nectin-3 y stress

Upload: rodolfo-rosas-martinez

Post on 06-Jul-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/17/2019 Nectin-3 y Stress

    1/10

    706 VOLUME 16 | NUMBER 6 | JUNE 2013 NATURE NEUROSCIENCE

    A R T I C L E S

    Synapses are specialized intercellular junctions that meditate the

    transmission of information between neurons. Glutamatergic exci-tatory synapses are established by presynaptic axonal terminals

    and postsynaptic dendritic spines, both of which anchor synapticcell adhesion molecules (CAMs)1,2. CAMs are not merely static

    constituents of synapses, but are dynamic modulators of synapticactivity and plasticity. During development, synaptic CAMs are

    involved in neurite growth, synaptogenesis and synapse maturation.In the adult brain, CAMs interact with various synaptic proteins

    and receptors to shape synaptic function3,4. A disruption of synaptic

    adhesion may lead to functional abnormalities. Recent evidenceindicates that the dysregulation of synaptic CAMs contribute to

    structural modifications and cognitive deficits5,6, including thoseinduced by stress7.

    Repeated exposure to severe stress exerts deleterious effects on cog-nition in different life stages8. Early adversities, such as an impover-

    ished environment, impair hippocampal integrity and function, whichis manifested by progressively deteriorated cognitive performance

    in the adult offspring9,10. As key mediators of neuroendocrine and

    behavioral responses to stress, CRH and CRHR1 have been shownto modulate the negative effects of early-life stress on cognition andstructural plasticity 11,12. In adult animals, the influence of chronic

    stress on cognition also involves hippocampal CRH-CRHR1 signal-ing13. Nonetheless, the molecular underpinnings of CRHR1-mediated

    cognitive effects remain to be elucidated.

    Nectin-3 is an immunoglobulin-like CAM, which primarily

    localizes at adherens junctions in adulthood, the sites adjacent tothe presynaptic active zone and postsynaptic density (PSD)14.

    Postsynaptic nectin-3 mediates heterophilic adhesion with presynap-tic nectin-1, and is indirectly connected to the actin cytoskeleton via

    L-afadin. The nectin-afadin complex colocalizes and cooperates withthe cadherin-catenin complex to organize adherens junctions, and

    participates in synaptic formation, maintenance and remodeling14–19.Some evidence suggests that impaired nectin-mediated adhesion

    disrupts hippocampal development16 and is associated with mental

    retardation20. Although expressed ubiquitously, nectin-3 is abundantin CA3 pyramidal neurons13,21 that are vulnerable to both acute22 

    and chronic23 stress challenge. Nectin-3 expression levels have beenshown to correlate with the observed cognitive phenotype following

    chronic stress13. However, it is still unclear whether nectin-3 is caus-ally involved in mediating the effect of stress via CRHR1 signaling on

    cognition and structural remodeling.We examined how stress and CRH-CRHR1 signaling might mod-

    ulate nectin-3 expression in the hippocampus. Using site-specific

    knockdown and overexpression of nectin-3, we then investigatedthe role of hippocampal nectin-3 in spatial learning and memoryand dendritic spine plasticity, and tested whether reinstating

    nectin-3 in the adult hippocampus could reverse the detrimentalconsequences of early adverse experience on cognitive function and

    structural plasticity.

    1Max Planck Institute of Psychiatry, Munich, Germany. 2Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental

    Health, Neuherberg, Germany. 3Technische Universität München, Lehrstuhl für Entwicklungsgenetik, Helmholtz Zentrum München, Neuherberg, Germany. 4Deutsches

    Zentrum für Neurodegenerative Erkrankungen, Munich, Germany. 5Present address: Institute of Mental Health, Peking University, Beijing, China, and Key Laboratory

    for Mental Health, Ministry of Health, Peking University, Beijing, China. Correspondence should be addressed to M.V.S. ([email protected]).

    Received 14 January; accepted 9 April; published online 5 May 2013; doi:10.1038/nn.3395

    Nectin-3 links CRHR1 signaling to stress-inducedmemory deficits and spine loss

    Xiao-Dong Wang1,5, Yun-Ai Su1,5, Klaus V Wagner1, Charilaos Avrabos1, Sebastian H Scharf 1,Jakob Hartmann1, Miriam Wolf 1, Claudia Liebl1, Claudia Kühne1, Wolfgang Wurst1–4, Florian Holsboer1,Matthias Eder1, Jan M Deussing1, Marianne B Müller1 & Mathias V Schmidt1

    Stress impairs cognition via corticotropin-releasing hormone receptor 1 (CRHR1), but the molecular link between abnormal

    CRHR1 signaling and stress-induced cognitive impairments remains unclear. We investigated whether the cell adhesion molecule

    nectin-3 is required for the effects of CRHR1 on cognition and structural remodeling after early-life stress exposure. Postnatally

    stressed adult mice had decreased hippocampal nectin-3 levels, which could be attenuated by CRHR1 inactivation and mimickedby corticotropin-releasing hormone (CRH) overexpression in forebrain neurons. Acute stress dynamically reduced hippocampal

    nectin-3 levels, which involved CRH-CRHR1, but not glucocorticoid receptor, signaling. Suppression of hippocampal nectin-3

    caused spatial memory deficits and dendritic spine loss, whereas enhancing hippocampal nectin-3 expression rescued the

    detrimental effects of early-life stress on memory and spine density in adulthood. Our findings suggest that hippocampal nectin-3

    is necessary for the effects of stress on memory and structural plasticity and indicate that the CRH-CRHR1 system interacts with

    the nectin-afadin complex to mediate such effects.

    http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395

  • 8/17/2019 Nectin-3 y Stress

    2/10

    NATURE NEUROSCIENCE  VOLUME 16 | NUMBER 6 | JUNE 201 3 70 7

    A R T I C L E S

    RESULTS

    Stress reduces nectin-3 levels via CRH-CRHR1 signaling

    As we reported previously 13, nectin-3 is enriched in hippocampalCA3 neurons (Fig. 1a). Using male Crhr1loxP/loxP  ; Camk2a-cre mice24,

    in which the Crhr1 gene is inactivated postnatally in forebrain princi-pal neurons (referred to as CRHR1-CKO hereafter), we investigated

    whether early-life stress (postnatal days 2–9) would lead to down-regulation of nectin-3 expression in a CRHR1-dependent manner. We

    examined nectin-3 (Pvrl3) mRNA and protein levels in adult CRHR1-CKO and wild-type mice exposed to either a standard or impover-

    ished environment early in life (Fig. 1a,b). In wild-type mice withearly stressful experiences, which exhibit impaired spatial learning

    and memory 11, both mRNA and protein levels of hippocampal nectin-

    3 were reduced. In comparison, control and stressed CRHR1-CKOmice showed comparable nectin-3 mRNA and protein levels. Because

    early-life stress increases CRH levels in the adult hippocampus12, weused adult male R26  flopCrh/f lopCrh ; Camk2a-cre mice25 ( flop refers to

    loxP -flanked stop) with postnatal overexpression of the Crh gene in

    forebrain principal neurons (referred to as CRH-COE hereafter) totest whether CRH overexpression would evoke similar effects onnectin-3 expression to early-life stress (Fig. 1c,d). Compared with

    the wild-type controls, stress-naive CRH-COE mice with prominentcognitive deficits11 had lower nectin-3 mRNA and protein levels in

    the hippocampus. These results indicate that early-life stress–inducedreductions of nectin-3 expression involve CRH-CRHR1 signaling.

    To further elucidate the effects of stress on nectin-3 expression, weevaluated nectin-3 levels following an acute severe stress challenge in

    adulthood (Fig. 1e,f ). A brief (5 min) exposure to social defeat stressdynamically regulated nectin-3 expression in the adult hippocampus.

    At 4 h, but not 1 h, 8 h or 24 h after the acute stress, hippocampalnectin-3 levels were substantially reduced. Notably, a single treatment

    with the glucocorticoid receptor agonist dexamethasone (10 mg perkg of body weight) failed to alter nectin-3 levels (Supplementary

    Fig. 1a), indicating that glucocorticoid receptor may not mediate theeffects of stress on nectin-3 expression.

    CRH-CRHR1 signaling regulates nectin-3 expression

    To dissect the involvement of the CRH-CRHR1 system in stress- regulated nectin-3 expression, we manipulated CRH-CRHR1 signaling

    in acute hippocampal slices. Consistent with the dynamic regulation

    pattern by acute stress, 4 h (Fig. 2a), but not 1 h (Supplementary

    Fig. 1b), of in vitro CRH (50 nM) treatment reduced hippocampal

    nectin-3 protein levels. Pre-incubating the slices with the selectivenonpeptide CRHR1 antagonist DMP696 (100 nM) reversed such

    effects (Fig. 2b).We continued to examine nectin-3 expression following CRH

    administration in vivo (Fig. 2c,d). To avoid the confounding effectsof mild stress induced by handling, we anesthetized mice in their

    home cages and delivered drugs intracerebroventricular (icv). At 4 h

    after icv CRH (0.2 mM) infusion, total and membrane nectin-3 lev-els in the hippocampus decreased (Fig. 2c). Notably, icv infusion ofcorticosterone (2.9 mM) did not alter nectin-3 levels (SupplementaryFig. 1c). Co-administration of CRH with DMP696 (1 mM) preventedCRH-induced reductions in both total and membrane nectin-3 levels

    (Fig. 2d). Moreover, the selective mitogen-activated protein kinase(MAPK) kinase (MEK) inhibitor U0126 (2.6 mM), but not the protein

    kinase A (PKA) inhibitor Rp-cAMPS (4 mM), attenuated the effects ofCRH. These results suggest that the CRH-CRHR1 system modulates

    hippocampal nectin-3 expression via the MAPK pathway.Next, we examined the colocalization of CRHR1 and nectin-3 in

    cortical and subcortical neurons. Because of the lack of reliable anti-bodies to CRHR1 (ref. 26), we used both CRHR1–enhanced green

    a

       W   T

       C   K   O

    CT ES

    0

    20

    40

    60

    80

    100

    120

    140

    WT CKO

    CTES

    CTES

    11 8   7   7

       N  o  r  m  a

       l   i  z  e

       d  n  e  c

       t   i  n  -

       3  m

       R   N   A

       (   %  o

       f   C   T  -   W

       T   )

       W   T

       C   O   E

    0

    20

    40

    60

    80

    100

    120

    140

    WT COE

    *

    9   9

       N  o  r  m  a

       l   i  z  e

       d  n  e  c   t   i  n

      -   3  m

       R   N   A

       (   %  o

       f   W   T

       )

    0

    20

    40

    60

    80

    100

    120

    140

    *

    WT CKO

    5   5   5   6

       R  a

       t   i  o  s  o

       f  n  e  c

       t   i  n  -   3

       t  o  a  c

       t   i  n

       (   %  o

       f   C   T  -   W

       T   )

    b

    Nectin-3

    Actin

    CT   ES

    WT CKO

    CT   ES

    0

    20

    40

    60

    80

    100

    120

    140

       R  a

       t   i  o  s  o

       f  n  e  c   t   i  n  -   3

       /  a  c

       t   i  n

       (   %  o

       f   W   T

       )

    **

    6   6

    WT COE

    1 4 8 24

    Time after acute social defeat stress (h)

       D  e

       f  e  a

       t   C  o  n

       t  r  o

       l

    e

    *

    1 4 8 24   N  o  r  m  a

       l   i  z  e

       d  n  e  c

       t   i  n  -   3  m   R

       N   A

       (   %  o

       f  c  o  n

       t  r  o

       l   )

    0

    20

    40

    60

    80

    100

    120

    140   Control

    Defeat

    Time after acute social defeat stress (h)

    9   9 1010   10 10 1010

    f

    c

    Nectin-3

    Actin

    WT COE

    d

    *

    1 4 8 24   R  a

       t   i  o  s  o

       f  n  e  c

       t   i  n  -   3

       t  o  a  c

       t   i  n

       (   %  o

       f  c  o  n

       t  r  o

       l   )

    0

    20

    40

    60

    80

    100120

    140   Control

    Defeat

    Time after acute social defeat stress (h)

    6   10 86   10 8 6   6

    1 4 8 24

    Time after acute social defeat stress (h)

    Nectin-3

    Actin

    Control Defeat Control Defeat Control Defeat Control Defeat

    Figure 1  Regulation of hippocampal nectin-3 expression by stress and the involvement of CRH-CRHR1

    signaling. (a) Early-life stress downregulated CA3 nectin-3 mRNA in 7–8-month-old male mice (stress

    effect: F 1,29 = 4.373, P  = 0.045, two-way ANOVA). CT, control; ES, early-life stress; WT, wild type;

    CKO, CRHR1-CKO; CT-WT, nonstressed and wild-type mice. (b) At protein levels, early-life stress reduced

    hippocampal nectin-3 expression in wild-type, but not CRHR1-CKO, mice (stress × genotype interaction:

    F 1,17 = 5.507, P  = 0.0313, two-way ANOVA; *P  < 0.05, Bonferroni’s test). (c,d) Conditional forebrain

    CRH overexpression mimicked the effects of early-life stress on nectin-3 expression. CA3 nectin-3 mRNA

    levels (c) and hippocampal nectin-3 protein levels (d) were reduced in 7–8-month-old male CRH-COE

    mice (t 16 = 2.869, *P  < 0.05; t 10 = 3.336, **P  < 0.01; unpaired t  test). (e,f) Acute social defeat stress

    disrupted hippocampal nectin-3 expression at both mRNA (stress effect: F 1,70 = 4.164, P  = 0.045,two-way ANOVA) and protein levels (stress effect: F 1,52 = 4.386, P  = 0.041, two-way ANOVA) in

    3-month-old male mice. At 4 h after the stress, nectin-3 mRNA levels ( e) were markedly decreased

    (t 17 = 2.144, *P  < 0.05, unpaired t  test), which were reflected at the protein levels (f, t 18 = 2.177,

    *P  < 0.05, unpaired t  test). Data represent mean ± s.e.m. Scale bars in the representative in situ  

    hybridization images represent 500 µm (a,c,e). For full-length blots (b,d,f), see Supplementary Figure 10.

    In this and all subsequent figures, the number of mice is indicated in the bar graphs.

  • 8/17/2019 Nectin-3 y Stress

    3/10

    708 VOLUME 16 | NUMBER 6 | JUNE 2013 NATURE NEUROSCIENCE

    A R T I C L E S

    fluorescent protein (EGFP) reporter mice26 and CRHR1 tau-lacZ  

    reporter mice27. We observed that CRHR1 partially colocalized withnectin-3 in hippocampal pyramidal neurons (Fig. 3) and neurons

    in various cortical and subcortical regions (Supplementary Fig. 2),which is suggestive of their functional interactions.

    Hippocampal nectin-3 knockdown impairs spatial memory

    Early-life stress and enhanced hippocampal CRH-CRHR1 signal-ing impair cognition11,12  and reduced nectin-3 expression levels.

    To gain insight into the behavioral and structural consequencesof nectin-3 downregulation in the adult hippocampus, we used

    adeno-associated virus (AAV)-mediated nectin-3 knockdown andinvestigated whether reduced levels of nectin-3 would impair cog-

    nition, thereby mimicking the early-life stress–induced phenotype.AAV-shSCR (contains a scrambled short hairpin RNA sequence)

    was chosen as the negative control and AAV-shNEC (contains thesequence for a short hairpin RNA specific

    for nectin-3) was used to suppress nectin-3

    expression in vivo.We found that the AAV-shNEC vector

    specifically reduced hippocampal nectin-3levels, whereas the levels of other nectins

    and related molecules remained unchanged

    (Supplementary Fig. 3). We also exam-ined the extent of viral transfection in the

    hippocampus (Supplementary Fig. 4). In AAV-shNEC–treated mice,

    nectin-3 expression levels were downregulated in CA3, dentate gyrusand CA1 throughout the dorsal hippocampus (Fig. 4a). Short-term

    spatial working memory was not altered in AAV-shNEC–treated mice,as seen by similar spontaneous alternation behavior in the Y-maze

    task compared with the controls (Fig. 4b). In the spatial object recog-nition test, AAV-shNEC–treated mice showed a markedly impaired

    performance (Fig. 4c  and Supplementary Fig. 5a,b). Moreover,AAV-shSCR–treated mice discriminated the novel object from the

    familiar one, whereas AAV-shNEC–treated mice failed to show objectdiscrimination (Supplementary Fig. 5b). In the spatial training ses-

    sions of the Morris water maze task (Fig. 4d), the spatial learning ofAAV-shNEC–treated mice was preserved. In the probe trial, how-

    ever, control mice, but not AAV-shNEC–treated mice, searched thetarget quadrant longer than the other quadrants. Together, these data

    indicate that suppression of hippocampal nectin-3 reproduces the

    Figure 3  Colocalization of CRHR1 and

    nectin-3 in hippocampal CA1 pyramidal neurons.

    (a,b) In 3-month-old female CRHR1-EGFP

    reporter mice and CRHR1 tau-lacZ  reporter mice,

    CRHR1-EGFP (a) and CRHR1-β-galactosidase (b)

    partially colocalized with nectin-3 in CA1

    pyramidal neurons. DAPI, 4′,6-diamidino-2-

    phenylindole. Arrowheads in the representative

    confocal images indicate neurons in which

    the colocalization of CRHR1 and nectin-3 was

    observed. Scale bars represent 20 µm.

    b

    CRHR1–β-gal Nectin-3 MergedDAPI

    a

    CRHR1-EGFP Nectin-3 MergedDAPI

    Figure 2  Regulation of hippocampal nectin-3 by CRH-CRHR1 signaling. (a) After 4 h of CRH application in vitro , hippocampal nectin-3 protein levels

    were reduced (t 8 = 2.56, *P  < 0.05, unpaired t  test). ACSF, artificial cerebrospinal fluid. (b) Adding the CRHR1 antagonist DMP696 to the brain slices

    at 10 min before the 4-h CRH treatment prevented the effects of CRH on nectin-3 expression in vitro  (interaction: F 1,16 = 5.446, P  = 0.033, two-way

    ANOVA; *P  < 0.05, Bonferroni’s test). (c) Intracerebroventricular administration of CRH downregulated total (t 20 = 2.472, *P  < 0.05, unpaired t  test)

    and membrane (t 8 = 4.632, **P  < 0.01, unpaired t  test) nectin-3 levels in the hippocampus 4 h later. (d) Co-administration of CRH with DMP696

    or U0126, but not Rp-cAMPS, attenuated the CRH-induced reduction of total nectin-3 in the hippocampus at 4 h after icv injection ( F 4,25 = 3.276,

    P  = 0.027, one-way ANOVA), whereas DMP696 prevented the effects of CRH on membrane nectin-3 expression ( F 4,25 = 2.796, P  = 0.048, one-wayANOVA). *P  < 0.05, Fisher’s LSD test. Data represent mean ± s.e.m. All tested mice were 3-month-old C57BL/6N males. For full-length blots, see

    Supplementary Figure 10.

    Nectin-3

    Actin

    ACSF   CRH

    a

    ACSF CRH

    0

    20

    40

    60

    80

    100120

    140

       R  a   t   i  o  s  o   f  n  e  c   t   i  n  -   3   t  o  a  c   t   i  n

       (   %   o

       f   A   C   S   F   )   *

    5   5

    Vehicle DMP696

    0

    20

    40

    60

    80

    100

    120

    140   ACSF

    CRH

       R  a   t   i  o  s  o   f  n  e  c   t   i  n  -   3   t  o  a  c   t   i  n

       (   %   o

       f   A   C   S   F  -  v  e   h

       i  c   l  e   )   *

    55 5   5

    Nectin-3

    Actin

    ACSF CRH ACSF CRH

    V eh ic le D MP 69 6

    b c

      A  C  S   F

      C   R   H

    0

    20

    40

    60

    80

    100120

    140

       R  a   t   i  o  s  o   f  n  e  c   t   i  n  -   3   t  o  a  c   t   i  n

       (   %   o

       f   A   C   S   F   )   *

    11 11

      A  C  S   F  C   R   H

    5  5

    **

    Nectin-3

    Actin

    ACSF CRH

    Total

    ACSF CRH

    Membrane

    d

    6 6 6   6 6

    0

    20

    40

    60

    80

    100120

    140

       R  a   t   i  o  s  o   f  n  e  c   t   i  n  -   3   t  o

      a  c   t   i  n

       (   %   o

       f  v  e   h   i  c   l  e   )

       V  e   h   i  c

       l  e

      C   R   H   C   R

       H  +

       D   M   P  6

      9  6

      C   R   H 

     +

       R  p  -  c  A

       M   P  S

      C   R   H 

     +

       U  0  1  2  6

       V  e   h   i  c

       l  e

      C   R   H   C   R

       H  +

       D   M   P  6

      9  6

      C   R   H 

     +

       R  p  -  c  A

       M   P  S

      C   R   H 

     +

       U  0  1  2  6

       V  e   h   i  c

       l  e  C   R

       H  C   R   H 

     +

       D   M   P  6

      9  6  C   R

       H  +

       R  p  -  c  A   M

       P  S  C   R   H 

     +

       U  0  1  2  6

       V  e   h   i  c

       l  e  C   R

       H  C   R   H 

     +

       D   M   P  6

      9  6  C   R

       H  +

       R  p  -  c  A   M

       P  S  C   R   H 

     +

       U  0  1  2  6

    *   *   *   *

    *

    6 6 6   6 6

    * *

    *

    Nectin-3

    Actin

    Total Membrane

  • 8/17/2019 Nectin-3 y Stress

    4/10

    NATURE NEUROSCIENCE  VOLUME 16 | NUMBER 6 | JUNE 201 3 70 9

    A R T I C L E S

    cognitive effects of early-life stress and that nectin-3 is essential forhippocampus-dependent long-term spatial memory.

    Suppression of nectin-3 evokes dendritic spine loss

    To assess the effects of long-term nect in-3 knockdown on structuralplasticity in vivo, we quantified the number and size of dendritic

    spines in EGFP-expressing hippocampal neurons in AAV-shSCR–and AAV-shNEC–treated mice. Similar to adult mice with a history

    of early-life stress11, AAV-shNEC–treated mice showed a markedreduction in spine density in CA3 (Fig. 5a,b), dentate gyrus and

    CA1 (Supplementary Fig. 6) principal neurons. Spine volumeand spine head diameter in CA3 neurons were unaffected by

    AAV-shNEC (Fig. 5c).Notably, we observed that approximately 17% of spines expressed

    nectin-3 (17.41 ±  0.89, n  = 2,480 spines analyzed). The density ofnectin-3–positive spines was reduced in AAV-shNEC–treated mice

      s   h   S   C   R

      s   h   N   E   C

    a

    –1.76   –2.08 –2.40 –2.72

    Bregma (mm)

    CA1

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    Bregma (mm)

    10

    15

    20

    0

    25

    ******

      *****   *

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

    DG

    10

    15

    20

    0

    25

    ***

    ***** **

    ** **

    Bregma (mm)

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    CA3

    20

    40

    60

    0

    80

    ***************

    *********

    shSCR

    shNEC

    Bregma (mm)

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    b

    shSCR shNEC   S  p  o

      n   t  a  n  e  o  u  s  a   l   t  e  r  n  a   t   i  o  n   (   %   )

    0

    20

    40

    60

    80

    Y-maze

    12121

    2

    3

    4

    5

    c

    s hS CR s hN EC

       N  o  v  e   l   t  o   k  n  o  w  n  r  a   t   i  o

    0

    Object recognition

    1212

    *

    d

       E  s  c  a  p  e   l  a   t  e  n  c  y   (  s   )

    0

    10

    20

    30

    40

    50

    60   shSCRshNEC

    Day 1 Day 2 Day 3

    Acquisition

    s hS CR s hN EC

       T   i  m  e   i  n  q  u  a   d  r  a  n   t  s   (  s   )

    0

    5

    10

    15

    20

    25

    30 TargetRightOppositeLeft

    ##

    Probe trial

    23 22

    Figure 4  Hippocampal nectin-3 knockdown impaired long-term spatial memory. (a) At 1 week after the behavioral tests , AAV-shNEC–induced

    hippocampal nectin-3 knockdown was verified by in situ  hybridization. Nectin-3 mRNA levels in CA3, dentate gyrus (DG) and CA1 were significantly

    reduced by AAV-shNEC (treatment effect: F 1,30 = 96.218, P  < 0.00001; F 1,29 = 22.193, P  = 0.00006; F 1,30 = 11.486, P  = 0.002; one-way

    repeated-measures ANOVA; n  = 16 mice per group). *P  < 0.05, **P  < 0.01, ***P  < 0.001, unpaired t  test. Scale bar in the representative in situ  

    hybridization images represents 500 µm. (b–d) Two cohorts of 3-month-old C57BL/6N males received an intrahippocampal viral injection and were

    tested in the Y-maze and spatial object recognition tasks (the first cohort only), and then in the Morris water maze task (both cohorts) 4 weeks

    later. (b) Spatial working memory was comparable between groups (t 22 = 0.996, P  = 0.33, unpaired t  test). (c) The ratio of time spent with the

    displaced (novel) object versus the non-displaced (known) object, a measure of spatial recognition memory, was significantly lower in AAV-shNEC

    mice compared to the controls (t 21.743 = 2.104, *P  < 0.05, Welch’s t  test). (d) In the Morris water maze test, AAV-shNEC mice showed similar spatial

    learning performance to AAV-shSCR mice (F 1,43 = 1.598, P  = 0.213, one-way repeated-measures ANOVA). In the probe trial, AAV-shSCR mice, but

    not AAV-shNEC mice, spent more time searching the target quadrant where the platform was previously placed than the other quadrants (AAV-shSCR:

    t 22 = 3.38,##P  < 0.01; AAV-shNEC: t 21 = 1.321, P  = 0.201; paired t  test). Data represent mean ± s.e.m.

    a

    EGFP Nectin-3 Merged

    shSCR

    shNEC

    shSCR

    shNEC

    0.1 0.2 0.3 0.4 0.5 0.60

    20

    40

    60

    80

    100

    Spine volume (µm3)

       C  u  m  u   l  a   t   i  v  e   d   i  s   t  r   i   b  u   t   i  o  n   (   %   )

    0.1 0.2 0.3 0.4 0.5 0.6 0.70

    20

    40

    60

    80

    100

    Spine head diameter (µm)

       C  u  m  u   l  a   t   i  v  e   d   i  s   t  r   i   b  u   t   i  o  n   (   %   )

    shSCR

    shNEC

    cb

    0

    2

    4

    6

    8

    10

       N  u  m   b  e  r  o   f  s  p   i  n  e  s  p  e  r   1   0     µ  m

      s   h   S  C

       R

      s   h   N   E  C

    **

    6   6

      s   h   S  C

       R

      s   h   N   E  C

    ***

    6   6

    Total   Nectin-3+

    Figure 5  Nectin-3 knockdown reduced dendritic

    spine density in CA3 pyramidal neurons.

    (a) Representative deconvolved z  stacks showing

    EGFP-filled dendrites and spines (green) and

    nectin-3–immunoreactive puncta (red) in

    the stratum lacunosum-moleculare of CA3.

    Arrowheads indicate nectin-3–positive spines.

    Scale bars represent 1 µm. (b) Suppression ofnectin-3–induced spine loss in CA3 pyramidal

    neurons (t 10 = 4.097, **P  < 0.01, unpaired

    t  test). The number of nectin-3–positive spines

    was significantly less in AAV-shNEC mice

    compared with AAV-shSCR mice (t 10 = 10.505,

    ***P  < 0.001, unpaired t  test). Data represent

    mean ± s.e.m. (c) Nectin-3 knockdown did not

    affect spine volume (t 1801.301 = 0.6693,

    P  = 0.5034, Welch’s t  test) or spine head

    diameter (t 2029.375 = 0.9195, P  = 0.358,

    Welch’s t  test). Male C57BL/6N mice were

    3 months old when they were injected with virus

    and were killed after 4 weeks of recovery. For

    each mouse, 8–14 dendrites were analyzed.

  • 8/17/2019 Nectin-3 y Stress

    5/10

    710 VOLUME 16 | NUMBER 6 | JUNE 2013 NATURE NEUROSCIENCE

    A R T I C L E S

    (Fig. 5b), whereas the numbers of spines lacking nectin-3 were compara-

    ble between groups (AAV-shSCR, 6.59± 0.34 spines per 10µm of dendrite;AAV-shNEC, 6.35± 0.15; t 10 = 0.637, P  = 0.539, unpaired t  test).

    Nectin-3 overexpression rescues stress-induced memory loss

    Because the suppression of hippocampal nectin-3 mimicked thecognitive impairments by early-life stress, we examined whether

    1

    2

    3

    4

    5

    6

    c

    0

       N

      o  v  e   l   /   K  n  o  w  n  r  a   t   i  o

    Null OE

    Object recognition

    *

    11 11   12 12

    d

    0

    10

    20

    30

    40

    50

    60CT-nullES-null

    CT-OEES-OE

       E  s  c  a  p  e

       l  a   t  e  n  c  y

       (  s   )

    Day 1 Day 2 Day 3

    Acquisition

    0

    10

    20

    30

    40TargetRight

    OppositeLeft

       T   i  m  e   i  n  q  u  a   d  r  a  n   t  s   (  s   )

    Probe trial

    ###

    ##

    CT ES CT ES

    Null OE

    9 911 12

    b

    0

    20

    40

    60

    80

       S  p  o  n   t  a  n  e  o  u  s  a   l   t  e  r  n  a   t   i  o  n   (   %   )

    Null OE

    Y-maze

    11 11   13 12

    a

    Null

    OE

    CT ES

    0

    20

    40

    120

    140

    160

    180

    CT-nullES-nullCT-OE

    CTES

    CTES

    ES-OE

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

    Bregma (mm)

    CA3

    *

    ###

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    0

    10

    80

    120

    160

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

    Bregma (mm)

    *

    ###

    DG

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    0

    10

    40

    80

    120

    160

       N  e  c   t   i  n  -   3  m   R   N   A

       (  a  r   b   i   t  r  a  r  y  u  n   i   t  s   )

    Bregma (mm)

    ###

    CA1

      –  1 .   6   0

      –  1 .   7   6

      –  1 .   9   2

      –   2 .   0   8

      –   2 .   2  4

      –   2 .  4   0

      –   2 .   5   6

      –   2 .   7   2

    Figure 6  Hippocampal nectin-3 overexpression reversed early-life stress–induced cognitive deficits. (a) Following the behavioral tests, hippocampalnectin-3 mRNA levels were determined. Nectin-3 mRNA levels in CA3, dentate gyrus and CA1 were significantly increased by AAV-OE (treatment

    effect: F 1,22 = 1740.24,###P  < 0.001; F 1,22 = 749.003, P  < 0.001; F 1,22 = 68.369, P  < 0.001; two-way repeated measures ANOVA). Compared with

    CT-null mice, nectin-3 mRNA levels were lower in the CA3 and dentate gyrus of ES-null mice (stress effect: F 1,10 = 5.581, *P  < 0.05; F 1,10 = 10.039,

    P  < 0.05; one-way repeated measures ANOVA; n  = 6–7 mice per group). Scale bar in the representative in situ  hybridization images represents 500 µm.

    (b–d) We injected 5-month-old C57BL/6N males, with or without early-life stress, intrahippocampally with virus and subjected them to cognitive testing

    after 4 weeks of recovery. (b) In the Y-maze test, early-life stress impaired spatial working memory (stress effect: F 1,43 = 4.808, P  = 0.0338, two-way

    ANOVA). (c) In the spatial object recognition task, nectin-3 overexpression restored spatial memory in stressed mice (treatment effect: F 1,42 = 4.127,

    P  = 0.0486; interaction: F 1,42 = 4.546, P  = 0.0389; two-way ANOVA; *P  < 0.05, Tukey’s test). (d) In the Morris water maze test, all groups of mice

    performed similarly in the spatial acquisition sessions. In the probe trial, nectin-3 overexpression increased the ratio of time spent exploring the target

    quadrant over non-target quadrants (treatment effect: F 1,36 = 5.96, P  = 0.02, two-way ANOVA). CT-null, CT-OE and ES-OE, but not ES-null, mice spent

    more time searching the target quadrant than the others (CT-null: t 8 = 2.72,#P  < 0.05; CT-OE: t 8 = 4.944,

    ##P  < 0.01; ES-OE: t 11 = 3.536, P  < 0.01;

    ES-null: t 10 = 1.684, P  = 0.123; paired t  test). Data represent mean ± s.e.m.

    0.1 0.2 0.3 0.4 0.5 0.6 0.7

    0

    20

    40

    60

    80

    100

    CT-null

    ES-null

    CT-OE

    ES-OE

    Spine head diameter (µm)

       C  u  m  u   l  a   t   i  v  e   d   i  s   t  r   i   b  u   t   i  o  n   (   %   )

    0.1 0.2 0.3 0.4 0.5 0.60

    20

    40

    60

    80

    100

    CT-null

    ES-null

    CT-OE

    ES-OE

    Spine volume (µm

    3

    )

       C  u  m  u   l  a   t   i  v  e   d   i  s   t  r   i   b  u   t   i  o  n   (   %   )

    ca

    EYFP Nectin-3 Merged

    CT-null

    ES-null

    CT-OE

    ES-OE

    b

    **

    Null OE

    *

       N  u  m   b  e  r  o   f  s  p   i  n  e  s  p  e  r   1   0     µ  m

    Null OE

    **

    CT

    ES

    *

    0

    2

    4

    6

    8

    10

    Total Nectin-3+

    3

    3

    45 5

    5   4  5

    Figure 7  Nectin-3 overexpression rescued early-life stress–evoked spine loss and spine volume changes.

    (a) Representative deconvolved z  stacks showing enhanced yellow fluorescent protein (EYFP)-filled dendrites and

    spines (green) and nectin-3–immunoreactive puncta (red) in the stratum lacunosum-moleculare of CA3. Scale

    bars represent 1 µm. (b) Early-life stress reduced spine density in control mice, which was reversed by nectin-3

    overexpression (stress effect: F 1,13 = 4.833, P  = 0.0466; interaction: F 1,13 = 9.827, P  = 0.0079; two-way ANOVA;

    *P  < 0.05, **P  < 0.01, Bonferroni’s test). Overexpression of nectin-3 increased the number of nectin-3–positive

    spines (treatment effect: F 1,13 = 23.94, P  = 0.0003, two-way ANOVA). Data represent mean ± s.e.m. (c) Nectin-3

    overexpression reversed early-life stress–induced spine volume changes (interaction: F 1,5365 = 15.191,

    P  = 0.0000984, two-way ANOVA). Spine volume in ES-null mice was larger than CT-null mice (P  < 0.05, Tukey’s

    test), whereas ES-OE mice had smaller spines compared with CT-OE mice (P  < 0.05, Tukey’s test). In addition, nectin-

    3 overexpression increased spine volume (CT-null versus CT-OE: P  < 0.001, Tukey’s test) and spine head diameter in

    control mice (treatment effect: F 1,5880 = 12.408, P  = 0.000431, two-way ANOVA; CT-null versus CT-OE: P  < 0.01, Tukey’s test). Male Thy1-YFPH mice

    were 5 months old when they were injected with virus and were killed after 4 weeks of recovery. For each mouse, 10–15 dendrites were analyzed.

  • 8/17/2019 Nectin-3 y Stress

    6/10

    NATURE NEUROSCIENCE  VOLUME 16 | NUMBER 6 | JUNE 201 3 71 1

    A R T I C L E S

    enhancing nectin-3 expression in the adult hippocampus would amel-iorate the deleterious effects of early-life stress. AAV-null (an empty

    AAV vector) was chosen as the control vector, whereas AAV-OE (con-tains the sequence for nectin-3) was used to overexpress nectin-3.

    We observed that AAV-OE increased hippocampal nectin-3 levelsat 4 weeks after injection, and the effects lasted for at least 8 weeks

    post-injection (Fig. 6a and Supplementary Fig. 7a). The specificity ofAAV-OE (Supplementary Fig. 7b–d) and the extent of viral transfec-

    tion (Supplementary Fig. 8) were validated.In the Y-maze test (Fig. 6b), early-life stress impaired short-term

    spatial memory, and AAV-OE failed to reverse such effects. In the spa-tial object recognition task, AAV-OE restored cognitive performance

    in postnatally stressed mice (Fig. 6c). In addition, early life–stressedmice injected with AAV-null (ES-null), but not control mice injected

    with either AAV-null (CT-null) or AAV-OE (CT-OE) or postnatallystressed mice injected with AAV-OE (ES-OE), failed to discriminate

    the novel object from the known one (Supplementary Fig. 9). In theMorris water maze test, although no difference in spatial acquisition

    was found among groups, ES-null mice searched the target quadrantand the other quadrants similarly in the probe trial, indicative of

    spatial memory impairments (Fig. 6d). Conversely, ES-OE miceshowed intact spatial memory.

    Nectin-3 overexpression reverses stress-induced spine loss

    In AAV-null– and AAV-OE–injected mice, dendrites and spines couldnot be visualized because the vectors did not express EGFP. Thus,

    we used Thy1-YFPH transgenic mice, in which a subpopulation ofhippocampal pyramidal neurons are selectively labeled by EYFP,

    and determined the effects of early-life stress and nectin-3 over-

    expression on dendritic spine plasticity (Fig. 7).Postnatally stressed adult mice had reduced spine density in CA3

    neurons, and nectin-3 overexpression restored the number of spines(Fig. 7b). Notably, although CT-OE and ES-OE mice had signifi-

    cantly more nectin-3–expressing spines than their respective controls,the number of nectin-3–negative spines was reduced (treatment effect,

    F 1,13 = 14.18, P  = 0.0024, two-way ANOVA). Compared with CT-nullmice, this resulted in a shift to a higher ratio of nectin-3–positive to

    nectin-3–negative spines, but the number of total spines remainedsimilar. In addition, overexpression of nectin-3 increased spine volume

    and spine head diameter in control mice (Fig. 7c). ES-null mice alsohad larger spine volume than CT-null mice, indicative of compensatory

    enlargements of the remaining spines. In contrast, stress-induced spineenlargement in ES-OE mice was reversed by nectin-3 overexpression.

    CRHR1 signaling and nectin-3 modulate L-afadin levels

    L-afadin, an F-actin–binding protein that connects nectin-3 to theactin cytoskeleton, has been shown to modulate spine formation

    and remodeling19,28,29. Thus, the effects of nectin-3 and possibly of

    stress and CRH-CRHR1 signaling on spine remodeling may be medi-ated by L-afadin. Consistent with this reasoning, we observed that

    hippocampal L-afadin levels decreased following 4 h of in vitro treat-ment with 50 nM CRH (Fig. 8a), which potentially induces spine loss

    under similar conditions22,30. Blockade of CRHR1 by DMP696 pre- vented the inhibitory effects of CRH on L-afadin levels (Fig. 8b).

    We observed that L-afadin immunoreactivity in the stratum radia-tum and stratum lacunosum-moleculare was reduced by nectin-3

    knockdown (Fig. 8c). Conversely, nectin-3 overexpression increased

    hippocampal L-afadin protein levels in stressed mice (Fig. 8d). Thus,these findings support L-afadin as a potential molecular substrate thatmediates nectin-3–dependent cognitive and structural changes, which

    in turn contribute to stress- and CRH-induced effects.

    DISCUSSION

    Unraveling the molecular machineries responsible for stress-induced

    cognitive dysfunction will provide insight into the neurobiology oflearning and memory and stress-related psychiatric disorders. Here,

    we identified the synaptic CAM nectin-3 as an important componentin stress- and CRH-evoked effects. Exposure to either acute severe

    stress or early-life stress downregulated nectin-3 expression levelsin the adult hippocampus via CRH-CRHR1 signaling. Suppression

    a

    L-afadin

    Actin

    ACSF CRH

    ACSF CRH0

    20

    40

    60

    80

    100

    120

    140*

       R  a   t   i  o  s  o   f   L  -  a   f  a   d   i  n   t  o

      a  c   t   i  n

       (   %   o

       f   A   C   S   F   )

    5   5

       N  o  r  m  a   l   i  z  e   d   L  -  a   f  a   d   i  n

      p  u  n  c   t  a   d  e  n  s   i   t  y

       (   %   o

       f  s   h   S   C   R   )   *

    0

    20

    40

    60

    80

    100

    120140

    shSCR shNEC

    6   6

    sr + slm

    shSCR shNEC

    slmsr

    cb

    Vehicle DMP696

    0

    20

    40

    60

    80

    100

    120

    140

       R  a   t   i  o  s  o   f   L  -  a   f  a   d   i  n   t  o  a  c   t   i  n

        (   %   o

       f   A   C   S   F  -  v  e   h   i  c   l  e   )   *

    55 5   5

    *

    L-afadin

    Actin

    ACSF CRH ACSF CRH

    Vehicle DMP696

    ACSF CRH

    d

    OE

    CT ES

    Null   slmsr

    0

    20

    40

    60

    80

    100

    120

    140

    Null OE

    *

    4 4   4 4

       N  o  r  m  a   l   i  z  e   d   L  -  a   f  a   d   i  n  p  u  n  c   t  a

       d  e  n  s   i   t  y   (   %   o

       f   C   T  -   N  u   l   l   )

    sr + slm CT

    ES

    Figure 8  Modulation of L-afadin levels by CRH-CRHR1 signaling and nectin-3. (a) Hippocampal L-afadin

    protein levels were reduced after 4 h of CRH application in vitro  (t 8 = 2.631, *P  < 0.05, unpaired t  test).

    (b) Adding DMP696 to the slices at 10 min before the 4-h CRH treatment prevented the effects of CRH

    on L-afadin levels (antagonism effect: F 1,16 = 4.525, P  = 0.0493; interaction: F 1,16 = 5.714, P  = 0.0295;

    two-way ANOVA; *P  < 0.05, Tukey’s test). (c) L-afadin immunoreactivity in the stratum radiatum (sr) and

    stratum lacunosum-moleculare (slm) of CA3 was significantly decreased in AAV-shNEC mice (t 7.028 = 3.302,

    *P  < 0.05, unpaired t  test). Representative coronal sections immunostained for L-afadin are shown.

    (d) Overexpression of nectin-3 increased L-afadin protein levels in the hippocampus (treatment effect: F 1,12 = 10.74, P  = 0.0066, two-way ANOVA;*P  < 0.05, Bonferroni’s test). Representative transverse sections immunostained for L-afadin are shown. Note the clustered L-afadin–immunoreactive

    puncta as indicated by arrows near the border between slm and sr in CT-OE and ES-OE mice. Scale bars represent 100 µm. Data represent mean ± s.e.m.

    Male mice were 3–6 months old. For full-length blots (a,b), see Supplementary Figure 10.

  • 8/17/2019 Nectin-3 y Stress

    7/10

    712 VOLUME 16 | NUMBER 6 | JUNE 2013 NATURE NEUROSCIENCE

    A R T I C L E S

    of hippocampal nectin-3 reproduced the effects of early-life stress,

    destabilized synaptic contacts and hampered spatial memory,whereas overexpression of hippocampal nectin-3 reversed such nega-

    tive effects of early-life stress. Together, our findings link impairednectin-3–driven synaptic adhesion to stress-induced structural and

    functional abnormalities.It has been shown by us and others that elevated hippocampal

    CRH-CRHR1 signaling modulates the negative effects of early-lifestress11,12 and chronic stress13 on hippocampus-dependent learning

    and memory in adult animals. One of the underlying mechanismsis that excessively released CRH acts through CRHR1 and evokes

    structural remodeling31,32, especially the elimination of thin den-

    dritic spines22,30. However, the molecules mediating these structuraleffects have not been fully clarified. Here, we found that hippocam-

    pal nectin-3 expression levels were regulated by early-life stress ina CRHR1-dependent manner: stress exposure during development

    or postnatal forebrain CRH overexpression reduced nectin-3 levels,whereas postnatal forebrain CRHR1 inactivation normalized nectin-3

    levels in early life–stressed mice. These findings greatly expand ourprevious observations that chronic stress during adulthood reduces

    hippocampal nectin-3 levels via CRHR1 (ref. 11), and pinpoint the

    role of the CRH-CRHR1 system in stress-induced downregulationof nectin-3. On the basis of our finding that acute severe stress tran-siently suppressed hippocampal nectin-3 expression, the influences of

    stress on nectin-3 expression may shift from short-term inhibition toenduring suppression after repeated exposure during development.

    Using both in vitro  and in vivo  approaches, we found that thedynamic regulation of nectin-3 expression by acute stress was medi-

    ated by CRH-CRHR1 signaling. Activation of central glucocorticoidreceptor by systemic administration of dexamethasone or intracer-

    ebroventricular infusion of corticosterone failed to reproduce theeffects of stress on nectin-3, indicating that such effects are gluco-

    corticoid receptor independent. Taking the functional relevance andspatial distribution patterns of CRHR1 and nectin-3 into account,

    CRHR1 hyperactivity likely disrupts synaptic adhesion through indi-rect interactions with nectin-3. Indeed, we found that the effects of

    CRH and CRHR1 could be modulated by the MAPK signaling path-way, but not by the cAMP-PKA pathway, as the inhibition of MEK,

    which phosphorylates MAPK, attenuated CRH-initiated nectin-3

    downregulation. Notably, CRH specifically increases the levels ofphosphorylated MAPK through CRHR1 in areas CA3 and CA1

    (ref. 33). The regional characteristics of the MAPK pathway activatedby CRHR1 and the expression pattern of nectin-3 in the hippocampus

    therefore provide a molecular basis for the circuit-specific effects ofstress. Nonetheless, although our data provide evidence that stress and

    excessive CRH-CRHR1 signaling reduce nectin-3 levels by inhibitinggene and protein expression, it is unclear whether other processes,

    such as the cleavage and degradation of the nectin-3 protein, are influ-

    enced by stress and CRHR1. Moreover, whether acute and chronicadult stress and early-life stress modulate nectin-3 levels through thesame molecular cascade remains an open question. Future studies are

    needed to disentangle the entire molecular mechanisms responsiblefor stress and CRHR1-induced downregulation of nectin-3.

    Synaptic CAMs have been implicated in hippocampus-dependentlearning and memory and cognitive disorders5–7. The disruption of

    synaptic adhesion mediated by N-cadherin, one of the key CAMsin adherens junctions, impairs long-term, but not short-term,

    hippocampus-dependent emotional memory 34. However, the roles ofnectins, the other group of CAMs that organize adherens junctions, in

    cognition remain unknown. We found that hippocampal nectin-3 knock-down disrupted long-term spatial memory, mimicking the cognitive

    consequences of early-life stress exposure. Notably, the protein levels

    of other nectins, N-cadherin and β-catenin were unaffected by thesuppression of nectin-3, indicating that the observed cognitive effects

    were nectin-3 specific. On the other hand, overexpressing nectin-3in the adult hippocampus did not improve cognitive performance

     per se , but reversed the negative effects of early-life stress on cogni-tion. These results highlight the crucial role of nectin-3–driven syn-

    aptic adhesion, which is susceptible to early-life stress, in long-termspatial memory.

    The cadherin-catenin complex and the nectin-afadin complexcooperate to modulate structural and synaptic plasticity 14–19.

    Although the roles of the cadherin-catenin complex in these pro-

    cesses have been investigated29,35,36, it was unclear whether nectin-3is involved in synaptic and structural remodeling. We found that the

    inhibition of nectin-3 specifically decreased the number of dendriticspines expressing nectin-3, resulting in a reduction in the total number

    of spines. Conversely, overexpression of nectin-3 reversed early-lifestress–induced spine loss. Nectin-3 knockdown did not change the

    overall size of the remaining spines, whereas nectin-3 overexpressionincreased spine volume and spine head diameter. Notably, early-life

    stress increased the volume, but not the head diameter, of the spines,

    possibly through a compensatory mechanism for the loss of spines.However, nectin-3 overexpression normalized spine dimensions instressed mice. These data suggest that, although reduced nectin-3

    levels do not fully mediate the effects of early-life stress on spinemorphology, enhancing its expression could rescue the abnormal

    changes by stress. In addition, it should be noted that the densityof spines and synapses remains unaltered in conventional nectin-3

    knockout mice, although the formation of adherens junctions ismarkedly impaired16. We ascribe this to the differences in the extent

    and duration of nectin-3 inhibition, as well as compensatory effectsamong various CAMs1.

    L-afadin links nectin-3 to the cadherin-catenin complex and actincytoskeleton1 and participates in spine formation and remodeling19,28,29.

    We observed that hippocampal L-afadin levels could be modulatedby CRH-CRHR1 signaling. Moreover, hippocampal nectin-3 knock-

    down reduced L-afadin levels, whereas nectin-3 overexpression couldincrease L-afadin levels in postnatally stressed mice. These findings

    imply that the effects of nectin-3 on structural remodeling and cog-nition are likely mediated by L-afadin. However, the involvement of

    L-afadin in learning and memory merits further investigations.In summary, our findings indicate that early-life stress exposure

    disrupts nectin-3–mediated axodendritic adhesion in hippocampalneurons through the enhancement of CRH-CRHR1 signaling, which

    in turn destabilizes dendritic spines and compromises hippocampus-dependent learning and memory. The discovery that restoring nectin-3

    levels ameliorates early-life stress–induced memory deficits maystimulate the development of therapeutic strategies for stress-related

    cognitive disorders.

    METHODS

    Methods and any associated references are available in the online  version of the paper.

    Note: Supplementary information is available in the online version of the paper .

    ACKNOWLEDGMENTS

    We are grateful to D. Harbich and B. Schmid for technical assistance. This workwas supported by the European Community’s Seventh Framework Program (FP7,Project No. 201600), the Bundesministerium für Bildung und Forschung withinthe framework of the NGFN-Plus (FKZ: 01GS08151 and 01GS08155) and by theInitiative and Networking Fund of the Helmholtz Association in the framework ofthe Helmholtz Alliance for Mental Health in an Ageing Society (HA-215).

    http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395http://www.nature.com/doifinder/10.1038/nn.3395

  • 8/17/2019 Nectin-3 y Stress

    8/10

    NATURE NEUROSCIENCE  VOLUME 16 | NUMBER 6 | JUNE 201 3 71 3

    A R T I C L E S

    AUTHOR CONTRIBUTIONS

    X.-D.W. and M.V.S. designed the experiments. X.-D.W., Y.-A.S., K.V.W., C.A.,S.H.S., J.H., M.W., C.L. and C.K. performed the experiments. X.-D.W., Y.-A.S.,C.A. and M.W. analyzed the data. M.E., J.M.D., M.B.M. and M.V.S. supervised theexperiments. X.-D.W., W.W., F.H., M.E., J.M.D., M.B.M. and M.V.S. wrote the paper.

    COMPETING FINANCIAL INTERESTS

    The authors declare no competing financial interests.

    Reprints and permissions information is available online at http://www.nature.com/ reprints/index.html.

    1. Giagtzoglou, N., Ly, C.V. & Bellen, H.J. Cell adhesion, the backbone of the synapse:

    “vertebrate” and “invertebrate” perspectives. Cold Spring Harb. Perspect. Biol. 1,

    a003079 (2009).

    2. Shapiro, L., Love, J. & Colman, D.R. Adhesion molecules in the nervous system: structural

    insights into function and diversity. Annu. Rev. Neurosci. 30, 451–474 (2007).

    3. Dalva, M.B., McClelland, A.C. & Kayser, M.S. Cell adhesion molecules: signaling

    functions at the synapse. Nat. Rev. Neurosci. 8, 206–220 (2007).

    4. Parrish, J.Z., Emoto, K., Kim, M.D. & Jan, Y.N. Mechanisms that regulate

    establishment, maintenance and remodeling of dendritic fields. Annu. Rev. Neurosci. 

    30, 399–423 (2007).

    5. Südhof, T.C. Neuroligins and neurexins link synaptic function to cognitive disease.

    Nature  455, 903–911 (2008).

    6. Lin, Y.C. & Koleske, A.J. Mechanisms of synapse and dendrite maintenance and

    their disruption in psychiatric and neurodegenerative disorders. Annu. Rev. Neurosci. 

    33, 349–378 (2010).

    7. Sandi, C. Stress, cognitive impairment and cell adhesion molecules. Nat. Rev.

    Neurosci. 5, 917–930 (2004).8. Lupien, S.J., McEwen, B.S., Gunnar, M.R. & Heim, C. Effects of stress throughout

    the lifespan on the brain, behavior and cognition. Nat. Rev. Neurosci. 10, 434–445

    (2009).

    9. Rice, C.J., Sandman, C.A., Lenjavi, M.R. & Baram, T.Z. A novel mouse model for acute

    and long-lasting consequences of early life stress. Endocrinology   149, 4892–4900

    (2008).

    10. Brunson, K.L. et al.  Mechanisms of late-onset cognitive decline after early-life

    stress. J. Neurosci. 25, 9328–9338 (2005).

    11. Wang, X.D. et al. Forebrain CRF1 modulates early-life stress–programmed cognitive

    deficits. J. Neurosci. 31, 13625–13634 (2011).

    12. Ivy, A.S. et al. Hippocampal dysfunction and cognitive impairments provoked by

    chronic early-life stress involve excessive activation of CRH receptors. J. Neurosci. 

    30, 13005–13015 (2010).

    13. Wang, X.D. et al.  Forebrain CRHR1 deficiency attenuates chronic stress–induced

    cognitive deficits and dendritic remodeling. Neurobiol. Dis. 42, 300–310 (2011).

    14. Mizoguchi, A. et al. Nectin: an adhesion molecule involved in formation of synapses.

    J. Cell Biol. 156, 555–565 (2002).

    15. Majima, T. et al. Involvement of afadin in the formation and remodeling of synapsesin the hippocampus. Biochem. Biophys. Res. Commun. 385, 539–544 (2009).

    16. Honda, T. et al.  Involvement of nectins in the formation of puncta adherentia

    junctions and the mossy fiber trajectory in the mouse hippocampus. Mol. Cell

    Neurosci. 31, 315–325 (2006).

    17. Togashi, H. et al.  Interneurite affinity is regulated by heterophilic nectin

    interactions in concert with the cadherin machinery. J. Cell Biol. 174, 141–151

    (2006).

    18. Lim, S.T., Lim, K.C., Giuliano, R.E. & Federoff, H.J. Temporal and spatial localization

    of nectin-1 and l-afadin during synaptogenesis in hippocampal neurons. J. Comp.

    Neurol. 507, 1228–1244 (2008).

    19. Beaudoin, G.M. III et al. Afadin, a Ras/Rap effector that controls cadherin function,

    promotes spine and excitatory synapse density in the hippocampus. J. Neurosci. 

    32, 99–110 (2012).

    20. Suzuki, K. et al.  Mutations of PVRL1, encoding a cell-cell adhesion molecule/ 

    herpesvirus receptor, in cleft lip/palate-ectodermal dysplasia. Nat. Genet.  25,427–430 (2000).

    21. Thompson, C.L. et al.  Genomic anatomy of the hippocampus. Neuron   60,

    1010–1021 (2008).

    22. Chen, Y., Dubé, C.M., Rice, C.J. & Baram, T.Z. Rapid loss of dendritic spines after

    stress involves derangement of spine dynamics by corticotropin-releasing hormone.

    J. Neurosci. 28, 2903–2911 (2008).

    23. Conrad, C.D. What is the functional significance of chronic stress–induced CA3

    dendritic retraction within the hippocampus? Behav. Cogn. Neurosci. Rev. 5, 41–60

    (2006).

    24. Müller, M.B. et al.  Limbic corticotropin-releasing hormone receptor 1 mediates

    anxiety-related behavior and hormonal adaptation to stress. Nat. Neurosci.  6,

    1100–1107 (2003).

    25. Lu, A. et al. Conditional mouse mutants highlight mechanisms of corticotropin-

    releasing hormone effects on stress-coping behavior. Mol. Psychiatry   13,

    1028–1042 (2008).

    26. Refojo, D. et al. Glutamatergic and dopaminergic neurons mediate anxiogenic and

    anxiolytic effects of CRHR1. Science  333, 1903–1907 (2011).

    27. Kühne, C. et al.  Visualizing corticotropin-releasing hormone receptor type 1

    expression and neuronal connectivities in the mouse using a novel multifunctionalallele. J. Comp. Neurol. 520, 3150–3180 (2012).

    28. Srivastava, D.P. et al. Afadin is required for maintenance of dendritic structure and

    excitatory tone. J. Biol. Chem. 287, 35964–35974 (2012).

    29. Xie, Z. et al. Coordination of synaptic adhesion with dendritic spine remodeling by

    AF-6 and kalirin-7. J. Neurosci. 28, 6079–6091 (2008).

    30. Chen, Y. et al. Impairment of synaptic plasticity by the stress mediator CRH involves

    selective destruction of thin dendritic spines via RhoA signaling. Mol. Psychiatry  

    18, 485–496 (2013).

    31. Maras, P.M. & Baram, T.Z. Sculpting the hippocampus from within: stress, spines

    and CRH. Trends Neurosci. 35, 315–324 (2012).

    32. Chen, Y., Andres, A.L., Frotscher, M. & Baram, T.Z. Tuning synaptic transmission

    in the hippocampus by stress: the CRH system. Front. Cell Neurosci.  6, 13

    (2012).

    33. Refojo, D. et al. Corticotropin-releasing hormone activates ERK1/2 MAPK in specific

    brain areas. Proc. Natl. Acad. Sci. USA 102, 6183–6188 (2005).

    34. Schrick, C. et al.  N-cadherin regulates cytoskeletally associated IQGAP1/ERK

    signaling and memory formation. Neuron  55, 786–798 (2007).

    35. Tan, Z.J., Peng, Y., Song, H.L., Zheng, J.J. & Yu, X. N-cadherin–dependent neuron-neuron interaction is required for the maintenance of activity-induced dendrite

    growth. Proc. Natl. Acad. Sci. USA 107, 9873–9878 (2010).

    36. Arikkath, J. & Reichardt, L.F. Cadherins and catenins at synapses: roles in

    synaptogenesis and synaptic plasticity. Trends Neurosci. 31, 487–494 (2008).

    http://www.nature.com/reprints/index.htmlhttp://www.nature.com/reprints/index.htmlhttp://www.nature.com/reprints/index.htmlhttp://www.nature.com/reprints/index.html

  • 8/17/2019 Nectin-3 y Stress

    9/10

    NATURE NEUROSCIENCE doi:10.1038/nn.3395

    ONLINE METHODSAnimals. Male Crhr1loxP/loxP  ;Camk2a-cre and R26  flopCrh/flopCrh ;Camk2a-cre mice

    were generated as described previously 24,25 and kept on a mixed 129S2/Sv ×

    C57BL/6J background. Female CRHR1-EGFP reporter mice26, female CRHR1

    tau-lacZ  reporter mice27, male CD1 and C57BL/6N mice (Charles River), and

    Thy1-YFPH mice (Jackson Laboratory) were used. All mice were housed under a

    12:12-h light/dark cycle (lights on at 7 a.m.) and constant temperature (22± 1 °C)

    conditions with ad libitum access to both food and water. The protocols were

    approved by the Committee for the Care and Use of Laboratory Animals of theGovernment of Upper Bavaria, Germany.

    Experiments. To examine the effects of early-life stress and postnatal CRH over-

    expression on nectin-3 expression, we killed control or stressed CRHR1-CKO

    mice, stress-naive CRH-COE mice and respective wild-type mice at 7–8 months

    of age. To study the effects of acute stress on nectin-3 expression, we killed male

    C57BL/6N mice (3 months old) at 1, 4, 8 or 24 h after a single social defeat

    stress. To assess the potential involvement of glucocorticoid receptor, we killed

    male CD1 mice (3 months old) at 1, 4, 8 or 24 h after a subcutaneous injection

    of a synthetic glucocorticoid receptor agonist dexamethasone (10 mg per kg,

    Ratiopharm) or 0.9% saline (wt/vol).

    To study the effects of CRH on nectin-3 expression in vitro, we incubated

    acute brain slices from male C57BL/6N mice (3 months old) with either arti-

    ficial cerebrospinal fluid (ACSF, containing 124 mM NaCl, 3 mM KCl, 26 mM

    NaHCO3, 2 mM CaCl2, 1 mM MgSO4, 10 mM -glucose and 1.25 mMNaH2PO4, pH 7.3) or 50 nM CRH (Bachem) in ACSF

    37 in the holding cham-

    bers. At 1 or 4 h after treatment, slices were collected. To address the role of

    CRHR1 in these processes, we pretreated slices with 0.01% dimethyl sulfoxide

    (DMSO, vol/vol) in ACSF (vehicle) or 100 nM (ref. 38) of a CRHR1 antagonist

    DMP696 (Bicoll GmbH) in vehicle for 10 min, and then incubated with or

    without 50 nM CRH for 4 h. To further examine the effects of CRH-CRHR1

    signaling and corticosterone on nectin-3 expression in vivo, we injected three

    cohorts of male C57BL/6N mice (3 months old) icv with either (1 µl per mouse)

    ACSF or CRH (0.2 mM in ACSF); vehicle (1% DMSO in ACSF), CRH (0.2 mM

    in vehicle), CRH (0.2 mM) and DMP696 (1 mM) in vehicle, CRH (0.2 mM) and

    Rp-cAMPS (4 mM) in vehicle, or CRH (0.2 mM) and U0126 (2.6 mM) in vehi-

    cle; vehicle (5% ethanol in ACSF, vol/vol) or corticosterone (2.9 mM in vehicle).

    Mice were killed and hippocampi were collected 4 h after the injection.

    For the colocalization studies of CRHR1 and nectin-3, female CRHR1-EGFP

    or CRHR1 tau-lacZ   reporter mice (3 months old) were anesthetized andtranscardially perfused with heparinized 0.9% saline followed by buffered 4%

    paraformaldehyde (wt/vol). Brains were processed for immunostaining.

    To study the effects of hippocampal nectin-3 knockdown, we injected two

    cohorts of male C57BL/6N mice (3 months old) with either the control or

    knockdown virus and tested them in the Y-maze and spatial object recognition

    tasks (the first cohort only) and the Morris water maze task (both cohorts) after

    4 weeks of recovery. At 1 week after behavioral testing, mice were killed. Another

    cohort (3 months old) was used to examine the roles of nectin-3 in dendritic

    spine plasticity. To study the effects of early-life stress and nectin-3 overexpres-

    sion, we microinjected male C57BL/6N or Thy1-YFPH mice (5 months old),

    with or without early-life stress, intrahippocampally with either the control or

    nectin-3–overexpressing virus. After 4 weeks of recovery, C57BL/6N mice were

    subjected to cognitive testing and killed 1 week later, whereas Thy1-YFPH mice

    were transcardially perfused and brains processed for structural analysis.

    Stress procedures.  The limited nesting and bedding material procedure was

    carried out as described previously 9,39. The day of birth was designated postna-

    tal day 0 (P0). On the morning of P2, control dams were provided with a suf-

    ficient amount of nesting material (two squares (4.8 g) of Nestlets, Indulab) and

    500 ml of standard sawdust bedding. In the ‘stress’ cages, dams were provided

    with a limited quantity of nesting material (one half of a square (1.2 g) of Nestlets),

    which was placed on a fine-gauge aluminum mesh platform (McNichols). All

    litters remained undisturbed from P2 to P9. On P9, all dams were provided with

    standard nesting and bedding material. Male offspring were weaned on P28, and

    tail tips were collected and genotyped.

    The acute social defeat stress procedure was performed by introducing each

    male C57BL/6N mouse into the home cage of an aggressive CD1 resident mouse

    with short attack latency for 5 min (ref. 40). Control mice were allowed to explore

    an empty novel cage similar to the resident cage for 5 min.

    Acute brain slice preparation. Serial coronal brain slices (350 µm thick) were

    prepared through the hippocampus using a vibrating microtome in ice-cold ACSF

    bubbled with a 95% O2/5% CO2 mixture. Slices were rapidly cut into halves and

    transferred to holding chambers. At 1 or 4 h after treatment, slices were collected,

    snap-frozen and stored at −20 °C. Hippocampal slices were later dissected on ice,

    pooled for each mouse (2–3 slices per treatment and time point), and homoge-nated for western blot analysis.

    Cannulation and intracerebroventricular injection. Stereotaxic surgery was

    performed as previously described41. A 23-gauge stainless steel cannula was

    placed in the right lateral ventricle (0.4 mm posterior to bregma, 1.0 mm lateral

    from midline, 1.6 mm dorsoventral from dura)42. Mice were allowed to recover

    for 1 week. On the day of the experiment, mice were anesthetized in the home

    cage by 1 ml of isoflurane, and the anesthesia was maintained using isoflurane-O2 

    (1~1.5:100) inhalation. Drugs (1 µl) were delivered via a Hamilton syringe over a

    1-min period followed by 1 min of rest. At 4 h after the injection, mice were killed,

    the cannula position was examined and hippocampi were dissected.

    Viral-mediated gene manipulation and intrahippocampal microinjection.  To

    suppress or enhance nectin-3 expression in the hippocampus, we used an adeno-

    associated bicistronic AAV1/2 vector (GeneDetect). AAV-shSCR (AAV1/2-U6-scrambled shRNA-terminator-CAG-EGFP-WPRE-BGH-polyA), AAV-shNEC

    (AAV1/2-U6-Nectin-3 shRNA-terminator-CAG-EGFP-WPRE-BGH-polyA),

    AAV-null (AAV1/2-CAG-null-WPRE-BGH-polyA), and AAV-OE (AAV1/2-CAG-

    Nectin-3-WPRE-BGH-polyA) were generated and purified by GeneDetect.

    Stereotaxic surgery and intrahippocampal microinjection was performed

    as previously described41. Briefly, 0.5 or 1 µl of the virus (1.2~11 × 1012 viral

    genomes per ml) was injected bilaterally, aiming at the stratum radiatum of the

    dorsal CA3 (1.9 mm posterior to bregma, 2.1 mm lateral from midline, 1.8 mm

    dorsoventral from dura)42. The virus was delivered over a 15-min period followed

    by 5 min of rest. Mice were given a 4-week period before experimentation to allow

    sufficient viral infection in the hippocampus.

    Behavioral testing. The tests were performed between 8 a.m. and 1 p.m. and

    scored by the ANY-maze 4.50 software (Stoelting). Short-term spatial work-

    ing memory was tested by recording spontaneous alternation behavior in theY-maze43. The apparatus was made of gray polyvinyl chloride with three sym-

    metrical arms (30 × 10 × 15 cm3) and evenly illuminated (30 lx). Prominent intra-

    and extra-maze spatial cues were provided. Mice were placed in the center of the

    maze and allowed to explore the arms freely for 5 min. Three consecutive choices

    of all three arms were counted as an alternation. The percentage of spontaneous

    alternation was determined by dividing the total number of alternations by the

    total number of choices minus 2.

    The spatial object recognition task was performed in an open field apparatus

    (50 × 50 × 50 cm3) under low illumination (30 lx)41. Prominent spatial cues were

    provided. Mice were habituated to the testing environment for 10 min on 2 con-

    secutive days before testing. During the acquisition trials, mice were presented

    with two identical aluminum cubes (5 × 5 × 5 cm3) and allowed to explore the

    objects twice for 10 min, separated by a 15-min intertrial interval (ITI). During

    the 5-min retrieval trial, 30 min following the last acquisition trial, mice were

    presented with a non-displaced object and a relocated one. The ratio of time spentwith the displaced (novel) object compared with the non-displaced (known)

    object and the percentages of time exploring the novel and known objects were

    calculated, with a higher preference for the novel object being rated as intact

    spatial recognition memory.

    The Morris water maze test was carried out in a circular tank (110 cm in diam-

    eter) filled with opaque colored water (22 ± 1 °C) and provided with prominent

    extra-maze visual cues11. After day 1 with a 60-s free swim trial, mice were trained

    to locate a visible platform (10 cm in diameter) above the surface of the water

    for four trials. In the following spatial training sessions, mice received four trials

    per day to locate the submerged platform in a fixed position over 3 consecutive

    days. On the next day, reference memory was assessed in a 60-s probe trial with

    platform removed, and the time spent in each quadrant was recorded. The trials in

  • 8/17/2019 Nectin-3 y Stress

    10/10

    NATURE NEUROSCIENCEdoi:10.1038/nn.3395

    spatial training sessions (ITI = 10 min) were terminated once the mouse found the

    platform or 60 s had elapsed, and the latency to reach the platform was recorded

    for each trial. Mice that did not employ a search strategy and floated in the tank

    in all trials were excluded from analysis.

    In situ hybridization. Coronal brain sections (20 µm thick) were prepared

    and in situ  hybridization performed as previously described13. The following

    primers were used to generate an antisense RNA hybridization probe (485 base

    pairs) that recognizes a shared sequence of alpha-, beta- and gamma-transcript variants of nectin-3: AGCCGTTACATTCCCACTTG (forward primer) and

    ATTGTCCATCCAACCTGCTC (reverse primer). The slides were apposed to

    Kodak Biomax MR films (Eastman Kodak) and developed. Autoradiographs

    were digitized, and relative expression (average optical density of the region of

    interest–average optical density of the background) was determined by Scion

    Image (Scion).

    Primary antibodies. For western blot, we used antibodies to nectin-3 (ab63931,

    1:2,000), nectin-2 (ab135246, 1:1,000), nectin-4 (ab110387, 1:1,000), N-cadherin

    (ab12221, 1:2,500),β-catenin (ab22656, 1:2,000) and L-afadin (ab11337, 1:1,000)

    from Abcam; to actin (sc-1616, 1:2,000), nectin-1 (sc-28639, 1:1,000) and PKR

    (sc-1702, 1:1,000) from Santa Cruz Biotechnology. For immunostaining, we used

    antibodies to EGFP (ab5450, 1:2,000),β-galactosidase (ab9361, 1:2,000), nectin-3

    (ab63931, 1:500) and L-afadin (ab11337, 1:500) from Abcam.

    Western blot. Total hippocampal protein extracts were prepared and western blot

    was performed as previously described13. Membrane fractions were extracted

    using the Calbiochem ProteoExtract kit (EMD Millipore)44. Samples were

    resolved by 10% sodium dodecyl sulfate–polyacrylamide gels, and transferred

    onto nitrocellulose membranes (Invitrogen). Membranes were labeled with

    primary antibodies overnight at 4 °C. Following incubation with horseradish

    peroxidase–conjugated secondary antibodies (1:2,000, DAKO) for 3 h, bands

    were visualized using an enhanced chemiluminescence system (Amersham

    Biosciences) and quantified by densitometry (Quantity One 4.2, Bio-Rad).

    Immunohistochemistry and image analysis. Double-labeling immunofluo-

    rescence was performed on free-floating coronal (20 µm thick) or transverse

    (30 µm thick) sections11. After incubation with primary antibodies overnight

    at 4 °C, sections were rinsed and labeled with Alexa Fluor 488– and Alexa

    Fluor 647–conjugated secondary antibodies (1:500, Invitrogen) for 3 h at22 ± 1 °C. After rinsing, sections were transferred onto slides and coverslipped

    with Vectashield mounting medium containing 4′,6-diamidino-2-phenylindole

    (Vector Laboratories).

    All images (1,600 × 1,600 pixels) were obtained with an Olympus IX81-FV1000

    laser-scanning confocal microscope (Olympus). A 10× objective (NA 0.40), a

    20× objective (NA 0.75) and a 60× water-immersion objective (NA 1.20) were

    used. Images were imported into the US National Institutes of Health ImageJ

    software, converted to 8-bit grayscale and thresholded uniformly. The density

    of L-afadin–immunoreactive puncta was measured (5–6 sections per mouse).

    Representative images were adjusted for better brightness and contrast using the

    FV10-ASW 2.0 software (Olympus).

    Quantitative morphological analysis of dendritic spines. Transverse sections

    were collected for analyzing the pyramidal neurons of CA3 and CA1, and coro-

    nal sections for dentate granule cells. In area CA3, as the stratum radiatum wasdensely packed with EGFP/EYFP-labeled commissural/associational fibers, only

    the dendritic segments (20–130 µm in length, 8–15 dendrites per mouse) in

    the stratum lacunosum-moleculare were analyzed. Dendrites were scanned at

    0.33-µm intervals along the z  axis using the 60× objective with a 2.5× digital

    zoom, yielding a voxel size of 0.053 × 0.053 × 0.33 µm3. The z   stack images

    were deconvolved using Huygens 4.2 software (Scientific Volume Imaging)45.

    Spine density (expressed as the number of spines per 10 µm of dendrite), spine

     volume and spine head diameter were analyzed with NeuronStudio software

    (http://research.mssm.edu/cnic/tools-ns.html )45,46. To quantify the density of

    nectin-3–positive spines, deconvolved dual-channel z  stacks (EGFP/EYFP andnectin-3) were merged and nectin-3–positive spines were counted manually

    using ImageJ.

    In AAV-shSCR and AAV-shNEC mice, the background in dentate gyrus and

    CA1 was higher than in CA3. Thus, only spine density in the outer molecular

    layer of the suprapyramidal blade of dentate gyrus and the stratum lacunosum-

    moleculare of CA1 was quantified. Dendrites (80–100 µm in length) were

    scanned at 1-µm intervals along the z   axis using the 60× objective with a

    2.5× digital zoom. For each mouse, eight dendrites from different CA1 pyramidal

    neurons or six dendrites from different dentate granule cells were selected. Spines

    were counted manually using ImageJ, and spine density was expressed as the

    number of spines per 10 µm of dendrite.

    Statistical analysis. SPSS 16 (SPSS), GraphPad Prism 5 (GraphPad Software),

    and R version 2.15 (http://www.r-project.org/) were used. Normally distributed

    data were analyzed by ANOVA followed by Bonferroni or Fisher’s LSD post hoc tests as necessary. Student’s t  test was used to compare pairs of means. Data that

    were not normally distributed were rank- and/or Box-Cox transformed to achieve

    a normal data distribution. After data transformation, data were analyzed by

    ANOVA followed by Tukey post hoc test when necessary, and Welch’s t  test was

    used to compare pairs of means. The level of statistical significance was set at

    P  < 0.05. Data are expressed as mean ± s.e.m.

    37. von Wolff, G. et al. Voltage-sensitive dye imaging demonstrates an enhancing effect

    of corticotropin-releasing hormone on neuronal activity propagation through the

    hippocampal formation. J. Psychiatr. Res. 45, 256–261 (2011).

    38. Li, Y.W. et al. Receptor occupancy of nonpeptide corticotropin-releasing factor 1

    antagonist DMP696: correlation with drug exposure and anxiolytic efficacy.

    J. Pharmacol. Exp. Ther. 305, 86–96 (2003).

    39. Wang, X.D. et al. Early-life stress-induced anxiety-related behavior in adult mice

    partially requires forebrain corticotropin-releasing hormone receptor 1. Eur. J.Neurosci. 36, 2360–2367 (2012).

    40. Wagner, K.V. et al. Homer1 mediates acute stress-induced cognitive deficits in the

    dorsal hippocampus. J. Neurosci. 33, 3857–3864 (2013).

    41. Schmidt, M.V. et al. Tumor suppressor down-regulated in renal cell carcinoma 1

    (DRR1) is a stress-induced actin bundling factor that modulates synaptic efficacy

    and cognition. Proc. Natl. Acad. Sci. USA 108, 17213–17218 (2011).

    42. Paxinos, G. & Watson, C. The Mouse Brain in Stereotaxic Coordinates  (Academic

    Press, San Diego, CA, USA, 2001).

    43. Schmidt, M.V. et al. Individual stress vulnerability is predicted by short-term memory

    and AMPA receptor subunit ratio in the hippocampus. J. Neurosci. 30, 16949–16958

    (2010).

    44. Wagner, K.V. et al. Differences in FKBP51 regulation following chronic social defeat

    stress correlate with individual stress sensitivity: influence of paroxetine treatment.

    Neuropsychopharmacology  37, 2797–2808 (2012).

    45. Heck, N., Betuing, S., Vanhoutte, P. & Caboche, J. A deconvolution method to

    improve automated 3D-analysis of dendritic spines: application to a mouse model

    of Huntington’s disease. Brain Struct. Funct. 217, 421–434 (2012).

    46. Dumitriu, D., Rodriguez, A. & Morrison, J.H. High-throughput, detailed, cell-specific

    neuroanatomy of dendritic spines using microinjection and confocal microscopy.

    Nat. Protoc. 6, 1391–1411 (2011).

    http://research.mssm.edu/cnic/tools-ns.htmlhttp://www.r-project.org/http://www.r-project.org/http://research.mssm.edu/cnic/tools-ns.html