Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Review Biological

Psychiatry

Emerging Role for Nucleus Accumbens Medium


Spiny Neuron Subtypes in Depression
T. Chase Francis and Mary Kay Lobo

ABSTRACT
The ventral striatum (nucleus accumbens) and its role in mood, reward, and motivation has been the focus of
significant research. Despite this interest, little work has addressed cell type–specific distinctions in medium spiny
neurons (MSNs), the main projection neurons in the nucleus accumbens and dorsal striatum, and their function in
relation to stress and depression. Previous work has shown opposing roles for D1 and D2 receptor MSN subtypes in
depression-like outcomes to stress, particularly in regard to repeated neuronal stimulation and excitatory trans-
mission. Yet the mechanisms of action are still unknown. We discuss potential mechanisms by which MSN subtype
function promotes dichotomous behavioral outcomes caused by differences in cellular plasticity, subcellular
signaling pathways, and genetic expression. This review aims to address our current understanding about the role
of nucleus accumbens MSN subtypes in stress-related depression behavior and speculates on how currently
understood mechanisms contribute to factors that control the activity of MSNs.
Keywords: Deep brain stimulation, Excitatory synaptic transmission, Medium spiny neuron, Social defeat, Stress,
Striatum
http://dx.doi.org/10.1016/j.biopsych.2016.09.007

Depression affects a significant portion of the world’s pop- eventually driving the dorsolateral striatum to facilitate action-
ulation each year, and according to the World Health Organ- related behaviors (14,15). These local and global NAc circuit
ization, depression is the leading cause of disability worldwide. interactions mediate motivated and emotional behavior, making
Studies examining antidepressant efficacy show that 30% of them an attractive target for therapeutics. However, the causes
depressed patients fail to respond to common antidepressant of depression in relation to the NAc are muddled because of the
treatments (1), suggesting a need for new targeted therapies. diversity of connections and cell types.
Treatments with enhanced spatial specificity, such as deep There is a growing understanding that morphologically
brain stimulation (DBS) or transcranial magnetic stimulation, similar but transcriptionally distinct neuronal subtypes show
aim to restore region-specific hypoactivity and show great differing patterns of information processing and integration.
promise in alleviating even the most severe cases of Despite this complication, expression dissimilarity provides an
treatment-resistant depression (2–10). Therefore, targeted exceptional opportunity to differentially target cell types using
therapy to affect activity of these regions may be effective in drug ligands for unique receptors and other molecular targets
treating depression. or expression constructs in both animals and humans. The
While acute stress can be adaptive, chronic stress can be NAc is composed of a vast array of transcriptionally distinct
biologically damaging, making it a leading factor in the develop- neuronal subtypes, where medium spiny neurons (MSNs)
ment of depression (11). The overwhelming majority of depres- make up the majority (.95%) of the cells (16). These neurons
sion models use repeated stress to produce depression-like are defined by their transcriptional profiles, including D1
symptomology in rodent and primate models. Stress promotes receptor versus D2 receptor expression, substance P and
central and peripheral adaptations in body systems via the enkephalin, and other enriched genes (16–18). NAc D1-MSN
hypothalamic-pituitary-adrenal axis, which feeds back on the and D2-MSNs are also distinguished by their projections to
brain through corticosteroid release (12). Significant focus has target regions: D1-MSNs target the ventral tegmental area
been placed on excitatory forebrain structures (13), including (VTA) and ventral pallidum (VP), whereas D2-MSNs specifically
the hippocampus, amygdala, and prefrontal cortex, among target the VP (Figure 1) (19–21). Input and output from the NAc
other regions that mediate emotional memory and anxiety. follows a dorsolateral to ventromedial topology (15). The NAc
After repeated stress, these regions show dramatic deficits in core and dorsolateral regions of the NAc project mainly to the
local and efferent excitatory signaling to integration regions, dorsolateral VP and substantia nigra, whereas the NAc medial
such as the nucleus accumbens (NAc) (11). NAc integration of shell projects mainly to the ventromedial VP and VTA (20).
this excitatory input triggers emotionally motivated behaviors Molecular expression, synaptic plasticity, and structural
through corticostriatal thalamic loops within the basal ganglia, plasticity changes in NAc MSNs are implicated in depression

SEE COMMENTARY ON PAGE 636

& 2017 Society of Biological Psychiatry. 645


ISSN: 0006-3223 Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal
Biological
Psychiatry NAc MSN Subtypes in Depression

D1-MSN
D2-MSN
Glutamatergic
mPFC GABAergic vHipp
Dopaminergic
Peptidergic VTA
SN

dHipp

Thalamus
NAcC
BLA VP

LH NAcS
Arc

Figure 1. Afferent and efferent connections of the nucleus accumbens (NAc). (Top left) The prelimbic cortex of the medial prefrontal cortex (mPFC)
innervates the NAc core (NAcC), and the infralimbic region of the mPFC innervates the NAc shell (NAcS). (Bottom left) Thalamic and basolateral amygdala
(BLA) inputs do not display preferential innervation for NAc subregions. The dorsal hippocampus (dHipp) preferentially innervates the lateral NAcS. The
arcuate nucleus (Arc) of the hypothalamus innervates shell regions of the NAc with melanocortin input. The lateral hypothalamus (LH) is innervated
preferentially by D1 receptor medium spiny neurons (D1-MSNs). (Center) NAc MSNs display a dorsolateral to ventromedial innervation and projection
topology. NAcC D1-MSNs project to the substantia nigra reticulata (SN), and D1-MSNs and D2-MSNs project to dorsolateral regions of the VP. NAcS
D1-MSNs project to the ventral tegmental area (VTA), and both subtypes in the NAcS project to the ventromedial ventral pallidum (VP). (Top right) Ventral
hippocampus (vHipp) projects preferentially to ventromedial NAc, and the VTA provides diffuse terminals to all of the NAc. (Bottom right) Lateral ventral
pallidum regions project to the SN, and medial ventral pallidum projections project to the VTA. GABAergic, gamma-aminobutyric acidergic.

symptomology (22,23). Despite a wide array of research transmission hypothesis of depression (22,32,33). Conven-
showing differential and often opposing roles of D1- and D2- tional antidepressant therapeutics, including selective seroto-
MSNs in motivation and reinforcement (24–31), little research nin reuptake inhibitors (SSRIs), or nontypical therapeutics,
has been devoted to understanding how theses MSN sub- including ketamine and brain stimulation, may alleviate depres-
types are regulated after stress (Table 1). We review the sion symptoms by acting on excitatory brain systems (33). It is
current literature discussing molecular and cellular investiga- known that excitatory synaptic transmission is impaired after
tion of these MSN subtypes to understand and treat stress in many frontal limbic brain regions, including the
depression-like symptoms after behavioral stress. We focus prefrontal cortex and hippocampus (22). Stress can lead to
on how these changes or treatments may influence excitatory functional decreases in activity across these regions through
plasticity at subtype-specific synapses and potentially alter structural dendritic spine and arbor atrophy and disruption in
the activity of these neurons in order to promote depression synaptic plasticity and intrinsic cellular mechanisms (22). These
symptomology. In addition, we discuss how brain stimulation excitatory deficits extend to the NAc in human depressed
may interact with excitatory and modulatory circuits to influ- patients, where reduced activity and overall volume is observed
ence this behavior. Finally, we touch on how modulatory in imaging studies (34). In addition, a recent study noted stress-
factors and underlying molecular expression patterns may induced volume reduction in the NAc of mice (35).
govern these cell type–specific changes and provide an array Electrophysiological studies of NAc excitatory transmission
of possible drug targets to alleviate depression. after stress reveal stress model–dependent and subregion-
specific outcomes. Lim et al. found that excitatory synaptic
transmission after repeated restraint stress promoted long-
STRESS-INDUCED ALTERATIONS IN NAc MSN term depression in D1-MSNs but not D2-MSNs of the NAc
SUBTYPE EXCITATORY TRANSMISSION core, suggesting that a D1-MSN–specific change in excitatory
Stress-induced dysfunction in forebrain excitatory transmis- transmission mediates anhedonic outcomes (36). Mice that
sion has led to the development of the excitatory synaptic have been subjected to chronic social defeat stress (CSDS)

646 Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal


Biological
NAc MSN Subtypes in Depression Psychiatry

Table 1. List of Current Studies Examining D1-MSNs and D2-MSNs in Stress-Related Behaviors
Strain Age,
Reference Genotype Background Sex Weeks Finding
Barik et al., 2013 (64) Nr3c1(loxP/loxP) 3 D1-Cre C57BL/6 Male 8–16 Glucocorticoid receptor knockout in D1-MSNs prevents
susceptibility to CSDS
Dias et al., 2014 (92) D1-Cre, D2-Cre, D1-GFP, and C57BL/6 Male 7–9 Studied role of β-catenin in promoting resilience to CSDS
D2-GFP
Francis et al., 2015 (41) D1-Cre and D2-Cre C57BL/6 Male 8–14 Optogenetic stimulation and chemogenetic inhibition oppositely
facilitates depressive-like behavior in MSN subtypes
Francis et al., 2015 (41) D1-GFP and D2-GFP C57BL/6 Male 8–14 Chronic stress oppositely regulates frequency of excitatory
transmission; stress increases intrinsic excitability in D1-MSNs
Joffe et al., 2016 (52) D1-Cre and A2a-Cre C57BL/6 Male and 6–12 Characterized behavioral despair in D1-MSN or D2-MSN
female NR1 knockout
Lim et al., 2013 (36) D1-tdTomato 3 D2-GFP C57BL/6 Male 6–8 Anhedonia associated with D1-MSN reduction in synaptic
transmission
Lobo et al., 2013 (84) D1-GFP and D2-GFP C57BL/6 Male 8–10 ΔFosB differentially regulated in D1-MSNs and D2-MSNs
after stress
Plattner et al., 2015 (63) D1-Cre 3 conditional Cdk5 C57BL/6 Male 12–14 Explored the role of D1-MSNs Cdk5 in stress
knockout
Vialou et al., 2010 (82) NSE-tTA 3 TetOp-ΔFosB C57BL/6 Male 8–11 Examined role of ΔFosB in mediating resilience to stress
(line 11A, D1-MSN
enriched)
CSDS, chronic social defeat stress; D1, D1 receptor; D2, D2 receptor; MSN, medium spiny neuron.

have a similar outcome in D1-MSNs. CSDS is a validated observed between interaction behavior and D1-MSN but not
stress model that results in mice displaying susceptibility (i.e., D2-MSN action potential firing (Figure 2C). Given the enhance-
depression-like behavior) or resilience (i.e., normal behavior) ment in excitability and despite the reduced frequency of input
(37,38). Susceptible mice have an increased number of stubby in susceptible mice, perhaps during a salient, NAc-stimulating
spines in NAc MSNs, which correlates with an increased event, D1-MSNs may display enhanced firing in a state-
frequency of miniature excitatory postsynaptic currents dependent fashion via an increase the probability of D1-MSN
(mEPSCs) (39). However, it is unknown which specific MSN firing. More research—particularly cell type–specific in vivo
subtype alone expresses this change—if either. Cell type– analysis of neuronal activity—is necessary to determine how
specific analysis of NAc shell thin and mushroom spines show these intrinsic and synaptic properties interact during specific
enhanced synaptic strength of D1-MSN mushroom spines behavioral events to mediate depressive-like outcomes.
from resilient animals and reduced strength of D2-MSN mush-
room spines (40). While the results suggest that resilient mice
display opposing adaptive mechanisms at single MSN sub-
DBS AND THE BALANCING ACT OF NAc MSN
types, more holistic studies are needed to analyze cell type– SUBTYPE NEURONAL STIMULATION IN
specific synaptic changes as they relate to morphological DEPRESSION BEHAVIOR
changes and cellular output. Researchers use brain stimulation techniques, such as trans-
We recently found that D1-MSNs have reduced mEPSC cranial magnetic stimulation and DBS, to target hypoactive
frequency and D2-MSNs have enhanced frequency of brain regions in depressed patients (2,42,43). High-frequency
mEPSCs in CSDS-susceptible mice (41). To better understand DBS of the NAc, among other regions, shows great promise in
how changes in mEPSC frequency were related to suscept- alleviating treatment-resistant depression, but its mechanism
ibility outcomes, we used previously collected data from is still unclear (5,8,10). Electrical DBS targets cells without
Francis et al. (41) and conducted correlational analyses regard to subtype specificity and may stimulate afferents
between excitatory transmission on MSN subtypes and social within these regions, such as medial prefrontal cortex (mPFC)
interaction times—the primary CSDS behavioral metric. We or ventral hippocampal (vHipp) inputs. Diffuse or nonspecific
found that D2-MSN mEPSC frequency negatively and strongly stimulation makes it particularly difficult to uncover neuronal
correlates with interaction time, while no correlation is mechanisms promoting antidepressant outcomes.
observed in D1-MSNs (Figure 2A). These data suggest that First, it is still unclear how synaptic plasticity, at specific
D2-MSNs, not D1-MSNs, may convey the change in fre- excitatory synapses, mediates stress-induced behavioral out-
quency of mEPSCs observed in non-cell type–specific MSN comes. These studies provide the initial means to dissect brain
recordings (39). In addition, increased frequency may enhance changes caused by DBS. Despite common cellular and
baseline activity of D2-MSNs in susceptible mice, while D1- physiological changes in excitatory NAc-projecting brain
MSN excitatory activity is dampened. A positive correlation is regions, studies examining specific excitatory inputs to the
observed between interaction time and intrinsic excitability of NAc produce varying behavioral outcomes, suggesting con-
D1-MSNs, while D2-MSNs show no change in intrinsic pro- trasting synaptic plasticity at these synapses. For instance,
perties (Figure 2B), linking D1-MSN intrinsic excitability to burst-like 100-Hz stimulation of the prefrontal cortex promotes
susceptibility. In addition, a strong negative correlation was antidepressive outcomes in mice (44,45) and “high-frequency”

Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal 647


Biological
Psychiatry NAc MSN Subtypes in Depression

Figure 2. Electrophysiological prop-


erties correlate with stress-induced
behavioral changes in medium spiny
neuron (MSN) subtypes. (A) D2 recep-
tor medium spiny neurons (D2-MSN)
miniature excitatory postsynaptic cur-
rent (mEPSC) frequency displays a
strong negative correlation with time
in interaction zone (Pearson’s r12 5
–.750, p 5 .003, n 5 13), and D1
receptor medium spiny neurons (D1-
MSNs) show no correlation (Pearson’s
r12 5 .180, p 5 .557, n 5 13).
(B) A moderate positive correlation
between D1-MSN rheobase and time
in interaction zone was observed
(Pearson’s r25 5 .573, p 5 .002,
n 5 26), and D2-MSNs displayed no
correlation (Pearson’s r15 5 –.195,
p 5 .470, n 5 16). (C) In agreement
with rheobase measures, correlations
between the number of spikes in
D1-MSNs (elicited by a 250-pA,
500-ms square pulse of current) and
time in the interaction zone negatively
correlated (Pearson’s r10 5 –.668,
p 5 .025, n 5 11); a correlation was
not observed between D2-MSNs
spiking and time in the interaction
zone (Pearson’s r105 –.049, p 5
.879, n 5 11). Each point represents
the average rheobase, miniature exci-
tatory postsynaptic current frequency,
or number of spikes for each indivi-
dual mouse. All mice in this study
were 8–14 weeks of age and were
D1-GFP or D2-GFP expressing male
mice on a C57 wild-type background.

20-Hz transcranial magnetic stimulation of the prefrontal weighted differently in a subregion-specific manner, where
cortex in humans is antidepressant (46). This stimulation in mice larger vHipp excitatory currents in the NAc shell are observed
is specific to mPFC-NAc, but not mPFC-amygdala input (45). (49). In addition, these synapses have larger N-methyl-
In comparison, increasing activity of mPFC-NAc inputs via D-aspartate currents that may enhance the probability of
4-Hz optogenetic stimulation promotes resilience to CSDS (47). plasticity because N-methyl-D-aspartate receptors drive spike
It is unclear which MSN subtype this activity affects or how timing–dependent calcium-dependent potentiation on MSN
these inputs interact with other synaptic inputs. subtypes (50). Interestingly, a variety of N-methyl-D-aspartate
Next, the stimulation of different inputs drives opposite receptor antagonists, including ketamine, have shown efficacy
behaviors. In contrast to prefrontal cortex-NAc afferents, in treating treatment-resistant depression (51), perhaps by
enhancing hippocampal and thalamic input to the NAc pro- disrupting plasticity in depression-related regions, including
motes depressive-like outcomes. Increased synaptic strength the vHipp (33). The antidepressant effects of N-methyl-
is observed on intralaminar thalamic NAc inputs, and driving D-aspartate receptor antagonism may partially be expressed
the thalamic inputs promotes susceptibility to CSDS (48). in the NAc by targeting excitatory input to distinct NAc
Enhancing the activity of vHipp inputs has similar out- circuits. For instance, knockout of NR1 from adenosine
comes (47). In contrast, 1-Hz–mediated long-term depression receptor type 2a–expressing cells (i.e., D2-MSNs in the NAc
of vHipp inputs to the NAc in CSDS mice restores normal but not D1-MSNs) reduces immobility in the forced swim
social interaction (47). Interestingly, vHipp-NAc projections are test (52). Taken together, resilience-promoting vHipp-NAc

648 Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal


Biological
NAc MSN Subtypes in Depression Psychiatry

long-term depression may preferentially target excitatory DBS itself may also act as a neuromodulator for excitatory
activity on NAc shell D2-MSNs. This manipulation may activity, facilitating or inhibiting neuronal transmission directly
balance transmission between MSN subtypes or weight input or by the release of modulatory neurotransmitters and
to D1-MSNs, thereby restoring normal behavior. peptides.
Our recent results indicate that specific NAc pathway
stimulation may best account for the effects of NAc-specific
DBS antidepressant outcomes. We based our stimulation
study on the idea that excitatory transmission was oppositely STRESS-INDUCED MODULATORY ALTERATIONS IN
changed in NAc MSN subtypes, and predicted that stimulation THE NAc
of D1-MSNs would promote antidepressant outcomes and Modulatory factors contribute to neuronal function and aid in
that the stimulation of D2-MSNs would promote depression amplifying or dampening neuronal excitatory transmission and
symptoms. Using optogenetics, NAc neurons were specifically eventually cellular output. The NAc and its targets are littered
targeted for stimulation (41). Like DBS, repeated high-frequency with potential modulatory drug targets, including peptides,
stimulation of all NAc neurons alleviated depression-like out- growth factors, and other neuromodulatory factors that modify
comes to CSDS. It is likely that D1-MSNs mediate this effect, cell firing properties and synaptic transmission (59). The
because cell type–specific stimulation of D1-MSNs mimicked balance of this chemical cocktail may produce drastic
nonspecific NAc neuronal stimulation (41). In addition, stress-dependent behavioral alterations. Pharmacological
repeated but not acute stimulation is necessary for this and optogenetic studies have attempted to tease out the
behavioral effect. It is unclear if direct elevation of D1-MSN roles of these substances and enlighten our understanding of
activity during stimulation mediates this effect. Several mech- cell type–specific control of stress-related behaviors.
anisms could exist to govern these changes. The most favored Dopamine (DA), a potent neuromodulator in the NAc, is
mechanism is one of homeostatic plasticity. Perhaps it is not important for the expression of depression-related symptoms
stimulation of D1-MSNs but rather a dampening of intrinsic (e.g., reward, hedonia, and motivation) (60). The NAc receives
excitability that negates the ill effects of stress. Intrinsic dense innervation from the VTA, which supplies the region
properties driving cell firing are complicated and governed with DA, gamma-aminobutyric acid, and brain-derived neuro-
by a variety of complex and interacting mechanisms, including trophic factor (BDNF). MSN enrichment with either D1 or D2
alterations in potassium channel function and homeostatic receptors and the opposing nature of DA receptor signaling
excitatory balance (53). For instance, repeated VTA optoge- cascades suggests that DA may act to oppositely regulate
netic stimulation is sufficient to dampen phasic firing of VTA subtype-specific MSN output after stress. Chronic mild stress
neurons through alterations in hyperpolarization activated promotes altered NAc D2 receptor sensitivity (61), and restraint
current potassium conductance (54), promoting resilience in stress studies have revealed that NAc D1-like receptor density
CSDS mice, and providing a potential parallel in mechanism. increases (62). In addition, cyclic adenosine monophosphate/
Alternatively, optogenetic stimulation allows for dendritic, protein kinase A signaling, activated by dopamine receptor
somatic, and axonal firing at high rates (55). In particular, signaling, plays a role in stress. NAc D1-MSN–specific knock-
axonal stimulation may drive higher fidelity firing rates by out of CDK5 reduces cyclic adenosine monophosphate levels,
resisting amplitude dampening seen in the soma of cells (56). increases protein kinase A activity, and promotes antidepres-
Therefore, stimulation may be promoting plasticity in down- sant outcomes (63). Enhanced DA release is observed after a
stream regions, such as the VP or VTA. Another possibility is single social defeat stress episode (64), suggesting that acute
that collateral inhibitory firing between D1-MSNs and D2-MSNs high-intensity stressors promote DA release. These results
may serve to balance D1-MSN and D2-MSN output (57,58). further suggest that enhancing D1 receptor signaling on D1-
D2-MSN stimulation promotes depressive-like outcomes (41), MSNs promotes stress-induced susceptibility and that high
suggesting that an imbalance in firing of MSN subtypes could DA tone may act as a catalyst for excitatory plasticity in the
mediate these symptoms. D2-MSN collaterals connect more NAc. The effect of chronic stress on VTA DA neuron firing is
strongly to D1-MSN than D1-MSNs connect to D2-MSN (57). lasting (37,65,66), is driven by the activity of intrinsic potas-
Therefore, depressive-like behavior induced by repeated D2- sium channels (54), and phasic but not tonic optogenetic
MSN stimulation may rebalance activity toward D2-MSNs. stimulation of VTA-NAc projecting neurons promotes suscept-
Consequently, repeated stimulation of these subtypes may act ibility to CSDS (54,66–68). It would therefore seem appropriate
through different but overlapping mechanisms in which a to conclude that DA tone is enhanced after chronic stress and
homeostatic mechanism is favored in antidepressant that lower-affinity D1 receptors would be activated by phasic
D1-MSN stimulation and an imbalanced excitatory signaling DA release, thereby preferentially enhancing activity of
mechanism favors depressant D2-MSN stimulation. Likewise, D1-MSNs. However, this effect is not as straightforward as it
repeated inhibition may facilitate a homeostatic process to seems. It may appear that DA, potentially through D1-MSNs,
enhance intrinsic excitability, because we were able to shift provides temporary and acute relief from depression sympto-
resilient mice to a depressed state with repeated D1-MSN mology, while other factors drive the long-lasting depressive
chemogenetic inhibition, but the inhibition of D2-MSNs state. First, phasic firing changes in VTA DA neurons are
had no effect (41). This bidirectional manipulation of inconsistent across stress models (66,69). In addition, a recent
behavior promotes the idea that D1-MSNs may provide a paper using DA voltammetry in the NAc discovered that VTA
reversible switch for depression-like behavior, while aberrant DA release was unaltered after CSDS and that the blockade of
D2-MSN activity may be involved in the transition to or DA receptors had no effect on depression-like outcomes
expression of a depression state. Brain stimulation or (67). Rather, BDNF was responsible for stress susceptibility;

Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal 649


Biological
Psychiatry NAc MSN Subtypes in Depression

antagonism of the BDNF receptor TrkB prevented suscepti- NAc subtypes. That is, dynorphin signaling may oppose and
bility (37,38,54,65,67,68). provide balance for D1-MSN firing and enkephalin signaling
Consistent with these findings, BDNF is observed to be may oppose enhanced D2-MSN activity. This interpretation
increased in the NAc of human depressed patients (37). would be consistent with observed excitatory changes on
Stress-induced BDNF release primarily occurs between the NAc MSN subtypes and stimulation-mediated antidepressant
VTA-NAc circuit, and VTA-specific knockout of BDNF pro- outcomes (41).
motes resilience to CSDS (38). Increased BDNF and TrkB
receptor activation are observed in NAc tissue from suscep-
tible mice (37). These results provide strong evidence for the
role of VTA-supplied NAc BDNF in depression. TrkB is MOLECULAR CORRELATES REGULATING STRESS-
enriched in D2-MSNs and BDNF signaling through TrkB has INDUCED ACTIVITY OF MSN SUBTYPES
different roles in NAc MSN subtypes in psychostimulant and Many of the aforementioned cellular signaling cascades con-
opiate motivation (26,70), but depression-related BDNF signal- verge on one another or are activated in conjunction or in
ing in MSN subtypes is unclear. BDNF is highly involved in parallel with other cascades to coordinate molecular expres-
structural remodeling and plasticity. Perhaps the structural sion and cellular output. The need for new molecular targets is
plasticity observed increases in NAc stubby spine growth (48) growing, and despite the emergence of various molecular
can be explained by a persistent release of BDNF and ongoing tools to probe specific cell subtypes, little research has been
remodeling of synapses, in a D2-MSN–specific manner. conducted examining molecular expression profiles in MSN
BDNF interacts with a variety of other signaling pathways subtypes after stress. Observed molecular expression in one
to promote these depression-like effects, including factors MSN subtype could be occluded by examining total NAc
involved in glucocorticoid signaling. The hypothalamic- expression. Despite the lack of research, information can be
pituitary-adrenal axis acts to release corticosterone in mice, gleaned from analysis of molecular manipulation in all MSNs.
which can feedback on neurons within the brain (12). Gluco- Significant research on the immediate early gene ΔFOSB
corticoid receptor knockout specifically in NAc D1-MSNs, but has demonstrated its role in resilience to stress. NAc ΔFOSB
not VTA-DA neurons, promotes resilience to CSDS (64), and expression is significantly reduced in human depressed
heightened DA release is blunted after an acute social defeat patients (82). In addition, NAc ΔFOSB expression is signifi-
stress episode, suggesting that glucocorticoid signaling cantly enhanced in mice that are resilient but not susceptible
plays a dynamic role in affecting VTA signaling in the NAc. to CSDS (82). CSDS mice treated with the common SSRI
In addition, another molecule involved in stress signaling, fluoxetine display enhanced ΔFOSB expression and enhanced
corticotropin-releasing factor, enhances the release of BDNF resilience (82). In addition, NAc overexpression of ΔFOSB
in the NAc and promotes susceptibility to stress caused by the using a bitransgenic mouse line 11A promotes resilience to
paired induction of phasic firing and social stress (68). These CSDS, which is thought to be through enhancement of GluR2
results suggest that BDNF and glucocorticoids may coordi- expression (82). Interestingly, the 11A mouse line serendip-
nate molecular programming on distinct MSN subtypes to itously expresses ΔFOSB in D1-MSNs, suggesting that this
affect overarching stress-related behavior. resilience effect is primarily caused by D1-MSN enrichment of
Among the many neuropeptides found within the NAc, cell ΔFOSB (83) and a facilitation of excitatory activity. Compara-
subtype–enriched peptides, such as dynorphin and encepha- tively, ΔFOSB expression is enhanced in D1-MSNs of resilient
lin, play a role in depression (71,72). Dynorphin is enriched in mice and enhanced in D2-MSNs of susceptible mice in both
D1-MSNs of the striatum. Restraint stress, the forced swim NAc core and shell regions (84). These results further suggest
test, and inescapable shock increases NAc dynorphin (73) and that ΔFOSB may mediate adaptive resilience through D1-
NAc-specific infusion of kappa opioid receptor (i.e., the MSNs. Despite enhancements in overall resilience and exci-
primary target of dynorphin) antagonists has antidepressant tatory transmission after expression of ΔFOSB in all NAc
effects (74). In addition, the inhibition of dynorphin in the NAc neurons, overexpression in stress-naïve mice decreased exci-
produces antidepressant effects (75). Activation of kappa tatory synaptic strength in D1-MSNs via an increased number
opioid receptors promotes aversion, potentially through the of silent synapses and stubby spines, while in D2-MSNs,
regulation of DA terminals in the NAc (76). However, it is not overexpression increased excitatory transmission in the shell
simply an enhancement of dynorphin that promotes depres- and decreased silent synapses (85). These results seem
sion. Rather, activation of dynorphin cells can produce either at odds with previously mentioned stress-mediated results
aversive or reinforcing outcomes in a region-specific manner revealing the complexity of signaling within the NAc and MSN
(77), suggesting that heterogeneous populations of striatal subtypes and may reflect a stress-driven shift in baseline
MSNs may drive depression symptomology differentially. electrophysiological properties. During a stress state, ΔFosB
In contrast, enkephalin, which is highly enriched in striatal could facilitate differential plasticity in stressed and non-
D2-MSNs (17), is thought to be antidepressant. Chronic mild stressed animals. RAC1 messenger RNA expression, which
stress reduces NAc enkephalin (78), and chronic antidepres- is significantly decreased in human depressed patients, has
sant treatment increases striatal enkephalin levels (79). Ago- been shown to mediate changes in stubby spine formation in
nism of the delta-opioid receptor, the primary receptor target mice via an epigenetic mechanism (86). It is possible that
of enkephalin, also promotes antidepressant outcomes (80), ΔFosB and signaling pathways involved in regulating Rac1
while delta receptor knockout promotes depressive-like messenger RNA are coordinated to produce these results.
behavior (81). These results indicate that peptide release from More research is necessary to tease out the cell type
MSN subtypes may actively oppose activity-driving factors on specificity of these results.

650 Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal


Biological
NAc MSN Subtypes in Depression Psychiatry

ΔFOSB has been found to interact with molecules that in these cell types will better provide a clearer picture of NAc
regulate plasticity. CAMKII, a protein kinase, is sufficient to subtype function in depression.
activate long-term synaptic potentiation (87) and could play a
role in the storage of aversive memories during stress states.
ΔFOSB binds to the promoter of CamkIIα, and CSDS enhan-
ACKNOWLEDGMENTS AND DISCLOSURES
ces its binding enrichment on this promoter (88). Chronic
treatment with the SSRI fluoxetine reduces binding of ΔFOSB This work was supported by the National Institute of Mental Health Grant
No. RO1MH106500 (to MKL).
to the CamkII promoter, and NAc overexpression of CamkII We thank the members of the Lobo laboratory for their feedback on this
blocks the antidepressant efficacy of fluoxetine in stressed article.
mice. These effects are through an epigenetic mechanism of The authors report no biomedical financial interests or potential conflicts
reducing acetylation and increasing H3meK9 methylation. of interest.
SSRI treatment results may seem inconsistent with previous
literature discussing increased ΔFOSB expression in resilient
animals. It is possible that ΔFOSB differentially affects Cam-
ARTICLE INFORMATION
kIIα expression in MSN subtypes and mediates this curious
From the Department of Anatomy and Neurobiology, University of Maryland
effect. In that regard, enhanced binding of ΔFOSB to the
School of Medicine, Baltimore, Maryland.
CamkIIα promoter in D2-MSNs may induce structurally plas- Address correspondence to Mary Kay Lobo, Ph.D., University of
ticity and potentiation in these cells, weighting excitatory Maryland School of Medicine, Department of Anatomy and Neurobiology,
signaling in D2-MSNs more heavily. This outcome would be 20 Penn Street, HSF2, Rm 265, Baltimore, MD 21201; E-mail: mklobo@
consistent with our results showing enhanced excitatory som.umaryland.edu.
signaling to D2-MSNs after stress and other results showing Received June 16, 2016; revised Sep 10, 2016; accepted Sep 12, 2016.
overexpression of a constitutively active CaMKIIα promotes
plasticity (89), while developmental inhibition of CaMKIIα REFERENCES
reduces functional numbers of synapses (90). 1. Gaynes BN, Warden D, Trivedi MH, Wisniewski SR, Fava M, Rush AJ
These changes extend to other parallel, susceptibility- (2009): What did STAR*D teach us? Results from a large-scale,
promoting signaling cascades in D2-MSNs. Disheveled practical, clinical trial for patients with depression. Psychiatr Serv 60:
1439–1445.
(DVL1) activity inhibition in the WNT-DVL-β-CATENIN signaling
2. Mayberg HS, Lozano AM, Voon V, McNeely HE, Seminowicz D,
cascade promotes susceptibility to CSDS, and DVL1 is found Hamani C, et al. (2005): Deep brain stimulation for treatment-resistant
to be significantly downregulated in the NAc of depressed depression. Neuron 45:651–660.
human patients. Comparatively, pharmacological and domi- 3. Mayberg HS (2009): Targeted electrode-based modulation of neural
nant negative activity inhibition of the downstream inhibition circuits for depression. J Clin Invest 119:717–725.
target of DVL1, GSK3β, promotes resilience to CSDS (91). 4. Cusin C, Dougherty DD (2012): Somatic therapies for treatment-
resistant depression: ECT, TMS, VNS, DBS. Biol Mood Anxiety Disord
β-CATENIN is activated by the nonphosphorylated form of
2; 14-5380-2-14.
GSK3β. In fact, β-CATENIN expression in NAc D2-MSNs but 5. Bewernick BH, Kayser S, Sturm V, Schlaepfer TE (2012): Long-term
not D1-MSNs promotes resilience to CSDS (92). This effect was effects of nucleus accumbens deep brain stimulation in treatment-
specific to expression of a microRNA synthesis related protein resistant depression: Evidence for sustained efficacy. Neuropsycho-
DICER1 downstream of β-CATENIN signaling, suggesting that pharmacology 37:1975–1985.
posttranscriptional regulation of a wide variety of mRNAs by 6. Schlaepfer TE, Bewernick BH (2014): Neuromodulation for treatment
resistant depression: State of the art and recommendations for clinical
β-CATENIN in D2-MSNs in part mediate outcomes to social
and scientific conduct. Brain Topogr 27:12–19.
stress. 7. Bewernick B, Schlaepfer TE (2015): Update on neuromodulation for
treatment-resistant depression. F1000Res 4:10.12688/f1000research.
6633.1; eCollection 2015
8. Schlaepfer TE, Cohen MX, Frick C, Kosel M, Brodesser D, Axmacher
CONCLUSIONS AND FUTURE DIRECTIONS N, et al. (2008): Deep brain stimulation to reward circuitry alleviates
anhedonia in refractory major depression. Neuropsychopharmacology
There is emerging evidence for functional changes in MSN
33:368–377.
subtypes to differentially and in some cases oppositely mediate 9. George MS, Wassermann EM, Williams WA, Callahan A, Ketter TA,
outcomes to stress. While both subtypes appear to be involved Basser P, et al. (1995): Daily repetitive transcranial magnetic stim-
in and control stress-mediated depression symptomology, the ulation (rTMS) improves mood in depression. Neuroreport 6:
degree to which each subtype is involved remains questionable. 1853–1856.
The majority of these studies were conducted in male rodents 10. Williams ZM (2015): Good vibrations with deep brain stimulation. Nat
Neurosci 18:618–619.
despite the higher female incidence of depression (93). Cur-
11. Pittenger C, Duman RS (2008): Stress, depression, and neuroplas-
rently, only a handful of studies addressed how stress differ- ticity: A convergence of mechanisms. Neuropsychopharmacology 33:
entially affects the NAc of female animals (52,94–97) and only 88–109.
one provided a MSN subtype–specific examination. It is 12. Pariante CM, Lightman SL (2008): The HPA axis in major depression:
necessary to determine if sex-specific alterations in these cells Classical theories and new developments. Trends Neurosci 31:
account for the larger incidence of depression in females. 464–468.
13. Drevets WC, Price JL, Furey ML (2008): Brain structural and functional
Overall, the development of a cohesive model of stress-
abnormalities in mood disorders: Implications for neurocircuitry
induced changes in NAc MSN subtypes will require a multitude models of depression. Brain Struct Funct 213:93–118.
of factors and analyses. These factors along with differences in 14. Floresco SB (2015): The nucleus accumbens: An interface between
circuit function, cell signaling, and molecular genetic expression cognition, emotion, and action. Annu Rev Psychol 66:25–52.

Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal 651


Biological
Psychiatry NAc MSN Subtypes in Depression

15. Voorn P, Vanderschuren LJ, Groenewegen HJ, Robbins TW, Pennartz 37. Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ,
CM (2004): Putting a spin on the dorsal-ventral divide of the striatum. et al. (2007): Molecular adaptations underlying susceptibility and
Trends Neurosci 27:468–474. resistance to social defeat in brain reward regions. Cell 131:
16. Gerfen CR, Engber TM, Mahan LC, Susel Z, Chase TN, Monsma FJ Jr, 391–404.
et al. (1990): D1 and D2 dopamine receptor-regulated gene expression 38. Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ,
of striatonigral and striatopallidal neurons. Science 250:1429–1432. et al. (2006): Essential role of BDNF in the mesolimbic dopamine
17. Lobo MK, Karsten SL, Gray M, Geschwind DH, Yang XW (2006): pathway in social defeat stress. Science 311:864–868.
FACS-array profiling of striatal projection neuron subtypes in juvenile 39. Christoffel DJ, Golden SA, Dumitriu D, Robison AJ, Janssen WG, Ahn
and adult mouse brains. Nat Neurosci 9:443–452. HF, et al. (2011): IkappaB kinase regulates social defeat stress-
18. Heiman M, Schaefer A, Gong S, Peterson JD, Day M, Ramsey KE, induced synaptic and behavioral plasticity. J Neurosci 31:314–321.
et al. (2008): A translational profiling approach for the molecular 40. Khibnik LA, Beaumont M, Doyle M, Heshmati M, Slesinger PA, Nestler
characterization of CNS cell types. Cell 135:738–748. EJ, et al. (2016): Stress and cocaine trigger divergent and cell type-
19. Koob GF, Swerdlow NR (1988): The functional output of the meso- specific regulation of synaptic transmission at single spines in nucleus
limbic dopamine system. Ann N Y Acad Sci 537:216–227. accumbens. Biol Psychiatry 79:898–905.
20. Smith RJ, Lobo MK, Spencer S, Kalivas PW (2013): Cocaine-induced 41. Francis TC, Chandra R, Friend DM, Finkel E, Dayrit G, Miranda J, et al.
adaptations in D1 and D2 accumbens projection neurons (a dichot- (2015): Nucleus accumbens medium spiny neuron subtypes mediate
omy not necessarily synonymous with direct and indirect pathways). depression-related outcomes to social defeat stress. Biol Psychiatry
Curr Opin Neurobiol 23:546–552. 77:212–222.
21. Kupchik YM, Brown RM, Heinsbroek JA, Lobo MK, Schwartz DJ, 42. Holtzheimer PE 3rd, Mayberg HS (2010): Deep brain stimulation for
Kalivas PW (2015): Coding the direct/indirect pathways by D1 and D2 treatment-resistant depression. Am J Psychiatry 167:1437–1444.
receptors is not valid for accumbens projections. Nat Neurosci 18: 43. Delaloye S, Holtzheimer PE (2014): Deep brain stimulation in the
1230–1232. treatment of depression. Dialogues Clin Neurosci 16:83–91.
22. McEwen BS, Bowles NP, Gray JD, Hill MN, Hunter RG, Karatsoreos 44. Covington HE 3rd, Lobo MK, Maze I, Vialou V, Hyman JM, Zaman S,
IN, et al. (2015): Mechanisms of stress in the brain. Nat Neurosci 18: et al. (2010): Antidepressant effect of optogenetic stimulation of the
1353–1363. medial prefrontal cortex. J Neurosci 30:16082–16090.
23. Krishnan V, Nestler EJ (2008): The molecular neurobiology of depres- 45. Vialou V, Bagot RC, Cahill ME, Ferguson D, Robison AJ, Dietz DM,
sion. Nature 455:894–902. et al. (2014): Prefrontal cortical circuit for depression- and anxiety-
24. Kravitz AV, Tye LD, Kreitzer AC (2012): Distinct roles for direct and related behaviors mediated by cholecystokinin: Role of DeltaFosB.
indirect pathway striatal neurons in reinforcement. Nat Neurosci 15: J Neurosci 34:3878–3887.
816–818. 46. Levkovitz Y, Harel EV, Roth Y, Braw Y, Most D, Katz LN, et al. (2009):
25. Kravitz AV, Freeze BS, Parker PR, Kay K, Thwin MT, Deisseroth K, Deep transcranial magnetic stimulation over the prefrontal cortex:
et al. (2010): Regulation of Parkinsonian motor behaviours by opto- Evaluation of antidepressant and cognitive effects in depressive
genetic control of basal ganglia circuitry. Nature 466:622–626. patients. Brain Stimul 2:188–200.
26. Lobo MK, Covington HE 3rd, Chaudhury D, Friedman AK, Sun H, 47. Bagot RC, Parise EM, Pena CJ, Zhang HX, Maze I, Chaudhury D, et al.
Damez-Werno D, et al. (2010): Cell type-specific loss of BDNF (2015): Ventral hippocampal afferents to the nucleus accumbens
signaling mimics optogenetic control of cocaine reward. Science regulate susceptibility to depression. Nat Commun 6:7062.
330:385–390. 48. Christoffel DJ, Golden SA, Walsh JJ, Guise KG, Heshmati M, Friedman
27. Ferguson SM, Phillips PE, Roth BL, Wess J, Neumaier JF (2013): AK, et al. (2015): Excitatory transmission at thalamo-striatal synapses
Direct-pathway striatal neurons regulate the retention of decision- mediates susceptibility to social stress. Nat Neurosci 18:962–964.
making strategies. J Neurosci 33:11668–11676. 49. Britt JP, Benaliouad F, McDevitt RA, Stuber GD, Wise RA, Bonci A
28. Tai LH, Lee AM, Benavidez N, Bonci A, Wilbrecht L (2012): Transient (2012): Synaptic and behavioral profile of multiple glutamatergic
stimulation of distinct subpopulations of striatal neurons mimics inputs to the nucleus accumbens. Neuron 76:790–803.
changes in action value. Nat Neurosci 15:1281–1289. 50. Shen W, Flajolet M, Greengard P, Surmeier DJ (2008): Dichotomous
29. Bock R, Shin JH, Kaplan AR, Dobi A, Markey E, Kramer PF, et al. dopaminergic control of striatal synaptic plasticity. Science 321:
(2013): Strengthening the accumbal indirect pathway promotes resil- 848–851.
ience to compulsive cocaine use. Nat Neurosci 16:632–638. 51. Dang YH, Ma XC, Zhang JC, Ren Q, Wu J, Gao CG, et al. (2014):
30. Chandra R, Lenz JD, Gancarz AM, Chaudhury D, Schroeder GL, Targeting of NMDA receptors in the treatment of major depression.
Han MH, et al. (2013): Optogenetic inhibition of D1R containing Curr Pharm Des 20:5151–5159.
nucleus accumbens neurons alters cocaine-mediated regulation of 52. Joffe ME, Vitter SR, Grueter BA (2016): GluN1 deletions in D1- and
Tiam1. Front Mol Neurosci 6:13. A2A-expressing cell types reveal distinct modes of behavioral regu-
31. Chandra R, Francis TC, Konkalmatt P, Amgalan A, Gancarz AM, Dietz lation. Neuropharmacology 112(Pt A):172–180.
DM, et al. (2015): Opposing role for Egr3 in nucleus accumbens cell 53. Bean BP (2007): The action potential in mammalian central neurons.
subtypes in cocaine action. J Neurosci 35:7927–7937. Nat Rev Neurosci 8:451–465.
32. Sanacora G, Treccani G, Popoli M (2012): Towards a glutamate 54. Friedman AK, Walsh JJ, Juarez B, Ku SM, Chaudhury D, Wang J, et al.
hypothesis of depression: An emerging frontier of neuropsychophar- (2014): Enhancing depression mechanisms in midbrain dopamine
macology for mood disorders. Neuropharmacology 62:63–77. neurons achieves homeostatic resilience. Science 344:313–319.
33. Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, 55. Yizhar O, Fenno LE, Davidson TJ, Mogri M, Deisseroth K (2011):
Cai X (2015): An excitatory synapse hypothesis of depression. Trends Optogenetics in neural systems. Neuron 71:9–34.
Neurosci 38:279–294. 56. Meeks JP, Jiang X, Mennerick S (2005): Action potential fidelity during
34. Drevets WC, Videen TO, Price JL, Preskorn SH, Carmichael ST, normal and epileptiform activity in paired soma-axon recordings from
Raichle ME (1992): A functional anatomical study of unipolar depres- rat hippocampus. J Physiol 566:425–441.
sion. J Neurosci 12:3628–3641. 57. Taverna S, Ilijic E, Surmeier DJ (2008): Recurrent collateral connec-
35. Anacker C, Scholz J, O’Donnell KJ, Allemang-Grand R, Diorio J, tions of striatal medium spiny neurons are disrupted in models of
Bagot RC, et al. (2016): Neuroanatomic differences associated with Parkinson’s disease. J Neurosci 28:5504–5512.
stress susceptibility and resilience. Biol Psychiatry 79:840–849. 58. Dobbs LK, Kaplan AR, Lemos JC, Matsui A, Rubinstein M, Alvarez VA
36. Lim BK, Huang KW, Grueter BA, Rothwell PE, Malenka RC (2012): (2016): Dopamine regulation of lateral inhibition between striatal
Anhedonia requires MC4R-mediated synaptic adaptations in nucleus neurons gates the stimulant actions of cocaine. Neuron 90:
accumbens. Nature 487:183–189. 1100–1113.

652 Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal


Biological
NAc MSN Subtypes in Depression Psychiatry

59. Covington HE 3rd, Vialou V, Nestler EJ (2010): From synapse to 79. De Felipe MC, De Ceballos ML, Gil C, Fuentes JA (1985): Chronic
nucleus: Novel targets for treating depression. Neuropharmacology antidepressant treatment increases enkephalin levels in n. accumbens
58:683–693. and striatum of the rat. Eur J Pharmacol 112:119–122.
60. Dunlop BW, Nemeroff CB (2007): The role of dopamine in the 80. Broom DC, Jutkiewicz EM, Folk JE, Traynor JR, Rice KC, Woods JH
pathophysiology of depression. Arch Gen Psychiatry 64:327–337. (2002): Nonpeptidic delta-opioid receptor agonists reduce immobility in
61. Willner P, Muscat R, Papp M (1992): Chronic mild stress-induced the forced swim assay in rats. Neuropsychopharmacology 26:744–755.
anhedonia: A realistic animal model of depression. Neurosci Biobehav 81. Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F,
Rev 16:525–534. et al. (2000): Mice deficient for delta- and mu-opioid receptors exhibit
62. Cabib S, Giardino L, Calza L, Zanni M, Mele A, Puglisi-Allegra (1998): opposing alterations of emotional responses. Nat Genet 25:195–200.
Stress promotes major changes in dopamine receptor densities within 82. Vialou V, Robison AJ, Laplant QC, Covington HE 3rd, Dietz DM,
the mesoaccumbens and nigrostriatal systems. Neuroscience 84: Ohnishi YN, et al. (2010): DeltaFosB in brain reward circuits mediates
193–200. resilience to stress and antidepressant responses. Nat Neurosci 13:
63. Plattner F, Hayashi K, Hernandez A, Benavides DR, Tassin TC, Tan C, 745–752.
et al. (2015): The role of ventral striatal cAMP signaling in stress- 83. Kelz MB, Chen J, Carlezon WA Jr, Whisler K, Gilden L, Beckmann AM,
induced behaviors. Nat Neurosci 18:1094–1100. et al. (1999): Expression of the transcription factor deltaFosB in the
64. Barik J, Marti F, Morel C, Fernandez SP, Lanteri C, Godeheu G, et al. brain controls sensitivity to cocaine. Nature 401:272–276.
(2013): Chronic stress triggers social aversion via glucocorticoid 84. Lobo MK, Zaman S, Damez-Werno DM, Koo JW, Bagot RC, DiNieri
receptor in dopaminoceptive neurons. Science 339:332–335. JA, et al. (2013): DeltaFosB induction in striatal medium spiny neuron
65. Cao JL, Covington HE 3rd, Friedman AK, Wilkinson MB, Walsh JJ, subtypes in response to chronic pharmacological, emotional, and
Cooper DC, et al. (2010): Mesolimbic dopamine neurons in the brain optogenetic stimuli. J Neurosci 33:18381–18395.
reward circuit mediate susceptibility to social defeat and antidepres- 85. Grueter BA, Robison AJ, Neve RL, Nestler EJ, Malenka RC (2013):
sant action. J Neurosci 30:16453–16458. FosB differentially modulates nucleus accumbens direct and indirect
66. Chaudhury D, Walsh JJ, Friedman AK, Juarez B, Ku SM, Koo JW, pathway function. Proc Natl Acad Sci U S A 110:1923–1928.
et al. (2013): Rapid regulation of depression-related behaviours by 86. Golden SA, Christoffel DJ, Heshmati M, Hodes GE, Magida J, Davis K,
control of midbrain dopamine neurons. Nature 493:532–536. et al. (2013): Epigenetic regulation of RAC1 induces synaptic remod-
67. Koo JW, Labonte B, Engmann O, Calipari ES, Juarez B, Lorsch Z, eling in stress disorders and depression. Nat Med 19:337–344.
et al. (2015): Essential role of mesolimbic brain-derived neurotrophic 87. Lisman J, Schulman H, Cline H (2002): The molecular basis of CaMKII
factor in chronic social stress-induced depressive behaviors. Biol function in synaptic and behavioural memory. Nat Rev Neurosci 3:
Psychiatry 80:469–478. 175–190.
68. Walsh JJ, Friedman AK, Sun H, Heller EA, Ku SM, Juarez B, et al. 88. Robison AJ, Vialou V, Sun HS, Labonte B, Golden SA, Dias C, et al.
(2014): Stress and CRF gate neural activation of BDNF in the (2014): Fluoxetine epigenetically alters the CaMKIIalpha promoter in
mesolimbic reward pathway. Nat Neurosci 17:27–29. nucleus accumbens to regulate DeltaFosB binding and antidepressant
69. Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai HC, effects. Neuropsychopharmacology 39:1178–1186.
Finkelstein J, et al. (2013): Dopamine neurons modulate neural 89. Pi HJ, Otmakhov N, Lemelin D, De Koninck P, Lisman J (2010):
encoding and expression of depression-related behaviour. Nature Autonomous CaMKII can promote either long-term potentiation or
493:537–541. long-term depression, depending on the state of T305/T306 phos-
70. Koo JW, Mazei-Robison MS, Chaudhury D, Juarez B, LaPlant Q, phorylation. J Neurosci 30:8704–8709.
Ferguson D, et al. (2012): BDNF is a negative modulator of morphine 90. Klug JR, Mathur BN, Kash TL, Wang HD, Matthews RT, Robison AJ,
action. Science 338:124–128. et al. (2012): Genetic inhibition of CaMKII in dorsal striatal medium
71. Knoll AT, Carlezon WA Jr (2010): Dynorphin, stress, and depression. spiny neurons reduces functional excitatory synapses and enhances
Brain Res 1314:56–73. intrinsic excitability. PLoS One 7:e45323.
72. Lutz PE, Kieffer BL (2013): Opioid receptors: Distinct roles in mood 91. Wilkinson MB, Dias C, Magida J, Mazei-Robison M, Lobo M, Kennedy
disorders. Trends Neurosci 36:195–206. P, et al. (2011): A novel role of the WNT-dishevelled-GSK3beta
73. Shirayama Y, Chaki S (2006): Neurochemistry of the nucleus accum- signaling cascade in the mouse nucleus accumbens in a social defeat
bens and its relevance to depression and antidepressant action in model of depression. J Neurosci 31:9084–9092.
rodents. Curr Neuropharmacol 4:277–291. 92. Dias C, Feng J, Sun H, Shao NY, Mazei-Robison MS, Damez-Werno
74. Mague SD, Pliakas AM, Todtenkopf MS, Tomasiewicz HC, Zhang Y, D, et al. (2014): β-catenin mediates stress resilience through Dicer1/
Stevens WC Jr, et al. (2003): Antidepressant-like effects of kappa- microRNA regulation. Nature 516:51–55.
opioid receptor antagonists in the forced swim test in rats. 93. Kessler RC (2003): Epidemiology of women and depression. J Affect
J Pharmacol Exp Ther 305:323–330. Disord 74:5–13.
75. Newton SS, Thome J, Wallace TL, Shirayama Y, Schlesinger L, Sakai 94. Hodes GE, Pfau ML, Purushothaman I, Ahn HF, Golden SA, Christoffel
N, et al. (2002): Inhibition of cAMP response element-binding protein DJ, et al. (2015): Sex differences in nucleus accumbens transcriptome
or dynorphin in the nucleus accumbens produces an antidepressant- profiles associated with susceptibility versus resilience to subchronic
like effect. J Neurosci 22:10883–10890. variable stress. J Neurosci 35:16362–16376.
76. Chefer VI, Backman CM, Gigante ED, Shippenberg TS (2013): 95. Javadifar TS, Sahraei H, Ketabi MA, Nasehi M, Zarrindast MR (2016):
Kappa opioid receptors on dopaminergic neurons are necessary for Transient inactivation of the nucleus accumbens (NAc) shell prom-
kappa-mediated place aversion. Neuropsychopharmacology 38: inently ameliorates responses to acute stress in female rats. Brain Res
2623–2631. 1649(Pt A):1–8.
77. Al-Hasani R, McCall JG, Shin G, Gomez AM, Schmitz GP, Bernardi 96. LaPlant Q, Chakravarty S, Vialou V, Mukherjee S, Koo JW, Kalahasti
JM, et al. (2015): Distinct subpopulations of nucleus accumbens G, et al. (2009): Role of nuclear factor kappaB in ovarian hormone-
dynorphin neurons drive aversion and reward. Neuron 87: mediated stress hypersensitivity in female mice. Biol Psychiatry 65:
1063–1077. 874–880.
78. Poulin JF, Laforest S, Drolet G (2014): Enkephalin downregulation in 97. Campi KL, Greenberg GD, Kapoor A, Ziegler TE, Trainor BC (2014):
the nucleus accumbens underlies chronic stress-induced anhedonia. Sex differences in effects of dopamine D1 receptors on social
Stress 17:88–96. withdrawal. Neuropharmacology 77:208–216.

Biological Psychiatry April 15, 2017; 81:645–653 www.sobp.org/journal 653

You might also like