contrast to adult rats, which show a habituation of
the stress response with repeated exposure to the same
stressor 54, juvenile rats have a potentiated release of
adrenocorticotropic hormone and glucocorticoids after
repeated exposure to stress55, suggesting that the HPA
axis responses to acute and chronic stress depend on
the developmental stage of the animal. Compared with
exposure to stress in adulthood alone, exposure to stress
as both a juvenile and an adult increases basal anxiety
levels in the adult 56. Moreover, exposure to juvenile
stress results in greater HPA axis activation than a dou-
ble exposure to stress during adulthood56, and this effect
is long-lasting. These results suggest that repeated stress
in adolescence leads to greater exposure of the brain to
glucocorticoids than similar experiences in adulthood.
The fact that the adolescent brain undergoes vigor-
ous maturation and the fact that, in rats, the hippocam-
pus continues to grow until adulthood suggest that the
adolescent brain may be more susceptible to stressors
and the concomitant exposure to high levels of gluco-
corticoids than the adult brain. Consistent with this
hypothesis are findings that increased glucocorticoid
levels before but not after puberty alter the expression of
genes for nMDA (N-methyl-d-aspartate) receptor sub-
units in the hippocampus57. In addition, chronic, vari-
able stress during the peripubertal juvenile period results
in reduced hippocampal volume in adulthood, which
is accompanied by impairments in Morris water maze
navigation and delayed shutdown of the HPA response
to acute stress58. These differences became evident only
in adulthood58, suggesting that stress in adolescence
reduces hippocampal growth. Finally, the effects of juve-
nile stress are long-lasting: adult rats exposed to juvenile
stress exhibit reduced exploratory behaviour and poor
avoidance learning 59. Moreover, stress in adolescence
increases susceptibility to drugs of abuse during the
adolescent period60 and in adulthood61.
Human studies. Interestingly, studies in human adoles-
cents also suggest that the adolescent period is associ -
ated with heightened basal and stress-induced activity
of the HPA axis62. This could be related to the dramatic
changes in sex steroid levels during this period, as these
steroids influence HPA axis activity50. However, studies
of stress in adolescent rats cannot be translated directly
to humans because the brain areas that are undergoing
development during adolescence differ between rats and
humans: although the rodent hippocampus continues to
Box 2 | The stress hyporesponsive period: from animals to
humans
Despite there being clear evidence that corticotropin-releasing
hormone-containing
neurons are present in the fetal rat139, in rodents noxious
stimuli evoke only a
subnormal hypothalamus-pituitary-adrenal (HPA) axis response
during the first 2 weeks
of life140. During this so-called stress hyporesponsive period
(SHRP), baseline plasma
glucocorticoid levels are lower than normal and are only
minimally increased by
exposure to a noxious stressor140. The SHRP is due to a rapid
regression of the HPA axis
after birth140 and may have evolved in rodents to protect the
rapidly developing brain
from the impact of elevated glucocorticoids.
Evidence is accumulating that in children there may be a
comparable hyporesponsive
period that emerges in infancy and extends throughout most of
childhood141. At birth,
glucocorticoid levels increase sharply in response to various
stressors, such as a
physical examination or a heel lance. However, over the course
of the first year the HPA
axis becomes more insensitive to stressors. No study has
assessed the exact period over
which this human SHRP may occur, but in adolescents
glucocorticoid levels can
become elevated in response to a psychosocial stressor141,
which suggests that the
SHRP could extend throughout childhood.
In rodents the SHRP is maintained primarily by maternal care
(that is, the presence of
the dam seems to suppress HPA axis activity); indeed, maternal
separation is a potent
inducer of a stress response, even during the SHRP. Similarly,
in humans the apparent
hyporesponsivity of the HPA axis might reflect the fact that
during the first year of life
the HPA axis comes under strong social regulation or parental
buffering141. Here again,
stressors that involve a lack of parental care or social contact
can induce a stress
response in children.
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 437
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
develop well into adulthood, in humans it is fully devel-
oped by 2 years of age63. The frontal cortex and amygdala
continue to develop in both species, but humans have
larger ontogenic bouts of development in frontal regions
than do rodents (BOX 3).
There are indications that the adolescent human brain
might be especially sensitive to the effects of elevated
levels of glucocorticoids and, by extension, to stress.
Recent studies on the ontogeny of MR and GR expres-
sion show that GR mRnA levels in the prefrontal cortex
are high in adolescence and late adulthood compared
with in infancy, young adulthood and senescence64. This
suggests that the cognitive and emotional processes that
are regulated by these brain areas might be sensitive to
GR-mediated regulation by glucocorticoids in an age-
dependent manner. Various forms of psychopathology,
including depression and anxiety, increase in prevalence
in adolescence65,66. Periods of heightened stress often
precede the first episodes of these disorders, raising the
possibility that heightened HPA reactivity during adoles-
cence increases sensitivity to the onset of stress-related
mental disorders.
Adolescence is also a period in which the long-
lasting effects of earlier exposures to stress become evi -
dent. Adolescents who grew up in poor economic condi-
tions have higher baseline glucocorticoid levels67, as do
adolescents whose mothers were depressed in the early
postnatal period44. High early-morning glucocorticoid
levels that vary markedly from day to day during the
transition to adolescence are not associated with depres-
sive symptoms at that time, but they predict increased
risk for depression by age 16 (ReF. 44).
Although early-life stress impairs hippocampal devel-
opment in rodents, there is currently little evidence
of comparable effects in humans. Children exposed
to physical or sexual abuse early in life do not exhibit
reduced hippocampal volume (relative to whole-brain
size) as adolescents, although adults with these histo-
ries do show volume reductions68. This finding holds
even when the abused children have been selected for
chronic post-traumatic stress disorder (PTSD), and even
though in some cases they exhibit overall reductions in
brain volume69. By contrast, alterations in grey matter
volume and the neuronal integrity of the frontal cortex,
and reduced size of the anterior cingulate cortex, have
been reported in adolescents exposed to early (and con-
tinued) adversity70. Together, these results suggest that in
humans the frontal cortex, which continues to develop
during adolescence, might be particularly vulnerable to
the effects of stress during adolescence. By contrast, the
hippocampus, which develops mainly in the first years
of life, might be less affected by exposure to adversity in
adolescence.
Stress in adulthood
Animal studies. Studies on adult stress in rodents have
delineated the effects of acute versus chronic stress
on brain and behaviour. The impact of acute stressors
depends on the level of glucocorticoid elevations, with
small increases resulting in enhanced hippocampus-
mediated learning and memory, and larger, prolonged
elevations impairing hippocampal function71. The
inverted-u-shaped effects of acute glucocorticoid ele-
vations might serve adaptive purposes by increasing
vigilance and learning processes during acute challenges.
The mechanism that underlies the acute bipha-
sic actions of glucocorticoids on cognition involves
the adrenergic system in the basolateral nucleus of the
amygdala. By enhancing noradrenergic function in
the amygdala, glucocorticoids have a permissive effect
on the priming of long-term potentiation in the den-
tate gyrus by the basolateral nucleus72. This modulation
of noradrenergic function by glucocorticoids has been
linked to the enhanced memory for emotional events
that occur under stress73.
Chronic stress or chronic exogenous administration
of glucocorticoids in rodents causes dendritic atrophy
in hippocampal CA3 pyramidal neurons74. However,
these changes take several weeks to develop and are
reversed by 10 days after the cessation of the stressor 75.
Chronic stress in adult rats also inhibits neurogenesis
in the dentate gyrus76 and causes hippocampal volume
loss77. Importantly, this volume decrease is not associ -
ated with reduced neuron numbers and is not limited
to the dentate gyrus78, suggesting that reduced neuro-
genesis might not be the only contributing factor. The
morphological changes that take place in the hippocam-
pus after chronic stress have been related to changes in
spatial learning79, which are reversed following 21 days
of withdrawal from stress80. Here, it is interesting to note
that in contrast to the effects of chronic or severe stress
on the brain and behaviour earlier in life, which are long-
lasting, effects of adulthood stress — even chronic stress
— are reversed after a few weeks of non-stress. These
differences between the effects of early and adulthood
Box 3 | Stress effects on the brain: timing is crucial
In animals that give birth to relatively mature young (for
example, primates, sheep and
guinea pigs), maximal brain growth and most of the
neuroendocrine maturation occurs
in utero. However, in rats, rabbits and mice the mother gives
birth to immature young
and most of the neuroendocrine development occurs in the
postnatal period17. In
humans the hypothalamus-pituitary-adrenal axis is highly
responsive at birth, but brain
development is not finished. The volume of the hippocampal
formation increases
sharply until the age of 2 years, whereas amygdala volume
continues to increase slowly
until the late 20s63. By contrast, the development of the frontal
cortex in humans takes
place mostly between 8 and 14 years of age63. The late increase
in prefrontal volumes is
consistent with data showing that this region develops latest in
terms of myelination
and synaptic density in humans136.
Prenatal and postnatal stress can both thus have contrasting
effects in different
species because perinatal manipulations will affect different
stages of development as
a function of the species studied. Consequently, stress in the
first week of the rodent’s
life is often developmentally equated with stress during the last
trimester of human
gestation.
Significant decreases in brain volume have been reported in
aged animals and
humans, although most of the studies performed are cross-
sectional. In men the
volume of the hippocampus starts to decrease by the second
decade of life, whereas in
women this decrease is delayed until around 40 years of age,
possibly owing to the
protective effects of oestrogen137. By contrast, amygdala
volume decreases around the
sixth decade of life in humans63. In the frontal cortex, different
subregions are
differentially affected by aging. For example, aging is
associated with shrinking of the
dorsolateral and inferior frontal cortices, but no age effects
have been reported for
the anterior cingulate cortex, the frontal pole or the precentral
gyrus138.
R E V I E W S
438 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
stress might be related to differences in the severity of
stressors to which pups and adult rats are exposed or
in the development of the hippocampus at the time of
exposure.
Pyramidal neurons in layers II/III of the prefron-
tal cortex also show dendritic retraction and a reduc-
tion in spine number 81 in response to chronic stress in
adulthood — this can be observed 24 h after a single
forced-swim stress82 — but remodelling occurs after ces-
sation of the stressor 83. In accordance with these find-
ings, glucocorticoid hypersecretion is associated with
reduced volume of at least the right anterior cingulate
cortex in rodents84. Contrary to the reduction in hip-
pocampal and frontal volumes, chronic stress in adult
rodents leads to dendritic hypertrophy in the baso lateral
amygdala85. Moreover, a recent study showed that
even a single acute administration of glucocorticoids
caused dend ritic hypertrophy in this area 12 days later 86.
The dendritic hypertrophy was correlated with anxi-
ety in both the acute86 and the chronic85 administration
paradigms.
Human studies. In humans, studies of the effects of acute
stress confirm animal studies and report the presence
of an inverted-u-shaped relationship between gluco-
corticoid levels and cognitive performance87. However,
contrary to animal studies, in which most laboratory
tests for learning and memory involve a fear and/or an
emotional process88, tests of learning and memory in
humans can differentiate the effects of glucocorticoids
on the processing of neutral versus emotional informa-
tion. Most studies to date have shown that acute gluco-
corticoid elevations significantly increase memory for
emotional information, whereas they impair the retrieval
of neutral information89.
only a few reports suggest that there is an association
between exposure to chronic stress and reduced hippo-
campal volume in individuals not suffering from men-
tal health disorders (for a review see ReF. 90). However,
a recent study reported that low self-esteem, a potent
predictor of increased reactivity to stress in humans91, is
associated with reduced hippocampal volume92.
Most of the studies of chronic-stress effects on the
adult human brain have concentrated either on stress-
related psychopathologies or on the impact of early-life
stress on adult psychopathology. A large number of stud-
ies have reported elevated basal glucocorticoid levels in
individuals with some forms of depression93, whereas
reduced basal glucocorticoid concentrations have been
reported in patients with PTSD94, although this finding
has been controversial95. Given that low glucocorticoid
concentrations seem to develop in early childhood in
response to neglect or trauma, it is possible that low
cortisol predicts vulnerability to developing PTSD in
response to trauma in adulthood.
Studies of adults who suffered childhood abuse
also reveal hyper-reactivity of the HPA axis in abused,
depressed individuals96 and hypoactivity in those with
PTSD94. The changes in abused, depressed adults have
been associated with CRH-induced ‘escape’ of gluco-
corticoid secretion from suppression by treatment with
dexamethasone97, suggesting that the glucocorticoid
feedback of the HPA axis is impaired under conditions of
increased hypothalamic drive. Thus, a decreased capac-
ity of glucocorticoids to inhibit the HPA axis when it is
stimulated could further accentuate CnS responses to
stressors. In agreement with this suggestion, increased
cerebrospinal fluid CRH levels have been reported
in individuals who reported childhood stress98 and
childhood abuse99.
Decreased hippocampal volume and function are
landmark features of depression and PTSD100,101. one
cross-sectional study 102 found that a smaller hippo-
campus in women with major depression was associ-
ated with experiences of childhood trauma, whereas
depressed women without such trauma had hippocam-
pal volumes similar to healthy controls. This supports
the notion that certain brain changes in patients with
depression or PTSD could represent markers of vulner-
ability for the disorder rather than markers of the dis-
order itself. This finding is in line with results from a
twin study of Vietnam veterans103 which showed that
decreased hippocampal volume is not a consequence of
combat exposure or PTSD: decreased volume was also
present in unexposed co-twins, and thus it might be a
pre-existing risk factor for PTSD that could be genetic
or rooted early in life.
Stress in aging
Animal studies. Approximately 30% of aged rats have
basal glucocorticoid hypersecretion, which is correlated
with memory impairments and reduced hippocampal
volume104. If a middle-aged rat is exposed for a long
period to high levels of exogenous glucocorticoids, it
will develop memory impairments and hippocampal
atrophy105 similar to those observed in these 30% of aged
rats. Conversely, artificially keeping glucocorticoid lev-
els low in middle-aged rats prevents the emergence of
both memory deficits and hippocampal atrophy in old
age106. Several groups have also found that chronic stress
in aged rats can accelerate the appearance of biomarkers
of hippocampal aging (for example, frequency potentia-
tion and synaptic excitability thresholds) and that excess
endogenous or exogenous glucocorticoids induce hip-
pocampal dendritic atrophy and inhibit neurogenesis107.
Finally, in aged monkeys108 chronic glucocorticoid treat-
ment can increase amyloid-β pathology, similar to that
reported in Alzheimer’s disease.
These results have given rise to the glucocorticoid
cascade hypothesis109, which suggests that there is a rela-
tionship between cumulative exposure to high glucocor-
ticoid levels and hippocampal atrophy. It was recently
renamed the neurotoxicity hypothesis103, because the
proposed explanation for this relationship is that pro-
longed exposure to stress hormones reduces the ability of
neurons to resist insults, thus increasing the rate at which
they are damaged by other toxic challenges or ordinary
attrition109. Glucocorticoids might have a similar neuro-
toxic effect in the prefrontal cortex. A study demon-
strated an enhanced elevation of extracellular glutamate
levels post-stress in the hippocampus and medial pre-
frontal cortex of aged rats compared with young rats110.
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 439
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
Stress in adolescence Stress in adulthood Stress in
agingPostnatal stressPrenatal stress
Birth 2 8 18 30 60 90
Amygdala
Frontal cortex
Hippocampus
Programming effects Differentiation effects
Outcome
Maintenance/
manifestation effects
Maintenance/
manifestation effects
↑ Glucocorticoids ↑↑ Glucocorticoids
↓↓ Glucocorticoids
Amygdala
Frontal cortex
Hippocampus
Nature Reviews | Neuroscience
↑ Glucocorticoids
(maternal separation)
↓ Glucocorticoids
(severe trauma)
↑ Glucocorticoids
(depression)
↓ Glucocorticoids
(PTSD)
Potentiation/
incubation effects
↓ Glucocorticoids
(PTSD)
↑ Glucocorticoids
(cognitive decline)
Effect on
HPA axis
Increased glutamate levels after stress, and perhaps other
neurotoxic insults, might thus increase the vulnerability
of the aging brain to neuronal damage.
Human studies. Aging, healthy humans exhibit higher
mean diurnal levels of cortisol than younger individu-
als111, and a longitudinal study has found that elevated
plasma glucocorticoid levels over years in older adults
negatively correlates with hippocampal volume and
memory112. Given that aged individuals with Alzheimer’s
disease present both memory impairments and hippo-
campal atrophy, studies have assessed basal glucocor-
ticoid levels in this population and found that they are
higher than in controls113. In addition, chronic glucocor-
ticoid treatment has been shown to worsen cognition in
people with Alzheimer’s disease114.
The frontal lobe also seems to be sensitive to glucocor -
ticoid effects during human aging. using a novel in vitro
post-mortem tracing method on human brain slices, Dai
et al.115 found an inverted-u-shaped effect of glucocor-
ticoids on axonal transport in prefrontal neurons with,
in most cases, a stimulating effect at low concentrations
and a depressing effect at high concentrations. Given
that axonal transport plays a crucial part in neuronal
survival and function, these results suggest that gluco-
corticoids potentially have negative effects on prefrontal
cortex neurons’ survival and/or function.
A model of stress effects throughout life
The data obtained in animals and humans suggest that
chronic or repeated exposure to stress has enduring
effects on the brain, through activation of the HPA axis
and the release of glucocorticoids, with the highest impact
on those structures that are developing at the time of the
stress exposure (in young individuals) and those that are
undergoing age-related changes (in adult and aged indi-
viduals). Stress in the prenatal period affects the devel -
opment of many of the brain regions that have a role in
regulating the HPA axis — that is, the hippocampus, the
frontal cortex and the amygdala (programming effects
(FIG. 2)). During childhood the hippocampus — which
continues to develop after birth — might be the brain
region that is most vulnerable to the effects of chronic
stress, possibly through a process of increased CRH
drive in the hippocampus116. Because it modulates HPA
axis activity, altered functioning of the hippocampus
Figure 2 | The life cycle model of stress. How the effects of
chronic or repeated exposure to stress (or a single exposure
to severe stress) at different stages in life depend on the brain
areas that are developing or declining at the time of the
exposure. Stress in the prenatal period affects the development
of many of the brain regions that are involved in regulating
the hypothalamus-pituitary-adrenal (HPA) axis — that is, the
hippocampus, the frontal cortex and the amygdala
(programming effects). Postnatal stress has varying effects:
exposure to maternal separation during childhood leads to
increased secretion of glucocorticoids, whereas exposure to
severe abuse is associated with decreased levels of
glucocorticoids. Thus, glucocorticoid production during
childhood differentiates as a function of the environment
(differentiation effects). From the prenatal period onwards, all
developing brain areas are sensitive to the effects of stress
hormones (broken blue bars); however, some areas undergo
rapid growth during a particular period (solid blue bars). From
birth to 2 years of age the hippocampus is developing; it might
therefore be the brain area that is most vulnerable to the
effects of stress at this time. By contrast, exposure to stress
from birth to late childhood might lead to changes in amygdala
volume, as this brain region continues to develop until the late
20s. During adolescence the hippocampus is fully organized,
the amygdala is still developing and there is an important
increase in frontal volume. Consequently, stress exposure
during
this period should have major effects on the frontal cortex.
Studies show that adolescents are highly vulnerable to stress,
possibly because of a protracted glucocorticoid response to
stress that persists into adulthood (potentiation/incubation
effects). In adulthood and during aging the brain regions that
undergo the most rapid decline as a result of aging (red bars)
are highly vulnerable to the effects of stress hormones. Stress
during these periods can lead to the manifestation of
incubated effects of early adversity on the brain (manifestation
effects) or to maintenance of chronic effects of stress
(maintenance effects). PTSD, post-traumatic stress disorder.
R E V I E W S
440 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
might cause glucocorticoid hyposecretion in cases
of severe abuse, or increased basal cortisol levels in
cases of maternal deprivation (differentiation effects
(FIG. 2)). By contrast, in adolescence the frontal cortex,
which undergoes major development at this stage, may be
most vulnerable to the effects of stress, possibly leading to
a protracted glucocorticoid response to stress that persists
into adulthood (potentiation/incubation effects (FIG. 2)).
In adulthood and old age the brain regions that undergo
the most rapid decline as a result of aging are highly vul -
nerable to the effects of stress hormones. For example,
in the hippocampus glucocorticoids affect neurogen-
esis, neuronal survival rate and dendritic arborization
(manifestation/maintenance effects (FIG. 2)).
The neurotoxicity and vulnerability hypotheses. The
data obtained in adults and older animals and humans
have led to the neurotoxicity hypothesis109, which sug-
gests that prolonged exposure to glucocorticoids reduces
the ability of neurons to resist insults, increasing the rate
at which they are damaged by other toxic challenges
or ordinary attrition109. This hypothesis implies that a
reduced hippocampal size is the end product of years
or decades of PTSD, depressive symptoms or chronic
stress. Although the neurotoxicity hypothesis has been
confirmed by various animal and human studies, it
does not explain the hyposecretion of glucocorticoids
that occurs in patients suffering from PTSD, who also
present reduced hippocampal volume.
Data obtained in children, adolescents or adult ani-
mals and humans exposed to acute or early-life trauma
have led to the vulnerability hypothesis103. In contrast to
the neurotoxicity hypothesis, the vulnerability hypoth-
esis suggests that reduced hippocampal volume in adult-
hood is not a consequence of chronic exposure to PTSD,
depression or chronic stress, but is a pre-existing risk fac-
tor for stress-related disorders that is induced by genetics
and/or early exposure to stress117. unlike the neurotoxicity
hypothesis, the vulnerability hypothesis can explain gluco-
corticoid hyposecretion in patients with PTSD. Indeed,
studies in children facing significant adversity, such as
abuse, report the development of glucocorticoid hypo-
secretion39, which might last until adulthood and confer
vulnerability to developing PTSD as a result of trauma.
we think that the two hypotheses are not mutually
exclusive when viewed from a developmental perspec-
tive. Indeed, the data summarized in this Review suggest
that there might be early windows of vulnerability (or
sensitive periods68) during which specific regions of the
developing brain are most susceptible to environmental
influences, through a neurotoxicity process. Exposure
to stress and/or adversity during these key vulnerable
periods might slow the development of those brain
regions for the duration of the adversity. when meas-
ured in adulthood, the reduced volumes of these brain
regions could be a strong marker of the time of exposure
to early adversity rather than of the effects of specific
traumas on various brain regions. These windows of vul-
nerability could also be used to predict the nature of the
psychopathology that will result from exposure to stress
at different ages. Exposure to adversity at the time of
hippocampal development could lead to hippocampus-
dependent emotional disorders, which would be differ-
ent from disorders arising from exposure to adversity at
times of frontal cortex development. Two recent studies
support this hypothesis. The first reported that women
who experienced trauma before the age of 12 years had
increased risk for major depression, whereas women who
experienced trauma between 12 and 18 years of age more
frequently developed PTSD118. The second study reported
that repeated episodes of sexual abuse were associated
with reduced hippocampal volume if the abuse occurred
early in childhood, but with reduced prefrontal cortex
volume if the abuse occurred during adolescence119. These
results suggest that, similar to what has been observed in
animals120, there may be distinct structural, neuropsy-
chological and neuropsychiatric sequelae of early abuse,
depending in part on the age or developmental stage of
the brain when the insult occurred.
Besides slowing down the development of the brain
during the time of adversity, leading to reduced
brain volumes in adulthood, stress in early life could
modify the developmental trajectory of the brain. The
potential immediate benefit of such modifications is that
they might increase acute survival probability, but they
could have negative long-term effects. During child-
hood and adolescence the brain undergoes a period
of overproduction and pruning of synapses121. one of
the brain regions that shows the slowest development
over the lifespan is the amygdala (BOX 3). It is interest-
ing to note that contrary to the hippocampus and the
frontal lobe — which show volume reduction as a result
of chronic stress — the amygdala increases in volume under
chronic stress, owing to increased dendritic arborization.
Given that the amygdala plays a significant part in the
detection of fear and threat, it is possible that throughout
evolution increases in amygdala volume in response to
stress might have improved the detection of threatening
information and so increased survival probability. If this
is indeed the case, young children exposed to adversity
should also have increased amygdala volume, but no
study has yet examined this important question.
This acute effect of adversity on brain organization
could have negative long-term consequences. Stress at
key periods of synaptic organization could modify the
trajectories of connections, leading to an incubation
period, such that the effects of stress would not be appar -
ent at the time of adversity but would emerge later, when
the synaptic organization has been completed. Studies
showing protracted effects of early-life stress that emerge
at puberty support this suggestion44. Furthermore,
although depression is the most extensively documented
outcome of exposure to chronic sexual abuse in adults,
it is not a common occurrence in children suffering
abuse. Indeed, the average time from the onset of abuse
to the emergence of clinical depression is 11.5 years, with
the first major episode occurring during adolescence122.
It is thus conceivable that in susceptible individuals expo-
sure to early adversity during a window of vulnerability
sets into motion a series of events that lead to a hetero-
typic reorganization of synaptic development, resulting
in a protracted expression of depression or PTSD.
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 441
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
This same process could also explain the develop-
ment of resilience in face of adversity. Environmental
enrichment in rodents is a potent inducer of changes
in neurogenesis and/or dendritic arborization in the
hippocampus, and has been documented to lead to
increases in hippocampal volume123. In children facing
early adversity, forms of environmental enrichment,
such as support from a family member, enriched day
care or school environment or social support from
members of the community, could induce a similar het-
erotypic reorganization of synaptic development, pro-
gramming of neurotrophic factors or changes in gene
expression that could lead to resilience later in life. If
this is the case, it could be suggested that any type of
intervention performed during the early years could not
only have a tremendous effect in preventing the deleteri -
ous impact of chronic stress and/or early abuse on the
developing brain, but could also help to prevent effects
on the brain of chronic stress occurring in adulthood or
during aging.
Conclusions and future directions
Although studies on stress have provided a wealth of
data delineating the effects of acute and chronic stress
on the developing brain, much remains to be done to
fully understand how the brain develops pathology or
resilience in the face of adversity. we believe that three
main factors should receive special consideration in
future studies on stress in both animals and humans.
The first factor is sex and gender. Sex refers to the bio-
logical differences between males and females, whereas
gender refers to the different roles (gender role and gen-
der identity) that men and women may have during their
lifetime. Both sex and gender might have potent influ-
ences on stress reactivity in humans of all ages. However,
most studies of the effects of stress on the brain, behav-
iour and cognition have tested only male animals or
humans. This is a major issue considering that studies
in both animals50 and humans124 report sex differences in
response to stress, and considering the gender gap ratio
(two girls for one boy) that emerges in early adoles-
cence for the risk of depression125. To this day, a consist-
ent finding in the endocrine literature is that the risk of
depression in adolescent girls increases with decreasing
age at menarche126. An increased sensitivity of girls to
environmental and/or family adversity, along with inter-
actions between glucocorticoids and gonadal steroids,
could be a potential explanation for the increased risk
of depressive disorders in females. Recent results show -
ing an earlier age at menarche in girls exposed to early
adversity127 support this suggestion.
The second factor that should be considered in future
studies is exposure to environmental toxins. Today, chil -
dren in many cities are chronically exposed, at back-
ground levels, to a range of common toxins that are
environmentally persistent and that tend to be lipophilic
and bioaccumulate, such as lead and bisphenol A128.
These agents reach humans mainly through food and
food additives, and they can be transferred to the fetus
through the placenta and to infants through maternal
milk129. They have been shown to affect the endocrine
system in laboratory animals and in wildlife, and conse-
quently have been called ‘endocrine-disrupting chemi-
cals’ (ReF. 130). A recent study showed that prenatal and
postnatal exposure to lead is associated with increased
glucocorticoid responses to acute stress in children131.
Also, perinatal exposure to endocrine-disrupting
chemicals is associated with an earlier age at menarche
among girls132. Taken together, these results suggest
that both the timing of sexual maturation and stress
reactivity may be sensitive to relatively low levels of
endocrine-disrupting chemicals in the environment.
The third factor that should receive greater attention
is circadian rhythmicity. Sleep deprivation, shift work
and jet lag all disrupt normal biological rhythms and
have major impacts on health. Interestingly, circadian
disorganization is often observed in stress-related dis-
orders such as depression133 and PTSD134. The discovery
of the molecular clock that is responsible for the genera-
tion of circadian rhythms135 provides new insights into
how rhythm abnormalities might lead to greater vulner-
ability to stress at various ages. Most studies performed
in animals and humans do not measure the circadian
fluctuations in glucocorticoid levels, but rather concen-
trate on specific time points across the day. Although
such measurements are easier, they do not provide
the full spectrum of circadian variations, which could
inform us about specific changes in circadian organi-
zation in response to chronic stress across the lifespan.
Consequently, studies assessing multiple time points
for glucocorticoid secretion across a whole day or sev-
eral days are needed in order to document the complex
relationships that exist between reactivity to stress and
circadian (dis)organization.
Animal and human studies have provided a wealth
of results showing the negative effects of chronic expo-
sure to stress and/or adversity on the developing brain.
However, stress is not and should not be considered
as a negative concept only. Stress is a physiological
response that is necessary for the survival of the spe-
cies. The stress response that today can have negative
consequences for brain development and mental health
may have conferred the necessary tools to our ances-
tors in prehistorical times for surviving in the presence
of predators. Studies of modern individuals who have
developed resilience by facing significant adversity
should inform us about the physiological and psy-
chological mechanisms at the basis of vulnerability or
resilience to stress. understanding these mechanisms,
which are possibly rooted in genes and modulated by
the family environment, is extremely important if one
wants to provide interventions early enough to individ-
uals who are the most likely to respond to them. This
article has reviewed the potential for early intervention
to prevent the deleterious effects of stress on the brain,
behaviour and cognition. After more than 30 years of
research on the negative effects of stress on the brain, it
is now time to turn our attention to the potential posi-
tive impact of early interventions on brain development.
These results could help us to develop social policies that
treat the problem of early-life stress at its root — that is,
in the family home.
R E V I E W S
442 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
1. Barker, D. J. The foetal and infant origins of
inequalities in health in Britain. J. Public Health Med.
13, 64–68 (1991).
2. Cadet, R., Pradier, P., Dalle, M. & Delost, P. Effects of
prenatal maternal stress on the pituitary
adrenocortical reactivity in guinea-pig pups. J. Dev.
Physiol. 8, 467–475 (1986).
3. Dean, F. & Matthews, S. G. Maternal dexamethasone
treatment in late gestation alters glucocorticoid and
mineralocorticoid receptor mRNA in the fetal guinea
pig brain. Brain Res. 846, 253–259 (1999).
4. Seckl, J. R. Glucocorticoids, developmental
‘programming’ and the risk of affective dysfunction.
Prog. Brain Res. 167, 17–34 (2008).
A superb review that summarized prenatal work
and linked it to clinical implications.
5. Koehl, M. et al. Prenatal stress alters circadian activity
of hypothalamo-pituitary-adrenal axis and
hippocampal corticosteroid receptors in adult rats of
both gender. J. Neurobiol. 40, 302–315 (1999).
6. Barbazanges, A., Piazza, P. V., Le Moal, M. &
Maccari, S. Maternal glucocorticoid secretion
mediates long-term effects of prenatal stress.
J. Neurosci. 16, 3943–3949 (1996).
7. Meyer, J. S. Early adrenalectomy stimulates
subsequent growth and development of the rat brain.
Exp. Neurol. 82, 432–446 (1983).
8. Weaver, I. C. et al. Epigenetic programming by maternal
behavior. Nature Neurosci. 7, 847–854 (2004).
The first paper to show that early experience has
epigenetic effects, altering methylation patterns.
9. Uno, H. et al. Brain damage induced by prenatal
exposure to dexamethasone in fetal rhesus macaques.
I. Hippocampus. Brain Res. Dev. Brain Res. 53,
157–167 (1990).
10. Murmu, M. S. et al. Changes of spine density and
dendritic complexity in the prefrontal cortex in
offspring of mothers exposed to stress during
pregnancy. Eur. J. Neurosci. 24, 1477–1487 (2006).
11. Cratty, M. S., Ward, H. E., Johnson, E. A., Azzaro, A. J.
& Birkle, D. L. Prenatal stress increases corticotropin-
releasing factor (CRF) content and release in rat
amygdala minces. Brain Res. 675, 297–302 (1995).
12. Vallee, M. et al. Long-term effects of prenatal stress
and postnatal handling on age-related glucocorticoid
secretion and cognitive performance: a longitudinal
study in the rat. Eur. J. Neurosci. 11, 2906–2916
(1999).
13. Deminiere, J. M. et al. Increased locomotor response
to novelty and propensity to intravenous amphetamine
self-administration in adult offspring of stressed
mothers. Brain Res. 586, 135–139 (1992).
14. Vallee, M. et al. Prenatal stress induces high anxiety
and postnatal handling induces low anxiety in adult
offspring: correlation with stress-induced
corticosterone secretion. J. Neurosci. 17,
2626–2636 (1997).
15. Lemaire, V., Koehl, M., Le Moal, M. & Abrous, D. N.
Prenatal stress produces learning deficits associated
with an inhibition of neurogenesis in the hippocampus.
Proc. Natl Acad. Sci. USA 97, 11032–11037 (2000).
16. Piazza, P. V. & Le Moal, M. L. Pathophysiological basis
of vulnerability to drug abuse: role of an interaction
between stress, glucocorticoids, and dopaminergic
neurons. Annu. Rev. Pharmacol. Toxicol. 36,
359–378 (1996).
17. Kapoor, A., Petropoulos, S. & Matthews, S. G. Fetal
programming of hypothalamic-pituitary-adrenal (HPA)
axis function and behavior by synthetic
glucocorticoids. Brain Res. Rev. 57, 586–595 (2008).
18. Hedegaard, M., Henriksen, T. B., Sabroe, S. & Secher,
N. J. Psychological distress in pregnancy and preterm
delivery. BMJ 307, 234–239 (1993).
19. Orr, S. T. & Miller, C. A. Maternal depressive
symptoms and the risk of poor pregnancy outcome.
Review of the literature and preliminary findings.
Epidemiol. Rev. 17, 165–171 (1995).
20. Lyons-Ruth, K., Wolfe, R. & Lyubchik, A. Depression
and the parenting of young children: making the case
for early preventive mental health services. Harv. Rev.
Psychiatry 8, 148–153 (2000).
21. Gutteling, B. M., de Weerth, C. & Buitelaar, J. K.
Prenatal stress and children’s cortisol reaction to the
first day of school. Psychoneuroendocrinology 30,
541–549 (2005).
22. O’Connor, T. G. et al. Prenatal anxiety predicts
individual differences in cortisol in pre-adolescent
children. Biol. Psychiatry 58, 211–217 (2005).
23. Glover, V. Maternal stress or anxiety in pregnancy and
emotional development of the child. Br. J. Psychiatry
171, 105–106 (1997).
24. Stott, D. H. Follow-up study from birth of the effects of
prenatal stresses. Dev. Med. Child. Neurol. 15,
770–787 (1973).
25. Trautman, P. D., Meyer-Bahlburg, H. F., Postelnek, J.
& New, M. I. Effects of early prenatal dexamethasone
on the cognitive and behavioral development of young
children: results of a pilot study.
Psychoneuroendocrinology 20, 439–449 (1995).
26. Buss, C. et al. Maternal care modulates the
relationship between prenatal risk and hippocampal
volume in women but not in men. J. Neurosci. 27,
2592–2595 (2007).
27. Levine, S. & Wiener, S. G. Psychoendocrine aspects of
mother-infant relationships in nonhuman primates.
Psychoneuroendocrinology 13, 143–154 (1988).
28. Anisman, H., Zaharia, M. D., Meaney, M. J. & Merali, Z.
Do early-life events permanently alter behavioral and
hormonal responses to stressors? Int. J. Dev.
Neurosci. 16, 149–164 (1998).
29. Liu, D. et al. Maternal care, hippocampal
glucocorticoid receptors, and
hypothalamic-pituitary-adrenal responses to stress.
Science 277, 1659–1662 (1997).
30. Fenoglio, K. A., Brunson, K. L. & Baram, T. Z.
Hippocampal neuroplasticity induced by early-life
stress: functional and molecular aspects. Front.
Neuroendocrinol. 27, 180–192 (2006).
31. Schulkin, J., Gold, P. W. & McEwen, B. S. Induction of
corticotropin-releasing hormone gene expression by
glucocorticoids: implication for understanding the
states of fear and anxiety and allostatic load.
Psychoneuroendocrinology 23, 219–243 (1998).
32. de Kloet, E. R. & Oitzl, M. S. Who cares for a stressed
brain? The mother, the kid or both? Neurobiol. Aging
24 (Suppl. 1), S61–S65; discussion S67–S68 (2003).
33. Sanchez, M. M. et al. Alterations in diurnal cortisol
rhythm and acoustic startle response in nonhuman
primates with adverse rearing. Biol. Psychiatry 57,
373–381 (2005).
34. Coplan, J. D. et al. Persistent elevations of
cerebrospinal fluid concentrations of corticotropin-
releasing factor in adult nonhuman primates exposed
to early-life stressors: implications for the
pathophysiology of mood and anxiety disorders. Proc.
Natl Acad. Sci. USA 93, 1619–1623 (1996).
35. Sanchez, M. M. The impact of early adverse care on
HPA axis development: nonhuman primate models.
Horm. Behav. 50, 623–631 (2006).
36. Rosenblum, L. A. et al. Differing concentrations of
corticotropin-releasing factor and oxytocin in the
cerebrospinal fluid of bonnet and pigtail macaques.
Psychoneuroendocrinology 27, 651–660 (2002).
37. Siegel, S. J. et al. Effects of social deprivation in
prepubescent rhesus monkeys: immunohistochemical
analysis of the neurofilament protein triplet in the
hippocampal formation. Brain Res. 619, 299–305
(1993).
38. Sanchez, M. M., Ladd, C. O. & Plotsky, P. M. Early
adverse experience as a developmental risk factor for
later psychopathology: evidence from rodent and primate
models. Dev. Psychopathol. 13, 419–449 (2001).
39. Gunnar, M. R. & Donzella, B. Social regulation of the
cortisol levels in early human development.
Psychoneuroendocrinology 27, 199–220 (2002).
40. Geoffroy, M. C., Cote, S. M., Parent, S. & Seguin, J. R.
Daycare attendance, stress, and mental health. Can.
J. Psychiatry 51, 607–615 (2006).
41. NICHD Early Child Care Research Network. Early child
care and children’s development prior to school entry:
results from the NICHD Study of Early Child Care. Am.
Educ. Res. J. 39, 133–164 (2002).
42. Albers, E. M., Riksen-Walraven, J. M., Sweep, F. C. &
de Weerth, C. Maternal behavior predicts infant
cortisol recovery from a mild everyday stressor.
J. Child. Psychol. Psychiatry 49, 97–103 (2008).
43. Lupien, S. J., King, S., Meaney, M. J. & McEwen, B. S.
Child’s stress hormone levels correlate with mother’s
socioeconomic status and depressive state. Biol.
Psychiatry 48, 976–980 (2000).
44. Halligan, S. L., Herbert, J., Goodyer, I. & Murray, L.
Disturbances in morning cortisol secretion in
association with maternal postnatal depression
predict subsequent depressive symptomatology in
adolescents. Biol. Psychiatry 62, 40–46 (2007).
Provided some of the first evidence that adverse
early life experiences in humans, in this case
rearing by a mother suffering from post-partum
depression, are associated with heightened HPA
activity years later, and that the HPA axis
hyperactivity mediates the association between
early risk exposure and later psychiatric symptoms.
45. Jones, N. A., Field, T. & Davalos, M. Right frontal EEG
asymmetry and lack of empathy in preschool children
of depressed mothers. Child. Psychiatry Hum. Dev.
30, 189–204 (2000).
46. Fries, E., Hesse, J., Hellhammer, J. & Hellhammer,
D. H. A new view on hypocortisolism.
Psychoneuroendocrinology 30, 1010–1016 (2005).
47. Yehuda, R., Yang, R. K., Buchsbaum, M. S. & Golier,
J. A. Alterations in cortisol negative feedback
inhibition as examined using the ACTH response to
cortisol administration in PTSD.
Psychoneuroendocrinology 31, 447–451 (2006).
48. Gunnar, M. R. & Quevedo, K. M. Early care
experiences and HPA axis regulation in children: a
mechanism for later trauma vulnerability. Prog. Brain
Res. 167, 137–149 (2008).
49. McGowan, P. O. et al. Epigenetic regulation of the
glucocorticoid receptor in human brain associates with
childhood abuse. Nature Neurosci. 12, 342–348
(2009).
This study examined epigenetic differences in a
neuron-specific glucocorticoid receptor (NR3C1)
promoter between post-mortem hippocampus
obtained from suicide victims with a history of
childhood abuse and hippocampus from either
suicide victims with no childhood abuse or controls.
It found decreased levels of glucocorticoid receptor
mRNA, as well as mRNA transcripts bearing the
glucocorticoid receptor 1F splice variant and
increased cytosine methylation of an NR3C1
promoter in suicide victims with early abuse.
50. McCormick, C. M. & Mathews, I. Z. HPA function in
adolescence: role of sex hormones in its regulation
and the enduring consequences of exposure to
stressors. Pharmacol. Biochem. Behav. 86, 220–233
(2007).
A very good review on the acute and chronic effects
of stress during adolescence.
51. O’Donnell, S., Noseworthy, M. D., Levine, B. &
Dennis, M. Cortical thickness of the frontopolar area
in typically developing children and adolescents.
Neuroimage 24, 948–954 (2005).
52. Vazquez, D. M. & Akil, H. Pituitary-adrenal response
to ether vapor in the weanling animal: characterization
of the inhibitory effect of glucocorticoids on
adrenocorticotropin secretion. Pediatr. Res. 34,
646–653 (1993).
53. Goldman, L., Winget, C., Hollingshead, G. W. &
Levine, S. Postweaning development of negative
feedback in the pituitary-adrenal system of the rat.
Neuroendocrinology 12, 199–211 (1973).
54. Girotti, M. et al. Habituation to repeated restraint
stress is associated with lack of stress-induced c-fos
expression in primary sensory processing areas of the
rat brain. Neuroscience 138, 1067–1081 (2006).
55. Romeo, R. D. et al. Stress history and pubertal
development interact to shape
hypothalamic-pituitary-adrenal axis plasticity.
Endocrinology 147, 1664–1674 (2006).
56. Avital, A. & Richter-Levin, G. Exposure to juvenile
stress exacerbates the behavioural consequences of
exposure to stress in the adult rat. Int.
J. Neuropsychopharmacol. 8, 163–173 (2005).
57. Lee, P. R., Brady, D. & Koenig, J. I. Corticosterone
alters N-methyl-d-aspartate receptor subunit mRNA
expression before puberty. Brain Res. Mol. Brain Res.
115, 55–62 (2003).
58. Isgor, C., Kabbaj, M., Akil, H. & Watson, S. J. Delayed
effects of chronic variable stress during peripubertal-
juvenile period on hippocampal morphology and on
cognitive and stress axis functions in rats.
Hippocampus 14, 636–648 (2004).
One of the first papers to show protracted effects
of adolescent stress on adulthood stress reactivity
in rodents.
59. Tsoory, M. & Richter-Levin, G. Learning under stress in
the adult rat is differentially affected by ‘juvenile’ or
‘adolescent’ stress. Int. J. Neuropsychopharmacol. 9,
713–728 (2006).
60. Kabbaj, M., Isgor, C., Watson, S. J. & Akil, H. Stress
during adolescence alters behavioral sensitization to
amphetamine. Neuroscience 113, 395–400 (2002).
61. McCormick, C. M., Robarts, D., Gleason, E. & Kelsey,
J. E. Stress during adolescence enhances locomotor
sensitization to nicotine in adulthood in female, but
not male, rats. Horm. Behav. 46, 458–466 (2004).
62. Gunnar, M. R., Wewerka, S., Frenn, K., Long, J. D. &
Griggs, C. Developmental changes in hypothalamus-
pituitary-adrenal activity over the transition to
adolescence: normative changes and associations with
puberty. Dev. Psychopathol. 21, 69–85 (2009).
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 443
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
63. Giedd, J. N. et al. Quantitative magnetic resonance
imaging of human brain development: ages 4–18.
Cereb. Cortex 6, 551–560 (1996).
64. Perlman, W. R., Webster, M. J., Herman, M. M.,
Kleinman, J. E. & Weickert, C. S. Age-related
differences in glucocorticoid receptor mRNA levels in
the human brain. Neurobiol. Aging 28, 447–458
(2007).
65. Dahl, R. E. Adolescent brain development: a period of
vulnerabilities and opportunities. Keynote address.
Ann. NY Acad. Sci. 1021, 1–22 (2004).
66. Paus, T., Keshavan, M. & Giedd, J. N. Why do many
psychiatric disorders emerge during adolescence?
Nature Rev. Neurosci. 9, 947–957 (2008).
A very interesting review on the state of research
into why adolescents have a greater vulnerability to
mental health disorders.
67. Evans, G. W. & English, K. The environment of poverty:
multiple stressor exposure, psychophysiological stress,
and socioemotional adjustment. Child Dev. 73,
1238–1248 (2002).
68. Andersen, S. L. & Teicher, M. H. Stress, sensitive
periods and maturational events in adolescent
depression. Trends Neurosci. 31, 183–191 (2008).
69. De Bellis, M. D. et al. A. E. Bennett Research Award.
Developmental traumatology. Part II: brain
development. Biol. Psychiatry 45, 1271–1284
(1999).
One of the first clear demonstrations that, in
children who were physically healthy at birth,
severe abuse in the early years of life is associated
with reduced brain volume. The reduction
correlates negatively with the age of onset and
positively with the duration of the maltreatment.
70. Cohen, R. A. et al. Early life stress and morphometry
of the adult anterior cingulate cortex and caudate
nuclei. Biol. Psychiatry 59, 975–982 (2006).
71. Diamond, D. M., Bennett, M. C., Fleshner, M. & Rose,
G. M. Inverted-U relationship between the level of
peripheral corticosterone and the magnitude of
hippocampal primed burst potentiation. Hippocampus
2, 421–430 (1992).
72. Vouimba, R. M., Yaniv, D. & Richter-Levin, G.
Glucocorticoid receptors and β-adrenoceptors in
basolateral amygdala modulate synaptic plasticity in
hippocampal dentate gyrus, but not in area CA1.
Neuropharmacology 52, 244–252 (2007).
73. Roozendaal, B., Brunson, K. L., Holloway, B. L.,
McGaugh, J. L. & Baram, T. Z. Involvement of stress-
released corticotropin-releasing hormone in the
basolateral amygdala in regulating memory
consolidation. Proc. Natl Acad. Sci. USA 99,
13908–13913 (2002).
74. Magarinos, A. M. & McEwen, B. S. Stress-induced
atrophy of apical dendrites of hippocampal CA3c
neurons: involvement of glucocorticoid secretion and
excitatory amino acid receptors. Neuroscience 69,
89–98 (1995).
75. Conrad, C. D., LeDoux, J. E., Magarinos, A. M. &
McEwen, B. S. Repeated restraint stress facilitates fear
conditioning independently of causing hippocampal
CA3 dendritic atrophy. Behav. Neurosci. 113,
902–913 (1999).
76. Gould, E., McEwen, B. S., Tanapat, P., Galea, L. A. &
Fuchs, E. Neurogenesis in the dentate gyrus of the
adult tree shrew is regulated by psychosocial stress
and NMDA receptor activation. J. Neurosci. 17,
2492–2498 (1997).
77. McEwen, B. S. Effects of adverse experiences for brain
structure and function. Biol. Psychiatry 48, 721–731
(2000).
78. Pham, K., Nacher, J., Hof, P. R. & McEwen, B. S.
Repeated restraint stress suppresses neurogenesis
and induces biphasic PSA-NCAM expression in the
adult rat dentate gyrus. Eur. J. Neurosci. 17,
879–886 (2003).
79. McEwen, B. S. Plasticity of the hippocampus:
adaptation to chronic stress and allostatic load. Ann.
NY Acad. Sci. 933, 265–277 (2001).
80. Luine, V., Villegas, M., Martinez, C. & McEwen, B. S.
Repeated stress causes reversible impairments of
spatial memory performance. Brain Res. 639,
167–170 (1994).
81. Joels, M., Karst, H., Krugers, H. J. & Lucassen, P. J.
Chronic stress: implications for neuronal morphology,
function and neurogenesis. Front. Neuroendocrinol.
28, 72–96 (2007).
82. Izquierdo, A., Wellman, C. L. & Holmes, A. Brief
uncontrollable stress causes dendritic retraction in
infralimbic cortex and resistance to fear extinction in
mice. J. Neurosci. 26, 5733–5738 (2006).
83. Shansky, R. M., Hamo, C., Hof, P. R., McEwen, B. S. &
Morrison, J. H. Stress-induced dendritic remodeling in
the prefrontal cortex is circuit specific. Cereb. Cortex 4
Feb 2009 (doi:10.1093/cercor/bhp003).
84. Cerqueira, J. J. et al. Corticosteroid status influences
the volume of the rat cingulate cortex - a magnetic
resonance imaging study. J. Psychiatr. Res. 39,
451–460 (2005).
85. Mitra, R., Jadhav, S., McEwen, B. S., Vyas, A. &
Chattarji, S. Stress duration modulates the
spatiotemporal patterns of spine formation in the
basolateral amygdala. Proc. Natl Acad. Sci. USA 102,
9371–9376 (2005).
86. Mitra, R. & Sapolsky, R. M. Acute corticosterone
treatment is sufficient to induce anxiety and
amygdaloid dendritic hypertrophy. Proc. Natl Acad.
Sci. USA 105, 5573–5578 (2008).
This interesting study addressed endocrine effects
on the brain, with a focus on the amygdala and
anxiety (rather than on hippocampus and memory).
Of note, a single dose of glucocorticoids was
sufficient to induce changes in amygdala structure
10 days later, which might be useful to model in
animals PTSD.
87. Lupien, S. J. & McEwen, B. S. The acute effects of
corticosteroids on cognition: integration of animal and
human model studies. Brain Res. Brain Res. Rev. 24,
1–27 (1997).
88. Roozendaal, B. Glucocorticoids and the regulation of
memory consolidation. Psychoneuroendocrinology
25, 213–238 (2000).
89. Lupien, S. J. et al. Stress hormones and human
memory function across the lifespan.
Psychoneuroendocrinology 30, 225–242 (2005).
90. Lupien, S. J. et al. Hippocampal volume is as variable
in young as in older adults: implications for the notion
of hippocampal atrophy in humans. Neuroimage 34,
479–485 (2007).
This study showed that ~25% of young adults
present hippocampal volumes as small as those of
older adults. The presence of small hippocampal
volumes in healthy young individuals supports the
vulnerability hypothesis.
91. Pruessner, J. C., Lord, C., Meaney, M. & Lupien, S.
Effects of self-esteem on age-related changes in
cognition and the regulation of the
hypothalamic-pituitary-adrenal axis. Ann. NY Acad.
Sci. 1032, 186–194 (2004).
92. Pruessner, J. C. et al. Self-esteem, locus of control,
hippocampal volume, and cortisol regulation in young
and old adulthood. Neuroimage 28, 815–826 (2005).
93. Burke, H. M., Davis, M. C., Otte, C. & Mohr, D. C.
Depression and cortisol responses to psychological
stress: a meta-analysis. Psychoneuroendocrinology
30, 846–856 (2005).
94. Yehuda, R., Golier, J. A. & Kaufman, S. Circadian
rhythm of salivary cortisol in Holocaust survivors with
and without PTSD. Am. J. Psychiatry 162, 998–1000
(2005).
95. Meewisse, M. L., Reitsma, J. B., de Vries, G. J.,
Gersons, B. P. & Olff, M. Cortisol and post-traumatic
stress disorder in adults: systematic review and meta-
analysis. Br. J. Psychiatry 191, 387–392 (2007).
This paper presented the first meta-analysis of
cortisol findings in PTSD, to elucidate the
determinants of hypocortisolism and resolve the
inconsistency in findings.
96. Heim, C. et al. Pituitary-adrenal and autonomic
responses to stress in women after sexual and physical
abuse in childhood. JAMA 284, 592–597 (2000).
97. Heim, C., Mletzko, T., Purselle, D., Musselman, D. L. &
Nemeroff, C. B. The dexamethasone/corticotropin-
releasing factor test in men with major depression:
role of childhood trauma. Biol. Psychiatry 63,
398–405 (2008).
98. Carpenter, L. L. et al. Cerebrospinal fluid corticotropin-
releasing factor and perceived early-life stress in
depressed patients and healthy control subjects.
Neuropsychopharmacology 29, 777–784 (2004).
99. Heim, C., Newport, D. J., Mletzko, T., Miller, A. H. &
Nemeroff, C. B. The link between childhood trauma and
depression: insights from HPA axis studies in humans.
Psychoneuroendocrinology 33, 693–710 (2008).
A crucially important review which documents that
the disturbances in the HPA axis that are observed
in many adults with depression may be specific to
those who experienced trauma or maltreatment in
childhood.
100. Videbech, P. & Ravnkilde, B. Hippocampal volume and
depression: a meta-analysis of MRI studies. Am.
J. Psychiatry 161, 1957–1966 (2004).
101. Smith, M. E. Bilateral hippocampal volume reduction
in adults with post-traumatic stress disorder: a meta-
analysis of structural MRI studies. Hippocampus 15,
798–807 (2005).
102. Vythilingam, M. et al. Childhood trauma associated
with smaller hippocampal volume in women with
major depression. Am. J. Psychiatry 159,
2072–2080 (2002).
103. Gilbertson, M. W. et al. Smaller hippocampal volume
predicts pathologic vulnerability to psychological
trauma. Nature Neurosci. 5, 1242–1247 (2002).
The first paper to study whether the reduced
hippocampal volume observed in PTSD patients is
due to the disorder, to trauma exposure or to a
pre-existing factor.
104. Issa, A. M., Rowe, W., Gauthier, S. & Meaney, M. J.
Hypothalamic-pituitary-adrenal activity in aged,
cognitively impaired and cognitively unimpaired rats.
J. Neurosci. 10, 3247–3254 (1990).
105. Landfield, P. W., Waymire, J. C. & Lynch, G.
Hippocampal aging and adrenocorticoids:
quantitative correlations. Science 202, 1098–1102
(1978).
106. Landfield, P. W., Baskin, R. K. & Pitler, T. A. Brain
aging correlates: retardation by hormonal-
pharmacological treatments. Science 214, 581–584
(1981).
The first study to show that chronic exposure to
high levels of glucocorticoids in rodents is
associated with memory impairments and reduced
hippocampal volume.
107. Landfield, P. W., Blalock, E. M., Chen, K. C. & Porter,
N. M. A new glucocorticoid hypothesis of brain aging:
implications for Alzheimer’s disease. Curr. Alzheimer
Res. 4, 205–212 (2007).
108. Kulstad, J. J. et al. Effects of chronic glucocorticoid
administration on insulin-degrading enzyme and
amyloid-β peptide in the aged macaque.
J. Neuropathol. Exp. Neurol. 64, 139–146 (2005).
109. Sapolsky, R. M., Krey, L. C. & McEwen, B. S. The
neuroendocrinology of stress and aging: the
glucocorticoid cascade hypothesis. Endocr. Rev. 7,
284–301 (1986).
The first paper to present the glucocorticoid
cascade hypothesis, now referred to as the
neurotoxicity hypothesis.
110. Lowy, M. T., Wittenberg, L. & Yamamoto, B. K. Effect of
acute stress on hippocampal glutamate levels and
spectrin proteolysis in young and aged rats.
J. Neurochem. 65, 268–274 (1995).
111. Raskind, M. A., Peskind, E. R. & Wilkinson, C. W.
Hypothalamic-pituitary-adrenal axis regulation and
human aging. Ann. NY Acad. Sci. 746, 327–335
(1994).
112. Lupien, S. J. et al. Cortisol levels during human aging
predict hippocampal atrophy and memory deficits.
Nature Neurosci. 1, 69–73 (1998).
113. Giubilei, F. et al. Altered circadian cortisol secretion in
Alzheimer’s disease: clinical and neuroradiological
aspects. J. Neurosci. Res. 66, 262–265 (2001).
114. Aisen, P. S. et al. A randomized controlled trial of
prednisone in Alzheimer’s disease. Alzheimer’s
Disease Cooperative Study. Neurology 54, 588–593
(2000).
115. Dai, J., Buijs, R. & Swaab, D. Glucocorticoid hormone
(cortisol) affects axonal transport in human cortex
neurons but shows resistance in Alzheimer’s disease.
Br. J. Pharmacol. 143, 606–610 (2004).
116. Chen, Y., Dube, C. M., Rice, C. J. & Baram, T. Z. Rapid
loss of dendritic spines after stress involves
derangement of spine dynamics by corticotropin-
releasing hormone. J. Neurosci. 28, 2903–2911
(2008).
117. Charney, D. S. & Manji, H. K. Life stress, genes, and
depression: multiple pathways lead to increased risk
and new opportunities for intervention. Sci. STKE
2004, re5 (2004).
118. Maercker, A., Michael, T., Fehm, L., Becker, E. S. &
Margraf, J. Age of traumatisation as a predictor of
post-traumatic stress disorder or major depression in
young women. Br. J. Psychiatry 184, 482–487
(2004).
119. Teicher, M. H., Tomoda, A. & Andersen, S. L.
Neurobiological consequences of early stress and
childhood maltreatment: are results from human and
animal studies comparable? Ann. NY Acad. Sci. 1071,
313–323 (2006).
120. Hall, F. S. Social deprivation of neonatal, adolescent,
and adult rats has distinct neurochemical and
behavioral consequences. Crit. Rev. Neurobiol. 12,
129–162 (1998).
R E V I E W S
444 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w
w.nature.com/reviews/neuro
R E V I E W S
© 2009 Macmillan Publishers Limited. All rights reserved
121. Andersen, S. L. Trajectories of brain development:
point of vulnerability or window of opportunity?
Neurosci. Biobehav. Rev. 27, 3–18 (2003).
A superb review paper which suggested that
trauma at different time points during early
development might be associated with different
outcomes, depending on the brain structure that
was affected at the time of exposure to adversity.
122. Widom, C. S., DuMont, K. & Czaja, S. J. A prospective
investigation of major depressive disorder and
comorbidity in abused and neglected children grown
up. Arch. Gen. Psychiatry 64, 49–56 (2007).
123. Clayton, N. S. & Krebs, J. R. Hippocampal growth and
attrition in birds affected by experience. Proc. Natl
Acad. Sci. USA 91, 7410–7414 (1994).
124. Kudielka, B. M., Buske-Kirschbaum, A., Hellhammer,
D. H. & Kirschbaum, C. HPA axis responses to
laboratory psychosocial stress in healthy elderly adults,
younger adults, and children: impact of age and gender.
Psychoneuroendocrinology 29, 83–98 (2004).
125. Kessler, R. C. Epidemiology of women and depression.
J. Affect. Disord. 74, 5–13 (2003).
126. Harlow, B. L., Cohen, L. S., Otto, M. W., Spiegelman, D.
& Cramer, D. W. Early life menstrual characteristics
and pregnancy experiences among women with and
without major depression: the Harvard study of moods
and cycles. J. Affect. Disord. 79, 167–176 (2004).
127. Zabin, L. S., Emerson, M. R. & Rowland, D. L.
Childhood sexual abuse and early menarche: the
direction of their relationship and its implications.
J. Adolesc. Health 36, 393–400 (2005).
128. Jones, K. C. & de Voogt, P. Persistent organic
pollutants (POPs): state of the science. Environ. Pollut.
100, 209–221 (1999).
129. Centers for Disease Control and Prevention. Second
National Report on Human Exposure to
Environmental Chemicals. (CDC, Atlanta, Georgia,
2003).
130. Daston, G. P., Cook, J. C. & Kavlock, R. J.
Uncertainties for endocrine disrupters: our view on
progress. Toxicol. Sci. 74, 245–252 (2003).
131. Gump, B. B. et al. Low-level prenatal and postnatal
blood lead exposure and adrenocortical responses to
acute stress in children. Environ. Health Perspect.
116, 249–255 (2008).
132. Denham, M. et al. Relationship of lead, mercury,
mirex, dichlorodiphenyldichloroethylene,
hexachlorobenzene, and polychlorinated biphenyls to
timing of menarche among Akwesasne Mohawk girls.
Pediatrics 115, e127–e134 (2005).
133. Turek, F. W. From circadian rhythms to clock genes in
depression. Int. Clin. Psychopharmacol. 22 (Suppl. 2),
S1–S8 (2007).
134. Lamarche, L. J. & De Koninck, J. Sleep disturbance
in adults with posttraumatic stress disorder:
a review. J. Clin. Psychiatry 68, 1257–1270
(2007).
135. Antoch, M. P. et al. Functional identification of the
mouse circadian Clock gene by transgenic BAC rescue.
Cell 89, 655–667 (1997).
136. Yakovlev, P. L. & Lecours, A. R. in Regional Development
of the Brain in Early Life (ed. Minkowski, A.) 3–70
(Blackwell, Oxford, 1967).
137. Pruessner, J. C. et al. Volumetry of hippocampus and
amygdala with high-resolution MRI and three-
dimensional analysis software: minimizing the
discrepancies between laboratories. Cereb. Cortex 10,
433–442 (2000).
138. Tisserand, D. J. et al. Regional frontal cortical
volumes decrease differentially in aging: an MRI
study to compare volumetric approaches and voxel-
based morphometry. Neuroimage 17, 657–669
(2002).
139. Insel, T. R., Battaglia, G., Fairbanks, D. W. &
De Souza, E. B. The ontogeny of brain receptors for
corticotropin-releasing factor and the development of
their functional association with adenylate cyclase.
J. Neurosci. 8, 4151–4158 (1988).
140. Levine, S. The ontogeny of the
hypothalamic-pituitary-adrenal axis. The influence of
maternal factors. Ann. NY Acad. Sci. 746, 275–288;
discussion 289–293 (1994).
141. Gunnar, M. R. & Cheatham, C. L. Brain and behavior
interfaces: stress and the developing brain. Infant
Ment. Health J. 24, 195–211 (2003).
A superb paper that summarized the effects of
stress during development and how this knowledge
can be used to develop effective interventions.
142. LeDoux, J. E. The Emotional Brain: The Mysterious
Underpinnings of Emotional Life (Simon & Schuster,
New York, 1996).
Acknowledgements
Sonia Lupien holds a Research Chair on Gender and Mental
Health by the Canadian Institutes of Health Research.
FURTHER INFORMATION
Sonia J. Lupien’s homepage: http://www.humanstress.ca
All liNks Are AcTive iN The oNliNe pdf
R E V I E W S
nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j
u n E 2 0 0 9 | 445
f o c u S o n S t R E S S
© 2009 Macmillan Publishers Limited. All rights reserved
http://www.humanstress.caEffects of stress throughout the
lifespan on the brain, behaviour and
cognitionMainAcknowledgementsReferences
University of Missouri – St. Louis
Department of Accounting
ACCTNG 4435 – Auditing – Summer 2021
Writing Assignment –
Risk Assessment and Management Assertions
at the Crafty Trinkets Company (“CTC”)
Extension of due date – the Writing Assignment is now
due by 1159 PM on 2 July.
Required:
Please read the following information about the Crafty Trinkets
Company (“CTC” or
“the Company”), and follow the instructions at the end of the
case. The requirements of
the case are focused and to-the-point.
While this case assignment is not intended to be some sort of
massive research paper, it
does account for 10% of your final grade. Please spend the time
and attention on this
assignment that 10% of your final grade would warrant. Take
the time to present well-
thought out responses to the requirements, written in your best,
polished writing style.
Please proofread your paper. If your paper appears to have
been dashed off in a few
minutes, unfortunately your grade on the paper will reflect that.
Do a professional job.
When reading the case, please pay careful attention to the dates.
Imagine it is now 28
November 2018. It may be helpful to create a timeline of
events, so you can put the
events in the proper context and chronology. (Please don’t
include any chronology that
you prepare in your submission.) Please note that, given the
dates cited below, none of
the events in the case are impacted by the covid pandemic.
I’m happy to answer any questions you have about the case or
the requirements.
Introduction As a senior auditor in a public accounting firm,
you have been assigned
to plan the audit of the financial statements of a privately-held
company called the
Crafty Trinkets Company (“CTC” or “the Company”).
Company Background CTC designs, manufactures, and
markets a variety of toys,
which are sold primarily to large national retailers like Target
and Wal-Mart. CTC is a
small company compared to competitors like Mattel and Hasbro;
nevertheless, CTC
managers believe their company’s toys are among the best in the
world. Unlike the
larger toy makers, CTC has enjoyed success with a small
portfolio of brands and
products, representing three categories: (1) soft toys, consisting
primarily of its Snuggle
Pets stuffed animals; (2) sturdy toys, including metal-cast and
plastic cast toys like
Speedster cars and Lightning action figures; and (3) digital
toys, consisting of video game
software under development. Like most toy makers, 60 percent
of CTC’s sales revenues
are generated in October and November, with the last two weeks
of November driving
half of those sales.
Your firm, Smith and Company, LLP (“Smith” or “the Firm”),
has been CTC’s public
accounting firm since 2013, providing audit and tax services to
the Company. The
primary external user of CTC’s audited financial statements is
its bank. Assume it is
now 28 November 2018. You have taken over audit senior
responsibilities for the
Company’s 30 November 2018 year-end financial statement
audit because the previous
audit senior has just unexpectedly left your Firm to accept a job
in another city. (The
Company’s year-end date is 30 November.)
Please review the following excerpts from relevant documents
involving the planning
and execution of the current year audit.
Excerpts from the document….
“Observations Noted in My Review of the
Previous Year (year-end 30 November 2017) Audit File”
(written by you, the new senior auditor…)
• In fiscal 2017, CTC exceeded its earning targets, reporting
operating income of
$3,026,100 and net income before taxes of $2,572,800. The
only large negatives
for the year 2017 were the substantial additions to allowances
for receivables and
inventories, including an extra $300,000 in the allowance for
doubtful accounts
related to the struggling Toy-Mart chain in the United States.
The increase to the
allowance for inventory was due to possible obsolete inventory.
The increase in
the Allowance for Doubtful Accounts was accomplished by a
debit to Bad Debt
Expense and a credit to the Allowance.
• CTC’s management advised our Firm that retailers
dramatically reduced the
quantity of toys they were willing to stock on its shelves at any
one time in fiscal
2017, and were expected to continue this trend into 2018. This
change did not (at
least in 2017) reduce the volume of the toys sold through
retailers, but it has
intensified competition among industry competitors for retail
shelf space, and
increased operating costs by increasing the frequency of
shipments to stores. (In
the competitive environment in which CTC operates, shipping
costs are usually
paid by the manufacturer, not the retailer.) It’s possible that the
restrictions on
shelf-space could result in lower sales in future periods.
• Ever since 2014, CTC executives have shared in a bonus pool
that is created
through CTC contributions of 10% of the first $750,000 of
operating income, plus
20% on the next $750,000, and an additional 30% of the next
$1,500,000. CTC’s
total contributions to the bonus pool are capped at a yearly
maximum of
$675,000.
• CTC does not have an internal audit group. In addition, they
have struggled to
implement the COSO Integrated Framework of Internal Control.
Excerpts from the document….
“Findings from Visit to Client and Interim (before year-end)
Audit Procedures Conducted in September 2018”
(written by the audit senior that you replaced)
• In January 2018, the long-time CEO and CFO of CTC retired,
and replacements
were hired and began work in March 2018. One senior manager
told me that the
pair are like “fire-breathing dragons,” and have indicated that
their “sole
number-one focus will be increasing sales and profitability, and
those who do
not contribute will be given the opportunity to continue their
career elsewhere.”
• We developed an understanding of controls over purchases and
payable and
found that controls in this area were not well designed and were
not operating
effectively. Therefore, following what I know about audit
strategy, I decided to
test the controls over purchases and payables in the interim
period and assigned
it to one of the staff auditors. Although this testing has not yet
been reviewed,
one item seemed unusual. It involved a payment of $30,000 to
the International
Toy Manufacturer Workers Union. The payment was initiated
by the CTC VP-
Operations and approved by the current CFO, and was properly
classified as a
non-operating expense. According to the VP-Operations, the
payment was “a
gesture of support for the toy factory workers – a gesture we
believe is important
since workers believe themselves to be underpaid and are
discussing the
possibility of work stoppages and strikes in the Fall of 2018.
We hope this
payment will assist in making it possible for union executives to
encourage their
members to resolve these issues before a work stoppage or
strike.”
• In the tests of controls over revenues and receivables, one of
the staff auditors
that conducted the testing noted that controls were effective. In
particular the
staff member noted that one thing that was very impressive was
that the CFO
was active in oversight of the area of bad debts and inventory
obsolescence.
Indeed, as an example, the current CFO herself approved the
reversal/recovery
of the $300,000 amount allowed for with respect to Toy-Mart,
and had even
initiated and approved the journal entry for the transaction,
reversing it into
income (debit Allowance for Doubtful Accounts, credit Bad
Debt Expense)
without the involvement of the credit manager.
Excerpts from the document….
“Audit Partner Memo to 30 November 2018 Audit Workpaper
File”
• CTC had been unable to produce enough Snuggle Pets for the
December 2017
year-end holiday season, due to raw material shortages in an
unstable stuffing
supplier market. The Company was able, however, to increase
production in
January 2018, which allowed for increased sales for Valentine’s
Day in February
2018. Soon after, at the insistence of the national retailers, all
unsold Snuggle Pets
were returned to CTC for a full refund. The retailers insisted
that the absence of
Snuggle Pets in stores after February 2018 would build demand
for the
Fall/Winter 2018-2019, as the retailers focused on the end-of-
the-year holidays
and gift-giving season.
• CTC has deferred their purchase of new, hi-tech
manufacturing equipment due
to a shortage of cash and the inability to obtain favorable
financing. This is the
second year in a row that CTC has deferred this investment.
• CTC executives entered into an agreement with Cartoon
Studios, Inc., who had
produced their very first animated movie, called “The Bronx
Zoo – Escape to
Manhattan!” for release on 30 November 2018. (The movie is
billed as “Jumanji”
meets “Babe: Pig in the City”!) For $900,000, CTC had won
the rights to produce a
line of soft and plastic toys based on characters from the movie.
(CTC plans to
amortize this fee over 9 years.) The toys would be sold through
CTC’s regular
retail customers. The toys were on schedule to be in stores on
30 November. The
agreement between the Company and Cartoon Studios indicated
that Cartoon
Studios would compensate CTC for the cost of the unsold toys
if sales of the toys
failed to reach $1,500,000 during the first two months after the
movie’s release.
CTC plans to accrue $1,500,000 of sales revenue on 30
November relating to this
provision of the agreement.
• CTC executives have carefully reviewed the pricing and
valuation of inventory
during early November 2018, and determined that the inventory
valuation
reserve established in the previous year is no longer necessary;
a journal entry
was made by the CFO on 15 November 2018 to reverse the
valuation allowance
into operating income in a manner similar to the reversal related
to the
Allowance for Doubtful Accounts described above.
• In October 2018, CTC announced that it was suspending it’s
partnership with the
charitable organization “Toys for Kids,” an organization that
distributes toys to
underprivileged children in less-developed countries around the
world. In the
past, CTC had donated a substantial number of toys to “Toys for
Kids.”
• On November 1, 2018, the Company’s Board of Directors
Compensation
Committee agreed to double the Company’s contribution to the
bonus pool, to
$1,350,000. This measure will be effective for the year-ending
30 November 2018.
Every day, parents observe the growing behavioural repertoir
Every day, parents observe the growing behavioural repertoir
Every day, parents observe the growing behavioural repertoir
Every day, parents observe the growing behavioural repertoir
Every day, parents observe the growing behavioural repertoir
Every day, parents observe the growing behavioural repertoir

More Related Content

Similar to Every day, parents observe the growing behavioural repertoir

Endocrinal response to stress
Endocrinal response to stressEndocrinal response to stress
Endocrinal response to stressAmira Ghiaty
 
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...Om Verma
 
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...Act addiction and evolutionary process, common aspects in pharmaco toxicologi...
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...M. Luisetto Pharm.D.Spec. Pharmacology
 
Stress And External Factors
Stress And External FactorsStress And External Factors
Stress And External Factorstraorefatima
 
Add adhd cause cure
Add adhd cause cureAdd adhd cause cure
Add adhd cause cureJohn Bergman
 
ADD and ADHD cause cure
ADD and ADHD cause cureADD and ADHD cause cure
ADD and ADHD cause cureJohn Bergman
 
Polina Kapoustina 1st student scholarship
Polina Kapoustina 1st student scholarshipPolina Kapoustina 1st student scholarship
Polina Kapoustina 1st student scholarshipPolina Kapoustina
 
Contributions of Neuroscience toOur Understanding of Cogniti
Contributions of Neuroscience toOur Understanding of CognitiContributions of Neuroscience toOur Understanding of Cogniti
Contributions of Neuroscience toOur Understanding of CognitiAlleneMcclendon878
 
Slide on neuroendocrinology
Slide on neuroendocrinologySlide on neuroendocrinology
Slide on neuroendocrinologyDishaSinha11
 
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxCapitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxhacksoni
 
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxCapitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxhumphrieskalyn
 
Fisiopatologia dello stress neonatale
Fisiopatologia dello stress neonataleFisiopatologia dello stress neonatale
Fisiopatologia dello stress neonataleCarlo Bellieni
 
psychoneuroimmunology in relation to psychiatric nursing
psychoneuroimmunology in relation to psychiatric nursingpsychoneuroimmunology in relation to psychiatric nursing
psychoneuroimmunology in relation to psychiatric nursingssuser8767171
 
Assignment #2 Neurobiology PTSD and Memory
Assignment #2 Neurobiology PTSD and MemoryAssignment #2 Neurobiology PTSD and Memory
Assignment #2 Neurobiology PTSD and MemoryCerin Takeuchi Venegas
 
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune function
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune functionLifelong impacts of moderate prenatal alcohol exposure on neuroimmune function
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune functionBARRY STANLEY 2 fasd
 
Infant brain development
Infant brain developmentInfant brain development
Infant brain developmentKhaled Saad
 
Maternal Stress And Offsprings Development
Maternal Stress And Offsprings DevelopmentMaternal Stress And Offsprings Development
Maternal Stress And Offsprings Developmenttraorefatima
 

Similar to Every day, parents observe the growing behavioural repertoir (20)

Endocrinal response to stress
Endocrinal response to stressEndocrinal response to stress
Endocrinal response to stress
 
Skaidres
SkaidresSkaidres
Skaidres
 
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...
Omega-3 Fatty Acids Increase Brain Volume While Reversing Many Aspects of Neu...
 
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...Act addiction and evolutionary process, common aspects in pharmaco toxicologi...
Act addiction and evolutionary process, common aspects in pharmaco toxicologi...
 
Abstract template addiction
Abstract template addiction Abstract template addiction
Abstract template addiction
 
A neurobiological model for the effects of early brainstem functioning
A neurobiological model for the effects of early brainstem functioningA neurobiological model for the effects of early brainstem functioning
A neurobiological model for the effects of early brainstem functioning
 
Stress And External Factors
Stress And External FactorsStress And External Factors
Stress And External Factors
 
Add adhd cause cure
Add adhd cause cureAdd adhd cause cure
Add adhd cause cure
 
ADD and ADHD cause cure
ADD and ADHD cause cureADD and ADHD cause cure
ADD and ADHD cause cure
 
Polina Kapoustina 1st student scholarship
Polina Kapoustina 1st student scholarshipPolina Kapoustina 1st student scholarship
Polina Kapoustina 1st student scholarship
 
Contributions of Neuroscience toOur Understanding of Cogniti
Contributions of Neuroscience toOur Understanding of CognitiContributions of Neuroscience toOur Understanding of Cogniti
Contributions of Neuroscience toOur Understanding of Cogniti
 
Slide on neuroendocrinology
Slide on neuroendocrinologySlide on neuroendocrinology
Slide on neuroendocrinology
 
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxCapitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
 
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docxCapitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
Capitão et al. Translational Psychiatry ( 2019) 930 httpsdoi.docx
 
Fisiopatologia dello stress neonatale
Fisiopatologia dello stress neonataleFisiopatologia dello stress neonatale
Fisiopatologia dello stress neonatale
 
psychoneuroimmunology in relation to psychiatric nursing
psychoneuroimmunology in relation to psychiatric nursingpsychoneuroimmunology in relation to psychiatric nursing
psychoneuroimmunology in relation to psychiatric nursing
 
Assignment #2 Neurobiology PTSD and Memory
Assignment #2 Neurobiology PTSD and MemoryAssignment #2 Neurobiology PTSD and Memory
Assignment #2 Neurobiology PTSD and Memory
 
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune function
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune functionLifelong impacts of moderate prenatal alcohol exposure on neuroimmune function
Lifelong impacts of moderate prenatal alcohol exposure on neuroimmune function
 
Infant brain development
Infant brain developmentInfant brain development
Infant brain development
 
Maternal Stress And Offsprings Development
Maternal Stress And Offsprings DevelopmentMaternal Stress And Offsprings Development
Maternal Stress And Offsprings Development
 

More from BetseyCalderon89

MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docxMANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docxBetseyCalderon89
 
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docxManage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docxBetseyCalderon89
 
Make sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docxMake sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docxBetseyCalderon89
 
Make sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docxMake sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docxBetseyCalderon89
 
make sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docxmake sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docxBetseyCalderon89
 
make two paragraphs on diffences and similiarties religous belifs .docx
make two paragraphs on diffences and similiarties  religous belifs .docxmake two paragraphs on diffences and similiarties  religous belifs .docx
make two paragraphs on diffences and similiarties religous belifs .docxBetseyCalderon89
 
Make a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docxMake a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docxBetseyCalderon89
 
Make a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docxMake a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docxBetseyCalderon89
 
Make a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docxMake a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docxBetseyCalderon89
 
Make sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docxMake sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docxBetseyCalderon89
 
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docxMajor Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docxBetseyCalderon89
 
Major earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docxMajor earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docxBetseyCalderon89
 
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docxMajor Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docxBetseyCalderon89
 
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docxMaintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docxBetseyCalderon89
 
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docxMaintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docxBetseyCalderon89
 
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docxMacro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docxBetseyCalderon89
 
Macromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docxMacromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docxBetseyCalderon89
 
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docxM7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docxBetseyCalderon89
 
Madison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docxMadison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docxBetseyCalderon89
 
Main content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docxMain content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docxBetseyCalderon89
 

More from BetseyCalderon89 (20)

MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docxMANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
 
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docxManage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
 
Make sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docxMake sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docx
 
Make sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docxMake sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docx
 
make sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docxmake sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docx
 
make two paragraphs on diffences and similiarties religous belifs .docx
make two paragraphs on diffences and similiarties  religous belifs .docxmake two paragraphs on diffences and similiarties  religous belifs .docx
make two paragraphs on diffences and similiarties religous belifs .docx
 
Make a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docxMake a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docx
 
Make a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docxMake a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docx
 
Make a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docxMake a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docx
 
Make sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docxMake sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docx
 
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docxMajor Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
 
Major earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docxMajor earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docx
 
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docxMajor Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
 
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docxMaintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
 
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docxMaintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
 
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docxMacro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
 
Macromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docxMacromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docx
 
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docxM7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
 
Madison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docxMadison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docx
 
Main content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docxMain content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docx
 

Recently uploaded

Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17Celine George
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Krashi Coaching
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentInMediaRes1
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfsanyamsingh5019
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Celine George
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppCeline George
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docxPoojaSen20
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxOH TEIK BIN
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdfssuser54595a
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Educationpboyjonauth
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 

Recently uploaded (20)

Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media Component
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdf
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
URLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website AppURLs and Routing in the Odoo 17 Website App
URLs and Routing in the Odoo 17 Website App
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docx
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptx
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Education
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 

Every day, parents observe the growing behavioural repertoir

  • 1. Every day, parents observe the growing behavioural repertoires of their infants and young children, and the corresponding changes in cognitive and emotional functions. These changes are thought to relate to normal brain development, particularly the development of the hippocampus, the amygdala and the frontal lobes, and the complex circuitry that connects these brain regions. At the other end of the age spectrum, we observe changes in cognition that accompany aging in our parents. These changes are related to both normal and pathological brain processes associated with aging. Studies in animals and humans have shown that during both early childhood and old age the brain is particularly sensitive to stress, probably because it undergoes such important changes during these periods. Furthermore, research now relates exposure to early-life stress with increased reactivity to stress and cognitive deficits in adulthood, indicating that the effects of stress at different periods of life interact. Stress triggers the activation of the hypothalamus- pituitary-adrenal (HPA) axis, culminating in the pro- duction of glucocorticoids by the adrenals (FIG. 1). Receptors for these steroids are expressed throughout the brain; they can act as transcription factors and so regulate gene expression. Thus, glucocorticoids can have potentially long-lasting effects on the functioning of the brain regions that regulate their release. This Review describes the effects of stress during pre- natal life, infancy, adolescence, adulthood and old age on
  • 2. the brain, behaviour and cognition, using data from ani- mal (BOX 1) and human studies. Here, we propose a model that integrates the effects of stress across the lifespan, along with future directions for stress research. Prenatal stress Animal studies. In animals, exposure to stress early in life has ‘programming’ effects on the HPA axis and the brain1. A single or repeated exposure of a pregnant female to stress2 or to glucocorticoids3 increases mater- nal glucocorticoid secretion. A portion of these gluco- corticoids passes through the placenta to reach the fetus, increasing fetal HPA axis activity and modifying brain development4. In rats prenatal stress leads to long-term increases in HPA axis activity 5. Controlling glucocor- ticoid levels in stressed dams by adrenalectomy and hormone replacement prevents these effects, indicating that elevations in maternal glucocorticoids mediate the prenatal programming of the HPA axis6. Glucocorticoids are important for normal brain maturation: they initiate terminal maturation, remodel axons and dendrites and affect cell survival7; both sup- pressed and elevated glucocorticoid levels impair brain development and functioning. For example, admin- istration of synthetic glucocorticoids to pregnant rats delays the maturation of neurons, myelination, glia and vasculature in the offspring, significantly altering neuronal structure and synapse formation and inhibit- ing neurogenesis4. Furthermore, juvenile and adult rats exposed to prenatal stress have decreased numbers of mineralocorticoid receptors (MRs) and glucocorticoid recep- tors (GRs) in the hippocampus, possibly because of epi - genetic effects on gene transcription8. The hippocampus
  • 3. *Université de Montréal, Mental Health Research Centre, Fernand Seguin Hôpital Louis‑ H Lafontaine, Montreal, Quebec, H1N 3V2, Canada. ‡Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, New York 10021, USA. §Institute of Child Development, University of Minnesota, Minneapolis, Minnesota 55455, USA. ||Department of Psychiatry, Emory University, 101 Woodruff Circle, Suite 4000, Atlanta, Georgia 30307, USA. Correspondence to S.J.L. e‑ mail: [email protected] umontreal.ca doi:10.1038/nrn2639 Published online 29 April 2009 Programming When an environmental factor that acts during a sensitive developmental period affects the structure and function of tissues, leading to effects that persist throughout life. Effects of stress throughout the lifespan on the brain, behaviour and cognition
  • 4. Sonia J. Lupien*, Bruce S. McEwen‡, Megan R. Gunnar § and Christine Heim|| Abstract | Chronic exposure to stress hormones, whether it occurs during the prenatal period, infancy, childhood, adolescence, adulthood or aging, has an impact on brain structures involved in cognition and mental health. However, the specific effects on the brain, behaviour and cognition emerge as a function of the timing and the duration of the exposure, and some also depend on the interaction between gene effects and previous exposure to environmental adversity. Advances in animal and human studies have made it possible to synthesize these findings, and in this Revi ew a model is developed to explain why different disorders emerge in individuals exposed to stress at different times in their lives. R E V I E W S 434 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S © 2009 Macmillan Publishers Limited. All rights reserved mailto:[email protected] mailto:[email protected] Nature Reviews | Neuroscience Hippocampus
  • 5. Hypothalamus Amygdala CRH AVP ACTH Glucocorticoids Adrenal cortex Anterior pituitary Frontal cortex GRs GRs GRs MRs and GRs Mineralocorticoid receptor A receptor that is activated by mineralocorticoids, such as aldosterone and deoxycorti- costerone, as well as glucocorticoids, such as cortisol and cortisone. It also responds to progestins.
  • 6. Glucocorticoid receptor A receptor that is activated by cortisol, corticosterone and other glucocorticoids and is expressed in almost every cell in the body. It regulates genes controlling development, metabolism and the immune response. inhibits HPA axis activity (FIG. 1), and a prenatal stress- induced reduction in hippocampal MRs and GRs could decrease this inhibition, with a resulting increase in basal and/or stress-induced glucocorticoid secretion. In rhe- sus monkeys, prenatal treatment with the synthetic GR agonist dexamethasone causes a dose-dependent degen- eration of hippocampal neurons, leading to a reduced hippocampal volume at 20 months of age9. Effects on other brain regions are also apparent. Rats exposed to stress during the last week of gestation have significantly decreased dendritic spine density in the anterior cingulate gyrus and orbitofrontal cortex10. Furthermore, prenatal exposure to glucocorticoids leads to increased adult corticotropin-releasing hormone (CRH) levels in the central nucleus of the amygdala, a key region in the regulation of fear and anxiety11. Exposure to prenatal stress has three major effects on adult behaviour: learning impairments, especially in aging rats12; enhanced sensitivity to drugs of abuse13; and increases in anxiety- and depression-related behav- iours14. The impaired learning is thought to result from the effects of prenatal stress on hippocampal function15, whereas the effects on anxiety are thought to be medi- ated by prenatal stress-induced increases in CRH in the
  • 7. amygdala11. Prenatal glucocorticoid exposure affects the developing dopaminergic system, which is involved in reward- or drug-seeking behaviour16, and it has been suggested that the increased sensitivity to drugs of abuse is related to the interaction between prenatal stress, glucocorticoids and dopaminergic neurons16. Human studies. In agreement with animal data, findings from retrospective studies on children whose mothers experienced psychological stress or adverse events or received exogenous glucocorticoids during pregnancy suggest that there are long-term neurodevelopmental effects17. First, maternal stress or anxiety18, depression19 and glucocorticoid treatment during pregnancy17 have been linked with lower birthweight or smaller size (for gestational age) of the baby. More importantly, mater- nal stress, depression and anxiety have been associated with increased basal HPA axis activity in the offspring at different ages, including 6 months20, 5 years21 and 10 years22. Disturbances in child development (both neurologi- cal and cognitive) and behaviour have been associated with maternal stress23 and maternal depression dur- ing pregnancy 24, and with fetal exposure to exogenous gluco corticoids in early pregnancy 25. These behavioural alterations include unsociable and inconsiderate behav- iours, attention deficit hyperactivity disorder and sleep disturbances as well as some psychiatric disorders, including depressive symptoms, drug abuse and mood and anxiety disorders. Very few studies have measured Figure 1 | The stress system. When the brain detects a threat, a coordinated physiological response involving autonomic, neuroendocrine, metabolic and immune
  • 8. system components is activated. A key system in the stress response that has been extensively studied is the hypothalamus-pituitary-adrenal (HPA) axis. Neurons in the medial parvocellular region of the paraventricular nucleus of the hypothalamus release corticotropin- releasing hormone (CRH) and arginine vasopressin (AVP). This triggers the subsequent secretion of adrenocortico- tropic hormone (ACTH) from the pituitary gland, leading to the production of glucocorticoids by the adrenal cortex. In addition, the adrenal medulla releases catecholamines (adrenaline and noradrenaline) (not shown). The responsiveness of the HPA axis to stress is in part determined by the ability of glucocorticoids to regulate ACTH and CRH release by binding to two corticosteroid receptors, the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). Following activation of the system, and once the perceived stressor has subsided, feedback loops are triggered at various levels of the system (that is, from the adrenal gland to the hypothalamus and other brain regions such as the hippocampus and the frontal cortex) in order to shut the HPA axis down and return to a set homeostatic point. By contrast, the amygdala, which is involved in fear processing142, activates the HPA axis in order to set in motion the stress response that is necessary to deal with the challenge. Not shown are the other major systems and factors that respond to stress, including the autonomic nervous system, the inflammatory cytokines and the metabolic hormones. All of these are affected by HPA activity and, in turn, affect HPA function, and they are also implicated in the pathophysiological changes that occur in response to chronic stress, from early experiences into adult life. R E V I E W S
  • 9. nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 435 f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved changes in the brain as a function of prenatal stress in humans. However, a recent study showed that low birth- weight combined with lower levels of maternal care was associated with reduced hippocampal volume in adult- hood26. This finding is consistent with evidence that effects of prenatal stress in humans are often moderated by the quality of postnatal care, which in turn is consist- ent with the protracted postnatal development of the human brain. Postnatal stress Animal studies. Although in rodents the postnatal period is relatively hyporesponsive to stress (BOX 2), one of the most potent stressors for pups is separation from the dam. long separation periods (3 h or more each day) activate the pups’ HPA axis, as evidenced by increased plasma levels of adrenocorticotropic hormone and glucocorticoids27. Protracted maternal separation also reduces pituitary CRH binding sites28, and low levels of maternal care reduce GR levels in the hippocampus29. The effects of maternal deprivation extend beyond the HPA axis. Early prolonged maternal separation in rats increases the density of CRH binding sites in the prefrontal cortex, amygdala, hypothalamus, hippo- campus and cerebellum, as measured post-infancy28. In the hippocampus CRH mediates stress-related loss of
  • 10. branches and spines30, and in the amygdala and hypotha- lamus elevated CRH levels are associated with increased anxiety and HPA axis activity, respectively31. Thus, the increase in CRH-binding sites induced by maternal sep- aration might have negative effects over time. The long- term effects of prolonged separation depend on the age of the pup and the duration of the deprivation, with the effects noted above generally being greater when these separations occur earlier in infancy and last for longer durations32. Although the rodent work provides a rich frame- work for conceptualizi ng the impact of early-life stress, the fact that the rodent brain is much less developed at birth than the primate brain makes translation of the findings to humans somewhat challenging (BOX 3). non- human primates have more human-like brain matura- tion at birth and patterns of parent–offspring relations, and so provide an important bridge in the translation of the rodent findings. Studies in monkeys have shown that repeated, unpredictable separations from the mother33, unpredictable maternal feedings34 or spontaneous mater- nal abusive behaviour35 increases CRH concentrations in the cerebrospinal fluid and alters the diurnal activity of the HPA axis for months or even years after the period of adversity: cortisol levels are lower than normal early in the morning (around wake-up) and slightly higher than normal later in the day, an effect that seems to reverse over time in the absence of continued, ongoing psy- chosocial stress35. These diurnal effects have not been noted in rodents, but the effects on higher brain regions seem to be comparable to the rodent findings and include heightened fear behaviour36, exaggerated startle responses33, hippocampal changes such as an increase in the intensity of non-phosphorylated neurofilament pro-
  • 11. tein immunoreactivity in the dentate gyrus granule cell layer37, and atypical development of prefrontal regions involved in emotion and behaviour control38. Human studies. A human equivalent of the rodent maternal separation paradigms might be studies of children who attend full-day, out-of-home day care centres. Studies have reported that glucocorticoid levels rise in these children over the day, more so in toddlers than in older preschool-aged children39,40. However, it is important to note that the elevated glucocorticoid levels observed are less pronounced than those observed in rodents and monkeys exposed to maternal separation. Moreover, although age accounts for most of the varia- tion in the rise in glucocorticoid levels by late afternoon, the quality of care is also important, with less supportive care producing larger increases, especially for children who are more emotionally negative and behaviour- ally disorganized39. So far, there is no evidence that the elevated glucocorticoid levels associated with being in day care affect development; however, children who are exposed to poor care for long hours early in develop- ment have an increased risk of behaviour problems later in development41. Parent–child interactions and the psychological state of the mother also influence the child’s HPA axis activity. Beginning early in the first year, when the HPA system of the infant is quite labile, sensitive parenting is associ - ated with either smaller increases in or less prolonged activations of the HPA axis to everyday perturbations42. Maternal depression often interferes with sensitive and supportive care of the infant and young child; there is increasing evidence that offspring of depressed mothers, Box 1 | Models to study stress in animals and humans
  • 12. The hypothalamus-pituitary-adrenal axis can be activated by a wide variety of stressors. Some of the most potent are psychological or processive stressors (that is, stressors that involve higher-order sensory cognitive processing), as opposed to physiological or systemic stressors. Many psychological stressors are anticipatory in nature — that is, they are based on expectation as the result of learning and memory (for example, conditioned stimuli in animals and the anticipation of threats, real or implied, in humans) or on species-specific predispositions (for example, avoidance of open space in rodents or the threat of social rejection and negative social evaluations in humans). Animal studies allow the development of experimental protocols in which animals are submitted to acute and/or chronic stress and the resulting effects on brain and behaviour are studied. Experimental stressful ‘early-life’ manipulations in animals can be broadly split into prenatal and postnatal manipulations. Prenatal manipulations involve maternal stress, exposing the mother to synthetic glucocorticoids or maternal nutrient restriction. Postnatal manipulations include depriving the animal of maternal contact, modifying maternal behaviour and exposing the animal to synthetic glucocorticoids. In these protocols, the cause–effects relationship between stress and its impact on the brain can be demonstrated. By contrast, and because of ethical issues,
  • 13. the cause–effects impact of stress on the brain cannot be studied in humans, and most human studies are correlational by nature. However, there are some ‘experiments of nature’ that can be used to inform scientists about the effects of chronic exposure to early adversity on brain development and of adulthood and late- life stress effects on the brain. Intrauterine under-growth and low birth weight are considered indices of prenatal stress (including malnutrition) in humans. In terms of postnatal stress, low socio-economic status, maltreatment and war are considered adverse events. In adults and older adults, studies of chronic caregivers (spouses of patients with brain degenerative disorders, parents of chronically sick children and health-care professionals) provide a human model of the impact of chronic stress on the brain, behaviour and cognition. R E V I E W S 436 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S © 2009 Macmillan Publishers Limited. All rights reserved especially those who were clinically depressed in the child’s early years, are at risk of heightened activity of the HPA axis43 or of developing depression during ado-
  • 14. lescence (controlling for maternal depression during adolescence)44. However, it should be noted that it can be difficult to exclude potentially confounding genetic factors in such studies. Furthermore, preschool-aged children of depressed mothers exhibit electroencephalo- graphic alterations in frontal lobe activity that corre- late with diminished empathy and other behavioural problems45. In contrast to findings of elevated glucocorticoid lev- els in conditions of low parental care, studies in human children exposed to severe deprivation (for example, in orphanages or other institutions), neglect or abuse report lower basal levels of glucocorticoids, similar to what has been observed in primates39. one proposed mechanism for the development of hypocortisolism is downregulation of the HPA axis at the level of the pitui- tary in response to chronic CRH drive from the hypoth- alamus46, whereas a second possible mechanism is target tissue hypersensitivity to glucocorticoids47. Importantly, this hypocortisolism in humans in response to severe stress may not be permanent: sensitive and supportive care of fostered children normalizes their basal gluco- corticoid levels after only 10 weeks48. Another impor- tant finding comes from a recent study which showed that exposure to early adversity is associated with epi - genetic regulation of the GR receptor, as measured in the post-mortem brains of suicide victims49. Stress in adolescence Animal studies. In rodents the period of adolescence has three stages: a prepubescent or early adolescent period from day 21 to 34, a mid-adolescent period from day 34 to 46 and a late adolescent period from day 46 to 59 (ReF. 50). In humans, adolescence is often considered
  • 15. to demarcate the period of sexual maturation (that is, starting with menarche in girls). Although adolescence is a time of significant brain development, particularly in the frontal lobe51, there has been relatively little research on stress during this period in rodents. In adolescent rodents, HPA function is char- acterized by a prolonged activation in response to stres- sors compared with adulthood. Moreover, prepubertal rats have a delayed rise of glucocorticoid levels and prolonged glucocorticoid release in response to several types of stressors compared with adult rats52, owing to incomplete maturation of negative-feedback systems53. In contrast to adult rats, which show a habituation of the stress response with repeated exposure to the same stressor 54, juvenile rats have a potentiated release of adrenocorticotropic hormone and glucocorticoids after repeated exposure to stress55, suggesting that the HPA axis responses to acute and chronic stress depend on the developmental stage of the animal. Compared with exposure to stress in adulthood alone, exposure to stress as both a juvenile and an adult increases basal anxiety levels in the adult 56. Moreover, exposure to juvenile stress results in greater HPA axis activation than a dou- ble exposure to stress during adulthood56, and this effect is long-lasting. These results suggest that repeated stress in adolescence leads to greater exposure of the brain to glucocorticoids than similar experiences in adulthood. The fact that the adolescent brain undergoes vigor- ous maturation and the fact that, in rats, the hippocam- pus continues to grow until adulthood suggest that the adolescent brain may be more susceptible to stressors and the concomitant exposure to high levels of gluco- corticoids than the adult brain. Consistent with this
  • 16. hypothesis are findings that increased glucocorticoid levels before but not after puberty alter the expression of genes for nMDA (N-methyl-d-aspartate) receptor sub- units in the hippocampus57. In addition, chronic, vari- able stress during the peripubertal juvenile period results in reduced hippocampal volume in adulthood, which is accompanied by impairments in Morris water maze navigation and delayed shutdown of the HPA response to acute stress58. These differences became evident only in adulthood58, suggesting that stress in adolescence reduces hippocampal growth. Finally, the effects of juve- nile stress are long-lasting: adult rats exposed to juvenile stress exhibit reduced exploratory behaviour and poor avoidance learning 59. Moreover, stress in adolescence increases susceptibility to drugs of abuse during the adolescent period60 and in adulthood61. Human studies. Interestingly, studies in human adoles- cents also suggest that the adolescent period is associ - ated with heightened basal and stress-induced activity of the HPA axis62. This could be related to the dramatic changes in sex steroid levels during this period, as these steroids influence HPA axis activity50. However, studies of stress in adolescent rats cannot be translated directly to humans because the brain areas that are undergoing development during adolescence differ between rats and humans: although the rodent hippocampus continues to Box 2 | The stress hyporesponsive period: from animals to humans Despite there being clear evidence that corticotropin-releasing hormone-containing neurons are present in the fetal rat139, in rodents noxious stimuli evoke only a subnormal hypothalamus-pituitary-adrenal (HPA) axis response
  • 17. during the first 2 weeks of life140. During this so-called stress hyporesponsive period (SHRP), baseline plasma glucocorticoid levels are lower than normal and are only minimally increased by exposure to a noxious stressor140. The SHRP is due to a rapid regression of the HPA axis after birth140 and may have evolved in rodents to protect the rapidly developing brain from the impact of elevated glucocorticoids. Evidence is accumulating that in children there may be a comparable hyporesponsive period that emerges in infancy and extends throughout most of childhood141. At birth, glucocorticoid levels increase sharply in response to various stressors, such as a physical examination or a heel lance. However, over the course of the first year the HPA axis becomes more insensitive to stressors. No study has assessed the exact period over which this human SHRP may occur, but in adolescents glucocorticoid levels can become elevated in response to a psychosocial stressor141, which suggests that the SHRP could extend throughout childhood. In rodents the SHRP is maintained primarily by maternal care (that is, the presence of the dam seems to suppress HPA axis activity); indeed, maternal separation is a potent inducer of a stress response, even during the SHRP. Similarly, in humans the apparent hyporesponsivity of the HPA axis might reflect the fact that during the first year of life the HPA axis comes under strong social regulation or parental
  • 18. buffering141. Here again, stressors that involve a lack of parental care or social contact can induce a stress response in children. R E V I E W S nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 437 f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved develop well into adulthood, in humans it is fully devel- oped by 2 years of age63. The frontal cortex and amygdala continue to develop in both species, but humans have larger ontogenic bouts of development in frontal regions than do rodents (BOX 3). There are indications that the adolescent human brain might be especially sensitive to the effects of elevated levels of glucocorticoids and, by extension, to stress. Recent studies on the ontogeny of MR and GR expres- sion show that GR mRnA levels in the prefrontal cortex are high in adolescence and late adulthood compared with in infancy, young adulthood and senescence64. This suggests that the cognitive and emotional processes that are regulated by these brain areas might be sensitive to GR-mediated regulation by glucocorticoids in an age- dependent manner. Various forms of psychopathology, including depression and anxiety, increase in prevalence in adolescence65,66. Periods of heightened stress often precede the first episodes of these disorders, raising the
  • 19. possibility that heightened HPA reactivity during adoles- cence increases sensitivity to the onset of stress-related mental disorders. Adolescence is also a period in which the long- lasting effects of earlier exposures to stress become evi - dent. Adolescents who grew up in poor economic condi- tions have higher baseline glucocorticoid levels67, as do adolescents whose mothers were depressed in the early postnatal period44. High early-morning glucocorticoid levels that vary markedly from day to day during the transition to adolescence are not associated with depres- sive symptoms at that time, but they predict increased risk for depression by age 16 (ReF. 44). Although early-life stress impairs hippocampal devel- opment in rodents, there is currently little evidence of comparable effects in humans. Children exposed to physical or sexual abuse early in life do not exhibit reduced hippocampal volume (relative to whole-brain size) as adolescents, although adults with these histo- ries do show volume reductions68. This finding holds even when the abused children have been selected for chronic post-traumatic stress disorder (PTSD), and even though in some cases they exhibit overall reductions in brain volume69. By contrast, alterations in grey matter volume and the neuronal integrity of the frontal cortex, and reduced size of the anterior cingulate cortex, have been reported in adolescents exposed to early (and con- tinued) adversity70. Together, these results suggest that in humans the frontal cortex, which continues to develop during adolescence, might be particularly vulnerable to the effects of stress during adolescence. By contrast, the hippocampus, which develops mainly in the first years of life, might be less affected by exposure to adversity in
  • 20. adolescence. Stress in adulthood Animal studies. Studies on adult stress in rodents have delineated the effects of acute versus chronic stress on brain and behaviour. The impact of acute stressors depends on the level of glucocorticoid elevations, with small increases resulting in enhanced hippocampus- mediated learning and memory, and larger, prolonged elevations impairing hippocampal function71. The inverted-u-shaped effects of acute glucocorticoid ele- vations might serve adaptive purposes by increasing vigilance and learning processes during acute challenges. The mechanism that underlies the acute bipha- sic actions of glucocorticoids on cognition involves the adrenergic system in the basolateral nucleus of the amygdala. By enhancing noradrenergic function in the amygdala, glucocorticoids have a permissive effect on the priming of long-term potentiation in the den- tate gyrus by the basolateral nucleus72. This modulation of noradrenergic function by glucocorticoids has been linked to the enhanced memory for emotional events that occur under stress73. Chronic stress or chronic exogenous administration of glucocorticoids in rodents causes dendritic atrophy in hippocampal CA3 pyramidal neurons74. However, these changes take several weeks to develop and are reversed by 10 days after the cessation of the stressor 75. Chronic stress in adult rats also inhibits neurogenesis in the dentate gyrus76 and causes hippocampal volume loss77. Importantly, this volume decrease is not associ - ated with reduced neuron numbers and is not limited to the dentate gyrus78, suggesting that reduced neuro- genesis might not be the only contributing factor. The
  • 21. morphological changes that take place in the hippocam- pus after chronic stress have been related to changes in spatial learning79, which are reversed following 21 days of withdrawal from stress80. Here, it is interesting to note that in contrast to the effects of chronic or severe stress on the brain and behaviour earlier in life, which are long- lasting, effects of adulthood stress — even chronic stress — are reversed after a few weeks of non-stress. These differences between the effects of early and adulthood Box 3 | Stress effects on the brain: timing is crucial In animals that give birth to relatively mature young (for example, primates, sheep and guinea pigs), maximal brain growth and most of the neuroendocrine maturation occurs in utero. However, in rats, rabbits and mice the mother gives birth to immature young and most of the neuroendocrine development occurs in the postnatal period17. In humans the hypothalamus-pituitary-adrenal axis is highly responsive at birth, but brain development is not finished. The volume of the hippocampal formation increases sharply until the age of 2 years, whereas amygdala volume continues to increase slowly until the late 20s63. By contrast, the development of the frontal cortex in humans takes place mostly between 8 and 14 years of age63. The late increase in prefrontal volumes is consistent with data showing that this region develops latest in terms of myelination and synaptic density in humans136. Prenatal and postnatal stress can both thus have contrasting effects in different
  • 22. species because perinatal manipulations will affect different stages of development as a function of the species studied. Consequently, stress in the first week of the rodent’s life is often developmentally equated with stress during the last trimester of human gestation. Significant decreases in brain volume have been reported in aged animals and humans, although most of the studies performed are cross- sectional. In men the volume of the hippocampus starts to decrease by the second decade of life, whereas in women this decrease is delayed until around 40 years of age, possibly owing to the protective effects of oestrogen137. By contrast, amygdala volume decreases around the sixth decade of life in humans63. In the frontal cortex, different subregions are differentially affected by aging. For example, aging is associated with shrinking of the dorsolateral and inferior frontal cortices, but no age effects have been reported for the anterior cingulate cortex, the frontal pole or the precentral gyrus138. R E V I E W S 438 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S © 2009 Macmillan Publishers Limited. All rights reserved
  • 23. stress might be related to differences in the severity of stressors to which pups and adult rats are exposed or in the development of the hippocampus at the time of exposure. Pyramidal neurons in layers II/III of the prefron- tal cortex also show dendritic retraction and a reduc- tion in spine number 81 in response to chronic stress in adulthood — this can be observed 24 h after a single forced-swim stress82 — but remodelling occurs after ces- sation of the stressor 83. In accordance with these find- ings, glucocorticoid hypersecretion is associated with reduced volume of at least the right anterior cingulate cortex in rodents84. Contrary to the reduction in hip- pocampal and frontal volumes, chronic stress in adult rodents leads to dendritic hypertrophy in the baso lateral amygdala85. Moreover, a recent study showed that even a single acute administration of glucocorticoids caused dend ritic hypertrophy in this area 12 days later 86. The dendritic hypertrophy was correlated with anxi- ety in both the acute86 and the chronic85 administration paradigms. Human studies. In humans, studies of the effects of acute stress confirm animal studies and report the presence of an inverted-u-shaped relationship between gluco- corticoid levels and cognitive performance87. However, contrary to animal studies, in which most laboratory tests for learning and memory involve a fear and/or an emotional process88, tests of learning and memory in humans can differentiate the effects of glucocorticoids on the processing of neutral versus emotional informa- tion. Most studies to date have shown that acute gluco- corticoid elevations significantly increase memory for
  • 24. emotional information, whereas they impair the retrieval of neutral information89. only a few reports suggest that there is an association between exposure to chronic stress and reduced hippo- campal volume in individuals not suffering from men- tal health disorders (for a review see ReF. 90). However, a recent study reported that low self-esteem, a potent predictor of increased reactivity to stress in humans91, is associated with reduced hippocampal volume92. Most of the studies of chronic-stress effects on the adult human brain have concentrated either on stress- related psychopathologies or on the impact of early-life stress on adult psychopathology. A large number of stud- ies have reported elevated basal glucocorticoid levels in individuals with some forms of depression93, whereas reduced basal glucocorticoid concentrations have been reported in patients with PTSD94, although this finding has been controversial95. Given that low glucocorticoid concentrations seem to develop in early childhood in response to neglect or trauma, it is possible that low cortisol predicts vulnerability to developing PTSD in response to trauma in adulthood. Studies of adults who suffered childhood abuse also reveal hyper-reactivity of the HPA axis in abused, depressed individuals96 and hypoactivity in those with PTSD94. The changes in abused, depressed adults have been associated with CRH-induced ‘escape’ of gluco- corticoid secretion from suppression by treatment with dexamethasone97, suggesting that the glucocorticoid feedback of the HPA axis is impaired under conditions of increased hypothalamic drive. Thus, a decreased capac- ity of glucocorticoids to inhibit the HPA axis when it is
  • 25. stimulated could further accentuate CnS responses to stressors. In agreement with this suggestion, increased cerebrospinal fluid CRH levels have been reported in individuals who reported childhood stress98 and childhood abuse99. Decreased hippocampal volume and function are landmark features of depression and PTSD100,101. one cross-sectional study 102 found that a smaller hippo- campus in women with major depression was associ- ated with experiences of childhood trauma, whereas depressed women without such trauma had hippocam- pal volumes similar to healthy controls. This supports the notion that certain brain changes in patients with depression or PTSD could represent markers of vulner- ability for the disorder rather than markers of the dis- order itself. This finding is in line with results from a twin study of Vietnam veterans103 which showed that decreased hippocampal volume is not a consequence of combat exposure or PTSD: decreased volume was also present in unexposed co-twins, and thus it might be a pre-existing risk factor for PTSD that could be genetic or rooted early in life. Stress in aging Animal studies. Approximately 30% of aged rats have basal glucocorticoid hypersecretion, which is correlated with memory impairments and reduced hippocampal volume104. If a middle-aged rat is exposed for a long period to high levels of exogenous glucocorticoids, it will develop memory impairments and hippocampal atrophy105 similar to those observed in these 30% of aged rats. Conversely, artificially keeping glucocorticoid lev- els low in middle-aged rats prevents the emergence of both memory deficits and hippocampal atrophy in old age106. Several groups have also found that chronic stress
  • 26. in aged rats can accelerate the appearance of biomarkers of hippocampal aging (for example, frequency potentia- tion and synaptic excitability thresholds) and that excess endogenous or exogenous glucocorticoids induce hip- pocampal dendritic atrophy and inhibit neurogenesis107. Finally, in aged monkeys108 chronic glucocorticoid treat- ment can increase amyloid-β pathology, similar to that reported in Alzheimer’s disease. These results have given rise to the glucocorticoid cascade hypothesis109, which suggests that there is a rela- tionship between cumulative exposure to high glucocor- ticoid levels and hippocampal atrophy. It was recently renamed the neurotoxicity hypothesis103, because the proposed explanation for this relationship is that pro- longed exposure to stress hormones reduces the ability of neurons to resist insults, thus increasing the rate at which they are damaged by other toxic challenges or ordinary attrition109. Glucocorticoids might have a similar neuro- toxic effect in the prefrontal cortex. A study demon- strated an enhanced elevation of extracellular glutamate levels post-stress in the hippocampus and medial pre- frontal cortex of aged rats compared with young rats110. R E V I E W S nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 439 f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved Stress in adolescence Stress in adulthood Stress in
  • 27. agingPostnatal stressPrenatal stress Birth 2 8 18 30 60 90 Amygdala Frontal cortex Hippocampus Programming effects Differentiation effects Outcome Maintenance/ manifestation effects Maintenance/ manifestation effects ↑ Glucocorticoids ↑↑ Glucocorticoids ↓↓ Glucocorticoids Amygdala Frontal cortex Hippocampus Nature Reviews | Neuroscience ↑ Glucocorticoids (maternal separation) ↓ Glucocorticoids
  • 28. (severe trauma) ↑ Glucocorticoids (depression) ↓ Glucocorticoids (PTSD) Potentiation/ incubation effects ↓ Glucocorticoids (PTSD) ↑ Glucocorticoids (cognitive decline) Effect on HPA axis Increased glutamate levels after stress, and perhaps other neurotoxic insults, might thus increase the vulnerability of the aging brain to neuronal damage. Human studies. Aging, healthy humans exhibit higher mean diurnal levels of cortisol than younger individu- als111, and a longitudinal study has found that elevated plasma glucocorticoid levels over years in older adults negatively correlates with hippocampal volume and memory112. Given that aged individuals with Alzheimer’s disease present both memory impairments and hippo- campal atrophy, studies have assessed basal glucocor- ticoid levels in this population and found that they are higher than in controls113. In addition, chronic glucocor- ticoid treatment has been shown to worsen cognition in people with Alzheimer’s disease114.
  • 29. The frontal lobe also seems to be sensitive to glucocor - ticoid effects during human aging. using a novel in vitro post-mortem tracing method on human brain slices, Dai et al.115 found an inverted-u-shaped effect of glucocor- ticoids on axonal transport in prefrontal neurons with, in most cases, a stimulating effect at low concentrations and a depressing effect at high concentrations. Given that axonal transport plays a crucial part in neuronal survival and function, these results suggest that gluco- corticoids potentially have negative effects on prefrontal cortex neurons’ survival and/or function. A model of stress effects throughout life The data obtained in animals and humans suggest that chronic or repeated exposure to stress has enduring effects on the brain, through activation of the HPA axis and the release of glucocorticoids, with the highest impact on those structures that are developing at the time of the stress exposure (in young individuals) and those that are undergoing age-related changes (in adult and aged indi- viduals). Stress in the prenatal period affects the devel - opment of many of the brain regions that have a role in regulating the HPA axis — that is, the hippocampus, the frontal cortex and the amygdala (programming effects (FIG. 2)). During childhood the hippocampus — which continues to develop after birth — might be the brain region that is most vulnerable to the effects of chronic stress, possibly through a process of increased CRH drive in the hippocampus116. Because it modulates HPA axis activity, altered functioning of the hippocampus Figure 2 | The life cycle model of stress. How the effects of chronic or repeated exposure to stress (or a single exposure to severe stress) at different stages in life depend on the brain
  • 30. areas that are developing or declining at the time of the exposure. Stress in the prenatal period affects the development of many of the brain regions that are involved in regulating the hypothalamus-pituitary-adrenal (HPA) axis — that is, the hippocampus, the frontal cortex and the amygdala (programming effects). Postnatal stress has varying effects: exposure to maternal separation during childhood leads to increased secretion of glucocorticoids, whereas exposure to severe abuse is associated with decreased levels of glucocorticoids. Thus, glucocorticoid production during childhood differentiates as a function of the environment (differentiation effects). From the prenatal period onwards, all developing brain areas are sensitive to the effects of stress hormones (broken blue bars); however, some areas undergo rapid growth during a particular period (solid blue bars). From birth to 2 years of age the hippocampus is developing; it might therefore be the brain area that is most vulnerable to the effects of stress at this time. By contrast, exposure to stress from birth to late childhood might lead to changes in amygdala volume, as this brain region continues to develop until the late 20s. During adolescence the hippocampus is fully organized, the amygdala is still developing and there is an important increase in frontal volume. Consequently, stress exposure during this period should have major effects on the frontal cortex. Studies show that adolescents are highly vulnerable to stress, possibly because of a protracted glucocorticoid response to stress that persists into adulthood (potentiation/incubation effects). In adulthood and during aging the brain regions that undergo the most rapid decline as a result of aging (red bars) are highly vulnerable to the effects of stress hormones. Stress during these periods can lead to the manifestation of incubated effects of early adversity on the brain (manifestation effects) or to maintenance of chronic effects of stress (maintenance effects). PTSD, post-traumatic stress disorder.
  • 31. R E V I E W S 440 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S © 2009 Macmillan Publishers Limited. All rights reserved might cause glucocorticoid hyposecretion in cases of severe abuse, or increased basal cortisol levels in cases of maternal deprivation (differentiation effects (FIG. 2)). By contrast, in adolescence the frontal cortex, which undergoes major development at this stage, may be most vulnerable to the effects of stress, possibly leading to a protracted glucocorticoid response to stress that persists into adulthood (potentiation/incubation effects (FIG. 2)). In adulthood and old age the brain regions that undergo the most rapid decline as a result of aging are highly vul - nerable to the effects of stress hormones. For example, in the hippocampus glucocorticoids affect neurogen- esis, neuronal survival rate and dendritic arborization (manifestation/maintenance effects (FIG. 2)). The neurotoxicity and vulnerability hypotheses. The data obtained in adults and older animals and humans have led to the neurotoxicity hypothesis109, which sug- gests that prolonged exposure to glucocorticoids reduces the ability of neurons to resist insults, increasing the rate at which they are damaged by other toxic challenges or ordinary attrition109. This hypothesis implies that a reduced hippocampal size is the end product of years or decades of PTSD, depressive symptoms or chronic stress. Although the neurotoxicity hypothesis has been
  • 32. confirmed by various animal and human studies, it does not explain the hyposecretion of glucocorticoids that occurs in patients suffering from PTSD, who also present reduced hippocampal volume. Data obtained in children, adolescents or adult ani- mals and humans exposed to acute or early-life trauma have led to the vulnerability hypothesis103. In contrast to the neurotoxicity hypothesis, the vulnerability hypoth- esis suggests that reduced hippocampal volume in adult- hood is not a consequence of chronic exposure to PTSD, depression or chronic stress, but is a pre-existing risk fac- tor for stress-related disorders that is induced by genetics and/or early exposure to stress117. unlike the neurotoxicity hypothesis, the vulnerability hypothesis can explain gluco- corticoid hyposecretion in patients with PTSD. Indeed, studies in children facing significant adversity, such as abuse, report the development of glucocorticoid hypo- secretion39, which might last until adulthood and confer vulnerability to developing PTSD as a result of trauma. we think that the two hypotheses are not mutually exclusive when viewed from a developmental perspec- tive. Indeed, the data summarized in this Review suggest that there might be early windows of vulnerability (or sensitive periods68) during which specific regions of the developing brain are most susceptible to environmental influences, through a neurotoxicity process. Exposure to stress and/or adversity during these key vulnerable periods might slow the development of those brain regions for the duration of the adversity. when meas- ured in adulthood, the reduced volumes of these brain regions could be a strong marker of the time of exposure to early adversity rather than of the effects of specific traumas on various brain regions. These windows of vul- nerability could also be used to predict the nature of the
  • 33. psychopathology that will result from exposure to stress at different ages. Exposure to adversity at the time of hippocampal development could lead to hippocampus- dependent emotional disorders, which would be differ- ent from disorders arising from exposure to adversity at times of frontal cortex development. Two recent studies support this hypothesis. The first reported that women who experienced trauma before the age of 12 years had increased risk for major depression, whereas women who experienced trauma between 12 and 18 years of age more frequently developed PTSD118. The second study reported that repeated episodes of sexual abuse were associated with reduced hippocampal volume if the abuse occurred early in childhood, but with reduced prefrontal cortex volume if the abuse occurred during adolescence119. These results suggest that, similar to what has been observed in animals120, there may be distinct structural, neuropsy- chological and neuropsychiatric sequelae of early abuse, depending in part on the age or developmental stage of the brain when the insult occurred. Besides slowing down the development of the brain during the time of adversity, leading to reduced brain volumes in adulthood, stress in early life could modify the developmental trajectory of the brain. The potential immediate benefit of such modifications is that they might increase acute survival probability, but they could have negative long-term effects. During child- hood and adolescence the brain undergoes a period of overproduction and pruning of synapses121. one of the brain regions that shows the slowest development over the lifespan is the amygdala (BOX 3). It is interest- ing to note that contrary to the hippocampus and the frontal lobe — which show volume reduction as a result of chronic stress — the amygdala increases in volume under
  • 34. chronic stress, owing to increased dendritic arborization. Given that the amygdala plays a significant part in the detection of fear and threat, it is possible that throughout evolution increases in amygdala volume in response to stress might have improved the detection of threatening information and so increased survival probability. If this is indeed the case, young children exposed to adversity should also have increased amygdala volume, but no study has yet examined this important question. This acute effect of adversity on brain organization could have negative long-term consequences. Stress at key periods of synaptic organization could modify the trajectories of connections, leading to an incubation period, such that the effects of stress would not be appar - ent at the time of adversity but would emerge later, when the synaptic organization has been completed. Studies showing protracted effects of early-life stress that emerge at puberty support this suggestion44. Furthermore, although depression is the most extensively documented outcome of exposure to chronic sexual abuse in adults, it is not a common occurrence in children suffering abuse. Indeed, the average time from the onset of abuse to the emergence of clinical depression is 11.5 years, with the first major episode occurring during adolescence122. It is thus conceivable that in susceptible individuals expo- sure to early adversity during a window of vulnerability sets into motion a series of events that lead to a hetero- typic reorganization of synaptic development, resulting in a protracted expression of depression or PTSD. R E V I E W S nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 441
  • 35. f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved This same process could also explain the develop- ment of resilience in face of adversity. Environmental enrichment in rodents is a potent inducer of changes in neurogenesis and/or dendritic arborization in the hippocampus, and has been documented to lead to increases in hippocampal volume123. In children facing early adversity, forms of environmental enrichment, such as support from a family member, enriched day care or school environment or social support from members of the community, could induce a similar het- erotypic reorganization of synaptic development, pro- gramming of neurotrophic factors or changes in gene expression that could lead to resilience later in life. If this is the case, it could be suggested that any type of intervention performed during the early years could not only have a tremendous effect in preventing the deleteri - ous impact of chronic stress and/or early abuse on the developing brain, but could also help to prevent effects on the brain of chronic stress occurring in adulthood or during aging. Conclusions and future directions Although studies on stress have provided a wealth of data delineating the effects of acute and chronic stress on the developing brain, much remains to be done to fully understand how the brain develops pathology or resilience in the face of adversity. we believe that three main factors should receive special consideration in future studies on stress in both animals and humans.
  • 36. The first factor is sex and gender. Sex refers to the bio- logical differences between males and females, whereas gender refers to the different roles (gender role and gen- der identity) that men and women may have during their lifetime. Both sex and gender might have potent influ- ences on stress reactivity in humans of all ages. However, most studies of the effects of stress on the brain, behav- iour and cognition have tested only male animals or humans. This is a major issue considering that studies in both animals50 and humans124 report sex differences in response to stress, and considering the gender gap ratio (two girls for one boy) that emerges in early adoles- cence for the risk of depression125. To this day, a consist- ent finding in the endocrine literature is that the risk of depression in adolescent girls increases with decreasing age at menarche126. An increased sensitivity of girls to environmental and/or family adversity, along with inter- actions between glucocorticoids and gonadal steroids, could be a potential explanation for the increased risk of depressive disorders in females. Recent results show - ing an earlier age at menarche in girls exposed to early adversity127 support this suggestion. The second factor that should be considered in future studies is exposure to environmental toxins. Today, chil - dren in many cities are chronically exposed, at back- ground levels, to a range of common toxins that are environmentally persistent and that tend to be lipophilic and bioaccumulate, such as lead and bisphenol A128. These agents reach humans mainly through food and food additives, and they can be transferred to the fetus through the placenta and to infants through maternal milk129. They have been shown to affect the endocrine system in laboratory animals and in wildlife, and conse- quently have been called ‘endocrine-disrupting chemi-
  • 37. cals’ (ReF. 130). A recent study showed that prenatal and postnatal exposure to lead is associated with increased glucocorticoid responses to acute stress in children131. Also, perinatal exposure to endocrine-disrupting chemicals is associated with an earlier age at menarche among girls132. Taken together, these results suggest that both the timing of sexual maturation and stress reactivity may be sensitive to relatively low levels of endocrine-disrupting chemicals in the environment. The third factor that should receive greater attention is circadian rhythmicity. Sleep deprivation, shift work and jet lag all disrupt normal biological rhythms and have major impacts on health. Interestingly, circadian disorganization is often observed in stress-related dis- orders such as depression133 and PTSD134. The discovery of the molecular clock that is responsible for the genera- tion of circadian rhythms135 provides new insights into how rhythm abnormalities might lead to greater vulner- ability to stress at various ages. Most studies performed in animals and humans do not measure the circadian fluctuations in glucocorticoid levels, but rather concen- trate on specific time points across the day. Although such measurements are easier, they do not provide the full spectrum of circadian variations, which could inform us about specific changes in circadian organi- zation in response to chronic stress across the lifespan. Consequently, studies assessing multiple time points for glucocorticoid secretion across a whole day or sev- eral days are needed in order to document the complex relationships that exist between reactivity to stress and circadian (dis)organization. Animal and human studies have provided a wealth of results showing the negative effects of chronic expo- sure to stress and/or adversity on the developing brain.
  • 38. However, stress is not and should not be considered as a negative concept only. Stress is a physiological response that is necessary for the survival of the spe- cies. The stress response that today can have negative consequences for brain development and mental health may have conferred the necessary tools to our ances- tors in prehistorical times for surviving in the presence of predators. Studies of modern individuals who have developed resilience by facing significant adversity should inform us about the physiological and psy- chological mechanisms at the basis of vulnerability or resilience to stress. understanding these mechanisms, which are possibly rooted in genes and modulated by the family environment, is extremely important if one wants to provide interventions early enough to individ- uals who are the most likely to respond to them. This article has reviewed the potential for early intervention to prevent the deleterious effects of stress on the brain, behaviour and cognition. After more than 30 years of research on the negative effects of stress on the brain, it is now time to turn our attention to the potential posi- tive impact of early interventions on brain development. These results could help us to develop social policies that treat the problem of early-life stress at its root — that is, in the family home. R E V I E W S 442 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S © 2009 Macmillan Publishers Limited. All rights reserved
  • 39. 1. Barker, D. J. The foetal and infant origins of inequalities in health in Britain. J. Public Health Med. 13, 64–68 (1991). 2. Cadet, R., Pradier, P., Dalle, M. & Delost, P. Effects of prenatal maternal stress on the pituitary adrenocortical reactivity in guinea-pig pups. J. Dev. Physiol. 8, 467–475 (1986). 3. Dean, F. & Matthews, S. G. Maternal dexamethasone treatment in late gestation alters glucocorticoid and mineralocorticoid receptor mRNA in the fetal guinea pig brain. Brain Res. 846, 253–259 (1999). 4. Seckl, J. R. Glucocorticoids, developmental ‘programming’ and the risk of affective dysfunction. Prog. Brain Res. 167, 17–34 (2008). A superb review that summarized prenatal work and linked it to clinical implications. 5. Koehl, M. et al. Prenatal stress alters circadian activity of hypothalamo-pituitary-adrenal axis and hippocampal corticosteroid receptors in adult rats of both gender. J. Neurobiol. 40, 302–315 (1999). 6. Barbazanges, A., Piazza, P. V., Le Moal, M. & Maccari, S. Maternal glucocorticoid secretion mediates long-term effects of prenatal stress. J. Neurosci. 16, 3943–3949 (1996). 7. Meyer, J. S. Early adrenalectomy stimulates subsequent growth and development of the rat brain. Exp. Neurol. 82, 432–446 (1983). 8. Weaver, I. C. et al. Epigenetic programming by maternal
  • 40. behavior. Nature Neurosci. 7, 847–854 (2004). The first paper to show that early experience has epigenetic effects, altering methylation patterns. 9. Uno, H. et al. Brain damage induced by prenatal exposure to dexamethasone in fetal rhesus macaques. I. Hippocampus. Brain Res. Dev. Brain Res. 53, 157–167 (1990). 10. Murmu, M. S. et al. Changes of spine density and dendritic complexity in the prefrontal cortex in offspring of mothers exposed to stress during pregnancy. Eur. J. Neurosci. 24, 1477–1487 (2006). 11. Cratty, M. S., Ward, H. E., Johnson, E. A., Azzaro, A. J. & Birkle, D. L. Prenatal stress increases corticotropin- releasing factor (CRF) content and release in rat amygdala minces. Brain Res. 675, 297–302 (1995). 12. Vallee, M. et al. Long-term effects of prenatal stress and postnatal handling on age-related glucocorticoid secretion and cognitive performance: a longitudinal study in the rat. Eur. J. Neurosci. 11, 2906–2916 (1999). 13. Deminiere, J. M. et al. Increased locomotor response to novelty and propensity to intravenous amphetamine self-administration in adult offspring of stressed mothers. Brain Res. 586, 135–139 (1992). 14. Vallee, M. et al. Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion. J. Neurosci. 17, 2626–2636 (1997).
  • 41. 15. Lemaire, V., Koehl, M., Le Moal, M. & Abrous, D. N. Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus. Proc. Natl Acad. Sci. USA 97, 11032–11037 (2000). 16. Piazza, P. V. & Le Moal, M. L. Pathophysiological basis of vulnerability to drug abuse: role of an interaction between stress, glucocorticoids, and dopaminergic neurons. Annu. Rev. Pharmacol. Toxicol. 36, 359–378 (1996). 17. Kapoor, A., Petropoulos, S. & Matthews, S. G. Fetal programming of hypothalamic-pituitary-adrenal (HPA) axis function and behavior by synthetic glucocorticoids. Brain Res. Rev. 57, 586–595 (2008). 18. Hedegaard, M., Henriksen, T. B., Sabroe, S. & Secher, N. J. Psychological distress in pregnancy and preterm delivery. BMJ 307, 234–239 (1993). 19. Orr, S. T. & Miller, C. A. Maternal depressive symptoms and the risk of poor pregnancy outcome. Review of the literature and preliminary findings. Epidemiol. Rev. 17, 165–171 (1995). 20. Lyons-Ruth, K., Wolfe, R. & Lyubchik, A. Depression and the parenting of young children: making the case for early preventive mental health services. Harv. Rev. Psychiatry 8, 148–153 (2000). 21. Gutteling, B. M., de Weerth, C. & Buitelaar, J. K. Prenatal stress and children’s cortisol reaction to the first day of school. Psychoneuroendocrinology 30, 541–549 (2005). 22. O’Connor, T. G. et al. Prenatal anxiety predicts
  • 42. individual differences in cortisol in pre-adolescent children. Biol. Psychiatry 58, 211–217 (2005). 23. Glover, V. Maternal stress or anxiety in pregnancy and emotional development of the child. Br. J. Psychiatry 171, 105–106 (1997). 24. Stott, D. H. Follow-up study from birth of the effects of prenatal stresses. Dev. Med. Child. Neurol. 15, 770–787 (1973). 25. Trautman, P. D., Meyer-Bahlburg, H. F., Postelnek, J. & New, M. I. Effects of early prenatal dexamethasone on the cognitive and behavioral development of young children: results of a pilot study. Psychoneuroendocrinology 20, 439–449 (1995). 26. Buss, C. et al. Maternal care modulates the relationship between prenatal risk and hippocampal volume in women but not in men. J. Neurosci. 27, 2592–2595 (2007). 27. Levine, S. & Wiener, S. G. Psychoendocrine aspects of mother-infant relationships in nonhuman primates. Psychoneuroendocrinology 13, 143–154 (1988). 28. Anisman, H., Zaharia, M. D., Meaney, M. J. & Merali, Z. Do early-life events permanently alter behavioral and hormonal responses to stressors? Int. J. Dev. Neurosci. 16, 149–164 (1998). 29. Liu, D. et al. Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science 277, 1659–1662 (1997).
  • 43. 30. Fenoglio, K. A., Brunson, K. L. & Baram, T. Z. Hippocampal neuroplasticity induced by early-life stress: functional and molecular aspects. Front. Neuroendocrinol. 27, 180–192 (2006). 31. Schulkin, J., Gold, P. W. & McEwen, B. S. Induction of corticotropin-releasing hormone gene expression by glucocorticoids: implication for understanding the states of fear and anxiety and allostatic load. Psychoneuroendocrinology 23, 219–243 (1998). 32. de Kloet, E. R. & Oitzl, M. S. Who cares for a stressed brain? The mother, the kid or both? Neurobiol. Aging 24 (Suppl. 1), S61–S65; discussion S67–S68 (2003). 33. Sanchez, M. M. et al. Alterations in diurnal cortisol rhythm and acoustic startle response in nonhuman primates with adverse rearing. Biol. Psychiatry 57, 373–381 (2005). 34. Coplan, J. D. et al. Persistent elevations of cerebrospinal fluid concentrations of corticotropin- releasing factor in adult nonhuman primates exposed to early-life stressors: implications for the pathophysiology of mood and anxiety disorders. Proc. Natl Acad. Sci. USA 93, 1619–1623 (1996). 35. Sanchez, M. M. The impact of early adverse care on HPA axis development: nonhuman primate models. Horm. Behav. 50, 623–631 (2006). 36. Rosenblum, L. A. et al. Differing concentrations of corticotropin-releasing factor and oxytocin in the cerebrospinal fluid of bonnet and pigtail macaques. Psychoneuroendocrinology 27, 651–660 (2002).
  • 44. 37. Siegel, S. J. et al. Effects of social deprivation in prepubescent rhesus monkeys: immunohistochemical analysis of the neurofilament protein triplet in the hippocampal formation. Brain Res. 619, 299–305 (1993). 38. Sanchez, M. M., Ladd, C. O. & Plotsky, P. M. Early adverse experience as a developmental risk factor for later psychopathology: evidence from rodent and primate models. Dev. Psychopathol. 13, 419–449 (2001). 39. Gunnar, M. R. & Donzella, B. Social regulation of the cortisol levels in early human development. Psychoneuroendocrinology 27, 199–220 (2002). 40. Geoffroy, M. C., Cote, S. M., Parent, S. & Seguin, J. R. Daycare attendance, stress, and mental health. Can. J. Psychiatry 51, 607–615 (2006). 41. NICHD Early Child Care Research Network. Early child care and children’s development prior to school entry: results from the NICHD Study of Early Child Care. Am. Educ. Res. J. 39, 133–164 (2002). 42. Albers, E. M., Riksen-Walraven, J. M., Sweep, F. C. & de Weerth, C. Maternal behavior predicts infant cortisol recovery from a mild everyday stressor. J. Child. Psychol. Psychiatry 49, 97–103 (2008). 43. Lupien, S. J., King, S., Meaney, M. J. & McEwen, B. S. Child’s stress hormone levels correlate with mother’s socioeconomic status and depressive state. Biol. Psychiatry 48, 976–980 (2000). 44. Halligan, S. L., Herbert, J., Goodyer, I. & Murray, L. Disturbances in morning cortisol secretion in
  • 45. association with maternal postnatal depression predict subsequent depressive symptomatology in adolescents. Biol. Psychiatry 62, 40–46 (2007). Provided some of the first evidence that adverse early life experiences in humans, in this case rearing by a mother suffering from post-partum depression, are associated with heightened HPA activity years later, and that the HPA axis hyperactivity mediates the association between early risk exposure and later psychiatric symptoms. 45. Jones, N. A., Field, T. & Davalos, M. Right frontal EEG asymmetry and lack of empathy in preschool children of depressed mothers. Child. Psychiatry Hum. Dev. 30, 189–204 (2000). 46. Fries, E., Hesse, J., Hellhammer, J. & Hellhammer, D. H. A new view on hypocortisolism. Psychoneuroendocrinology 30, 1010–1016 (2005). 47. Yehuda, R., Yang, R. K., Buchsbaum, M. S. & Golier, J. A. Alterations in cortisol negative feedback inhibition as examined using the ACTH response to cortisol administration in PTSD. Psychoneuroendocrinology 31, 447–451 (2006). 48. Gunnar, M. R. & Quevedo, K. M. Early care experiences and HPA axis regulation in children: a mechanism for later trauma vulnerability. Prog. Brain Res. 167, 137–149 (2008). 49. McGowan, P. O. et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nature Neurosci. 12, 342–348 (2009). This study examined epigenetic differences in a
  • 46. neuron-specific glucocorticoid receptor (NR3C1) promoter between post-mortem hippocampus obtained from suicide victims with a history of childhood abuse and hippocampus from either suicide victims with no childhood abuse or controls. It found decreased levels of glucocorticoid receptor mRNA, as well as mRNA transcripts bearing the glucocorticoid receptor 1F splice variant and increased cytosine methylation of an NR3C1 promoter in suicide victims with early abuse. 50. McCormick, C. M. & Mathews, I. Z. HPA function in adolescence: role of sex hormones in its regulation and the enduring consequences of exposure to stressors. Pharmacol. Biochem. Behav. 86, 220–233 (2007). A very good review on the acute and chronic effects of stress during adolescence. 51. O’Donnell, S., Noseworthy, M. D., Levine, B. & Dennis, M. Cortical thickness of the frontopolar area in typically developing children and adolescents. Neuroimage 24, 948–954 (2005). 52. Vazquez, D. M. & Akil, H. Pituitary-adrenal response to ether vapor in the weanling animal: characterization of the inhibitory effect of glucocorticoids on adrenocorticotropin secretion. Pediatr. Res. 34, 646–653 (1993). 53. Goldman, L., Winget, C., Hollingshead, G. W. & Levine, S. Postweaning development of negative feedback in the pituitary-adrenal system of the rat. Neuroendocrinology 12, 199–211 (1973). 54. Girotti, M. et al. Habituation to repeated restraint
  • 47. stress is associated with lack of stress-induced c-fos expression in primary sensory processing areas of the rat brain. Neuroscience 138, 1067–1081 (2006). 55. Romeo, R. D. et al. Stress history and pubertal development interact to shape hypothalamic-pituitary-adrenal axis plasticity. Endocrinology 147, 1664–1674 (2006). 56. Avital, A. & Richter-Levin, G. Exposure to juvenile stress exacerbates the behavioural consequences of exposure to stress in the adult rat. Int. J. Neuropsychopharmacol. 8, 163–173 (2005). 57. Lee, P. R., Brady, D. & Koenig, J. I. Corticosterone alters N-methyl-d-aspartate receptor subunit mRNA expression before puberty. Brain Res. Mol. Brain Res. 115, 55–62 (2003). 58. Isgor, C., Kabbaj, M., Akil, H. & Watson, S. J. Delayed effects of chronic variable stress during peripubertal- juvenile period on hippocampal morphology and on cognitive and stress axis functions in rats. Hippocampus 14, 636–648 (2004). One of the first papers to show protracted effects of adolescent stress on adulthood stress reactivity in rodents. 59. Tsoory, M. & Richter-Levin, G. Learning under stress in the adult rat is differentially affected by ‘juvenile’ or ‘adolescent’ stress. Int. J. Neuropsychopharmacol. 9, 713–728 (2006). 60. Kabbaj, M., Isgor, C., Watson, S. J. & Akil, H. Stress during adolescence alters behavioral sensitization to amphetamine. Neuroscience 113, 395–400 (2002).
  • 48. 61. McCormick, C. M., Robarts, D., Gleason, E. & Kelsey, J. E. Stress during adolescence enhances locomotor sensitization to nicotine in adulthood in female, but not male, rats. Horm. Behav. 46, 458–466 (2004). 62. Gunnar, M. R., Wewerka, S., Frenn, K., Long, J. D. & Griggs, C. Developmental changes in hypothalamus- pituitary-adrenal activity over the transition to adolescence: normative changes and associations with puberty. Dev. Psychopathol. 21, 69–85 (2009). R E V I E W S nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 443 f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved 63. Giedd, J. N. et al. Quantitative magnetic resonance imaging of human brain development: ages 4–18. Cereb. Cortex 6, 551–560 (1996). 64. Perlman, W. R., Webster, M. J., Herman, M. M., Kleinman, J. E. & Weickert, C. S. Age-related differences in glucocorticoid receptor mRNA levels in the human brain. Neurobiol. Aging 28, 447–458 (2007). 65. Dahl, R. E. Adolescent brain development: a period of vulnerabilities and opportunities. Keynote address. Ann. NY Acad. Sci. 1021, 1–22 (2004).
  • 49. 66. Paus, T., Keshavan, M. & Giedd, J. N. Why do many psychiatric disorders emerge during adolescence? Nature Rev. Neurosci. 9, 947–957 (2008). A very interesting review on the state of research into why adolescents have a greater vulnerability to mental health disorders. 67. Evans, G. W. & English, K. The environment of poverty: multiple stressor exposure, psychophysiological stress, and socioemotional adjustment. Child Dev. 73, 1238–1248 (2002). 68. Andersen, S. L. & Teicher, M. H. Stress, sensitive periods and maturational events in adolescent depression. Trends Neurosci. 31, 183–191 (2008). 69. De Bellis, M. D. et al. A. E. Bennett Research Award. Developmental traumatology. Part II: brain development. Biol. Psychiatry 45, 1271–1284 (1999). One of the first clear demonstrations that, in children who were physically healthy at birth, severe abuse in the early years of life is associated with reduced brain volume. The reduction correlates negatively with the age of onset and positively with the duration of the maltreatment. 70. Cohen, R. A. et al. Early life stress and morphometry of the adult anterior cingulate cortex and caudate nuclei. Biol. Psychiatry 59, 975–982 (2006). 71. Diamond, D. M., Bennett, M. C., Fleshner, M. & Rose, G. M. Inverted-U relationship between the level of peripheral corticosterone and the magnitude of hippocampal primed burst potentiation. Hippocampus
  • 50. 2, 421–430 (1992). 72. Vouimba, R. M., Yaniv, D. & Richter-Levin, G. Glucocorticoid receptors and β-adrenoceptors in basolateral amygdala modulate synaptic plasticity in hippocampal dentate gyrus, but not in area CA1. Neuropharmacology 52, 244–252 (2007). 73. Roozendaal, B., Brunson, K. L., Holloway, B. L., McGaugh, J. L. & Baram, T. Z. Involvement of stress- released corticotropin-releasing hormone in the basolateral amygdala in regulating memory consolidation. Proc. Natl Acad. Sci. USA 99, 13908–13913 (2002). 74. Magarinos, A. M. & McEwen, B. S. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory amino acid receptors. Neuroscience 69, 89–98 (1995). 75. Conrad, C. D., LeDoux, J. E., Magarinos, A. M. & McEwen, B. S. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behav. Neurosci. 113, 902–913 (1999). 76. Gould, E., McEwen, B. S., Tanapat, P., Galea, L. A. & Fuchs, E. Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J. Neurosci. 17, 2492–2498 (1997). 77. McEwen, B. S. Effects of adverse experiences for brain structure and function. Biol. Psychiatry 48, 721–731 (2000).
  • 51. 78. Pham, K., Nacher, J., Hof, P. R. & McEwen, B. S. Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. Eur. J. Neurosci. 17, 879–886 (2003). 79. McEwen, B. S. Plasticity of the hippocampus: adaptation to chronic stress and allostatic load. Ann. NY Acad. Sci. 933, 265–277 (2001). 80. Luine, V., Villegas, M., Martinez, C. & McEwen, B. S. Repeated stress causes reversible impairments of spatial memory performance. Brain Res. 639, 167–170 (1994). 81. Joels, M., Karst, H., Krugers, H. J. & Lucassen, P. J. Chronic stress: implications for neuronal morphology, function and neurogenesis. Front. Neuroendocrinol. 28, 72–96 (2007). 82. Izquierdo, A., Wellman, C. L. & Holmes, A. Brief uncontrollable stress causes dendritic retraction in infralimbic cortex and resistance to fear extinction in mice. J. Neurosci. 26, 5733–5738 (2006). 83. Shansky, R. M., Hamo, C., Hof, P. R., McEwen, B. S. & Morrison, J. H. Stress-induced dendritic remodeling in the prefrontal cortex is circuit specific. Cereb. Cortex 4 Feb 2009 (doi:10.1093/cercor/bhp003). 84. Cerqueira, J. J. et al. Corticosteroid status influences the volume of the rat cingulate cortex - a magnetic resonance imaging study. J. Psychiatr. Res. 39, 451–460 (2005).
  • 52. 85. Mitra, R., Jadhav, S., McEwen, B. S., Vyas, A. & Chattarji, S. Stress duration modulates the spatiotemporal patterns of spine formation in the basolateral amygdala. Proc. Natl Acad. Sci. USA 102, 9371–9376 (2005). 86. Mitra, R. & Sapolsky, R. M. Acute corticosterone treatment is sufficient to induce anxiety and amygdaloid dendritic hypertrophy. Proc. Natl Acad. Sci. USA 105, 5573–5578 (2008). This interesting study addressed endocrine effects on the brain, with a focus on the amygdala and anxiety (rather than on hippocampus and memory). Of note, a single dose of glucocorticoids was sufficient to induce changes in amygdala structure 10 days later, which might be useful to model in animals PTSD. 87. Lupien, S. J. & McEwen, B. S. The acute effects of corticosteroids on cognition: integration of animal and human model studies. Brain Res. Brain Res. Rev. 24, 1–27 (1997). 88. Roozendaal, B. Glucocorticoids and the regulation of memory consolidation. Psychoneuroendocrinology 25, 213–238 (2000). 89. Lupien, S. J. et al. Stress hormones and human memory function across the lifespan. Psychoneuroendocrinology 30, 225–242 (2005). 90. Lupien, S. J. et al. Hippocampal volume is as variable in young as in older adults: implications for the notion of hippocampal atrophy in humans. Neuroimage 34, 479–485 (2007). This study showed that ~25% of young adults
  • 53. present hippocampal volumes as small as those of older adults. The presence of small hippocampal volumes in healthy young individuals supports the vulnerability hypothesis. 91. Pruessner, J. C., Lord, C., Meaney, M. & Lupien, S. Effects of self-esteem on age-related changes in cognition and the regulation of the hypothalamic-pituitary-adrenal axis. Ann. NY Acad. Sci. 1032, 186–194 (2004). 92. Pruessner, J. C. et al. Self-esteem, locus of control, hippocampal volume, and cortisol regulation in young and old adulthood. Neuroimage 28, 815–826 (2005). 93. Burke, H. M., Davis, M. C., Otte, C. & Mohr, D. C. Depression and cortisol responses to psychological stress: a meta-analysis. Psychoneuroendocrinology 30, 846–856 (2005). 94. Yehuda, R., Golier, J. A. & Kaufman, S. Circadian rhythm of salivary cortisol in Holocaust survivors with and without PTSD. Am. J. Psychiatry 162, 998–1000 (2005). 95. Meewisse, M. L., Reitsma, J. B., de Vries, G. J., Gersons, B. P. & Olff, M. Cortisol and post-traumatic stress disorder in adults: systematic review and meta- analysis. Br. J. Psychiatry 191, 387–392 (2007). This paper presented the first meta-analysis of cortisol findings in PTSD, to elucidate the determinants of hypocortisolism and resolve the inconsistency in findings. 96. Heim, C. et al. Pituitary-adrenal and autonomic responses to stress in women after sexual and physical
  • 54. abuse in childhood. JAMA 284, 592–597 (2000). 97. Heim, C., Mletzko, T., Purselle, D., Musselman, D. L. & Nemeroff, C. B. The dexamethasone/corticotropin- releasing factor test in men with major depression: role of childhood trauma. Biol. Psychiatry 63, 398–405 (2008). 98. Carpenter, L. L. et al. Cerebrospinal fluid corticotropin- releasing factor and perceived early-life stress in depressed patients and healthy control subjects. Neuropsychopharmacology 29, 777–784 (2004). 99. Heim, C., Newport, D. J., Mletzko, T., Miller, A. H. & Nemeroff, C. B. The link between childhood trauma and depression: insights from HPA axis studies in humans. Psychoneuroendocrinology 33, 693–710 (2008). A crucially important review which documents that the disturbances in the HPA axis that are observed in many adults with depression may be specific to those who experienced trauma or maltreatment in childhood. 100. Videbech, P. & Ravnkilde, B. Hippocampal volume and depression: a meta-analysis of MRI studies. Am. J. Psychiatry 161, 1957–1966 (2004). 101. Smith, M. E. Bilateral hippocampal volume reduction in adults with post-traumatic stress disorder: a meta- analysis of structural MRI studies. Hippocampus 15, 798–807 (2005). 102. Vythilingam, M. et al. Childhood trauma associated with smaller hippocampal volume in women with major depression. Am. J. Psychiatry 159, 2072–2080 (2002).
  • 55. 103. Gilbertson, M. W. et al. Smaller hippocampal volume predicts pathologic vulnerability to psychological trauma. Nature Neurosci. 5, 1242–1247 (2002). The first paper to study whether the reduced hippocampal volume observed in PTSD patients is due to the disorder, to trauma exposure or to a pre-existing factor. 104. Issa, A. M., Rowe, W., Gauthier, S. & Meaney, M. J. Hypothalamic-pituitary-adrenal activity in aged, cognitively impaired and cognitively unimpaired rats. J. Neurosci. 10, 3247–3254 (1990). 105. Landfield, P. W., Waymire, J. C. & Lynch, G. Hippocampal aging and adrenocorticoids: quantitative correlations. Science 202, 1098–1102 (1978). 106. Landfield, P. W., Baskin, R. K. & Pitler, T. A. Brain aging correlates: retardation by hormonal- pharmacological treatments. Science 214, 581–584 (1981). The first study to show that chronic exposure to high levels of glucocorticoids in rodents is associated with memory impairments and reduced hippocampal volume. 107. Landfield, P. W., Blalock, E. M., Chen, K. C. & Porter, N. M. A new glucocorticoid hypothesis of brain aging: implications for Alzheimer’s disease. Curr. Alzheimer Res. 4, 205–212 (2007). 108. Kulstad, J. J. et al. Effects of chronic glucocorticoid administration on insulin-degrading enzyme and amyloid-β peptide in the aged macaque.
  • 56. J. Neuropathol. Exp. Neurol. 64, 139–146 (2005). 109. Sapolsky, R. M., Krey, L. C. & McEwen, B. S. The neuroendocrinology of stress and aging: the glucocorticoid cascade hypothesis. Endocr. Rev. 7, 284–301 (1986). The first paper to present the glucocorticoid cascade hypothesis, now referred to as the neurotoxicity hypothesis. 110. Lowy, M. T., Wittenberg, L. & Yamamoto, B. K. Effect of acute stress on hippocampal glutamate levels and spectrin proteolysis in young and aged rats. J. Neurochem. 65, 268–274 (1995). 111. Raskind, M. A., Peskind, E. R. & Wilkinson, C. W. Hypothalamic-pituitary-adrenal axis regulation and human aging. Ann. NY Acad. Sci. 746, 327–335 (1994). 112. Lupien, S. J. et al. Cortisol levels during human aging predict hippocampal atrophy and memory deficits. Nature Neurosci. 1, 69–73 (1998). 113. Giubilei, F. et al. Altered circadian cortisol secretion in Alzheimer’s disease: clinical and neuroradiological aspects. J. Neurosci. Res. 66, 262–265 (2001). 114. Aisen, P. S. et al. A randomized controlled trial of prednisone in Alzheimer’s disease. Alzheimer’s Disease Cooperative Study. Neurology 54, 588–593 (2000). 115. Dai, J., Buijs, R. & Swaab, D. Glucocorticoid hormone (cortisol) affects axonal transport in human cortex neurons but shows resistance in Alzheimer’s disease.
  • 57. Br. J. Pharmacol. 143, 606–610 (2004). 116. Chen, Y., Dube, C. M., Rice, C. J. & Baram, T. Z. Rapid loss of dendritic spines after stress involves derangement of spine dynamics by corticotropin- releasing hormone. J. Neurosci. 28, 2903–2911 (2008). 117. Charney, D. S. & Manji, H. K. Life stress, genes, and depression: multiple pathways lead to increased risk and new opportunities for intervention. Sci. STKE 2004, re5 (2004). 118. Maercker, A., Michael, T., Fehm, L., Becker, E. S. & Margraf, J. Age of traumatisation as a predictor of post-traumatic stress disorder or major depression in young women. Br. J. Psychiatry 184, 482–487 (2004). 119. Teicher, M. H., Tomoda, A. & Andersen, S. L. Neurobiological consequences of early stress and childhood maltreatment: are results from human and animal studies comparable? Ann. NY Acad. Sci. 1071, 313–323 (2006). 120. Hall, F. S. Social deprivation of neonatal, adolescent, and adult rats has distinct neurochemical and behavioral consequences. Crit. Rev. Neurobiol. 12, 129–162 (1998). R E V I E W S 444 | j u n E 2 0 0 9 | Vo lu M E 1 0 w w w.nature.com/reviews/neuro R E V I E W S
  • 58. © 2009 Macmillan Publishers Limited. All rights reserved 121. Andersen, S. L. Trajectories of brain development: point of vulnerability or window of opportunity? Neurosci. Biobehav. Rev. 27, 3–18 (2003). A superb review paper which suggested that trauma at different time points during early development might be associated with different outcomes, depending on the brain structure that was affected at the time of exposure to adversity. 122. Widom, C. S., DuMont, K. & Czaja, S. J. A prospective investigation of major depressive disorder and comorbidity in abused and neglected children grown up. Arch. Gen. Psychiatry 64, 49–56 (2007). 123. Clayton, N. S. & Krebs, J. R. Hippocampal growth and attrition in birds affected by experience. Proc. Natl Acad. Sci. USA 91, 7410–7414 (1994). 124. Kudielka, B. M., Buske-Kirschbaum, A., Hellhammer, D. H. & Kirschbaum, C. HPA axis responses to laboratory psychosocial stress in healthy elderly adults, younger adults, and children: impact of age and gender. Psychoneuroendocrinology 29, 83–98 (2004). 125. Kessler, R. C. Epidemiology of women and depression. J. Affect. Disord. 74, 5–13 (2003). 126. Harlow, B. L., Cohen, L. S., Otto, M. W., Spiegelman, D. & Cramer, D. W. Early life menstrual characteristics and pregnancy experiences among women with and without major depression: the Harvard study of moods
  • 59. and cycles. J. Affect. Disord. 79, 167–176 (2004). 127. Zabin, L. S., Emerson, M. R. & Rowland, D. L. Childhood sexual abuse and early menarche: the direction of their relationship and its implications. J. Adolesc. Health 36, 393–400 (2005). 128. Jones, K. C. & de Voogt, P. Persistent organic pollutants (POPs): state of the science. Environ. Pollut. 100, 209–221 (1999). 129. Centers for Disease Control and Prevention. Second National Report on Human Exposure to Environmental Chemicals. (CDC, Atlanta, Georgia, 2003). 130. Daston, G. P., Cook, J. C. & Kavlock, R. J. Uncertainties for endocrine disrupters: our view on progress. Toxicol. Sci. 74, 245–252 (2003). 131. Gump, B. B. et al. Low-level prenatal and postnatal blood lead exposure and adrenocortical responses to acute stress in children. Environ. Health Perspect. 116, 249–255 (2008). 132. Denham, M. et al. Relationship of lead, mercury, mirex, dichlorodiphenyldichloroethylene, hexachlorobenzene, and polychlorinated biphenyls to timing of menarche among Akwesasne Mohawk girls. Pediatrics 115, e127–e134 (2005). 133. Turek, F. W. From circadian rhythms to clock genes in depression. Int. Clin. Psychopharmacol. 22 (Suppl. 2), S1–S8 (2007). 134. Lamarche, L. J. & De Koninck, J. Sleep disturbance
  • 60. in adults with posttraumatic stress disorder: a review. J. Clin. Psychiatry 68, 1257–1270 (2007). 135. Antoch, M. P. et al. Functional identification of the mouse circadian Clock gene by transgenic BAC rescue. Cell 89, 655–667 (1997). 136. Yakovlev, P. L. & Lecours, A. R. in Regional Development of the Brain in Early Life (ed. Minkowski, A.) 3–70 (Blackwell, Oxford, 1967). 137. Pruessner, J. C. et al. Volumetry of hippocampus and amygdala with high-resolution MRI and three- dimensional analysis software: minimizing the discrepancies between laboratories. Cereb. Cortex 10, 433–442 (2000). 138. Tisserand, D. J. et al. Regional frontal cortical volumes decrease differentially in aging: an MRI study to compare volumetric approaches and voxel- based morphometry. Neuroimage 17, 657–669 (2002). 139. Insel, T. R., Battaglia, G., Fairbanks, D. W. & De Souza, E. B. The ontogeny of brain receptors for corticotropin-releasing factor and the development of their functional association with adenylate cyclase. J. Neurosci. 8, 4151–4158 (1988). 140. Levine, S. The ontogeny of the hypothalamic-pituitary-adrenal axis. The influence of maternal factors. Ann. NY Acad. Sci. 746, 275–288; discussion 289–293 (1994). 141. Gunnar, M. R. & Cheatham, C. L. Brain and behavior
  • 61. interfaces: stress and the developing brain. Infant Ment. Health J. 24, 195–211 (2003). A superb paper that summarized the effects of stress during development and how this knowledge can be used to develop effective interventions. 142. LeDoux, J. E. The Emotional Brain: The Mysterious Underpinnings of Emotional Life (Simon & Schuster, New York, 1996). Acknowledgements Sonia Lupien holds a Research Chair on Gender and Mental Health by the Canadian Institutes of Health Research. FURTHER INFORMATION Sonia J. Lupien’s homepage: http://www.humanstress.ca All liNks Are AcTive iN The oNliNe pdf R E V I E W S nATuRE REVIEwS | N e u r o s c i e N c e Vo lu M E 1 0 | j u n E 2 0 0 9 | 445 f o c u S o n S t R E S S © 2009 Macmillan Publishers Limited. All rights reserved http://www.humanstress.caEffects of stress throughout the lifespan on the brain, behaviour and cognitionMainAcknowledgementsReferences University of Missouri – St. Louis
  • 62. Department of Accounting ACCTNG 4435 – Auditing – Summer 2021 Writing Assignment – Risk Assessment and Management Assertions at the Crafty Trinkets Company (“CTC”) Extension of due date – the Writing Assignment is now due by 1159 PM on 2 July. Required: Please read the following information about the Crafty Trinkets Company (“CTC” or “the Company”), and follow the instructions at the end of the case. The requirements of the case are focused and to-the-point. While this case assignment is not intended to be some sort of massive research paper, it does account for 10% of your final grade. Please spend the time and attention on this
  • 63. assignment that 10% of your final grade would warrant. Take the time to present well- thought out responses to the requirements, written in your best, polished writing style. Please proofread your paper. If your paper appears to have been dashed off in a few minutes, unfortunately your grade on the paper will reflect that. Do a professional job. When reading the case, please pay careful attention to the dates. Imagine it is now 28 November 2018. It may be helpful to create a timeline of events, so you can put the events in the proper context and chronology. (Please don’t include any chronology that you prepare in your submission.) Please note that, given the dates cited below, none of the events in the case are impacted by the covid pandemic. I’m happy to answer any questions you have about the case or the requirements. Introduction As a senior auditor in a public accounting firm, you have been assigned
  • 64. to plan the audit of the financial statements of a privately-held company called the Crafty Trinkets Company (“CTC” or “the Company”). Company Background CTC designs, manufactures, and markets a variety of toys, which are sold primarily to large national retailers like Target and Wal-Mart. CTC is a small company compared to competitors like Mattel and Hasbro; nevertheless, CTC managers believe their company’s toys are among the best in the world. Unlike the larger toy makers, CTC has enjoyed success with a small portfolio of brands and products, representing three categories: (1) soft toys, consisting primarily of its Snuggle Pets stuffed animals; (2) sturdy toys, including metal-cast and plastic cast toys like Speedster cars and Lightning action figures; and (3) digital toys, consisting of video game software under development. Like most toy makers, 60 percent of CTC’s sales revenues are generated in October and November, with the last two weeks
  • 65. of November driving half of those sales. Your firm, Smith and Company, LLP (“Smith” or “the Firm”), has been CTC’s public accounting firm since 2013, providing audit and tax services to the Company. The primary external user of CTC’s audited financial statements is its bank. Assume it is now 28 November 2018. You have taken over audit senior responsibilities for the Company’s 30 November 2018 year-end financial statement audit because the previous audit senior has just unexpectedly left your Firm to accept a job in another city. (The Company’s year-end date is 30 November.) Please review the following excerpts from relevant documents involving the planning and execution of the current year audit. Excerpts from the document…. “Observations Noted in My Review of the
  • 66. Previous Year (year-end 30 November 2017) Audit File” (written by you, the new senior auditor…) • In fiscal 2017, CTC exceeded its earning targets, reporting operating income of $3,026,100 and net income before taxes of $2,572,800. The only large negatives for the year 2017 were the substantial additions to allowances for receivables and inventories, including an extra $300,000 in the allowance for doubtful accounts related to the struggling Toy-Mart chain in the United States. The increase to the allowance for inventory was due to possible obsolete inventory. The increase in the Allowance for Doubtful Accounts was accomplished by a debit to Bad Debt Expense and a credit to the Allowance. • CTC’s management advised our Firm that retailers dramatically reduced the quantity of toys they were willing to stock on its shelves at any one time in fiscal 2017, and were expected to continue this trend into 2018. This change did not (at
  • 67. least in 2017) reduce the volume of the toys sold through retailers, but it has intensified competition among industry competitors for retail shelf space, and increased operating costs by increasing the frequency of shipments to stores. (In the competitive environment in which CTC operates, shipping costs are usually paid by the manufacturer, not the retailer.) It’s possible that the restrictions on shelf-space could result in lower sales in future periods. • Ever since 2014, CTC executives have shared in a bonus pool that is created through CTC contributions of 10% of the first $750,000 of operating income, plus 20% on the next $750,000, and an additional 30% of the next $1,500,000. CTC’s total contributions to the bonus pool are capped at a yearly maximum of $675,000. • CTC does not have an internal audit group. In addition, they have struggled to
  • 68. implement the COSO Integrated Framework of Internal Control. Excerpts from the document…. “Findings from Visit to Client and Interim (before year-end) Audit Procedures Conducted in September 2018” (written by the audit senior that you replaced) • In January 2018, the long-time CEO and CFO of CTC retired, and replacements were hired and began work in March 2018. One senior manager told me that the pair are like “fire-breathing dragons,” and have indicated that their “sole number-one focus will be increasing sales and profitability, and those who do not contribute will be given the opportunity to continue their career elsewhere.” • We developed an understanding of controls over purchases and payable and found that controls in this area were not well designed and were not operating effectively. Therefore, following what I know about audit
  • 69. strategy, I decided to test the controls over purchases and payables in the interim period and assigned it to one of the staff auditors. Although this testing has not yet been reviewed, one item seemed unusual. It involved a payment of $30,000 to the International Toy Manufacturer Workers Union. The payment was initiated by the CTC VP- Operations and approved by the current CFO, and was properly classified as a non-operating expense. According to the VP-Operations, the payment was “a gesture of support for the toy factory workers – a gesture we believe is important since workers believe themselves to be underpaid and are discussing the possibility of work stoppages and strikes in the Fall of 2018. We hope this payment will assist in making it possible for union executives to encourage their members to resolve these issues before a work stoppage or strike.” • In the tests of controls over revenues and receivables, one of
  • 70. the staff auditors that conducted the testing noted that controls were effective. In particular the staff member noted that one thing that was very impressive was that the CFO was active in oversight of the area of bad debts and inventory obsolescence. Indeed, as an example, the current CFO herself approved the reversal/recovery of the $300,000 amount allowed for with respect to Toy-Mart, and had even initiated and approved the journal entry for the transaction, reversing it into income (debit Allowance for Doubtful Accounts, credit Bad Debt Expense) without the involvement of the credit manager. Excerpts from the document…. “Audit Partner Memo to 30 November 2018 Audit Workpaper File” • CTC had been unable to produce enough Snuggle Pets for the
  • 71. December 2017 year-end holiday season, due to raw material shortages in an unstable stuffing supplier market. The Company was able, however, to increase production in January 2018, which allowed for increased sales for Valentine’s Day in February 2018. Soon after, at the insistence of the national retailers, all unsold Snuggle Pets were returned to CTC for a full refund. The retailers insisted that the absence of Snuggle Pets in stores after February 2018 would build demand for the Fall/Winter 2018-2019, as the retailers focused on the end-of- the-year holidays and gift-giving season. • CTC has deferred their purchase of new, hi-tech manufacturing equipment due to a shortage of cash and the inability to obtain favorable financing. This is the second year in a row that CTC has deferred this investment. • CTC executives entered into an agreement with Cartoon Studios, Inc., who had
  • 72. produced their very first animated movie, called “The Bronx Zoo – Escape to Manhattan!” for release on 30 November 2018. (The movie is billed as “Jumanji” meets “Babe: Pig in the City”!) For $900,000, CTC had won the rights to produce a line of soft and plastic toys based on characters from the movie. (CTC plans to amortize this fee over 9 years.) The toys would be sold through CTC’s regular retail customers. The toys were on schedule to be in stores on 30 November. The agreement between the Company and Cartoon Studios indicated that Cartoon Studios would compensate CTC for the cost of the unsold toys if sales of the toys failed to reach $1,500,000 during the first two months after the movie’s release. CTC plans to accrue $1,500,000 of sales revenue on 30 November relating to this provision of the agreement. • CTC executives have carefully reviewed the pricing and valuation of inventory during early November 2018, and determined that the inventory
  • 73. valuation reserve established in the previous year is no longer necessary; a journal entry was made by the CFO on 15 November 2018 to reverse the valuation allowance into operating income in a manner similar to the reversal related to the Allowance for Doubtful Accounts described above. • In October 2018, CTC announced that it was suspending it’s partnership with the charitable organization “Toys for Kids,” an organization that distributes toys to underprivileged children in less-developed countries around the world. In the past, CTC had donated a substantial number of toys to “Toys for Kids.” • On November 1, 2018, the Company’s Board of Directors Compensation Committee agreed to double the Company’s contribution to the bonus pool, to $1,350,000. This measure will be effective for the year-ending 30 November 2018.