identification of the cortical neurons that mediate ... · mood is governed by the actions of a...

12
Identification of the Cortical Neurons that Mediate Antidepressant Responses Eric F. Schmidt, 1 Jennifer L. Warner-Schmidt, 2 Benjamin G. Otopalik, 1 Sarah B. Pickett, 1 Paul Greengard, 2 and Nathaniel Heintz 1, * 1 Laboratory of Molecular Biology, Howard Hughes Medical Institute 2 Laboratory of Molecular and Cellular Neuroscience The Rockefeller University, 1230 York Ave., New York, NY 10065, USA *Correspondence: [email protected] DOI 10.1016/j.cell.2012.03.038 SUMMARY Our understanding of current treatments for depres- sion, and the development of more specific thera- pies, is limited by the complexity of the circuits controlling mood and the distributed actions of anti- depressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically respon- sive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific dele- tion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepres- sants, and suggest that the regulation of seroto- nergic tone in this single cell type plays a pivotal role in antidepressant therapy. INTRODUCTION Mood is governed by the actions of a complex, anatomically dispersed circuit comprised of specific subpopulations of neurons arranged into functional units. Functional imaging studies of patients suffering from major depressive disorder (MDD), bipolar disorder, and anxiety indicate that a fundamental neural circuit controls emotion, with different elements of the circuit contributing to specific aspects of emotive behavior (Drevets et al., 2008; Mayberg, 2009). For example, fMRI and PET imaging of patients suffering from MDD has consistently demonstrated decreased prefrontal cortex function, and alterations in the activity of subcortical structures that include the basal ganglia, amygdala, and thalamus (Drevets, 2000; Mayberg et al., 1999). There is general agreement that in MDD, disease and treatment mechanisms impact the same basic circuitry controlling emotional state. However, it is evident from deep brain stimulation studies in a number of neurological and psychiatric disorders that the pathological mechanisms causing dysfunction and the immediate targets of clinical interventions most effective in alleviating symptoms may involve different brain structures. Given this realization, our under- standing of the pathophysiology of depression as well as the development of improved therapies for this disorder will be advanced by identification of cell types and molecular mechanisms responsible for generating depression-like pheno- types, as well as those mediating responses to antidepressant treatment. Recently, p11 (the protein product of the S100a10 gene) was found to be an important factor mediating antidepressant responses and depression-like states (Svenningsson et al., 2006). P11 is an adaptor protein that is expressed specifically in the central nervous system (CNS) (Egeland et al., 2011). It regulates serotonin signaling by binding to serotonin receptors (Htrs) 1b, 1d, and 4 and stabilizing the localization of these receptors at the cell surface (Svenningsson et al., 2006; Warner-Schmidt et al., 2009). Decreased p11 levels were found in the cortex of MDD patients, suicide victims, and a mouse model of depression (Anisman et al., 2008; Svenningsson et al., 2006). Chronic antidepressant treatment, electroconvul- sive therapy, and brain-derived neurotrophic factor treatment all result in increased p11 expression in the cerebral cortex (Svenningsson et al., 2006; Warner-Schmidt et al., 2010). Importantly, mice lacking p11 exhibit depressive-like behaviors, increased anxiety, and a blunted behavioral response to antidepressant treatment (Svenningsson et al., 2006; Warner- Schmidt et al., 2009). Antidepressant drugs target neuromodulatory systems that have widespread effects throughout the CNS, engaging recep- tors that are broadly expressed in the brain. Although the pharmacological effect of these drugs is immediate, there is a therapeutic delay of weeks to months before antidepressant activity is evident. This delay is thought to reflect neuroadaptive changes in pre- and postsynaptic cells, including long-term changes in gene expression and protein translation (Krishnan and Nestler, 2008). Although the cell types and precise molecular mechanisms mediating the efficacy of antidepressant drugs have not been identified, neuroimaging studies have shown that the clinical effects of antidepressant drug therapy and deep brain stimulation correlate with increased activity in the 1152 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

Upload: others

Post on 08-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

Identification of the Cortical Neuronsthat Mediate Antidepressant ResponsesEric F. Schmidt,1 Jennifer L. Warner-Schmidt,2 Benjamin G. Otopalik,1 Sarah B. Pickett,1 Paul Greengard,2

and Nathaniel Heintz1,*1Laboratory of Molecular Biology, Howard Hughes Medical Institute2Laboratory of Molecular and Cellular Neuroscience

The Rockefeller University, 1230 York Ave., New York, NY 10065, USA*Correspondence: [email protected]

DOI 10.1016/j.cell.2012.03.038

SUMMARY

Our understanding of current treatments for depres-sion, and the development of more specific thera-pies, is limited by the complexity of the circuitscontrolling mood and the distributed actions of anti-depressants. Although the therapeutic efficacy ofserotonin-specific reuptake inhibitors (SSRIs) iscorrelated with increases in cortical activity, the celltypes crucial for their action remain unknown. Herewe employ bacTRAP translational profiling to showthat layer 5 corticostriatal pyramidal cells expressingp11 (S100a10) are strongly and specifically respon-sive to chronic antidepressant treatment. Thisresponse requires p11 and includes the specificinduction of Htr4 expression. Cortex-specific dele-tion of p11 abolishes behavioral responses to SSRIs,but does not lead to increased depression-likebehaviors. Our data identify corticostriatal projectionneurons as critical for the response to antidepres-sants, and suggest that the regulation of seroto-nergic tone in this single cell type plays a pivotalrole in antidepressant therapy.

INTRODUCTION

Mood is governed by the actions of a complex, anatomically

dispersed circuit comprised of specific subpopulations of

neurons arranged into functional units. Functional imaging

studies of patients suffering from major depressive disorder

(MDD), bipolar disorder, and anxiety indicate that a fundamental

neural circuit controls emotion, with different elements of the

circuit contributing to specific aspects of emotive behavior

(Drevets et al., 2008; Mayberg, 2009). For example, fMRI and

PET imaging of patients suffering from MDD has consistently

demonstrated decreased prefrontal cortex function, and

alterations in the activity of subcortical structures that include

the basal ganglia, amygdala, and thalamus (Drevets, 2000;

Mayberg et al., 1999). There is general agreement that in MDD,

disease and treatment mechanisms impact the same basic

circuitry controlling emotional state. However, it is evident from

1152 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

deep brain stimulation studies in a number of neurological

and psychiatric disorders that the pathological mechanisms

causing dysfunction and the immediate targets of clinical

interventions most effective in alleviating symptoms may involve

different brain structures. Given this realization, our under-

standing of the pathophysiology of depression as well as

the development of improved therapies for this disorder will

be advanced by identification of cell types and molecular

mechanisms responsible for generating depression-like pheno-

types, as well as those mediating responses to antidepressant

treatment.

Recently, p11 (the protein product of the S100a10 gene) was

found to be an important factor mediating antidepressant

responses and depression-like states (Svenningsson et al.,

2006). P11 is an adaptor protein that is expressed specifically

in the central nervous system (CNS) (Egeland et al., 2011). It

regulates serotonin signaling by binding to serotonin receptors

(Htrs) 1b, 1d, and 4 and stabilizing the localization of these

receptors at the cell surface (Svenningsson et al., 2006;

Warner-Schmidt et al., 2009). Decreased p11 levels were found

in the cortex of MDD patients, suicide victims, and a mouse

model of depression (Anisman et al., 2008; Svenningsson

et al., 2006). Chronic antidepressant treatment, electroconvul-

sive therapy, and brain-derived neurotrophic factor treatment

all result in increased p11 expression in the cerebral cortex

(Svenningsson et al., 2006; Warner-Schmidt et al., 2010).

Importantly, mice lacking p11 exhibit depressive-like behaviors,

increased anxiety, and a blunted behavioral response to

antidepressant treatment (Svenningsson et al., 2006; Warner-

Schmidt et al., 2009).

Antidepressant drugs target neuromodulatory systems that

have widespread effects throughout the CNS, engaging recep-

tors that are broadly expressed in the brain. Although the

pharmacological effect of these drugs is immediate, there is

a therapeutic delay of weeks to months before antidepressant

activity is evident. This delay is thought to reflect neuroadaptive

changes in pre- and postsynaptic cells, including long-term

changes in gene expression and protein translation (Krishnan

andNestler, 2008). Although the cell types and precisemolecular

mechanisms mediating the efficacy of antidepressant drugs

have not been identified, neuroimaging studies have shown

that the clinical effects of antidepressant drug therapy and

deep brain stimulation correlate with increased activity in the

Page 2: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

cerebral cortex (Drevets et al., 2008; Mayberg, 2009). Given

these observations, and the dramatic regulation of p11 in the

frontal cortex of depressed patients, we wished to determine

whether p11-expressing cells in the cerebral cortex are critically

important for antidepressant action, and to search for beneficial

adaptations that may occur in these cells in response to chronic

antidepressant treatment.

For this study, we created an S100a10 bacTRAPmouse line to

characterize p11-expressing cells and measure their responses

to antidepressant treatment (Doyle et al., 2008; Heiman et al.,

2008). We report that p11 is highly enriched in layer 5 cortico-

striatal (CStr) projection neurons, that these cells respond prefer-

entially to chronic SSRI treatment by altering serotonergic tone,

and that loss of p11 in the cortex results in the inability to respond

to an SSRI. Our data demonstrate that the beneficial actions of

antidepressant therapy can be mediated by a single cell type

in the cerebral cortex, and suggest that development of drugs

that specifically target the activity of CStr neurons can result in

improved therapies for depression.

RESULTS

An S100a10 bacTRAP Transgenic LinePrevious in situ hybridization (ISH) studies have shown that the

S100a10 gene is expressed in the cerebral cortex (Arlotta

et al., 2005; Svenningsson et al., 2006), although the cell types

expressing S100a10 in layer 5 and their position in CNS circuitry

have not been determined. To identify this cell population and

study its properties using the translating ribosome affinity

purification (TRAP) approach, bacTRAP transgenic mice were

generated using a BAC clone containing the S100a10 gene.

The EGFP-L10a ribosomal protein fusion (Heiman et al., 2008)

was engineered into the S100a10 BAC by homologous recombi-

nation in Escherichia coli (Gong et al., 2002) so that it would be

expressed under the control of S100a10 regulatory elements

(Figure 1A). S100a10 bacTRAP founder line ES691 was chosen

for further analysis because the pattern of transgene expression

reliably recapitulated endogenous expression patterns of the

S100a10 gene throughout the brain (Figure S1 available online).

In the cortex, EGFP-L10a was expressed in a laminar

pattern (Figures 1B and 1C and S1A), consistent with previous

reports (Arlotta et al., 2005; http://www.gensat.org; http://

www.brain-map.org; http://www.stjudebgem.org). Transgene

expression was primarily restricted to superficial layer 5 (layer

5a), although a few cells with low levels of EGFP-L10a expres-

sion could be found in layers 4 and 5b. EGFP-L10a expression

was localized to the soma, proximal dendrites, and the nucle-

olus, as previously described for other bacTRAP mouse lines

(Doyle et al., 2008; Heiman et al., 2008). At high magnification,

it was evident that somas of EGFP-L10a+ cells were pyramidal,

with a clear apical dendrite (Figure 1D). As shown in Figure 1E, all

EGFP+ cells were also labeled with anti-p11, and the vast

majority of p11+ cells also expressed the fusion protein. These

results confirm previous studies demonstrating laminar expres-

sion of the S100a10 gene in the cortex, identify these cells as

layer 5a pyramidal cells, and demonstrate that the ES691

bacTRAP transgenic line faithfully reproduces expression of

endogenous S100a10 in the mouse cortex.

S100a10-Expressing Pyramidal Neurons Projectto the Dorsal Striatum and Contralateral CortexInspection of an S100a10 EGFP transgenic reporter line (http://

www.gensat.org) revealed axons within the corpus callosum

and striatum, and no labeling in the thalamus or pyramidal tract,

suggesting that the S100a10-expressing cells project primarily

to the striatum. To determine definitively the axonal targets of

the pyramidal cells labeled in the S100a10 bacTRAP line, retro-

grade tracing experiments were performed using fluorescently-

tagged cholera toxin b subunit (CTb). These studies demonstrate

that pyramidal cells labeled in the S100a10 bacTRAP line

provide a major afferent input to the dorsal striatum (caudate

putamen [CPu]). Thus, 72 ± 5% of the cortical neurons labeled

by injecting CTb into CPu were also EGFP-L10a+ (Figures 2A

and 2C). Injections into different areas of the CPu resulted in

labeling of S100a10 neurons in diverse areas of cortex, reflecting

the topographic map of CStr projections (Figure S2D). A large

proportion of cells that were double positive for CTb and

EGFP-L10a were also found in contralateral cortex following

injections into CPu (Figure 2C), indicating that the S100a10 cells

are capable of making both ipsilateral and contralateral projec-

tions to the dorsal striatum. The laminar distribution of EGFP-

L10a+ andCTb+ substantially overlapped (Figure 2B), consistent

with previous studies demonstrating that a majority of striatally

projecting cortical neurons are present in layer 5a.

Because the CPu and nucleus accumbens (NAc) are

known to be functionally distinct, and because the majority of

cortical inputs to the NAc arise from medial prefrontal cortical

areas, we employed CTb retrograde tracing to ask whether

S100a10-expressing neurons project to the ventral striatum.

Although CTb- and EGFP-L10a-labeled cell bodies were inter-

mingled in frontal cortex, there was minimal overlap between

accumbal projecting neurons and S100a10 cells (1.9 ± 1.7%

ipsilaterally and 3.9 ± 1.4% contralaterally; Figures 2A and 2C).

Rather, medial prefrontal S100a10 cells colabeled with injections

to anteromedial CPu whereas those in lateral regions sent

projections primarily to anterolateral CPu (Figure S2 and data

not shown). We conclude that the major afferent input from the

cortex to the CPu originates in S100a10-expressing pyramidal

cells, but that direct projections from the frontal cortex to the

NAc do not arise from this cell population.

As it is common for CStr neurons to send collateral projections

to contralateral cortex, we examined whether this was the

case for S100a10 cells. When CTb was injected into somato-

sensory cortex, CTb+ commissural projecting neurons (CPNs)

were widely distributed across the cortex and showed no

laminar patterning (Figures 2A and 2B). However, we found

that 66 ± 8% of CTb+ cells in layer 5 of contralateral cortex

were also EGFP+ (Figure 2C). Although we cannot definitively

conclude that individual S100a10-expressing neurons that

project to the striatum also send collaterals to contralateral

cortex, strong support for this suggestion comes from the fact

that the majority of layer 5 cells labeled by retrograde tracing

with CTb from either the dorsal striatum or the contralateral

cortex are EGFP-L10a+.

Finally, we note that retrograde tracing from the cervical spinal

cord revealed little overlap between corticospinal tract (CST)

projection neurons in layer 5b and S100a10-expressing cells

Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc. 1153

Page 3: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

WT

ba

cT

RA

P

GFP p11 Merge

1

Prefrontal Motor Somatosensory Visual

GFP

NeuN

A

C

D

B

E

p11

KO

/ba

cT

RA

P

A

’A/A’A/A

1 2 3

A’ EGFPL10a Ampr

R6kγ

pS296 vector

S100a10

// //

// //

BAC vector

Modified BAC vector

1 1

4

2/3

5a

5b

2/3

4

5a

6

2/3

1

5

6

5b

2/35

RecA +

Figure 1. The S100a10 bacTRAP Mouse Line Labels a Subpopulation of Pyramidal Cells in the Cerebral Cortex

(A) Schematic depicting themodification of the BAC vector containing the S100a10 gene by RecA-mediated homologous recombination in bacteria. Exons 1–3 of

the S100a10 gene are represented by black bars. (A) and (A0) refer to the homology region used for recombination. Upstream and downstream transcriptional

regulatory elements are represented by white bars on the BAC vector. The red box following the EGFP-L10a transgene represents the polyA site.

(B) Low magnification anti-EGFP IF image of EGFP-L10a transgene expression in the brain of S100a10 bacTRAP mouse. Scale bar represents 2 mm. See also

Figure S1.

(C) EGFP-L10a expression in multiple regions of neocortex. EGFP (green) and NeuN (red) staining are shown. Layers are indicated. Scale bar represents 500 mm.

(D) High magnification image of cell bodies of EGFP+ cells. Apical dendrites are indicated by double arrowheads (where visible). In cases where the apical

dendrite is out of the plane of section, single arrowheads indicate the base. Scale bar, 25 mm.

(E) Double-labeling with anti-EGFP and anti-p11 antibodies in WT (top row) and constitutive p11 KO (bottom row) in sensorimotor cortex of S100a10 bacTRAP

mice. Scale bar, 50 mm.

(Figures 2B and S2B), despite the fact that long distance projec-

tion neurons of this class are known to send axon collaterals

to the striatum. Taken together, our results demonstrate that

S100a10-expressing pyramidal cells constitute a major class of

layer 5 neurons whose projections are limited to forebrain struc-

tures (Figure 2D). The fact that these neurons send afferents

specifically to medial and lateral CPu, and that they do not

provide inputs to the NAc, is interesting given the concept of

multiple, parallel, and functionally segregated pathways from

the cerebral cortex to the striatum that has emerged from rodent

1154 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

and primate neuroanatomical studies over the last several

decades (Voorn et al., 2004).

S100a10-Expressing Cells and Glt25d2-ExpressingCells Are Molecularly DistinctTo compare themolecular properties of a secondwell character-

ized pyramidal cell type to the S100a10 CStr cell population, we

chose Glt25d2-expressing corticopontine neurons. Glt25d2-

expressing pyramidal cells are long-distance projection neurons

that reside in layer 5b and send axons to pons, thalamus, and

Page 4: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

Cell d

ep

th

(p

ia/w

m ratio

)

0

10

20

30

40

50

60

70

80

90

CPu NAc Cortex

Ipsilateral

Contralateral

Do

ub

le lab

eled

cells (%

)

CPu CT EGFP Merge

**

*

*

Contralateral cortex

**

*

*

*

NAc*

*

**

*

*

**

*

* ***

**

A

CBMotor Sensory

0 20 40 60

0

0.2

0.4

0.6

0.8

1

EGFP

CPu

SpC

EGFP

Cortex

Number of cells (%)

0 10 20 30 40

D

β

Figure 2. Retrograde Tracing Reveals Axonal Targets of S100a10-Expressing Cells(A) Representative images from CTb injections into dorsal striatum (CPu; top row), nucleus accumbens (NAc, middle row), and contralateral cortex (bottom row).

Left panels are low magnification images of cortex with the injection site shown in inset, and right three panels are higher magnification IF images of double-

labeling with anti-EGFP and anti-CTb antibodies. Arrows indicate double-labeled cells; asterisks indicate CTb+ cells that are not colabeled with EGFP.

(B) Histogram of the depth of the cell bodies of EGFP+ cells (EGFP), and CTb+ cells projecting to CPu or spinal cord (SpC) in motor cortex (left), and CTb+

commissural projection neurons (Cortex) in sensory cortex (right).

(C) Quantification (mean ± SEM) of the number of CTb+ cells that were colabeled with EGFP following injections into CPu, NAcc, and contralateral cortex.

(D) Schematic demonstrating the projection targets of S100a10-expressing cortical projection neurons. Brain diagram was modified from Franklin and Paxinos

(2008).

See also Figure S2.

spinal cord (Doyle et al., 2008; Groh et al., 2010). Their dendritic

architecture and electrophysiological properties distinguish

them fromCStr cells (Groh et al., 2010). Glt25d2 cells are present

deeper in layer 5 than S100a10 neurons in multiple areas of

cortex and cell somas of Glt25d2 cells are larger than those of

S100a10 cells (Figure S3), consistent with previous observations

(Groh et al., 2010).

The TRAP strategy allows identification of all proteins being

synthesized in any genetically targeted cell population, and alter-

ations of this translational profile in response to pharmacological

perturbations (Doyle et al., 2008; Heiman et al., 2008). To identify

moleculardistinctionsbetweenS100a10andGlt25d2-expressing

cells, we isolated polysome-bound mRNA from these cell types

andassayed global differences in gene expression usingmicroar-

rays. Because the S100a10 gene and the corresponding EGFP-

L10a bacTRAP construct are also expressed in the pia mater

and some blood vessels in the brain (Figures S4A–S4D), we

limited our analysis to a subset of 12,235 probe sets (representing

8,390genes) that areexpressedspecifically in neurons (TableS1).

As shown in Figure 3A, comparative analysis of mRNAs immuno-

precipitated (IP) from S100a10 cells versus those present in the

whole cortex (input) demonstrates selective enrichment of

Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc. 1155

Page 5: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

e671usW6D21pakA Depdc6

kiR32I050033DbmEixeD

101

102

103

104

101

102

103

S100a10 IP (normalized expression)Co

rte

x in

pu

t (n

orm

alize

d e

xp

re

ss

io

n)

S100a10

101

102

103

104

101

102

103

104

Glt25d2 IP (normalized expression)

S1

00

a1

0 IP

(n

orm

alize

d e

xp

re

ss

io

n)

S100a10

Glt25d2

JIH

A

C

E

B

D

F G

6

4

2

0

2

4

6S

100a10

Akap

12

Dep

dc6

D6W

su

176e

Dexi

Em

b

Glt25d

2

D330050123R

ik

Lo

g2 F

old

C

ha

ng

e

S100a10/Glt25d2

IP/UB

-4

-3

-2

-1

0

1

Ald

h1l1

Cn

p

Gad

1

Slc17a7

Etv1

Ntsr1

Lo

g2 F

old

C

ha

ng

e (IP

/U

B)

* * *

*

**

Figure 3. S100a10 and Glt25d2 Are Molecu-

larly Distinct Cell Populations

(A) Scatter plot comparing normalized expression

values of neuronal genes (see Figure S4 and Table

S1) in S100a10 IP (x axis) or whole cortex input

(y axis). Blue dots represent individual probe sets.

Outer gray lines represent 1.5-fold enrichment in

either sample. Probe sets representing the

S100a10 gene are circled.

(B) Quantification (mean ± SEM) of expression of

control genes (Aldh1l1, Cnp, Gad1, and Slc17a7)

and previously reported BAC driver genes (Etv1

and Ntsr1, gray bars) by qRT-PCR in S100a10 IP

versus whole cortex (UB) RNA. Dotted lines mark

2-fold regulation. *p < 0.05; **p < 0.01.

(C) Scatter plot comparing normalized expression

values of CST (red dots) or CPN (blue dots)

markers (see Figure S3 and Table S2) in Glt25d2

(x axis) or S100a10 (y axis) IP samples. Outer gray

lines represent 1.5-fold enrichment in either cell

population. Probe sets representing S100a10

and Glt25d2 are indicated.

(D) qRT-PCR quantification (mean ± SEM) of the

expression of cortex layer 5 genes in S100a10 IP

compared to either Glt25d2 IP (white bars) or

whole cortex (UB, gray bars). Positive values

indicate enrichment in S100a10 IP whereas

negative values indicate enrichment in UB or

Glt25d2 IP. Dotted lines mark 2-fold regulation.

p < 0.05 for all genes shown.

(E–J) In situ hybridizations showing expression

patterns in cortex of genes enriched in S100a10

(E–I) or Glt25d2 (J) cells. All images are from the

Allen Brain Atlas.

S100a10 probe sets in the IP (7.5- and 5.9-fold). Quantitative RT-

PCR (qRT-PCR) of TRAP RNA showed that mRNA for the vesic-

ular glutamate transporter, Slc17a7, was expressed (although

not enriched compared to neighboring cells), confirming that the

S100a10 cells are indeed excitatory neurons (Figure 3B). In

contrast, mRNAs specific to astrocytes (Aldh1l1), oligodendro-

cytes (Cnp), and GABAergic interneurons (Gad1) were expressed

at very low levels compared to the input (Figure 3B). mRNA for

genes known to label other pyramidal cell types (Doyle et al.,

2008), including layer 6 corticothalamic (Ntsr1), a subpopulation

of layer 5 CStr (Etv1), and Glt25d2 were not enriched in IP

mRNA (Figures 3B and 3D), suggesting that the S100a10 cells

are a unique, previously uncharacterized cell population.

To assess whether other genes enriched in the S100a10 IP

distinguish these cells from Glt25d2-expressing pyramidal cells,

1156 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

mRNAs purified from these two cell types

using the TRAP protocol were compared.

This analysis resulted in identification

of 1,112 differentially expressed mRNAs

(data not shown). Genes previously

shown to be expressed in CST projection

neurons were more highly enriched in the

Glt25d2 cells, whereas those demon-

strated to be expressed in CPNs in

cortical layer 5 were preferentially ex-

pressed in the S100a10 cells (Figure 3C and Table S2). To

demonstrate that the bacTRAP data collected from the

S100a10 and Glt25d2 pyramidal cell populations provide cell

type specific information even for those genes that have previ-

ously been identified as expressed in layer 5 of the cerebral

cortex, and to identify useful additional markers for CStr

neurons, we compared our data with genes annotated as specif-

ically expressed in layer 5 by the Allen Brain Atlas. Although

the ISH data demonstrate laminar expression in layer 5 for

each of the six examples shown (Figures 3E–3J), comparative

analysis of the translational profiles of S100a10 and Glt25d2

pyramidal cells identify Akap12, Depdc6, D6Wsu176e, Dexi,

and Emb as specifically expressed in CStr cells, and

D330050I23Rik as enriched in corticopontine neurons (Fig-

ure 3D). These results demonstrate the value of bacTRAP

Page 6: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

B

C

5

0

5

10

15

20

25

30

35

40

vehicle fluoxetine

*p

11 m

RN

A (%

ch

an

ge)

A

D

102

103

104

102

103

104

Normalized expression

No

rm

alized

exp

ressio

n

FLX

VEH

S100a10

102

103

104

102

103

104

Normalized expression

FLX

VEH

Glt25d2

-3

-2

-1

0

1

2

3

4

5S100a10

Glt25d2

Actin 1a 1b 1d 1f 2c 4 5a 7

Serotonin Receptor

Lo

g2

F

old

C

ha

ng

e (flx

/v

eh

)

-2

-1.5

-1

-0.5

0

0.5

1Whole cortex

1a 1b 1d 1f 2c 4 5a 7

Serotonin Receptor

Lo

g2

F

old

C

ha

ng

e (flx

/v

eh

)

*

ns

Figure 4. Cell Type-Specific Molecular Responses to SSRI Treatment

(A) The level of p11 mRNA was quantified by ISH and expressed as percent of control (mean ± SEM). Representative images are shown below the graph.

*p < 0.05.

(B) Scatter plot comparing normalized expression values of FLX- (x axis) or VEH (y axis)-treated cortex IP samples from S100a10 (left) or Glt25d2 (right) bacTRAP

mice. Dots represent probe sets that are significantly increased (red) or decreased (green) >1.4-fold by drug treatment (see Table S3). Outer gray lines represent

1.4-fold enrichment in either sample.

(C) Quantification of Htr expression following chronic FLX in S100a10 and Glt25d2 cells by qRT-PCR (mean ± SEM). Positive values indicate upregulation and

negative values indicate downregulation by FLX. Dotted lines show a 2-fold change in either direction. *p < 0.05.

(D) Quantification of Htr expression following chronic FLX in whole cortex by qRT-PCR (mean ± SEM).

See also Figure S4.

data for definitively identifying the specific cell type expressing

a gene of interest and for determining the distinguishing molec-

ular characteristics of closely related cell types, as well as illus-

trate the utility of ISH databases as tools for validation of

bacTRAP data sets.

S100a10 Corticostriatal Neurons Respond Specificallyto Antidepressant TreatmentGiven human neuroimaging studies implicating CStr circuits in

the response to antidepressants (Drevets et al., 2008), previous

studies demonstrating the induction of p11 in the cerebral cortex

by antidepressants and electroconvulsive therapy (Svennings-

son et al., 2006; Warner-Schmidt et al., 2010), and the present

demonstration that S100a10-expressing cells provide a major

and specific pathway for communication between the cortex

and dorsal striatum, we were interested in examining the possi-

bility that these cells respond preferentially and specifically to

antidepressant treatment. We chose the selective serotonin

reuptake inhibitor (SSRI) fluoxetine (FLX) because it is one of

the most highly prescribed antidepressants and is known to

have antidepressant activity in rodent models of depression

(Cryan et al., 2005; Dulawa et al., 2004). It was previously

reported that chronic administration of tricyclic antidepressants

imipramine or tranylcypromine increases the amount of p11 in

the cortex inmice (Svenningsson et al., 2006). To assesswhether

FLX had a similar effect, we treated wild-type (WT) mice with

FLX or saline for 14 days, and measured levels of p11 mRNA in

the cortex by ISH. As expected, p11 expression increased

�25% in response to FLX (Figure 4A), confirming the previous

observation that p11 expression in cortex is sensitive to antide-

pressant action (Svenningsson et al., 2006).

To identify long-term neuroadaptations that occur in response

to SSRI treatment, we administered FLX (0.167mg/ml) or vehicle

(VEH) to S100a10 and Glt25d2 bacTRAP mice in their drinking

water for 15–18 days. No change in the distribution of cells

expressing EGFP-L10a occurred in either bacTRAP line in

response to treatment (data not shown). Polysome-bound RNA

from the cortex of each line was collected using TRAP, and

microarray analysis was used to examine the effect of chronic

FLX on the profile of proteins being synthesized by each cell

type. Sixty-two genes (65 probe sets) were differentially regu-

lated by FLX in the S100a10 cells, as opposed to only four in

Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc. 1157

Page 7: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

B

C

Low mag IF image ofEGFP in KO brain.

A

D

LowLoLowLowLowLLowLowLowLowowLowLowwoLowowwowLowLowwwLowLowLowowowLoLowLowLowLowowwLowwowwowwLoLowLLowLLowLowoowLowLowLowLowLowowLowLowwwowLowLLLLowLowoLowLowLowLowLowLowLowLowLowLowLLLLowLowLowLowLowLowLowowwowLowLowLLowLowowowwLowwLowoLowwwwwLowwwwLowwwwLowLowowwwowwwwwwLowwwwLowowowowwowwowwwwowwLowoowowwowwwwwLowwwwwLowowLowLoowoowwowLowLowLowLowooLoowoLLooLooLowLowLowLowLooowwwwwLowLowowoowowwLowwwwwwLLowwwLowowLLLowwwLLLLLoowwwLLLLooowwwwLLLLowooLoLowwwwwLLooowwLLLooLoooLow mmamamammamaamamamamamamamammmmmmmmamammmmmmmmamammamaaaamamaammmmmmmmmmmmmmamaaaamaaamamamamammmmmmmammmmmmmmmmmmaamamamamaaammmmmmmmaaamaaaammamaamammmmmamamaamaaammmmammmamaaamammamamamamammmmammmmmmmmmmmmmmammmmmmmmmmmmmmmmmmm g Ig Ig Ig Iggg Iggggggg Igggg Iggg gg Ig Ig IIIg Igg Ig Ig Ig Ig Igg IIg Igg Ig Ig Igg I Igg Ig ggggg Igggggg IIgggggggggggg Ig ggg Ig Igggg Igg Iggggg IIg Ig Igggggggggg FF iF iF iF iF iF iF iFFF iF iF iFF iF iF iF iFFF iFFFFFFF iFFFFFF iF iFFFF iFFFFF FF iF iF FFF iFFFF iFF iF iiF iiFF magmagmagmagmagmagmagmagmagmagmagagmagmagmagmagmagmmmagmagaggmagm gggmagmmmagmmmmmammmmmmmmmagm gmmmmmm gmmmmmm ggmmamaaammmm e oe oe oe oe oe oe oe oo oe oe oe oe oe oe ooe oe oe oee oooooe oe ooooofffffffffffffffffffffffffffEGFEGFEGFEGFEGFEGEGFEGFEGGGGEGFEGEGFEGFGGGGGGFFFFEGFEGFEGEGFGGGGEGEGEGFEGFFFGFEGEEGEGGGGEGFEGFEGFEGEGGGGFGFGFEEGGGFGFGFFFEEGEGGGFFFEGEGFGGGGFGGGFGFEEGGGEGEGGGGGEGGFGGGGGEGGGGGGGGGFFFE FFFFFFEEGFFE FEGGEGEGEGGGGEEEGGGGFFFGFEEGGGGEGFGFPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPPP inininninininininininninininnininnnnnninnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnnn KOKOKOKOKOKOKOKOKOKOKOKOKOKOKOKOOOOOKOKOOKOKOKOKOOKOOKKKOKOKOKKKKKKOKOKOKOKKOOKOOKKKKKKK brbrbrbrbrbrbrbbbrbrbrbrbbrrbbbrrrbbbbbbbrb ainainainainainainainainainainainnnaininainaina naainainainainaiainainn...

2/3

5a

5b

6

wm

102

103

102

103

Normalized expression

No

rm

alize

d e

xp

re

ss

io

n

FLX

VEHp11 KO

104

104

GFPNeuN

-3

-2

-1

0

1

2

3

4

5

Actin 1a 1b 1d 1f 2c 4 5a 7

Serotonin Receptor

*

Lo

g2 F

old

C

ha

ng

e (flx

/v

eh

)

-3

-2

-1

0

1WT

KO

Lo

g2 F

old

C

ha

ng

e (K

O/W

T)

1a 1b 1d 1f 2c 4 5a 7

Serotonin Receptor

* * **

**

Figure 5. Molecular Changes Are Depen-

dent on p11 Expression

(A) Anti-EGFP IF image of EGFP-L10a expression

in constitutive p11 KO mice crossed to the

S100a10 bacTRAP line. See Figure S5 for further

anatomical analysis.

(B) Quantification of Htr expression in IP RNA from

S100a10 bacTRAP/p11 KO relative to WT cortex

by qRT-PCR (mean ± SEM). Dotted line indicates

a 2-fold change. Negative values indicate

downregulation in KO compared to WT. *p < 0.05,

**p < 0.01.

(C) Scatter plot comparing normalized expression

values of FLX- (x axis) or VEH (y axis)-treated

cortex IP samples from S100a10 bacTRAP/p11

KO mice. Dots represent probe sets that are

significantly increased (red) or decreased (green)

>1.4-fold by drug treatment in WT cortex (see

Figure 4B and Table S3). Outer gray lines represent

1.4-fold enrichment in either sample.

(D) Quantification of Htr expression following

chronic FLX in S100a10 bacTRAP/p11 KO cells

by qRT-PCR (mean ± SEM). Positive values

indicate upregulation and negative values

indicate downregulation by FLX treatment. Dotted

lines indicate a 2-fold change in either direction.

*p < 0.05.

the Glt25d2 cells (Figure 4B and Table S3). These data confirmed

the hypothesis that the S100a10 (and thus p11)-expressing cells

are preferentially sensitive to antidepressant treatment, and

suggest that this population of CStr cells might play an important

role in antidepressant therapy.

Given the facts that p11modulates Htr function (Svenningsson

et al., 2006; Warner-Schmidt et al., 2009), and that chronic

treatment with SSRIs results in elevated levels of serotonin in

the nervous system, we asked whether the response of

S100a10-expressing cells to FLX included long-term changes

in the expression of Htrs. We restricted our analysis to Htrs

expressed in cortical neurons (see Table S1). As shown in Fig-

ure 4C, a dramatic and specific increase in Htr4 mRNA

(16-fold) occurred in S100a10 cells following chronic FLX treat-

ment. Elevated Htr4 expression was not observed in Glt25d2

neurons and no increases in the expression of other Htr subtypes

were observed in either cell type. Further, this increase in Htr4

expression was not detectable in whole cortex (input) samples

(Figure 4D). Taken together, our data both demonstrate that layer

5 CStr projection neurons are highly responsive to antidepres-

sant treatment, and suggest that the induction of Htr4 expres-

sion in this critical cell population may play an important role in

restoring the balance between cortical and striatal activity that

occurs in response to SSRIs.

Serotonin Receptors Are Dysregulated inS100a10-Expressing Cells in p11 Null Mutant Micep11 directly interacts with multiple Htrs, and its loss leads to

reduced responses of cortical cells to serotonin at striatal

synapses (Svenningsson et al., 2006). To determine whether

p11 is involved in the molecular adaptations that we have

1158 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

observed in CStr neurons in response to SSRIs, we crossed

S100a10 bacTRAP mice with p11 knockout (KO) animals

(Svenningsson et al., 2006) to obtain animals (S100a10

bacTRAP/p11KO) in which the molecular consequences of p11

deletion could be assessed in CStr pyramidal cells. EGFP-

L10a transgene expression was not significantly altered in the

cortex of KO mice (Figures 1E and 5A), and retrograde tracing

revealed that EGFP-expressing cells still accounted for amajority

of projections to CPu and contralateral cortex (Figure S5A). As

shown in Figure 5B, deletion of p11 resulted in severe dysregu-

lation of Htrs in these neurons. Htrs 1a, 1b, 2c, 5a, and 7 were all

significantly downregulated in the KOs. No significant change

was observed in the translation of Htr1d or Htr4, both of which

bind directly to p11. However, given previous studies demon-

strating that the cell surface expression of Htr1b and Htr4 is

critically dependent on p11 (Svenningsson et al., 2006;

Warner-Schmidt et al., 2009), we expect that signaling through

these receptors is also reduced. Taken together, our data

suggest that deletion of p11 results in a loss of serotonergic

tone in CStr pyramidal cells.

We next tested whether the alterations in Htr expression in

S100a10 cortical neurons in p11 KOs resulted in dampened

responses to FLX. S100a10 bacTRAP/p11KO mice received

chronic administration of FLX or VEH as described above, and

translational profiles from these cells were assayed on microar-

rays. As shown in Figure 5C, p11 KO animals failed to show the

robust response to FLX observed in S100a10 CStr neurons.

Thus, the majority of genes shown to be significantly regulated

in the S100a10 cells of WT animals were no longer regulated

by FLX in the p11 KO, and the induction of Htr4 noted in WT cells

was significantly blunted (Figure 5D; p < 0.05, Student’s t test).

Page 8: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

0

0.2

0.4

0.6

0.8

1

WTCortex

KO

Th

igm

ota

xis

0

20

40

60

80

100

120

WTCortex

KO

Ta

il S

us

pe

ns

ion

Te

st

Imm

ob

ilit

y (%

c

on

tro

l)

*

VEH FLX

CB

ED

0

100

200

300

400

500

600

700

WTCortex

KO

No

ve

lty

Su

pp

re

ss

ed

Fe

ed

ing

La

te

nc

y (s

ec

)

**

VEH FLX

A

WT

Cortex

KO

Cortex Striatum

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

*

**

*

*

*

*

*

*

*

*

*

0

0.5

1

1.5

2

2.5

3

WTCortex

KO

Su

cro

se

P

re

fe

re

nc

e

Ra

tio

(s

uc

ro

se

/w

ate

r)

*

**

*

*

*********

****

******

******

*****

****

**

*****

*

*

*

*

*

Dentate Gyrus

*

*

*

*

*

*

*

*

Figure 6. Cortical Expression of p11 Is Required

for Behavioral Responses to Chronic SSRI

Administration

(A) Anti-p11 IF in brain sections from floxed p11 mice (WT,

top row) and Emx1/p11KO mice (bottom row) from layer 5

of sensorimotor cortex (left panels), striatum (center), or

dentate gyrus (right). Inset shows anti-NeuN staining from

the same field. Asterisks label p11-positive cells.

(B) Bar graph representing the effect of chronic FLX

treatment on the latency to feed in WT and Emx1/p11 KO

(Cortex KO) mice in the novelty NSF paradigm (mean ±

SEM). **p < 0.01.

(C) Bar graph representing the effect of chronic FLX

treatment on immobility in WT and Emx1/p11 KO (Cortex

KO) mice in TST (mean ± SEM). *p < 0.05.

(D) Bar graph representing thigmotaxis in an open field in

WT or Emx1/p11 KO (Cortex KO) mice (mean ± SEM).

(E) Bar graph representing sucrose preference in WT or

Emx1/p11 KO (Cortex KO) mice (mean ± SEM). Dotted line

indicates no preference.

See also Figure S5.

We conclude that the actions of SSRIs on S100a10 CStr projec-

tion neurons are critically dependent on p11.

Behavioral Responses to Antidepressants Depend onthe Presence of p11 in the Cerebral CortexGiven our bacTRAP molecular phenotyping data establishing

S100a10 CStr pyramidal cells as strongly and specifically

responsive to chronic SSRI treatment, and our demonstration

that these cells provide the majority of afferents to the dorsal

striatum from the cortex, we were interested in testing the role

of S100a10 cortical neurons in the therapeutic actions of antide-

Cell 149, 11

pressants. Because CStr cells in p11 KOs failed

to show molecular responses to FLX and have

altered expression of Htrs, we generated

cortex-specific p11 KO animals (Emx1/p11KO)

to assess the functional consequences of loss

of these adaptations on behavior. Immunohisto-

chemistry was employed to confirm the loss of

p11 from the cortex, and its continued expres-

sion in the striatum and in other subcortical sites

(Figure 6A; data not shown).

We employed the novelty suppressed feeding

(NSF) paradigm and the tail suspension test

(TST) to assess the therapeutic actions of

chronic FLX in Emx1/p11KO mice. In NSF,

food-deprived mice are placed in an open field

that contains food in the center. The mice

must decide to approach and consume the

food, or avoid the novel environment. Animals

treated with antidepressants for several weeks

before testing consistently exhibit shorter

feeding latencies than those that received acute

or no treatment, making NSF a valuable assay

for evaluating responses to chronic antidepres-

sant administration (Dulawa and Hen, 2005). We

found that Emx1/p11KO mice failed to exhibit

behavioral responses to chronic FLX treatment

in NSF (Figure 6B). WT mice (carrying the floxed p11 allele but

not Cre) showed a significantly decreased latency to feed (436

± 36 s) following chronic FLX administration compared to VEH-

treated controls (561 ± 25 s). In contrast, there was no difference

in latency between VEH and FLX administration (523 ± 32 versus

469 ± 41 s) in Emx1/p11KO mice. VEH-treated Emx1/p11KOs

showed no difference in baseline anxiety (as measured by

latency to feed) compared to WT mice (Figure 6B, white bars).

Further supporting the conclusion that the behavioral differences

in the NSF test likely reflect an inability of the cortex-specific KOs

to respond to drug treatment rather than a change in generalized

52–1163, May 25, 2012 ª2012 Elsevier Inc. 1159

Page 9: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

anxiety, mice of all genotypes displayed no difference in thigmo-

taxis in an open field test.

We also tested Emx1/p11 KO mice in the TST, which is

a classic behavioral test widely employed to screen antidepres-

sant efficacy in mice (Cryan et al., 2005). The TST measures the

timemice spend immobile while being suspended by their tail, an

inescapable stress. Following chronic FLX treatment, WT mice

exhibited a significant reduction (�20%) in immobility in the

TST and this effect was abolished in Emx1/p11KOs (Figure 6C).

There was no difference between genotypes in locomotor

activity (Figure S5B), and importantly there was also no indica-

tion of a depressive-like effect in Emx1/p11 KO on immobility

in the TST (Figure 6C, white bars), or anhedonia in the sucrose

preference test (Figure 6E). These data further support the notion

that the behavioral phenotype of Emx1/p11KO mice lies in their

lack of response to FLX treatment and not to an elevated depres-

sive-like state.

Emx1/p11KOmice also failed to display a behavioral response

to chronic FLX in the forced swim test (FST; Figure S5C). WT

animals showed the expected decrease in immobility following

chronic FLX treatment, but it did not reach statistical significance

(p = 0.08). This is likely due to genetic background, because our

mice were backcrossed onto the C57BL/6 strain and it has been

shown that chronic, but not acute, FLX treatment in drinking

water reduced immobility in the FST in BALB/c mice but not

C57BL/6 mice (Dulawa et al., 2004). We conclude that proper

function of S100a10 CStr pyramidal cells is required for normal

behavioral responses to chronic antidepressant treatment.

DISCUSSION

Human neuroimaging studies have consistently demonstrated

alterations in cortical activity in depressed patients and normali-

zation of this activity in response to antidepressants, although

neither the cell types altered in response to treatment nor the

nature of the neuroadaptations that occur in these cell types

have been identified. In this study, we show that CStr projection

neurons expressing S100a10 (p11) are essential for both the

molecular andbehavioral responses to theSSRIFLX.Wedemon-

strate that S100a10 pyramidal cells project primarily to the dorsal

striatum and contralateral cortex, and that they experience long-

term adaptive changes in response to chronic administration of

FLX that do not occur in other layer 5 projection neurons. These

adaptations require p11 and include the induction of Htr4

receptor expression. Finally, we show that deletion of p11 in

these cells abolishes behavioral responses to FLX, but does not

lead to a depressive-like phenotype. Our data identify S100a10

CStr neurons as essential for the actions of SSRI antidepres-

sants, reveal specific molecular adaptations that occur in these

cells in response to chronic treatment, and suggest that modula-

tion of serotonergic tone in this single cortical cell population can

result in effective antidepressant therapy.

Impact of bacTRAP Translational Profilingof Disease-Relevant Cell TypesMolecular phenotyping of cell types that comprise the neural

circuits underlying pathological neuropsychiatric conditions

and their responses to therapy provides one avenue for develop-

1160 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

ment of more specific and effective treatments. Until recently,

the histological complexity of the CNS has provided a barrier

for such studies, especially in the cerebral cortex where few

molecular markers exist to distinguish between subpopulations

of morphologically similar pyramidal cells. In this study, we

took advantage of the discovery that p11 is regulated in the

cortex following antidepressant treatment (Svenningsson et al.,

2006) and of the recently developed bacTRAP approach for

translational profiling of specific CNS cell types (Doyle et al.,

2008; Heiman et al., 2008) to study long-term changes that occur

in specific cortical neuron populations in response to antidepres-

sants. Our results demonstrate that bacTRAP translational

profiling can be used to identify cell types that respond to thera-

peutic drugs, and provide a first illustration that those cells most

responsive to treatment may also participate in the therapeutic

actions of the drug. It will be of great interest to employ this

strategy in other mouse models of psychiatric and neurological

disease to explore further the relationship between molecular

phenotype and cellular function in the setting of complex, circuit

based disorders.

Our data argue strongly that the specific molecular changes

that occur in relevant cell types in the context of effective treat-

ment of disease may provide critical insights into mechanisms

that determine therapeutic efficacy. For example, we have

shown here that chronic treatment with FLX results in the specific

induction of Htr4 expression in S100a10 CStr pyramidal cells,

and that loss of p11 causes both a decrease in the expression

of Htrs in this cell type and a failure to respond behaviorally to

antidepressants. The induction of Htr4 expression is consistent

with previous studies showing that Htr4 agonists have antide-

pressant effects (Lucas et al., 2007) that are dependent on p11

expression (Warner-Schmidt et al., 2009). This suggests the

specific hypothesis that the increase of Htr4 expression

following chronic FLX treatment enhances the sensitivity of

CStr neurons to the elevated serotonin levels, thereby increasing

outflow from the cerebral cortex to the striatum and contributing

to the efficacy of antidepressants. The fact that the loss of p11

from these cells resulted in a downregulation of many Htrs

suggests a wider impact of p11 on serotonergic tone. Although

additional experimentation is needed to test this hypothesis,

our data strongly argue that the information obtained from

TRAP profiling of individual cell types in mouse models of

disease can significantly advance our understanding of molec-

ular and cellular mechanisms of disease.

Corticostriatal Neurocircuitry MediatingAntidepressant ResponsesA confluence of evidence has emerged from studies as diverse

as human neuroimaging (Drevets et al., 2008; Mayberg et al.,

1999) and immediate early gene transcription (Covington et al.,

2010) to implicate altered activity in several brain areas in

depression. These studies form the basis of our current under-

standing of depression, in which mood is governed by a distrib-

uted brain circuit that does not function normally in depressed

individuals (Ressler and Mayberg, 2007). Given this under-

standing, how is it that the molecular adaptations that occur in

a single circuit element (S100a10 CStr projection neurons) can

be so important for the antidepressant activity of SSRIs? Our

Page 10: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

data identify several factors that may help to answer this query.

First, S100a10 pyramidal cells occupy a critical position in the

mood circuitry. We have shown by retrograde tracing that this

cell population provides a major conduit for communication

between the cortex and the dorsal striatum. Although early

studies emphasized a role for the ventral striatum in reward pro-

cessing and depression, recent structural and functional imaging

studies have revealed that the dorsal striatum is both active

during reward related tasks and altered in MDD (Delgado,

2007; Pizzagalli et al., 2009). In particular, reduced prediction

error signals in both the bilateral caudate and the NAc correlate

with increased anhedonia (a core feature of MDD) in depressed

subjects (Gradin et al., 2011). In addition, MDD subjects with

elevated anhedonic symptoms or late-onset depression showed

a reduced caudate volume (Krishnan et al., 1992; Pizzagalli et al.,

2009). It is possible that S100a10 CStr cells projecting to dorso-

medial striatum (DMS) underlie antidepressant responses. This

idea is supported by a growing body of evidence demonstrating

that the DMS plays a significant role in reward learning and moti-

vated behavior (Voorn et al., 2004).

The adaptations that occur in this cell type in response to FLX,

including the increase in Htr4 expression, occur specifically in

S100a10 CStr cells. Thus, we were not able to detect these

changes in TRAP data from Glt25d2 corticopontine neurons or

in data collected fromwhole cortex samples. These results high-

light the unique biochemical properties of distinct classes of

pyramidal cells, and confirm our previous demonstration that

the genome wide pharmacokinetic responses of even closely

related cell types can vary dramatically (Heiman et al., 2008).

Finally, it is important to emphasize that we chose to analyze

p11-expressing cortical neurons because previous studies had

shown that this protein is expressed in a laminar pattern in the

cortex, and that its expression in cortex is reduced in depressed

individuals and restored in response to treatment (Anisman et al.,

2008; Svenningsson et al., 2006). We report here that p11 is

required for the molecular adaptations seen in CStr cells in

response to an SSRI, and that loss of p11 expression in the

cortex abolishes behavioral responses to the SSRI FLX.

Although the large number of distinct serotonin receptors and

their broad distribution across the nervous system (and

periphery) suggest that serotonin elicits a variety of responses

in vivo, it seems possible that effective long-term, functional

adaptation to SSRIs may require a constellation of factors

present only in S100a10 cortical neurons.

Our datademonstrate that the beneficial actions of SSRI antide-

pressant therapy may be mediated by a single cell type in the

cerebral cortex, and suggest that development of drugs that

specifically target the activity of CStr neurons may result in

improved therapies for depression. However, it is important to

note that our results do not predict that the pathophysiological

changes that elicit depression occur in this cell type, or thatmodu-

lation of other critical cell types might not provide equally effective

therapies. The observation that deletion of p11 in the cerebral

cortex results in a lack of responsiveness to FLX but does not elicit

depressive-like or anxiety behaviors strongly suggests that loss of

p11 in other cell typesmust be involved in generation of the behav-

ioral phenotypesevident in constitutive p11KOmice. Although the

separation of the anatomical locations responsible for generation

of the behavioral phenotypes from those governing treatment

responses in this animal model of depression may need clarifica-

tion, we believe that such a dichotomy is typical of complex

circuit-based disorders. For example, deep brain stimulation

studies of Parkinson’s disease have shown that even circuits

that are irreversibly altered as a result of neurodegeneration can

be phenotypically normalized by stimulation of circuit elements

that are not lost as a result of the pathophysiology of the

disease (Limousin and Martinez-Torres, 2008). Given the genetic

complexity of MDD and the circuitry that is involved in the regula-

tion of mood, we expect that the causes of MDDwill alter function

atmany points in themood circuitry. However, the identification of

a single cell type in the cerebral cortex that is both positioned to

normalize activity between cortical and subcortical sites, and

that is required for the response to SSRI antidepressants, is an

exciting step forward in the march toward development of more

effective treatments for depression.

EXPERIMENTAL PROCEDURES

Animals and Drug Treatments

All procedures involving animals were approved by the Rockefeller University

Institutional Animal Care and Use Committee and were in accordance with the

National Institutes of Health guidelines. The Glt25d2 bacTRAP mice, constitu-

tive p11 KOmice, and Snap25 bacTRAPmice were generated and maintained

at Rockefeller University and have been described previously (Doyle et al.,

2008; Svenningsson et al., 2006). The S100a10 bacTRAP transgenic mouse

line was generated by modifying a BAC clone containing the S100a10 gene

to insert an EGFP-L10a fusion protein into the translation start site, and the

modified BAC was purified and used for transgenesis as described previously

(Gong et al., 2002). See Extended Experimental Procedures for details.

Cortex-specific p11 KO mice were generated by breeding floxed p11 mice

(Warner-Schmidt et al., 2011) with Emx1-Cre mice purchased from The Jack-

son Laboratory (Stock 005628). All mice were maintained on a 12-hr light/dark

cycle and given ad libitum access to food and water. For chronic drug treat-

ments, mice were housed one to two per cage and a FLX (0.167 mg/ml)/

saccharine (1%) mixture or saccharine alone (VEH) was administered in the

drinking water for 15–18 days and replaced with fresh solution every 3–4 days.

Retrograde Labeling and Immunohistochemistry

Alexa-555-conjugated cholera toxin b-subunit (Invitrogen) was stereotaxically

injected into adult (8–12 weeks old) transgenic mice under ketamine/xylazine

(100/10 mg/kg) anesthesia as described in Supplemental Information. Quanti-

fication of double-labeled cells and cell depth was done using Image J (NIH) as

detailed in Extended Experimental Procedures.

Immunohistochemistry and In Situ Hybridization

Tissue was prepared as described in Extended Experimental Procedures.

Immunofluorescent staining was carried out on 35-mm thick sections using

chicken anti-GFP (1:2,000, Abcam), mouse anti-EGFP (1:500; Heiman et al.,

2008), rabbit anti-CTb (1:1,000, Abcam), goat anti-S100a10 (1:200, R&D

Systems), and/or mouse anti-NeuN (1:1,000, Millipore) followed by Alexa-fluor

conjugated secondary antibodies (Invitrogen). All sections were imaged on

a Zeiss LSM700 confocal microscope.

S100a10 (p11) in situ hybridization was performed as described previously

(Warner-Schmidt et al., 2010) and in Extended Experimental Procedures.

Translating Ribosome Affinity Purification

Three male mice were pooled for each sample and three biological replicates

were collected for each condition. All polysome purifications and mRNA

extractions were carried out as described previously (Heiman et al., 2008)

and in Extended Experimental Procedures. RNA quantity was measured

with a Nanodrop 1000 spectrophotometer and quality was assayed on an

Agilent 2100 Bioanalyzer.

Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc. 1161

Page 11: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

Microarray Analysis

For microarrays, RNA was amplified and labeled using the GeneChip Expres-

sion 30 Amplification 2-Cycle cDNA Synthesis Kit (Affymetrix) and hybridized to

GeneChip Mouse Genome 430 2.0 microarrays (Affymetrix) according to

manufacturers’ protocols. CEL files were imported into GeneSpring GX 7.3.1

and preprocessed with the GCRMA algorithm. For each experiment, data

were filtered to remove probe sets with low intensities (normalized expression

<50 in at least one condition) and those identified as background (Doyle et al.,

2008). Only probe sets expressed in neuronal cells were used for analysis (see

Supplemental Information). Differentially expressed genes were identified by

performing a moderated t test (Smyth, 2004) as described in Extended Exper-

imental Procedures. The p values were adjusted for multiple hypothesis

testing, controlling the false discovery rate with the Benjamini-Hochberg

procedure. Only genes with an FDR < 0.05 (5%) and a fold change > 1.4

were used for analyses. MIAME-compliant raw data are available from Gene

Expression Omnibus with accession number GSE35766.

Quantitative RT-PCR

cDNA was synthesized using the WT-Ovation RNA Amplification kit (NuGEN

Technologies) according to manufacturer’s protocol. For Htrs, PCR was

carried out in an Applied Biosystems StepOnePlus RT-PCR System using

TaqMan expression assays. All other reactions were performed in a Bio-Rad

iQ5 Multicolor RT-PCR Detection System using the SYBR green method

(BioRad). Data were normalized to b-actin and relative expression changes

between conditions were calculated by the comparative CT (2�DDCT) method.

Student’s t test was used for all statistical analyses. Detailed methods are

provided in Extended Experimental Procedures.

Behavioral Analysis

Detailed descriptions of all behavioral methods can be found in Extended

Experimental Procedures. NSF was performed exactly as described (Egeland

et al., 2010) and the tail suspension test, forced swim, sucrose preference, and

open field were performed as described previously (Warner-Schmidt et al.,

2009). Statistical comparisons were made by ANOVA using Prism 5 software

(GraphPad). In experiments comprised of more than two groups, data were

first analyzed by two-way ANOVA followed by post hoc Bonferroni test.

Statistical significance was set at p < 0.05.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

five figures, and five tables and can be found with this article online at

doi:10.1016/j.cell.2012.03.038.

ACKNOWLEDGMENTS

This work was supported by the Howard Hughes Medical Institute (N.H.),

NIH/NIDA ARRA Grand Opportunity Grant PHS DA028968 (N.H., P.G.), Conte

Center PHS MH090963 (P.G., N.H.), USAMRAA grant W81XWH-09-0401

(J.W.S.), the JPB Foundation (P.G.), and the Fisher Foundation (P.G.). We

thank the Rockefeller University Genomics Resource Center, and Neurosci-

ence Associates. We would further like to thank Tanya Stevens, Sujata

Bupp, and George Skabardonis for their excellent advice and assistance,

and Joseph Dougherty for providing the Snap25 bacTRAP mice. N.H. and

P.G. are cofounders of, and consults for, Envoy Therapeutics.

Received: September 30, 2011

Revised: January 17, 2012

Accepted: March 7, 2012

Published: May 24, 2012

REFERENCES

Anisman, H., Du, L., Palkovits, M., Faludi, G., Kovacs, G.G., Szontagh-Kishazi,

P., Merali, Z., and Poulter, M.O. (2008). Serotonin receptor subtype and p11

mRNA expression in stress-relevant brain regions of suicide and control

subjects. J. Psychiatry Neurosci. 33, 131–141.

1162 Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc.

Arlotta, P., Molyneaux, B.J., Chen, J., Inoue, J., Kominami, R., and Macklis,

J.D. (2005). Neuronal subtype-specific genes that control corticospinal motor

neuron development in vivo. Neuron 45, 207–221.

Covington, H.E., III, Lobo, M.K., Maze, I., Vialou, V., Hyman, J.M., Zaman, S.,

LaPlant, Q., Mouzon, E., Ghose, S., Tamminga, C.A., et al. (2010). Antidepres-

sant effect of optogenetic stimulation of the medial prefrontal cortex.

J. Neurosci. 30, 16082–16090.

Cryan, J.F., Mombereau, C., and Vassout, A. (2005). The tail suspension test

as a model for assessing antidepressant activity: review of pharmacological

and genetic studies in mice. Neurosci. Biobehav. Rev. 29, 571–625.

Delgado, M.R. (2007). Reward-related responses in the human striatum.

Ann. N Y Acad. Sci. 1104, 70–88.

Doyle, J.P., Dougherty, J.D., Heiman, M., Schmidt, E.F., Stevens, T.R., Ma, G.,

Bupp, S., Shrestha, P., Shah, R.D., Doughty, M.L., et al. (2008). Application of

a translational profiling approach for the comparative analysis of CNS cell

types. Cell 135, 749–762.

Drevets, W.C. (2000). Functional anatomical abnormalities in limbic and

prefrontal cortical structures in major depression. Prog. Brain Res. 126,

413–431.

Drevets, W.C., Price, J.L., and Furey, M.L. (2008). Brain structural and func-

tional abnormalities in mood disorders: implications for neurocircuitry models

of depression. Brain Struct. Funct. 213, 93–118.

Dulawa, S.C., and Hen, R. (2005). Recent advances in animal models of

chronic antidepressant effects: the novelty-induced hypophagia test.

Neurosci. Biobehav. Rev. 29, 771–783.

Dulawa, S.C., Holick, K.A., Gundersen, B., and Hen, R. (2004). Effects of

chronic fluoxetine in animal models of anxiety and depression. Neuropsycho-

pharmacology 29, 1321–1330.

Egeland, M., Warner-Schmidt, J., Greengard, P., and Svenningsson, P. (2010).

Neurogenic effects of fluoxetine are attenuated in p11 (S100A10) knockout

mice. Biol. Psychiatry 67, 1048–1056.

Egeland, M., Warner-Schmidt, J., Greengard, P., and Svenningsson, P. (2011).

Co-expression of serotonin 5-HT(1B) and 5-HT(4) receptors in p11 containing

cells in cerebral cortex, hippocampus, caudate-putamen and cerebellum.

Neuropharmacology 61, 442–450.

Franklin, K.B.J., and Paxinos, G. (2008). The Mouse Brain in Stereotaxic Coor-

dinates, Third Edition (Amsterdam: Elsevier Academic Press).

Gong, S., Yang, X.W., Li, C., and Heintz, N. (2002). Highly efficient modification

of bacterial artificial chromosomes (BACs) using novel shuttle vectors contain-

ing the R6Kgamma origin of replication. Genome Res. 12, 1992–1998.

Gradin, V.B., Kumar, P., Waiter, G., Ahearn, T., Stickle, C., Milders, M., Reid, I.,

Hall, J., and Steele, J.D. (2011). Expected value and prediction error abnormal-

ities in depression and schizophrenia. Brain 134, 1751–1764.

Groh, A., Meyer, H.S., Schmidt, E.F., Heintz, N., Sakmann, B., and Krieger, P.

(2010). Cell-type specific properties of pyramidal neurons in neocortex under-

lying a layout that is modifiable depending on the cortical area. Cereb. Cortex

20, 826–836.

Heiman, M., Schaefer, A., Gong, S., Peterson, J.D., Day, M., Ramsey, K.E.,

Suarez-Farinas, M., Schwarz, C., Stephan, D.A., Surmeier, D.J., et al. (2008).

A translational profiling approach for the molecular characterization of CNS

cell types. Cell 135, 738–748.

Krishnan, K.R., McDonald, W.M., Escalona, P.R., Doraiswamy, P.M., Na, C.,

Husain, M.M., Figiel, G.S., Boyko, O.B., Ellinwood, E.H., and Nemeroff, C.B.

(1992). Magnetic resonance imaging of the caudate nuclei in depression.

Preliminary observations. Arch. Gen. Psychiatry 49, 553–557.

Krishnan, V., and Nestler, E.J. (2008). The molecular neurobiology of depres-

sion. Nature 455, 894–902.

Limousin, P., and Martinez-Torres, I. (2008). Deep brain stimulation for

Parkinson’s disease. Neurotherapeutics 5, 309–319.

Lucas, G., Rymar, V.V., Du, J., Mnie-Filali, O., Bisgaard, C., Manta, S.,

Lambas-Senas, L., Wiborg, O., Haddjeri, N., Pineyro, G., et al. (2007). Sero-

tonin(4) (5-HT(4)) receptor agonists are putative antidepressants with a rapid

onset of action. Neuron 55, 712–725.

Page 12: Identification of the Cortical Neurons that Mediate ... · Mood is governed by the actions of a complex, anatomically ... deep brain stimulation correlate with increased activity

Mayberg, H.S. (2009). Targeted electrode-based modulation of neural circuits

for depression. J. Clin. Invest. 119, 717–725.

Mayberg, H.S., Liotti, M., Brannan, S.K., McGinnis, S., Mahurin, R.K., Jerabek,

P.A., Silva, J.A., Tekell, J.L., Martin, C.C., Lancaster, J.L., and Fox, P.T. (1999).

Reciprocal limbic-cortical function and negative mood: converging PET

findings in depression and normal sadness. Am. J. Psychiatry 156, 675–682.

Pizzagalli, D.A., Holmes, A.J., Dillon, D.G., Goetz, E.L., Birk, J.L., Bogdan, R.,

Dougherty, D.D., Iosifescu, D.V., Rauch, S.L., and Fava, M. (2009).

Reduced caudate and nucleus accumbens response to rewards in unmedi-

cated individuals with major depressive disorder. Am. J. Psychiatry 166,

702–710.

Ressler, K.J., and Mayberg, H.S. (2007). Targeting abnormal neural circuits in

mood and anxiety disorders: from the laboratory to the clinic. Nat. Neurosci.

10, 1116–1124.

Smyth, G.K. (2004). Linear models and empirical bayes methods for assessing

differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol.

3, Article3.

Svenningsson, P., Chergui, K., Rachleff, I., Flajolet, M., Zhang, X., El Yacoubi,

M., Vaugeois, J.M., Nomikos, G.G., and Greengard, P. (2006). Alterations in

5-HT1B receptor function by p11 in depression-like states. Science 311,

77–80.

Voorn, P., Vanderschuren, L.J., Groenewegen, H.J., Robbins, T.W., and

Pennartz, C.M. (2004). Putting a spin on the dorsal-ventral divide of the stria-

tum. Trends Neurosci. 27, 468–474.

Warner-Schmidt, J.L., Flajolet, M., Maller, A., Chen, E.Y., Qi, H., Svennings-

son, P., and Greengard, P. (2009). Role of p11 in cellular and behavioral effects

of 5-HT4 receptor stimulation. J. Neurosci. 29, 1937–1946.

Warner-Schmidt, J.L., Chen, E.Y., Zhang, X., Marshall, J.J., Morozov, A.,

Svenningsson, P., and Greengard, P. (2010). A role for p11 in the antidepres-

sant action of brain-derived neurotrophic factor. Biol. Psychiatry 68, 528–535.

Warner-Schmidt, J.L., Vanover, K.E., Chen, E.Y., Marshall, J.J., and

Greengard, P. (2011). Antidepressant effects of selective serotonin reuptake

inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and

humans. Proc. Natl. Acad. Sci. USA 108, 9262–9267.

Cell 149, 1152–1163, May 25, 2012 ª2012 Elsevier Inc. 1163