Anda di halaman 1dari 10

review

focus on stress

Stress weakens prefrontal networks:


molecular insults to higher cognition
Amy F T Arnsten

A variety of cognitive disorders are worsened by stress exposure and involve dysfunction of the newly evolved prefrontal cortex
(PFC). Exposure to acute, uncontrollable stress increases catecholamine release in PFC, reducing neuronal firing and impairing
cognitive abilities. High levels of noradrenergic a1-adrenoceptor and dopaminergic D1 receptor stimulation activate feedforward
calciumprotein kinase C and cyclic AMPprotein kinase A signaling, which open potassium channels to weaken synaptic efficacy
in spines. In contrast, high levels of catecholamines strengthen the primary sensory cortices, amygdala and striatum, rapidly
2015 Nature America, Inc. All rights reserved.

flipping the brain from reflective to reflexive control of behavior. These mechanisms are exaggerated by chronic stress exposure,
where architectural changes lead to persistent loss of PFC function. Understanding these mechanisms has led to the successful
translation of prazosin and guanfacine for treating stress-related disorders. Dysregulation of stress signaling pathways by genetic
insults likely contributes to PFC deficits in schizophrenia, while age-related insults initiate interacting vicious cycles that increase
vulnerability to Alzheimers degeneration.

Exposure to uncontrollable stress rapidly evokes chemical changes striatum, hypothalamus and brainstem for control of the autonomic
in brain that impair the higher cognitive functions of the PFC while nervous system2 (Fig. 1b). These PFC areas, along with the insular
strengthening primitive brain reactions. This flip from reflective to cortex, are thought to be critical for the mental suffering aspects of
reflexive brain state may have survival value when we are in danger, pain3. These areas receive projections from more rostral and lateral
but it can be ruinous for life in the Information Age, when we need PFC, providing opportunities for the integration of cognitive and
higher cognitive abilities to thrive. It has been appreciated for decades emotional processing. PFC circuits are usually positioned to either
that uncontrollable stress drives mental illness, including cognitive facilitate or inhibit processing, thus allowing flexible, top-down con-
disorders such as schizophrenia, and new evidence suggests it may trol. Data from humans suggest that the right hemisphere may be
also contribute to the cognitive deterioration of Alzheimers disease. particularly important for inhibitory control3.
These disorders particularly afflict the most newly evolved pyramidal The topographic organization of the PFC in humans is reflected in
cell circuits in association cortex, circuits that are uniquely regulated the sites of dysfunction in neuropsychiatric disorders. For example,
at the molecular level. The following reviews the effects of stress on there is loss of dlPFC gray matter in schizophrenia4,5 and Alzheimers
npg

PFC circuits and its relevance to degenerative changes in stress-related disease6, while changes in more ventral and medial PFC regions are
cognitive disorders. evident in mood disorders7 and in post-traumatic-stress disorder
(PTSD)8. In bipolar disorder, the disinhibitory symptoms of mania
The newly evolved prefrontal cortex are associated with dysfunction of the right hemisphere7, consistent
The evolution and organization of the PFC. The PFC subserves our with the specialized inhibitory role of this hemisphere.
highest order cognitive abilities, generating the mental representa- The integrity of dlPFC function is often tested in working mem-
tions that are the foundation of abstract thought and the basis for ory tasks, where information must be held in mind and constantly
flexible, goal-directed behavior. In primates, the PFC is topographi- updated to guide accurate, flexible responding. Studies of nonhuman
cally organized: the dorsolateral PFC (dlPFC) guides thoughts, atten- primate PFC have shown that the pyramidal cell microcircuits that
tion and actions1, while the orbital and ventromedial PFC (vmPFC) subserve visual spatial working memory reside in deep layer III of
regulate emotion2 (Fig. 1a). The dlPFC has extensive connections the dlPFC1 (Fig. 2). These are the circuits that have expanded most
with the association cortices and the dorsal aspects of the striatum1 in mammalian evolution, with increasing numbers of basal dendrites
for the regulation of thought and action. In contrast, the most caudal and spines9. This huge increase in dendritic spines allows the extraor-
and medial aspects of the PFC (for example, Brodmanns areas 24 and dinary number of neural connections needed for high-order cogni-
25, also called the anterior cingulate cortex and the subgenual cortex, tion, where representations of representations expand the repertoire
respectively) project to limbic structures such as the amygdala, ventral of cognitive abilities9.

Microcircuits for the generation of mental representations in


Department of Neurobiology, Yale University School of Medicine,
New Haven, Connecticut, USA. Correspondence should be addressed primate dlPFC. Delay cells in the primate dlPFC are able to gener-
to A.F.T.A. (amy.arnsten@yale.edu). ate mental representations in the absence of sensory stimulation1:
Received 17 April; accepted 15 July; published online 25 September 2015; for example, the representation of the 90 direction from a central
doi:10.1038/nn.4087 fixation point (Fig. 2). This persistent firing across a delay period

1376 VOLUME 18 | NUMBER 10 | OCTOBER 2015 nature neuroscience


review

Figure 1 Changes in brain systems controlling


behavior under conditions of alert safety
a Alert b Stressed

versus uncontrollable stress. (a) Under


conditions when a subject feels alert, safe PFC PFC
and interested, phasic release of Top down
Basal ganglia
regulation of:
catecholamines strengthens the higher Habitual responses
Thought
cognitive functioning of the PFC, thus
allowing top-down regulation of thought, Action
Sensory cortex
action and emotion. In primates, Emotion Amygdala
the PFC is topographically organized, with Conditioned
the dorsal and lateral surfaces mediating emotional
PAG responses
attention, thought and action while the LC
Reflexive/habitual
ventral and medial aspects mediate emotion. responding by more
The anatomical projections of these areas reflect these primitive brain circuits
specializations. (b) During stress exposure, high levels
To autonomic
of catecholamines take the PFC off-line while strengthening the functions of more primitive circuits nervous system
for example, the conditioned emotional responses of the amygdala and the habitual actions of the basal ganglia.
The amygdala activates brainstem stress systems, which in turn activate the sympathetic nervous system.

arises from the recurrent excitation of pyramidal cells with shared A variety of stressors have been used to observe how stress affects
spatial tuningfor example, a group of cells that all receive informa- functioning in the rodent brain. Early studies often used restraint
tion from the parietal association cortex for the location 90, their stress23 and/or inescapable shock18, as well as conditioned fear (for
preferred direction. The spatial tuning of delay cells is refined by example, a tone previously paired with shock)24. Biochemical and
2015 Nature America, Inc. All rights reserved.

lateral inhibition from GABAergic interneurons (Fig. 2). Pyramidal then behavioral studies also used a pharmacological stressor, FG7142,
cells interconnect on dendritic spines through glutamatergic NMDA a benzodiazepine inverse agonist (that is, a compound with an action
receptor (NMDAR) type NR2B synapses10 (Fig. 3). The permissive opposite to that of Valium) that generates a classic glucocorticoid
depolarization of the postsynaptic density needed for NMDAR open- response and increases catecholamine release in the PFC19. Studies of
ing is provided by cholinergic stimulation of nicotinic 7 receptors stress effects in monkeys as well have employed FG7142, or loud white
in the postsynaptic density11 with only minor contributions from noise, a stressor used in early studies of humans20. More recent stress
AMPA-type glutamate receptors (AMPAR)10, consistent with the research in humans has employed a variety of stressors, including
lower expression of AMPAR in layer III (ref. 12). social stress, watching an upsetting video and listening to an account
The functional strength of these NMDAR synapses is dynami- of stressful effects in ones own life.
cally modulated to rapidly enhance or weaken connections and thus Exposure to an acute, uncontrollable stress impairs the performance
helps to shape the contents and strength of working memory (Fig. 3). of PFC cognitive tasks in rodents, monkeys and humans25. For example,
These very rapid changes in synapse strength, called dynamic network rats exposed to either 2 h of restraint stress or administered the phar-
connectivity13, are mediated by feedforward, cAMPCa2+ signaling, macological stressor, FG7142, are impaired on the spatial delayed
which opens K+ channels (HCN, KCNQ) near the synapse to weaken alternation task, a test of spatial working memory that depends on
the connection. A constellation of cAMP-related proteins are observed the medial PFC19,26. Performance of this task also requires decision-
next to the Ca2+-containing spine apparatus, where they can increase making capabilities and the ability to inhibit a prepotent but inap-
or decrease feedforward, cAMPCa2+ signaling14 (Fig. 3) propriate response, functions linked to the PFC. Stressed rats make
The dlPFC also contains response cells, neurons that fire just before more perseverative errors on the task, consistent with the inflexible
npg

or during the motor response (Fig. 2). These neurons are modulated behavior patterns that often occur under conditions of PFC dysfunc-
in a more classical mannerfor example, with a reliance on AMPAR tion19. In contrast, the performance of a visual spatial discrimination
actions10consistent with the higher expression of AMPAR in layer V task with similar sensory, motor and motivational demands, but no
of monkey dlPFC12. Layer V response-like cells appear to be the type of need for PFC abilities, is unchanged by stress exposure 19. A similar
neuron most common in rodent PFC15. Thus, even within the dlPFC, pattern is seen in monkeys, where acute exposure to loud white noise
delay cells have distinct molecular signatures compared to surrounding stress20 or FG7142 (ref. 19) impairs performance of a spatial delayed
neurons that make them especially vulnerable to stress exposure. response working memory task, but has no effect on performance of a
spatial discrimination task. Human subjects exposed to an acute social
Acute stress exposure rapidly impairs higher PFC functions in stress also exhibit impairments in working memory and attention, for
animals and humans example27, indicating that this effect is found across many species.
Exposure to uncontrollable stress impairs the higher cognitive The effects of acute stress on hippocampal physiology and function
functions of the PFC. The study of stress effects on cognitive abilities are more complex. Acute stress appears to enhance hippocampus-
began after the Second World War, when it was realized that highly dependent fear-related memory consolidation (for example, contex-
skilled pilots crashed their planes in the stress of battle as a result of tual fear conditioning), but impairs spatial learning that is unrelated to
mental errors (reviewed in ref. 16). A key aspect of these findings the fear-inducing conditions28. The severity of the acute stressor also
was that the subject had to feel a lack of control over the stressor17, appears to influence whether hippocampal physiology is affected. In
a factor also found in animal studies18. Later research in animals many studies, acute, mild restraint stress had subtle or no effects on
demonstrated that exposure to acute, uncontrollable stress impairs LTP23, or if brief, could even enhance LTP29. However, the addition
the working memory abilities of the PFC19,20, while tasks that rely of inescapable shocks to the restraint paradigm impairs LTP28. As
on the habitual functions of basal ganglia circuits, for example20,21, restraint stress alone is sufficient to impair the spatial working memory
or the emotional conditioning of the amygdala22 are spared or even functions of the PFC26, it appears that the hippocampus is less
enhanced by stress exposure (Fig. 1b). sensitive to impairment by acute stress exposure than is the PFC.

nature neuroscience VOLUME 18 | NUMBER 10 | OCTOBER 2015 1377


Review

Figure 2 The cellular basis of working memory, as a


ODR task b
discovered by Goldman-Rakic. (a) The oculomotor Cue
delayed response (ODR) task. A monkey fixates 0
45 315 Visual spatial information from
on a central spot while a cue is briefly lit at one of dlPFC physiology and microcircuitry
parietal association cortex
eight locations. The monkey must remember that 90 270
location over a delay period while maintaining I
fixation. At the end of the delay, the fixation point 135 225
is extinguished and the monkey can move its eyes 180
II
to the remembered location for juice reward. The 0.5 s
cue location constantly changes over hundreds of
Delay Delay cells
trials, requiring the constant updating of working
memory. (b) The physiology and microcircuitry III
45 0 315
G
of the primate dlPFC. Delay cells maintain 90 270
90
90
45
45
135 180 225
persistent firing across the delay period for their 90 D1R
45

preferred location, but not other locations. The


persistent firing is generated by the recurrent NMDA
excitation of pyramidal cells with shared preferred NR2B
IV
directions, likely receiving their information from Perisaccadic
area 7 of the parietal association cortex. These 2.57 s Response cells
G Peri-
pyramidal cells excite each other via NMDAR
NR2B synapses on spines; there are only subtle Cue Delay Response D2R
V
influences of AMPARs. The spatial tuning of delay Respond
NMDA Post-
To motor system D2R
cells is enhanced via lateral inhibition from GABA
(G) interneurons. Delay-cell microcircuits reside Corollary discharge AMPA
2015 Nature America, Inc. All rights reserved.

in deep layer III and possibly superficial layer V. Postsaccadic


VI
Delay cells are modulated by dopamine actions
at D1R but not D2R. In contrast, response cells
are modulated by D2R but not D1R and likely Cue Delay Response Feedback from basal
reside in layer V. Perisaccadic response cells fire ganglia/motor circuits
via thalamus
immediately before the motor response and likely
convey orders to the motor system, while postsaccadic response cells convey feedback (corollary discharge) about the response. Some response cells show
both pre- and postsaccadic firing; that is, both motor and feedback characteristics. Postsaccadic response cell firing relies on AMPAR as well as NMDAR
stimulation. Response cells are what are most common in rodent PFC, which has a very large layer V.

With the advent of brain imaging, stress studies have now begun catecholamine actions35 (see below), accentuating how chemical
to examine the neural circuit activity altered by acute stress in changes during acute stress exposure can switch neural orchestration
humans30,31. Functional MRI studies have shown that listening to of behavior from top-down to more primitive brain states (Fig. 1b).
a stressful account of ones own life, compared to listening to a neu-
tral passage, increases the blood oxygen leveldependent (BOLD) Relevance to mental disorders. It has been appreciated for many years
response in the medial PFC (anterior cingulate cortex), especially in that stress exacerbates mental illness36for example, the initial
the right hemisphere30. These results are consistent with the role of descent into schizophrenia37 or the switch from euthymia to illness in
the anterior cingulate in processing mental suffering3. Studies have bipolar disorder38. Prolonged or traumatic stress exposure can lead to
also shown evidence of acute stress impairing dlPFC function in depression or PTSD, disorders that are more prevalent in women39,40.
npg

humans. Subjects who watched an upsetting video showed impaired Data from animal studies indicate that estrogen can exaggerate stress-
performance of an N-back working memory task and reduced BOLD induced PFC dysfunction in female rats26,41. Similar mechanisms in
activity over the dlPFC31.This study also found that acute stress expo- humans may contribute to the increased vulnerability of women of
sure diminished the normal deactivation of the default mode network, cycling age for stress-induced mental disorders42. Notably, there is
including relative increases in the BOLD signal in the vmPFC and recent evidence that women exposed to serious stressors in middle
insula, circuits that normally deactivate during cognition and activate age have an increased incidence of Alzheimers disease 20 years later43.
with stress31. The stress-induced impairment in working memory This study is consistent with others showing that distress may hasten
performance and reduction in dlPFC activity were particularly evi- dementia44. Thus, stress exposure may increase risk of a variety of
dent in subjects with greater catecholamine actions; that is, in those mental or cognitive disorders.
subjects with a methionine substitution in the catabolic enzyme,
COMT, which weakens catecholamine degradation. These results are Rapid molecular events with acute stress exposure
consistent with stress-induced catecholamine release impairing dlPFC Increased catecholamine release in PFC. Exposure to acute,
working memory function32 (see below). Stress-induced impairment uncontrollable stress induces a number of chemical changes in brain
of working memory during an N-back task has also been linked to that rapidly impair PFC function. In addition to global increases in
electrophysiological signs of PFC dysfunction: cognitive impairment glucocorticoids, stress increases catecholamine release in PFC19,24,45.
correlated with reduced PFC theta activity33. In primates, even a very mild stress can activate the dopaminergic
In contrast to the impairments in dlPFC working memory, an salience neurons that respond to both aversive and rewarding
earlier study showed that watching an upsetting video enhanced the events46 and can increase dopamine release in dlPFC 47. Stress
memory consolidation of the emotionally charged events in the film34. also activates the noradrenergic neurons of the locus coeruleus
This improvement in memory consolidation correlated with increased via stimulation by the amygdala of corticotropin-releasing factor
activity in the amygdala while the subject watched the video34. (CRF) receptors on locus coeruleus neurons 48, increasing nore-
The increased activity in the amygdala also involved increased pinephrine release in PFC49. Indeed, a subset of locus coeruleus

1378 VOLUME 18 | NUMBER 10 | OCTOBER 2015 nature neuroscience


review

Figure 3 Dynamic network connectivity (DNC) in


DNC regulation of a layer III dlPFC glutamatergic synapse
the primate dlPFC. Layer III NMDAR synapses Dendrite
on spines in the primate dlPFC are powerfully
modulated by the arousal systems (acetylcholine Stress
(ACh), norepinephrine, dopamine). ACh has
permissive effects on NMDAR opening via

Marina Corral Spence/Nature Publishing Group


mGluR5
D1R 2A-AR
nicotinic 7 receptors (nic-7R) in the synapse. 1-AR? HCN KCNQ
1-AR? mGluR2/3
Feedforward Ca2+cAMP signaling, as driven by
stress exposure, can rapidly weaken synaptic
efficacy and network connectivity by opening K+ 2+ Ca2+ AC
Glutamatergic 2+ Ca
channels (HCN, KCNQ) near the synapse and in Ca AKAP6
axon terminal IP3R cAMP PDE4A-DISC1
the spine neck (red). Conversely, inhibition of PKA
Spine
feedforward Ca2+cAMP signaling strengthens
connections (green). The ultrastructural locations
of 1-AR and 1-AR in primate dlPFC are not yet NMDAR ACh Nic-7R
known. Asterisk indicates the spine apparatus, (NR2B)
the extension of the smooth endoplasmic
reticulum into the spine. AC, adenylyl cyclase.

neurons project selectively to PFC50, which may accentuate the stress requires further study. Physiological, behavioral and immunoelectron
response in this region. Catecholamine levels are further increased microscopic evidence suggest that these pathways interact: feedfor-
by glucocorticoids, which block the transporters on glia that ward Ca2+cAMP signaling opens nearby HCN and KCNQ K+ chan-
2015 Nature America, Inc. All rights reserved.

normally remove catecholamines from the extracellular space51. nels to weaken the efficacy of nearby NMDAR synaptic connections13.
These catecholamine actions may be increased in females by estrogen. This reduces the persistent firing of the dlPFC neurons that generate
For example, CRF activation of the locus coeruleus is accentuated in the mental representations needed for working memory and top-
females52 and dopamine in the PFC is increased by estrogen53, sug- down control. Conversely, inhibition of Ca2+PKC or cAMPPKA
gesting mechanisms that may underlie the increased vulnerability of signaling, or blockade of HCN channels, can rescue PFC delay-cell
females to stress exposure. firing and working memory functions57,60,62.
High levels of catecholamine release in PFC lead to cognitive In contrast to delay cells, which reduce firing with high levels of
deficits. For example, the degree of cognitive impairment during dopamine D1R stimulation, layer V sensory-motor response cells in
stress exposure correlates with levels of dopamine release in the rat dlPFC show increased firing with high levels of dopamine D2 recep-
PFC19. In both rats and monkeys, stress-induced PFC dysfunction tor stimulation63. As response cells are inhibited by delay cells during
can be blocked by dopamine D1 receptor (D1R) or norepinephrine the delay epoch, they also may become disinhibited as a result of loss
1-adrenoceptor (AR) antagonists19,54, and conversely, it can be of this top-down regulation. As layer V response-like cells appear
mimicked by high levels of D1R55 or 1-AR56,57 stimulation in PFC. to predominate in rodents, recordings from rodent PFC may give a
For example, infusion of an 1-AR agonist into the monkey dlPFC misleading view of what occurs in primate dlPFC, where the higher
or rat medial PFC produces a marked impairment in spatial working cognitive circuits in layer III show reduced rather than elevated levels
memory performance56. of firing with high levels of catecholamines.
Higher catecholamine levels have been linked to stress-induced In contrast to that in PFC, high levels of catecholamines strengthen
impairment of PFC function and changes in brain state in humans the affective responses of the amygdala22,64, the habitual or compul-
npg

as well. As mentioned above, those with a methionine substitution sive responses of the striatum65 and sensory processing in the primary
in COMT have weaker enzymatic activity and thus higher levels of somatosensory cortex66. Similarly, PKC signaling excites sensory
catecholamines. These people show much greater working memory processing in the barrel cortex67 and reinforces fear conditioning in
impairment and dlPFC hypoactivity during stress than those subjects the amygdala68. Glucocorticoids have been shown to accentuate the
with the more effective enzyme32. High levels of norepinephrine -AR effects of catecholamines in both the PFC and the amygdala69, thus
stimulation during acute stress increase the coupling of the vmPFC coordinating and exaggerating the switch from thoughtful to habitual
to subcortical limbic areas58 and enhance the memory consolidation responding during exposure to stress (Fig. 1).
processing of the amygdala35. High levels of norepinephrine com-
bined with glucocorticoids have also been shown to promote habitual Exaggeration of changes with chronic stress exposure
responding and reduce the sensitivity of the vmPFC to changes in out- Circuit-specific, architectural changes with chronic stress. Chronic
come value59. Thus, the importance of norepinephrine in switching stress exposure accentuates many of the effects of acute stress expo-
control from reflective, dlPFC circuits to more reflexive subcortical sure, as architectural changes exaggerate the switch from highly
circuits can be seen in humans as well as in animals. evolved to more primitive brain circuits. Sustained stress exposure
induces loss of dendrites and spines in layer II/III pyramidal cells of
Intracellular signaling pathways that weaken PFC function. We rodent PFC7073 and loss of the dendritic tufts of layer V pyramidal
have begun to understand the intracellular actions that impair cells74. Dendritic spine loss from layer II/III pyramidal cells in the
PFC function during stress16 (Fig. 3). Norepinephrine 1-ARs prelimbic medial PFC correlates with impaired working memory
activate Ca2+protein kinase C (PKC) signaling, which reduces on the delayed alternation task75. Similarly, dendritic retraction
delay-cell firing in the primate dlPFC57, while high levels of dopamine from layer II/III pyramidal cells in the dorsal medial PFC correlates
D1R stimulation reduce dlPFC delay-cell firing by increasing with weaker attentional flexibility on a perceptual set-shifting task71.
cAMPprotein kinase A (PKA) signaling60. Norepinephrine may These findings indicate that architectural changes have functional
also drive cAMP signaling via the 1-AR61, although this pathway relevance. In young adult rodents, layer II/III PFC pyramidal cell

nature neuroscience VOLUME 18 | NUMBER 10 | OCTOBER 2015 1379


Review

dendrites can regrow with sufficient time spent under safe conditions, One possible link between stress signaling pathways and actin regu-
but this plasticity is lost with advanced age76. lation involves PKC phosphorylation of MARCKS (myristoylated,
The changes in dendrites and spines with chronic stress are circuit alanine-rich C-kinase substrate), which normally anchors the actin
specific. In contrast to the PFC, chronic stress exposure increases skeleton to the cell membrane. In vitro studies of hippocampal
dendritic growth in the amygdala77, thus accentuating the imbal- pyramidal cell cultures have shown that PKC phosphorylation of
ance of amygdala over PFC function. Even within the PFC, there MARCKS induces collapse of the actin cytoskeleton by disconnect-
are circuit-specific alterations that lead to amygdala dominance with ing actin from the neuronal membrane96 (Fig. 4, gold). Inhibition of
chronic stress: the subset of PFC neurons that activate the amygdala PKC signaling before daily stress exposure in rats prevents the loss of
do not atrophy during stress (indeed, in females, these dendrites can spines from layer II/III PFC pyramidal cells normally observed with
be extended with stress), while the PFC neurons engaged in cortico- chronic stress75. The protection of dendritic spines correlates with
cortical connections show the expected loss of dendritic material73. preserved working memory function75. Future studies could examine
Similarly, the dendrites of pyramidal cells in the rodent orbital PFC whether the preservation of spines involves MARCKS stabilization of
extend rather than retract with chronic stress71. Chronic stress has no the actin cytoskeleton and whether medications that similarly inhibit
effect on performance of a reward reversal task that depends on orbital PKC signaling (for example, lithium, valproic acid, atypical anti
PFC function in rats71, further delineating this dissociation. Overall, psychotics) similarly rescue PFC dendritic spines from the effects
a simplistic interpretation of this body of work is that pyramidal cells of stress exposure.
in cognitive circuits lose dendrites with chronic stress while those A more recent study found that inhibition of cAMPPKA signal-
in emotional circuits are unchanged or strengthened. In contrast to ing with the 2A-AR agonist guanfacine is also protective of PFC
pyramidal cells, the dendrites of GABAergic Martinotti interneurons dendritic spines and cognitive function in rats97. Guanfacines ben-
hypertrophy with chronic stress in mouse PFC78, which may further eficial effects during chronic stress likely arise from a number of
reduce pyramidal cell excitation in cognitive circuits. interrelated mechanisms. Guanfacine strengthens dlPFC connectiv-
2015 Nature America, Inc. All rights reserved.

The loss of PFC gray matter with chronic stress has also been docu- ity via stimulation of postsynaptic 2A-ARs on layer III dendritic
mented in humans. Structural imaging has shown that lower PFC gray spines, inhibiting cAMP opening of HCN channels near the synapse98
matter volume correlates with exposure to adverse events79. Chronic (Fig. 4). Guanfacine may also diminish the harmful effects of stress
stress has also been shown to weaken PFC functional connectivity80 through actions outside the PFC. Stimulation of 2A-ARs weakens
and PFC regulation of the amygdala81, and to increase the volume of the amygdala function99, reduces stress-induced dopamine release in
putamen, thus accentuating the switch from flexible goal-directed to the PFC100 and reduces the tonic firing of locus coeruleus neurons
habitual responding82. Thus, sustained stress exposure in both animals and thus reduces norepinephrine release87,101. Guanfacine may also
and humans maintains the brain in a more primitive, reactive state. prevent spine loss by reducing inflammation in the brain (Fig. 4,
purple). Microglia and astrocytes are activated by -AR stimulation,
Molecular changes with chronic stress that contribute to spine loss. while activated microglia are deactivated by 2A-AR stimulation102.
The actions of norepinephrine are exaggerated with chronic stress As guanfacine is approved by the US Food and Drug Administration
exposure: there is increased expression of the synthetic enzymes for use in adolescents103, it may offer a practical approach for reducing
tyrosine hydroxylase and dopamine -hydroxylase in noradren- the inflammatory response and gray matter loss found in prodromal
ergic neurons and axons in both rats8385 and primates86. Chronic schizophrenia5.
stress also increases the tonic firing of locus coeruleus neurons via Stress may also reduce the number of dendritic spines in the PFC
increased CRFPKA activation of pacemaker cation channels87. by suppressing new spine formation. Recent studies have shown that
Interestingly, CRF is increased in the locus coeruleus of patients mTor (mammalian target of rapamycin) signaling increases spine gen-
with depression88, suggesting that this mechanism may be central eration in the apical tuft of layer V pyramidal cells in the rat PFC and
npg

to a chronic stress response in humans as well. Physical exercise can that stress exposure inhibits this effect by increasing the expression
be protective during stress by increasing the expression of galanin in of REDD1 (regulated in development and DNA damage responses 1)
the locus coeruleus, which reduces locus coeruleus firing, decreases (Fig. 4, pink)74. Norepinephrine stimulation of -ARcAMPPKA
stress-induced catecholamine release and protects PFC spines 89. In signaling increases the expression of REDD1 in macrophages104.
contrast to noradrenergic neurons, the dopaminergic axons project- Similar events in PFC neurons could provide a bridge between cat-
ing to rodent PFC become depleted with chronic stress exposure90,91. echolamine-induced increases in cAMP and reductions in mTor
However, remaining dopamine release appears sufficient for detri- signaling during stress exposure. It is not known, however, whether
mental actions, as D1R blockade during chronic stress prevents den- the mechanisms underlying spine loss are universal or are specific
dritic retraction in rat PFC92. to particular brain regions or circuits, or why stress causes dendritic
The mechanisms underlying stress-induced spine loss are just expansion in some neurons and atrophy in others. These are impor-
beginning to be understood and are an important arena for further tant areas for future research.
research given their relevance to cognitive disorders. How do stress Emerging data also suggest that different kinds of stress (physiolog-
pathways interact with the normal processes of spine pruning, for ical or psychological) may evoke similar signaling pathways to lead
example, during adolescence93? Are they related to spine loss with to PFC dysfunction and spine loss. For example, hypoxia increases
advancing age94? More specifically, how do the feedforward Ca2+ REDD1 expression105 and also induces spine loss in PFC and impaired
cAMP signaling mechanisms induced by stress exposure interact with PFC cognitive function106. As with psychological stressors, these
inflammatory events and with signaling pathways that regulate actin effects are prevented by treatment with guanfacine106. Similarly, trau-
dynamics in spines? Studies of the developing visual system show that matic brain injury (TBI) induces elevated catecholamine signaling in
activation of complement signaling induces phagocytosis of spines the PFC107 and elevated 1-AR expression that contributes to work-
and synapses by astroctyes95, but this may be a cleanup system that ing memory impairment108. As TBI increases the risk of PTSD109 and
works in tandem with other mechanismsfor example, mechanisms Alzheimers disease110, these data may help us understand the factors
that actively disassemble the actin cytoskeleton. that make higher brain circuits so vulnerable to insult.

1380 VOLUME 18 | NUMBER 10 | OCTOBER 2015 nature neuroscience


review

Figure 4 Hypothetical interactions between the Hypothetical interactions between stress-, actin- and inflammatory-
signaling pathways leading to loss of synapses on spines
intracellular signaling pathways activated by Dendrite
stress exposure and pathways that regulate actin
dynamics and inflammation. Stress signaling Stress
Unanchoring of actin
pathways are shown in red, regulatory pathways from membrane
and mechanisms that strengthen connectivity D1R
1R? 2A-AR
are shown in green. Inflammatory pathways are
shown in purple; calcium-related signaling events pMARCKS Spine
1-AR?
are shown in yellow; Rac1 constitutive activation CDC42
CDC42 PKC
by PKA is shown in gold; REDD1 inhibition of

Marina Corral Spence/Nature Publishing Group


CaMK
mTor signaling is shown in pink. Note that the 2+ IQGAP
Ca
CDCEP3 2+ AC
regulation of actin is often studied in cultured Ca
Glutamatergic
neurons and rarely in PFC neurons. Thus, future axon SEPT7 IP3R PDE4A --- DISC1
cAMP
research will be needed to see stress signaling NMDAR
PKA
events alter spine number in PFC pyramidal cells Actin DISC1- REDD1 mTor MK2
disassembly Kal7
through activation of these pathways. Rac1 PAK LIMK Cofilin
Kal7
Actin

Translation to humans. At least some of Constitutive activation of Rac1 by PKA leads to


these mechanisms studied in animals are actin depolymerization and spine loss Inflammation

immediately relevant to stress-related disor-


ders in humans. For example, increases in REDD1 have been found in with high rates of mental illness 120,121, and more subtle polymor-
the dlPFC of depressed patients, which is similar to what is seen in the phisms are associated with decreased PFC gray matter and impaired
2015 Nature America, Inc. All rights reserved.

stressed rat PFC74. Notably, there is evidence that treatment strategies working memory122,123. DISC1 anchors many proteins and thus
arising from basic research are effective in stress-related disorders111. regulates their functional localization and molecular interactions 124.
The 1-AR antagonist prazosin is now in widespread use to treat Particularly relevant to stress signaling in PFC, DISC1 anchors the
PTSD in veterans, active duty soldiers and civilians (reviewed in ref. phosphodiesterases (PDE4s) that catabolize cAMP and regulate its
111). Prazosin reduces flashbacks, improves concentration and think- signaling121,125. Immunoelectron microscopy studies of human126 and
ing, and reduces substance abuse, signs of improved PFC function. rhesus monkey13,127 dlPFC show that DISC1 is located in layer III
Guanfacine is now in widespread use for the treatment of PFC spines, where it anchors PDE4A next to the spine apparatus, critically
disorders on the basis of research in animals, and it has been shown positioned to regulate feedforward cAMPCa2+ stress signaling
to improve PFC functions and reduce cravings in subjects with stress- pathways13,127. Notably, genetic insults in PDE4A are also linked to
induced substance abuse112,113. Guanfacine also appears to help chil- schizophrenia128. The onset of schizophrenia is associated with signs
dren who have been traumatized, one of the few medications helpful of increased inflammation and PFC gray matter loss5, and biochemi-
in this arena114. The positive findings with guanfacine and prazosin cal studies in vitro have shown that inflammation reduces the abil-
are reassuring, as they validate the mission of basic research. ity of DISC1 to anchor PDE4A via increases in MK2 signaling125
(Fig. 4, purple). Loss of DISC1 anchoring of PDE4A due to inflamma-
Potential relevance to spine loss in mental disorders tion or genetic insults would thus disinhibit the stress response and
A major goal of current research is to understand how activation of lower the threshold for stress-induced PFC dysfunction.
stress signaling pathways in PFC contributes to psychiatric symp- Studies in rodents with genetic alterations of DISC1 are consistent
toms and to dendritic spine loss in mental illness. There is evidence with this hypothesis. Knockdown of DISC1 in rodent PFC increases
npg

of vmPFC gray matter loss in mood disorders7,115,116, which implies cAMP signaling in PFC neurons129 and increases sensitivity to stress-
dendritic atrophy. However, there have been no direct studies of induced PFC cognitive deficits130. DISC1 also regulates the integrity
changes in spine numbers in these circuits. Similarly, there have been of PFC spines by anchoring kalirin-7 (Kal7, the rodent homolog of
no studies of the molecular regulation of vmPFC circuits in primates, Duo) and preventing its stimulation of Rac1 signaling131. Loss of
and so we do not know whether these circuits are modulated in a DISC1 leads to constituent activation of Rac1 signaling and spine
manner similar to that of dlPFC. These are both important arenas for loss via p21-activated kinase (PAK) signaling131,132 (Fig. 4, orange).
future research. However, there have been several studies of dendritic Interestingly, PKA can form a complex with Rac1 that induces
spine changes in the dlPFC in schizophrenia. Neuronal cell bodies constitutive Rac1 activity133, a mechanism that may contribute to
are preserved, but there is extensive loss of dendrites and spines from stress-induced spine loss. As loss-of-function mutations in DISC1 are
layers III and possibly layer V pyramidal cells4,117,118. Indeed, the associated with a variety of mental illnesses, especially an increased
onset of schizophrenia is accompanied by waves of PFC gray matter incidence of depression120, loss of spines may contribute to a variety of
loss, as well as increased signs of inflammation5. The loss of spines disorders, with symptoms related to the subcircuit(s) most affected
from newly evolved cognitive circuits in schizophrenia likely contrib- for example, changes in vmPFC increasing risk of depression and
utes to their profound hypoactivity119. Understanding the causes of impairment of dlPFC circuits increasing risk of schizophrenia.
dendritic spine loss may help identify treatments to slow or prevent
the descent into disease. Molecular differences in the dlPFC of patients with schizophrenia.
Molecular analyses of the dlPFC from patients with schizophrenia
Clues from DISC1. Emerging data indicate that the scaffolding pro- have also begun to provide clues regarding potential mechanisms
tein Disrupted In Schizophrenia 1 (DISC1) is critical for regulation driving dendritic spine loss. Tissue analyses have found reductions in
of the stress response in PFC, suggesting that genetic insults that mRNAs for CDC42 and Duo that correlate with decreased spines134.
interfere with the function of this protein increase the risk for stress- A later study found increased expression of the CDC42 effector pro-
related psychiatric disorders. Mutations in DISC1 are associated tein CDC42EP3 specifically in layer III, as well as reduced septin-7

nature neuroscience VOLUME 18 | NUMBER 10 | OCTOBER 2015 1381


Review

(SEPT7), suggesting altered regulation of septin filaments in layer Feedforward stress signaling pathways are dysregulated by
III synapses135. In vitro data indicate that high levels of Ca2+ advancing age. The phosphodiesterase PDE4A is critically posi-
calmodulin signaling can disrupt CDC42IQGAP interactions needed tioned to regulate stress signaling pathways in the dlPFC pyramidal
for actin regulation136 (Fig. 4, gold), suggesting another possible link cell circuits needed for higher cognition14. PDE4A is anchored to
between stress signaling and actin dynamics. However, it is not known the spine apparatus, where it can catabolize cAMP and reduce feed-
whether such interactions occur in layer III dlPFC spines. Bridging forward Ca2+cAMP signaling in spines (Figs. 35). Studies of the
signaling events in dlPFC neuronal circuits with molecular changes aging monkey cortex have found that PDE4A is lost from these spines
in the neurons of patients with mental illness is an important goal with advancing age, perhaps as a result of age-related increases in
for further research. inflammation that may unanchor PDE4A125. Age-related reductions
in PDE4A are associated with increased pTau in the dlPFC but not
Potential relevance to degeneration in Alzheimers disease the primary visual cortex, a pattern similar to the pattern of neurofi-
Dysregulation of stress signaling pathways with advancing age may brillary tangles in Alzheimers disease14. Increased tau phosphoryla-
also increase vulnerability to degeneration in Alzheimers diseasefor tion occurs at sites phosphorylated by PKA and by Ca2+-activated
example, due to an age-related loss of PDE4A. Alzheimers disease is kinases14 (Fig. 5, brown). pTau accumulates over the spine apparatus
characterized by amyloid- (A) plaques and by neurofibrillary tan- and in the postsynaptic density of putative glutamatergic-like (but
gles composed of hyperphosphorylated tau (pTau). Cognitive impair- not inhibitory) synapses on spines, where there is evidence of pTau
ment correlates with the number of neurofibrillary tangles137, which trafficking in vesicles. In the nearby dendrite, pTau aggregates on
selectively affect highly connected pyramidal cells in association cor- microtubules, where it may interfere with intracellular trafficking,
tex but not in primary sensory cortex6,138,139. Research is beginning to including the trafficking of amyloid precursor protein (APP).
uncover why pyramidal cells in association cortex are so vulnerable,
why advancing age is such a large risk factor for neurodegeneration Multiple, interacting vicious cycles increase risk of neuro
2015 Nature America, Inc. All rights reserved.

and why stress may drive disease. degeneration. Dysregulation of feedforward Ca2+cAMP signaling
Although the largest risk factor for Alzheimers disease is advanced in dlPFC spines could drive multiple, interacting, vicious cycles that
age, TBI is also an established risk factor140, and new evidence suggests increase vulnerability to degeneration (Fig. 5, red). APP can be cleaved
that psychological distress43 and female sex141 are also risk factors for to A (Fig. 5, magenta) when it is trapped in endosomes that con-
Alzheimers degeneration. Indeed, the increased risk of Alzheimers tain -secretase (BACE)143, a process exacerbated by the APOE E4
disease associated with the E4 allele of the APOE gene is especially genotype144,145. The aggregation of pTau on microtubules may simi-
pronounced in women and is associated with increased pTau141. As larly trap APP-containing endosomes and lead to the generation of
described above, TBI and psychological distress share signaling events A oligomers. A oligomers can drive additional vicious cycles by
in PFC, and females have an exaggerated stress response. Intriguingly, stimulating metabotropic glutamate receptor 5 (mGluR5)146,147,
animal studies have shown that stress exposure increases the phosphor- which activates feedforward Ca2+cAMP signaling and drive tau
ylation of tau142. Thus, these seemingly disparate risk factors may share phosphorylation (Fig. 5, brown). A fibrils also increase inflamma-
underlying molecular mechanisms that confer risk of degeneration. tion148 (Fig. 5, purple), which may unanchor residual PDE4A125, and

Figure 5 The multiple, interacting, feedforward


Interacting vicious cycles with advancing age Pyramidal
vicious cycles that may be disinhibited in cell dendrite
the aging dlPFC, contributing to increased SER
APP
vulnerability to degeneration. Red: stress
apoE4
activates feedforward Ca2+cAMP signaling
npg


pathways near the glutamate NMDAR synapses APP
Stress
on spines. In the young adult dlPFC, the A
BACE
D1R
phosphodiesterase PDE4A is anchored by DISC1 1R?
mGluR5 Inflammation
next to the spine apparatus (*), an extension

Marina Corral Spence/Nature Publishing Group


of the smooth endoplasmic reticulum (SER),
CaMKII
critically positioned to regulate feedforward 2+
2+ Ca AC
Ca cAMP signaling in dlPFC spines. NMDAR Ca 2+
pTau
PDE4A is lost from spines with advancing Glutamatergic
axon IP3R cAMP MK2
age, dysregulating Ca2+cAMP signaling and PKA Microtubules
increasing the activation of kinases (for example,
pTau m
PKA and calcium/calmodulin-dependent kinase PDE4A DISC1
14
II (CaMKII)) that phosphorylate tau . IP3R, ROS
pCOXIV
inositol-1,4,5-trisphosphate receptor. Brown: pTau aggregates over the
spine apparatus, at glutamatergic synapses, and over microtubules in dendrites and traffics in vesicles between
neurons14. The aggregation of pTau on microtubules in dendrites likely interferes with intracellular trafficking,
including the trafficking of APP, the precursor to A. Magenta: APP is cleaved to A when it is trapped in endosomes
that contain -secretase (BACE)for example, when there is interference with APP endosomal trafficking 143. Indeed, the increased risk of Alzheimers
disease conferred by the apoE4 variant is thought to involve increased localization of APP into endosomes 145. The aggregation of pTau on microtubules may
similarly trap APP-containing endosomes and lead to the increased generation of A oligomers. The generation of A oligomers can drive additional vicious
cycles by stimulating mGluR5 (ref. 147). mGluR5 are localized near the synapse on spines in dlPFC, positioned to activate feedforward Ca 2+cAMP signaling
and thus drive more tau phosphorylation. Purple: A fibrils drive inflammation148, which can unanchor residual PDE4A125 and further disinhibit stress
signaling pathways. Orange: increased stress signaling may also dysregulate mitochondrial function, as PKA can phosphorylate cyclooxygenase IV (COX IV) to
increase reactive oxygen species (ROS)149, which also increase tau phosphorylation and A production150, leading to additional mitochondrial dysfunction.
Thus, dysregulation of stress signaling pathways in the dlPFC with advancing age may contribute to many deleterious molecular events that increase
vulnerability to degeneration. Alzheimers disease pathology may begin anywhere along these pathways (for example, genetic alterations in APP processing or
environmental stressors promoting pTau) and, by driving these interacting cycles, lead to the same degenerative phenotype.

1382 VOLUME 18 | NUMBER 10 | OCTOBER 2015 nature neuroscience


review

further disinhibit stress signaling pathways. Increased stress signaling 7. Blumberg, H.P. et al. Age, rapid-cycling, and pharmacotherapy effects on ventral
prefrontal cortex in bipolar disorder: a cross-sectional study. Biol. Psychiatry 59,
may also dysregulate mitochondrial function, which also leads to tau
611618 (2006).
phosphorylation149 and A production150 (Fig. 5, orange). These in 8. Khn, S. & Gallinat, J. Gray matter correlates of posttraumatic stress disorder: a
turn cause more mitochondrial dysfunction, thus feeding yet another quantitative meta-analysis. Biol. Psychiatry 73, 7074 (2013).
9. Elston, G.N. et al. Specializations of the granular prefrontal cortex of primates:
intracellular vicious cycle. implications for cognitive processing. Anat. Rec. A Discov. Mol. Cell. Evol. Biol.
The presence of so many interacting vicious cycles suggests that the 288, 2635 (2006).
degenerative process could be initiated by a variety of precipitating 10. Wang, M. et al. NMDA receptors subserve working memory persistent neuronal
firing In dorsolateral prefrontal cortex. Neuron 77, 736749 (2013).
events, any of which could set the entire process in motion. For exam- 11. Yang, Y. et al. Nicotinic 7 receptors enhance NMDA cognitive circuits in
ple, genetic errors in APP processing such as presenilin mutations can dorsolateral prefrontal cortex. Proc. Natl. Acad. Sci. USA 110, 1207812083
(2013).
increase the production of A early in life and thus cause early-onset
12. Datta, D., Arion, D. & Lewis, D.A. Developmental expression patterns of GABAA
illness, or the loss of PDE4A regulation of the stress response with receptor subunits in layer 3 and 5 pyramidal cells of monkey prefrontal cortex.
advancing age can drive the phosphorylation of tau and lead to late- Cereb. Cortex 25, 22952305 (2015).
13. Arnsten, A.F.T., Wang, M. & Paspalas, C.D. Neuromodulation of thought:
onset disease. Future research may determine whether this signature of Flexibilities and vulnerabilities in prefrontal cortical network synapses. Neuron
vulnerability observed in the dlPFC is also evident in other association 76, 223239 (2012).
cortices that degenerate in Alzheimers disease (for example, entorhinal 14. Carlyle, B.C. et al. cAMP-PKA phosphorylation of tau confers risk for degeneration
in aging association cortex. Proc. Natl. Acad. Sci. USA 111, 50365041
cortex, parietal association cortex) and whether inhibition of stress (2014).
signaling events (for example, with 2A-AR or mGluR3 agonists, or 15. Caetano, M.S. et al. Lost in transition: Aging-related changes in executive control
by the medial prefrontal cortex. J. Neurosci. 32, 37653777 (2012).
mGluR5 antagonists) can provide strategies for prevention.
16. Arnsten, A.F.T. Stress signaling pathways that impair prefrontal cortex structure
and function. Nat. Rev. Neurosci. 10, 410422 (2009).
Closing 17. Glass, D.C., Reim, B. & Singer, J.E. Behavioral consequences of adaptation to
controllable and uncontrollable noise. J. Exp. Soc. Psychol. 7, 244257
Studies of the molecular pathways activated by stress exposure have
2015 Nature America, Inc. All rights reserved.

(1971).
begun to explain why PFC circuits deteriorate in so many cognitive 18. Minor, T.R., Jackson, R.L. & Maier, S.F. Effects of task-irrelevant cues and
disorders. The presence of intrinsic mechanisms to actively weaken reinforcement delay on choice-escape learning following inescapable shock:
evidence for a deficit in selective attention. J. Exp. Psychol. Anim. Behav. Process.
connections during stress exposure in these newly evolved circuits 10, 543556 (1984).
renders them particularly vulnerable when they are dysregulated 19. Murphy, B.L., Arnsten, A.F.T., Goldman-Rakic, P.S. & Roth, R.H. Increased
dopamine turnover in the prefrontal cortex impairs spatial working memory
owing to genetic or environmental insults. This contrasts with the
performance in rats and monkeys. Proc. Natl. Acad. Sci. USA 93, 13251329
stress effects on subcortical regions such as the amygdala that are (1996).
strengthened by stress exposure, thus switching the brain into a more 20. Arnsten, A.F.T. & Goldman-Rakic, P.S. Noise stress impairs prefrontal cortical
cognitive function in monkeys: evidence for a hyperdopaminergic mechanism.
primitive, reflexive state. Much more research is needed to understand Arch. Gen. Psychiatry 55, 362368 (1998).
the mechanics of spine loss, the generality of the stress response to This study and ref. 19 above were the first to show that stress exposure impairs
other high-order association cortices, and how genetic insults interact PFC function in animals and that it is caused by high levels of stress-induced
catecholamine release in the PFC.
with stress signaling pathways to hasten disease. However, the benefits 21. Elliott, A.E. & Packard, M.G. Intra-amygdala anxiogenic drug infusion prior to
of this basic research are already evident in new, effective treatments retrieval biases rats towards the use of habit memory. Neurobiol. Learn. Mem.
for stress-related cognitive disorders. 90, 616623 (2008).
22. Rodrigues, S.M., LeDoux, J.E. & Sapolsky, R.M. The influence of stress hormones
on fear circuitry. Annu. Rev. Neurosci. 32, 289313 (2009).
Acknowledgments 23. Yamada, K., McEwen, B.S. & Pavlides, C. Site and time dependent effects of
Gratitude to S. Katrancha for discussions regarding spine dynamics. A.F.T.A. acute stress on hippocampal long-term potentiation in freely behaving rats. Exp.
is supported by a US National Institutes of Health Directors Pioneer Award Brain Res. 152, 5259 (2003).
DP1AG047744-01, R01AG043430-01A1 and RO1MH100064-01A1. 24. Goldstein, L.E., Rasmusson, A.M., Bunney, S.B. & Roth, R.H. Role of the
amygdala in the coordination of behavioral, neuroendocrine and prefrontal cortical
npg

COMPETING FINANCIAL INTERESTS monoamine responses to psychological stress in the rat. J. Neurosci. 16,
The author declares competing financial interests: details are available in the online 47874798 (1996).
25. Arnsten, A.F.T. The biology of feeling frazzled. Science 280, 17111712
version of the paper.
(1998).
26. Shansky, R.M., Rubinow, K., Brennan, A. & Arnsten, A.F. The effects of sex and
Reprints and permissions information is available online at http://www.nature.com/ hormonal status on restraint-stress-induced working memory impairment. Behav.
reprints/index.html. Brain Funct. 2, 8 (2006).
27. Olver, J.S., Pinney, M., Maruff, P. & Norman, T.R. Impairments of spatial working
1. Goldman-Rakic, P.S. Cellular basis of working memory. Neuron 14, 477485 memory and attention following acute psychosocial stress. Stress Health 31,
(1995). 115123 (2015).
This is an excellent review of the dlPFC microcircuits that underlie spatial working 28. Kim, J.J. & Diamond, D.M. The stressed hippocampus, synaptic plasticity and
memory in primates and of how they generate the mental representations that lost memories. Nat. Rev. Neurosci. 3, 453462 (2002).
are the foundation of abstract thought. 29. Spyrka, J., Danielewicz, J. & Hess, G. Brief neck restraint stress enhances long-
2. Ongr, D. & Price, J.L. The organization of networks within the orbital and medial term potentiation and suppresses long-term depression in the dentate gyrus of
prefrontal cortex of rats, monkeys and humans. Cereb. Cortex 10, 206219 (2000). the mouse. Brain Res. Bull. 85, 363367 (2011).
This is an excellent review of the connections and functions of the ventral and 30. Sinha, R., Lacadie, C.M., Skudlarski, P. & Wexler, B.E. Neural circuits
medial PFC in primates. underlying emotional distress in humans. Ann. NY Acad. Sci. 1032, 254257
3. Opler, L.A., Opler, M.G. & Arnsten, A.F.T. Ameliorating treatment-refractory (2004).
depression with intranasal ketamine: potential NMDA receptor actions in the 31. Qin, S., Hermans, E.J., van Marle, H.J.F., Lou, J. & Fernandez, G. Acute
pain circuitry representing mental anguish. CNS Spectr. doi:10.1017/ psychological stress reduces working memory-related activity in the dorsolateral
S1092852914000686 (26 January 2015). prefrontal cortex. Biol. Psychiatry 66, 2532 (2009).
4. Selemon, L.D. & Goldman-Rakic, P.S. The reduced neuropil hypothesis: a circuit This study was the first to show that acute stress exposure impairs the working
based model of schizophrenia. Biol. Psychiatry 45, 1725 (1999). memory functions of the dlPFC in human subjects, using fMRI to document the
5. Cannon, T.D. et al. Progressive reduction in cortical thickness as psychosis pattern of brain activity changes that occur with exposure to chronic stress. Its
develops: A multisite longitudinal neuroimaging study of youth at elevated clinical companion study (ref. 32) showed that stress-induced cognitive impairment in
risk. Biol. Psychiatry 77, 147157 (2015). dlPFC is associated with genetic signatures of higher catecholamine levels, thus
6. Bussire, T. et al. Progressive degeneration of nonphosphorylated neurofilament bridging the research in animals and human subjects.
protein-enriched pyramidal neurons predicts cognitive impairment in Alzheimers 32. Qin, S. et al. The effect of moderate acute psychological stress on working
disease: stereologic analysis of prefrontal cortex area 9. J. Comp. Neurol. 463, memory-related neural activity is modulated by a genetic variation in
281302 (2003). catecholaminergic function in humans. Front. Integr. Neurosci. 6, 16 (2012).

nature neuroscience VOLUME 18 | NUMBER 10 | OCTOBER 2015 1383


Review

33. Grtner, M., Rohde-Liebenau, L., Grimm, S. & Bajbouj, M. Working memory- 65. Everitt, B.J. et al. Neural mechanisms underlying the vulnerability to develop
related frontal theta activity is decreased under acute stress. Psychoneuro compulsive drug-seeking habits and addiction. Phil. Trans. R. Soc. Lond. B 363,
endocrinology 43, 105113 (2014). 31253135 (2008).
34. Cahill, L. & McGaugh, J.L. Modulation of memory storage. Curr. Opin. Neurobiol. 66. Waterhouse, B.D., Moises, H.C. & Woodward, D.J. Noradrenergic modulation of
6, 237242 (1996). somatosensory cortical neuronal responses to iontophoretically applied putative
35. Cahill, L., Prins, B., Weber, M. & McGaugh, J.L. Beta-adrenergic activation and transmitters. Exp. Neurol. 69, 3049 (1980).
memory for emotional events. Nature 371, 702704 (1994). 67. Mouradian, R.D., Seller, F.M. & Waterhouse, B.D. Noradrenergic potentiation of
36. Mazure, C.M. ed. Does Stress Cause Psychiatric Illness? (American Psychiatric excitatory transmitter action in cerebrocortical slices: evidence of mediation by
Press, Washington, DC, 1995). an alpha1-receptor-linked second messenger pathway. Brain Res. 546, 8395
37. Raune, D., Kuipers, E. & Bebbington, P. Stressful and intrusive life events (1991).
preceding first episode psychosis. Epidemiol. Psichiatr. Soc. 18, 221228 68. Bonini, J.S., Cammarota, M., Kerr, D.S., Bevilaqua, L.R. & Izquierdo, I. Inhibition
(2009). of PKC in basolateral amygdala and posterior parietal cortex impairs consolidation
38. Hammen, C. & Gitlin, M. Stress reactivity in bipolar patients and its relation to of inhibitory avoidance memory. Pharmacol. Biochem. Behav. 80, 6367
prior history of disorder. Am. J. Psychiatry 154, 856857 (1997). (2005).
39. Weissman, M.M. et al. Cross-national epidemiology of major depression and 69. Barsegyan, A., Mackenzie, S.M., Kurose, B.D., McGaugh, J.L. & Roozendaal, B.
bipolar disorder. J. Am. Med. Assoc. 276, 293299 (1996). Glucocorticoids in the prefrontal cortex enhance memory consolidation and impair
40. Breslau, N. The epidemiology of trauma, PTSD, and other posttrauma disorders. working memory by a common neural mechanism. Proc. Natl. Acad. Sci. USA
Trauma Violence Abuse 10, 198210 (2009). 107, 1665516660 (2010).
41. Shansky, R.M. et al. Estrogen mediates sex differences in stress-induced prefrontal 70. Seib, L.M. & Wellman, C.L. Daily injections alter spine density in rat medial
cortex dysfunction. Mol. Psychiatry 9, 531538 (2004). prefrontal cortex. Neurosci. Lett. 337, 2932 (2003).
42. Bebbington, P.E. et al. The influence of age and sex on the prevalence of 71. Liston, C. et al. Stress-induced alterations in prefrontal cortical dendritic
depressive conditions: report from the National Survey of Psychiatric Morbidity. morphology predict selective impairments in perceptual attentional set-shifting.
Psychol. Med. 28, 919 (1998). J. Neurosci. 26, 78707874 (2006).
43. Johansson, L. et al. Common psychosocial stressors in middle-aged women related 72. Radley, J.J. et al. Reversibility of apical dendritic retraction in the rat medial
to longstanding distress and increased risk of Alzheimers disease: a 38-year prefrontal cortex following repeated stress. Exp. Neurol. 196, 199203 (2005).
longitudinal population study. BMJ Open 3, e003142 (2013). 73. Shansky, R.M., Hamo, C., Hof, P.R., McEwen, B.S. & Morrison, J.H.
44. Wilson, R.S. et al. Chronic psychological distress and risk of Alzheimers disease Stress-induced dendritic remodeling in the prefrontal cortex is circuit specific.
in old age. Neuroepidemiology 27, 143153 (2006). Cereb. Cortex 19, 24792484 (2009)
This study used tract tracing to be able to identify the connections of neurons
2015 Nature America, Inc. All rights reserved.

45. Deutch, A.Y. & Roth, R.H. The determinants of stress-induced activation of the
prefrontal cortical dopamine system. Prog. Brain Res. 85, 367402 (1990). in rat medial PFC and showed that those positioned to excite the amygdala had
46. Bromberg-Martin, E.S., Matsumoto, M. & Hikosaka, O. Dopamine in motivational expanded dendrites following chronic stress, while those with cortical projections
control: rewarding, aversive, and alerting. Neuron 68, 815834 (2010). showed the expected atrophy with chronic stress exposure. These effects were
47. Kodama, T., Hikosaka, K., Honda, Y., Kojima, T. & Watanabe, M. Higher dopamine particularly prevalent in females, consistent with the greater stress response in
release induced by less rather than more preferred reward during a working memory females with circulating estrogen. This study thus revealed an important
task in the primate prefrontal cortex. Behav. Brain Res. 266, 104107 (2014). heterogeneity within the PFC, reminding us that some circuits within the PFC
48. Van Bockstaele, E.J., Colago, E.E. & Valentino, R.J. Amygdaloid corticotropin- actually promote the stress response.
releasing factor targets locus coeruleus dendrites: substrate for the co-ordination 74. Ota, K.T. et al. REDD1 is essential for stress-induced synaptic loss and depressive
of emotional and cognitive limbs of the stress response. J. Neuroendocrinol. 10, behavior. Nat. Med. 20, 531535 (2014).
743757 (1998). 75. Hains, A.B. et al. Inhibition of protein kinase C signaling protects prefrontal cortex
49. Nakane, H., Shimizu, N. & Hori, T. Stress-induced norepinephrine release in the dendritic spines and cognition from the effects of chronic stress. Proc. Natl. Acad.
rat prefrontal cortex measured by microdialysis. Am. J. Physiol. 267, Sci. USA 106, 1795717962 (2009).
R1559R1566 (1994). 76. Bloss, E.B. et al. Evidence for reduced experience-dependent dendritic spine
50. Chandler, D.J., Gao, W.J. & Waterhouse, B.D. Heterogeneous organization of the plasticity in the aging prefrontal cortex. J. Neurosci. 31, 78317839 (2011).
locus coeruleus projections to prefrontal and motor cortices. Proc. Natl. Acad. 77. Vyas, A., Mitra, R., Shankaranarayana Rao, B.S. & Chattarji, S. Chronic stress
Sci. USA 111, 68166821 (2014). induces contrasting patterns of dendritic remodeling in hippocampal and
51. Grndemann, D., Schechinger, B., Rappold, G.A. & Schomig, E. Molecular amygdaloid neurons. J. Neurosci. 22, 68106818 (2002).
identification of the cortisone-sensitive extraneuronal catecholamine transporter. 78. Gilabert-Juan, J., Castillo-Gomez, E., Guirado, R., Molt, M.D. & Nacher, J.
Nat. Neurosci. 1, 349351 (1998). Chronic stress alters inhibitory networks in the medial prefrontal cortex of adult
52. Bangasser, D.A. & Valentino, R.J. Sex differences in molecular and cellular mice. Brain Struct. Funct. 218, 15911605 (2013).
substrates of stress. Cell. Mol. Neurobiol. 32, 709723 (2012). 79. Ansell, E.B., Rando, K., Tuit, K., Guarnaccia, J. & Sinha, R. Cumulative adversity
53. Kritzer, M.F. & Kohama, S.G. Ovarian hormones influence morphology, distribution and smaller gray matter volume in medial prefrontal, anterior cingulate, and insula
and density of tyrosine hydroxylase immunoreactive axons in the dorsolateral regions. Biol. Psychiatry 72, 5764 (2012).
prefrontal cortex of adult rhesus monkeys. J. Comp. Neurol. 395, 117 80. Liston, C., McEwen, B.S. & Casey, B.J. Psychosocial stress reversibly disrupts
npg

(1998). prefrontal processing and attentional control. Proc. Natl. Acad. Sci. USA 106,
54. Birnbaum, S., Gobeske, K.T., Auerbach, J., Taylor, J.R. & Arnsten, A.F.T. A role 912917 (2009).
for norepinephrine in stress-induced cognitive deficits: -1-adrenoceptor mediation 81. Kim, P. et al. Effects of childhood poverty and chronic stress on emotion regulatory
in prefrontal cortex. Biol. Psychiatry 46, 12661274 (1999). brain function in adulthood. Proc. Natl. Acad. Sci. USA 110, 1844218447
55. Zahrt, J., Taylor, J.R., Mathew, R.G. & Arnsten, A.F.T. Supranormal stimulation (2013).
of dopamine D1 receptors in the rodent prefrontal cortex impairs spatial working 82. Soares, J.M. et al. Stress-induced changes in human decision-making are
memory performance. J. Neurosci. 17, 85288535 (1997). reversible. Transl. Psychiatry 2, e131 (2012).
56. Arnsten, A.F.T., Mathew, R., Ubriani, R., Taylor, J.R. & Li, B.-M. Alpha-1 83. Melia, K.R. et al. Coordinate regulation of the cyclic AMP system with firing rate
noradrenergic receptor stimulation impairs prefrontal cortical cognitive function. and expression of tyrosine hydroxylase in the rat locus coeruleus: effects of chronic
Biol. Psychiatry 45, 2631 (1999). stress and drug treatments. J. Neurochem. 58, 494502 (1992).
57. Birnbaum, S.G. et al. Protein kinase C overactivity impairs prefrontal cortical 84. Miner, L.H. et al. Chronic stress increases the plasmalemmal distribution of the
regulation of working memory. Science 306, 882884 (2004). norepinephrine transporter and the coexpression of tyrosine hydroxylase in
58. Hermans, E.J. et al. Stress-related noradrenergic activity prompts large-scale norepinephrine axons in the prefrontal cortex. J. Neurosci. 26, 15711578 (2006).
neural network reconfiguration. Science 334, 11511153 (2011). 85. Fan, Y., Chen, P., Li, Y. & Zhu, M.Y. Effects of chronic social defeat on expression
59. Schwabe, L., Tegenthoff, M., Hffken, O. & Wolf, O.T. Simultaneous glucocorticoid of dopamine -hydroxylase in rat brains. Synapse 67, 300312 (2013).
and noradrenergic activity disrupts the neural basis of goal-directed action in the 86. Bethea, C.L., Kim, A. & Cameron, J.L. Function and innervation of the locus
human brain. J. Neurosci. 32, 1014610155 (2012). ceruleus in a macaque model of functional hypothalamic amenorrhea. Neurobiol.
60. Vijayraghavan, S. et al. Inverted-U dopamine D1 receptor actions on prefrontal Dis. 50, 96106 (2013).
neurons engaged in working memory. Nat. Neurosci. 10, 376384 (2007). 87. Nestler, E.J., Alreja, M. & Aghajanian, G.K. Molecular control of locus coeruleus
61. Ramos, B.P. et al. The beta-1 adrenergic antagonist, betaxolol, improves working neurotransmission. Biol. Psychiatry 46, 11311139 (1999).
memory performance in rats and monkeys. Biol. Psychiatry 58, 894900 (2005). 88. Bissette, G., Klimek, V., Pan, J., Stockmeier, C. & Ordway, G. Elevated
62. Gamo, N.J. et al. Stress impairs prefrontal cortical function via D1 dopamine concentrations of CRF in the locus coeruleus of depressed subjects.
receptor interactions with HCN channels. Biol. Psychiatry doi:10.1016/j. Neuropsychopharmacology 28, 13281335 (2003).
biopsych.2015.01.009 (4 February 2015). 89. Sciolino, N.R. et al. Galanin mediates features of neural and behavioral stress
63. Wang, M., Vijayraghavan, S. & Goldman-Rakic, P.S. Selective D2 receptor resilience afforded by exercise. Neuropharmacology 89, 255264 (2015).
actions on the functional circuitry of working memory. Science 303, 853856 90. Mizoguchi, K. et al. Chronic stress induces impairment of spatial working memory
(2004). due to prefrontal dopaminergic dysfunction. J. Neurosci. 20, 15681574 (2000).
64. Ferry, B., Roozendaal, B. & McGaugh, J.L. Basolateral amygdala noradrenergic 91. Matuszewich, L., McFadden, L.M., Friedman, R.D. & Frye, C.A. Neurochemical
influences on memory storage are mediated by an interaction between - and and behavioral effects of chronic unpredictable stress. Behav. Pharmacol. 25,
1-adrenoceptors. J. Neurosci. 19, 51195123 (1999). 557566 (2014).

1384 VOLUME 18 | NUMBER 10 | OCTOBER 2015 nature neuroscience


review

92. Lin, G.L., Borders, C.B., Lundewall, L.J. & Wellman, C.L. D1 receptors regulate 120. Chubb, J.E., Bradshaw, N.J., Soares, D.C., Porteous, D.J. & Millar, J.K. The DISC
dendritic morphology in normal and stressed prelimbic cortex. locus in psychiatric illness. Mol. Psychiatry 13, 3664 (2008).
Psychoneuroendocrinology 51, 101111 (2015). 121. Millar, J.K. et al. DISC1 and PDE4B are interacting genetic factors in schizophrenia
93. Rakic, P., Bourgeois, J.P. & Goldman-Rakic, P.S. Synaptic development of the that regulate cAMP signaling. Science 310, 11871191 (2005).
cerebral cortex: implications for learning, memory, and mental illness. Prog. Brain 122. Cannon, T.D. et al. Association of DISC1/TRAX haplotypes with schizophrenia,
Res. 102, 227243 (1994). reduced prefrontal gray matter, and impaired short- and long-term memory. Arch.
94. Dumitriu, D. et al. Selective changes in thin spine density and morphology in Gen. Psychiatry 62, 12051213 (2005).
monkey prefrontal cortex correlate with aging-related cognitive impairment. 123. Szeszko, P.R. et al. DISC1 is associated with prefrontal cortical gray matter and
J. Neurosci. 30, 75077515 (2010). positive symptoms in schizophrenia. Biol. Psychol. 79, 103110 (2008).
95. Chung, W.S. et al. Astrocytes mediate synapse elimination through MEGF10 and 124. Camargo, L.M. et al. Disrupted in Schizophrenia 1 interactome: evidence for the
MERTK pathways. Nature 504, 394400 (2013). close connectivity of risk genes and a potential synaptic basis for schizophrenia.
96. Calabrese, B. & Halpain, S. Essential role for the PKC target MARCKS in Mol. Psychiatry 12, 7486 (2007).
maintaining dendritic spine morphology. Neuron 48, 7790 (2005). 125. MacKenzie, K.F. et al. Phosphorylation of cAMP-specific PDE4A5
97. Hains, A.B., Yabe, Y. & Arnsten, A.F.T. Chronic stimulation of alpha-2A-adrenoceptors (phosphodiesterase-4A5) by MK2 (MAPKAPK2) attenuates its activation through
with guanfacine protects rodent prefrontal cortex dendritic spines and cognition protein kinase A phosphorylation. Biochem. J. 435, 755769 (2011).
from the effects of chronic stress. Neurobiol. Stress 2, 19 (2015). 126. Kirkpatrick, B. et al. DISC1 immunoreactivity at the light and ultrastructural level
98. Wang, M. et al. 2A-adrenoceptor stimulation strengthens working memory in the human neocortex. J. Comp. Neurol. 497, 436450 (2006).
networks by inhibiting cAMP-HCN channel signaling in prefrontal cortex. 127. Paspalas, C.D., Min Wang, M. & Arnsten, A.F.T. Constellation of HCN channels
Cell 129, 397410 (2007). and cAMP regulating proteins in dendritic spines of the primate prefrontal cortex
99. DeBock, F. et al. 2-adrenoreceptor activation inhibits LTP and LTD in the potential substrate for working memory deficits in schizophrenia. Cereb. Cortex
basolateral amygdala: involvement of Gi/o-protein-mediated modulation of Ca2+- 23, 16431654 (2013).
channels and inwardly rectifying K+-channels in LTD. Eur. J. Neurosci. 17, 128. Deng, X. et al. Positive association of phencyclidine-responsive genes, PDE4A
14111424 (2003). and PLAT, with schizophrenia. Am. J. Med. Genet. B. Neuropsychiatr. Genet.
100. Morrow, B.A., George, T.P. & Roth, R.H. Noradrenergic -2 agonists have 156B, 850858 (2011).
anxiolytic-like actions on stress-related behavior and mesoprefrontal dopamine 129. El-Hassar, L. et al. Disrupted in Schizophrenia 1 modulates medial prefrontal
biochemistry. Brain Res. 1027, 173178 (2004). cortex pyramidal neuron activity through cAMP regulation of transient receptor
101. Engberg, G. & Eriksson, E. Effects of alpha 2-adrenoceptor agonists on locus potential C and small-conductance K+ channels. Biol. Psychiatry 76, 476485
coeruleus firing rate and brain noradrenaline turnover in N-ethoxycarbonyl-2- (2014).
ethoxy-1,2-dihydroquinoline (EEDQ)-treated rats. Naunyn Schmiedebergs Arch. 130. Gamo, N.J. et al. Role of Disrupted in Schizophrenia 1 (DISC1) in stress-induced
2015 Nature America, Inc. All rights reserved.

Pharmacol. 343, 472477 (1991). prefrontal cognitive dysfunction. Transl. Psychiatry 3, e328 (2013).
102. Gyoneva, S. & Traynelis, S.F. Norepinephrine modulates the motility of resting 131. Hayashi-Takagi, A. et al. Disrupted-in-Schizophrenia 1 (DISC1) regulates spines
and activated microglia via different adrenergic receptors. J. Biol. Chem. 288, of the glutamate synapse via Rac1. Nat. Neurosci. 13, 327332 (2010).
1529115302 (2013). 132. Hayashi-Takagi, A. et al. PAKs inhibitors ameliorate schizophrenia-associated
103. Childress, A.C. & Berry, S.A. Pharmacotherapy of attention-deficit hyperactivity dendritic spine deterioration in vitro and in vivo during late adolescence. Proc.
disorder in adolescents. Drugs 72, 309325 (2012). Natl. Acad. Sci. USA 111, 64616466 (2014).
104. Yanagawa, Y., Hiraide, S., Matsumoto, M. & Togashi, H. Rapid induction of REDD1 133. Bachmann, V.A., Bister, K. & Stefan, E. Interplay of PKA and Rac: fine-tuning
gene expression in macrophages in response to stress-related catecholamines. of Rac localization and signaling. Small GTPases 4, 247251 (2013).
Immunol. Lett. 158, 109115 (2014). 134. Hill, J.J., Hashimoto, T. & Lewis, D.A. Molecular mechanisms contributing to
105. Katiyar, S. et al. REDD1, an inhibitor of mTOR signalling, is regulated by the dendritic spine alterations in the prefrontal cortex of subjects with schizophrenia.
CUL4ADDB1 ubiquitin ligase. EMBO Rep. 10, 866872 (2009). Mol. Psychiatry 11, 557566 (2006).
106. Kauser, H., Sahu, S., Kumar, S. & Panjwani, U. Guanfacine is an effective 135. Ide, M. & Lewis, D.A. Altered cortical CDC42 signaling pathways in schizophrenia:
countermeasure for hypobaric hypoxia-induced cognitive decline. Neuroscience implications for dendritic spine deficits. Biol. Psychiatry 68, 2532 (2010).
254, 110119 (2013). 136. Briggs, M.W. & Sacks, D.B. IQGAP1 as signal integrator: Ca2+, calmodulin, Cdc42
107. Kobori, N., Clifton, G.L. & Dash, P.K. Enhanced catecholamine synthesis in the and the cytoskeleton. FEBS Lett. 542, 711 (2003).
prefrontal cortex after traumatic brain injury: implications for prefrontal 137. Giannakopoulos, P. et al. Tangle and neuron numbers, but not amyloid load,
dysfunction. J. Neurotrauma 23, 10941102 (2006). predict cognitive status in Alzheimers disease. Neurology 60, 14951500
108. Kobori, N., Hu, B. & Dash, P.K. Altered adrenergic receptor signaling following (2003).
traumatic brain injury contributes to working memory dysfunction. Neuroscience 138. Pearson, R.C.A., Esiri, M.M., Hiorns, R.W., Wilcock, G.K. & Powell, T.P.S. Anatomical
172, 293302 (2011). correlates of the distribution of the pathological changes in the neocortex in
109. Bryant, R. Post-traumatic stress disorder vs traumatic brain injury. Dialogues Clin. Alzheimer disease. Proc. Natl. Acad. Sci. USA 82, 45314534 (1985).
Neurosci. 13, 251262 (2011). 139. Lewis, D.A., Campbell, M.J., Terry, R.D. & Morrison, J.H. Laminar and regional
110. Sivanandam, T.M. & Thakur, M.K. Traumatic brain injury: a risk factor for distributions of neurofibrillary tangles and neuritic plaques in Alzheimers disease:
Alzheimers disease. Neurosci. Biobehav. Rev. 36, 13761381 (2012). a quantitative study of visual and auditory cortices. J. Neurosci. 7, 17991808
npg

111. Arnsten, A.F.T., Raskind, M., Taylor, F.B. & Connor, D.F. The effects of stress (1987).
exposure on prefrontal cortex: translating basic research into successful treatments 140. Guo, Z. et al. Head injury and the risk of AD in the MIRAGE study. Neurology
for post-traumatic stress disorder. Neurobiol. Stress 1, 8999 (2015). 54, 13161323 (2000).
This paper reviews how studies of stress effects in animals have led to successful 141. Altmann, A., Tian, L., Henderson, V.W., Greicius, M.D. & Alzheimers Disease
treatments for PTSD in humans. Neuroimaging Initiative Investigators. Sex modifies the APOE-related risk of
112. McKee, S. et al. A translational investigation targeting stress-reactivity and developing Alzheimer disease. Ann. Neurol. 75, 563573 (2014).
prefrontal cognitive control with guanfacine for smoking cessation. 142. Okawa, Y., Ishiguro, K. & Fujita, S.C. Stress-induced hyperphosphorylation of tau
J. Psychopharmacol. 29, 300311 (2015). in the mouse brain. FEBS Lett. 535, 183189 (2003).
113. Fox, H., Sofuoglu, M. & Sinha, R. Guanfacine enhances inhibitory control and 143. Bhalla, A. et al. The location and trafficking routes of the neuronal retromer and its
attentional shifting in early abstinent cocaine-dependent individuals. role in amyloid precursor protein transport. Neurobiol. Dis. 47, 126134 (2012).
J. Psychopharmacol. 29, 312323 (7 January 2015). 144. Cam, J.A. & Bu, G. Modulation of -amyloid precursor protein trafficking and
114. Connor, D.F., Grasso, D.J., Slivinsky, M.D., Pearson, G.S. & Banga, A. An open- processing by the low density lipoprotein receptor family. Mol. Neurodegener. 1,
label study of guanfacine extended release for traumatic stress related symptoms 8 (2006).
in children and adolescents. J. Child Adolesc. Psychopharmacol. 23, 244251 145. Yajima, R. et al. ApoE-isoform-dependent cellular uptake of amyloid- is mediated
(2013). by lipoprotein receptor LR11/SorLA. Biochem. Biophys. Res. Commun. 456,
115. Licznerski, P. & Duman, R.S. Remodeling of axo-spinous synapses in the 482488 (2015).
pathophysiology and treatment of depression. Neuroscience 251, 3350 (2013). 146. Renner, M. et al. Deleterious effects of amyloid oligomers acting as an
116. Savitz, J.B., Price, J.L. & Drevets, W.C. Neuropathological and neuromorphometric extracellular scaffold for mGluR5. Neuron 66, 739754 (2010).
abnormalities in bipolar disorder: view from the medial prefrontal cortical network. 147. Um, J.W. et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer
Neurosci. Biobehav. Rev. 42, 132147 (2014). a oligomer bound to cellular prion protein. Neuron 79, 887902 (2013).
117. Glantz, L.A. & Lewis, D.A. Decreased dendritic spine density on prefrontal 148. Doens, D. & Fernndez, P.L. Microglia receptors and their implications in the
cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 57, 6573 response to amyloid for Alzheimers disease pathogenesis. J. Neuroinflammation
(2000). 11, 48 (2014).
118. Black, J.E. et al. Pathology of layer V pyramidal neurons in the prefrontal cortex 149. Acin-Perez, R., Gatti, D.L., Bai, Y. & Manfredi, G. Protein phosphorylation and
of patients with schizophrenia. Am. J. Psychiatry 161, 742744 (2004). prevention of cytochrome oxidase inhibition by ATP: coupled mechanisms of
119. Arion, D. et al. Distinctive transcriptome alterations of prefrontal pyramidal energy metabolism regulation. Cell Metab. 13, 712719 (2011).
neurons in schizophrenia and schizoaffective disorder. Mol. Psychiatry 150. Melov, S. et al. Mitochondrial oxidative stress causes hyperphosphorylation of tau.
doi:10.1038/mp.2014.171 (6 January 2015). PLoS ONE 2, e536 (2007).

nature neuroscience VOLUME 18 | NUMBER 10 | OCTOBER 2015 1385

Anda mungkin juga menyukai