DDR 400

You might also like

You are on page 1of 18

Human Molecular Genetics, 2011, Vol. 20, No.

23 4666–4683
doi:10.1093/hmg/ddr400
Advance Access published on September 8, 2011

Behavioral alterations associated with targeted


disruption of exons 2 and 3 of the Disc1 gene
in the mouse
Keisuke Kuroda 1,5,{, Shinnosuke Yamada 2,5,{, Motoki Tanaka 3,{, Michiro Iizuka 1,6, Hisashi Yano 1,
Daisuke Mori 1,5, Daisuke Tsuboi 1,5, Tomoki Nishioka 1,5, Takashi Namba 1,5, Yukihiko Iizuka 1,5,
Shimpei Kubota 1, Taku Nagai 2,5, Daisuke Ibi 2,5, Rui Wang 2,5, Atsushi Enomoto 4,

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Mayu Isotani-Sakakibara 4, Naoya Asai 4, Kazushi Kimura 6, Hiroshi Kiyonari 7, Takaya Abe 7,
Akira Mizoguchi 6, Masahiro Sokabe 3,8, Masahide Takahashi 4, Kiyofumi Yamada 2,5
and Kozo Kaibuchi 1,5,∗
1
Department of Cell Pharmacology, 2Department of Neuropsychopharmacology and Hospital Pharmacy, 3Department
of Physiology and 4Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya,
Aichi 466-8550, Japan, 5JST, CREST, Kawaguchi, Saitama 322-0012, Japan, 6Department of Neural Regeneration
and Cell Communication, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan, 7Laboratory for
Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047,
Japan and 8FIRST Research Center for Innovative Nanobiodevice, Nagoya University, Nagoya, Aichi 466-8550,
Japan

Received June 27, 2011; Revised and Accepted August 31, 2011

Disrupted-In-Schizophrenia 1 (DISC1) is a promising candidate gene for susceptibility to psychiatric


disorders, including schizophrenia. DISC1 appears to be involved in neurogenesis, neuronal migration,
axon/dendrite formation and synapse formation; during these processes, DISC1 acts as a scaffold protein
by interacting with various partners. However, the lack of Disc1 knockout mice and a well-characterized
antibody to DISC1 has made it difficult to determine the exact role of DISC1 in vivo. In this study, we gen-
erated mice lacking exons 2 and 3 of the Disc1 gene and prepared specific antibodies to the N- and C-ter-
mini of DISC1. The Disc1 mutant mice are viable and fertile, and no gross phenotypes, such as
disorganization of the brain’s cytoarchitecture, were observed. Western blot analysis revealed that the
DISC1-specific antibodies recognize a protein with an apparent molecular mass of ∼100 kDa in brain
extracts from wild-type mice but not in brain extracts from DISC1 mutant mice. Immunochemical studies
demonstrated that DISC1 is mainly localized to the vicinity of the Golgi apparatus in hippocampal neurons
and astrocytes. A deficiency of full-length Disc1 induced a threshold shift in the induction of long-term
potentiation in the dentate gyrus. The Disc1 mutant mice displayed abnormal emotional behavior as
assessed by the elevated plus-maze and cliff-avoidance tests, thereby suggesting that a deficiency of
full-length DISC1 may result in lower anxiety and/or higher impulsivity. Based on these results, we suggest
that full-length Disc1-deficient mice and DISC1-specific antibodies are powerful tools for dissecting the
pathophysiological functions of DISC1.


To whom correspondence should be addressed at: Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai,
Showa, Nagoya, Aichi 466-8550, Japan. Tel: +81 527442075; Fax: +81 527442083; Email: kaibuchi@med.nagoya-u.ac.jp

These three authors contributed equally to this work.

# The Author 2011. Published by Oxford University Press. All rights reserved.
For Permissions, please email: journals.permissions@oup.com
Human Molecular Genetics, 2011, Vol. 20, No. 23 4667

INTRODUCTION hippocampal neurons and astrocytes. Gross anatomical abnor-


malities were not evident in the Disc1 (D2-3) mice, but elec-
Although the etiological mechanisms of psychiatric disorders, trophysiological analyses revealed that the threshold for the
such as schizophrenia, remain largely esoteric, multiple induction of long-term potentiation (LTP) was significantly
hypotheses (e.g. dysfunction of dopamine, glutamate or sero- higher than in normal mice. Additionally, behavioral tests
tonin, neurodevelopmental disorders, stress during pregnancy revealed a phenotype characterized by gender-independent
and viral infection) have been proposed (1,2). The dissection of higher impulsivity as well as gender-dependent deficits of pre-
molecular pathways centered on promising risk factors, such as pulse inhibition (PPI) and increased responsiveness to meth-
Disrupted-In-Schizophrenia 1 (DISC1), may help us achieve a amphetamine (METH).
better understanding of the pathogenesis of these disorders.
The DISC1 gene locus was originally identified at the breakpoint
of a balanced chromosomal translocation t(1;11)(q42.1;q14.3) RESULTS
that co-segregated with schizophrenia, bipolar disorder and re-

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


current major depression in a large Scottish family (3,4). Generation of mice lacking exons 2 and 3 of Disc1
Further analysis has revealed that the inheritance of the transloca- Although many splice variants of Disc1 at the protein and mRNA
tion is causal and increases the risk of these psychiatric disorders levels have been reported (3,15,30–32), the major form of DISC1
50-fold (5). Genetic association studies of several ethnic groups appears to have a molecular mass of 100 kDa, which corre-
indicate that DISC1 is a major risk factor for psychiatric disor- sponds to the size of the mouse full-length Disc1 mRNA.
ders, including schizophrenia and mood disorders (6). In add- Because each of the putative isoforms contain exons 2 and 3
ition, clinical studies have shown that genetic variations in (30), exons 2 and 3 of Disc1 were replaced with a loxP/
DISC1 influence brain function and anatomy (7). PGK-Neo-pA/loxP cassette, and an in-frame stop codon was
Several groups have isolated DISC1-interacting proteins. inserted between the remaining exon 2 and the loxP site in
These proteins include nuclear distribution gene E homolog-like mouse TT2 ES cells from an F1 embryo between a C57BL/6
1 (NDEL1), lissencephaly-1 (LIS1), fasciculation and elong- and CBA cross and HK3i ES cells from a C57BL/6N embryo
ation protein zeta 1 (FEZ1), phosphodiesterase 4B (PDE4B), (Fig. 1A and B; http://www.cdb.riken.jp/arg/ES_cells.html).
kinesin family member 5B (KIF5B), growth factor receptor- Many proteins, including PDE4B, KIF5B, GSK3b, girdin and
bound protein 2 (GRB2), glycogen synthase kinase 3b TNIK, associate with DISC1 through the region encoded by
(GSK3b), girdin, kalirin, TRAF2 and NCK-interacting kinase exons 2 and 3 (8,10,12,13,17). Thus, the function of DISC1 as a
(TNIK) and DIX domain-containing 1 (DIXDC1) (8 – 19). scaffold protein that interacts with these proteins is lost in mice
These studies suggest that DISC1 is involved in neurogenesis, lacking exons 2 and 3 of Disc1. Homologous recombination
neuronal migration, axon/dendrite formation and synapse was confirmed by southern blot analysis (Fig. 1C) and genomic
formation through interactions with the abovementioned pro- polymerase chain reaction (PCR) (Fig. 1D). Mice heterozygous
teins (20). DISC1 is thus considered to be a scaffold protein for the Disc1 mutation (hereafter termed Disc1 +/D2-3 mice)
that regulates specific molecular pathways (21). were backcrossed onto the C57BL/6JJmsSlc background for at
To aid our understanding of the physiological function of least five generations to generate Disc1 +/D2-3 mice with a pre-
DISC1, mouse models with various alterations in DISC1 dominantly C57BL/6J genetic background (100/100 marker at
have been developed. These models include mice with mis- N5). The Disc1 +/D2-3 mice appeared normal and were healthy
sense mutations (22), overexpression of truncated forms of and fertile. Mice homozygous for the Disc1 mutation (hereafter
DISC1 (23 – 26) and mice that have a 25 bp deletion in exon termed Disc1 D2-3/D2-3 mice) were produced at the expected Men-
6 of the Disc1 gene [hereafter termed Disc1 (D6) mice] delian ratio by intercrossing Disc1 +/D2-3 mice. The Disc1 D2-3/D2-3
(27,28). Anatomical and behavioral analyses of these mice mice were also viable and fertile and showed no obvious differ-
have revealed abnormal phenotypes, such as schizophrenia ences in physical characteristics from the Disc1 +/D2-3 mice.
and/or depression-related phenotypes (21). However, the Reverse transcription (RT)–PCR analysis revealed that the
pathophysiological and behavioral functions of DISC1 are mRNA derived from exons 2 and 3 of Disc1 were absent in the
controversial, and possible unknown splice variants of Disc1 Disc1 D2-3/D2-3 mice, whereas exons 7–10 of Disc1 were detected
hinder efforts to resolve this problem (29). To further under- in both wild-type (hereafter termed Disc1 +/+ ) and Disc1 D2-3/D2-3
stand the physiological role of DISC1 and its relationship to mice (Fig. 1B and E). Because both commercial and previously
psychiatric disorders, Disc1 knockout mice and a well- generated antibodies to DISC1 showed high background levels
characterized antibody to DISC1 are required. when used for western blot analysis, two different polyclonal anti-
In this study, we generated mice lacking exons 2 and 3 of bodies were developed (Fig. 1B). The N-terminal antibody
the Disc1 gene by homologous recombination in embryonic detected two major protein bands with molecular masses of
stem (ES) cells. These mice are hereafter to be termed Disc1 100 and 45 kDa as assessed by western blot analysis (Fig. 1F
(D2-3) mice. We also made new antibodies against DISC1 and Supplementary Material, Fig. S1A). The 100 kDa band was
for use in evaluating the Disc1 (D2-3) mice and analyzing absent in the Disc1 D2-3/D2-3 mouse brain lysate but not in the
DISC1 function. Western blot analysis revealed that the Disc1 +/+ mouse brain lysate. In contrast, the 45 kDa band was
DISC1 antibodies recognize a polypeptide with a molecular present in lysates from both Disc1 +/+ and Disc1 D2-3/D2-3 mice.
mass of 100 kDa in wild-type mice but not in the Disc1 This result indicated that the 45 kDa band may represent a non-
(D2-3) mice. Immunochemical analysis revealed that these specific signal. The C-terminal antibody detected a protein
DISC1 antibodies recognized endogenous DISC1 that was band with a molecular mass of 100 kDa. Similarly, the
mainly localized to the vicinity of the Golgi apparatus in 100 kDa band was absent in the Disc1 D2-3/D2-3 mouse brain
4668 Human Molecular Genetics, 2011, Vol. 20, No. 23

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Figure 1. Targeted disruption of Disc1. (A) Summary of the strategy to target the Disc1 locus. The Neo cassette was inserted by homologous recombination,
replacing the region spanning exons 2– 3 of Disc1. Arrows indicate primers used for PCR-based genotyping; asterisk indicates stop codon; the probe used for
southern blot analysis is shown as a bar. (B) Schematic representation of the mouse Disc1 transcript. The gray portion indicates a deleted region. Regions targeted
by RT –PCR and anti-DISC1 antibodies are shown as bars. (C) Southern blot analysis of tail genomic DNA from wild-type (Disc1 +/+ ; +/+), heterozygous
mutant (Disc1 +/D2-3; +/D) and homozygous mutant (Disc1 D2-3/D2-3; D/D) mice. Genomic DNA was digested with ApaI and subjected to hybridization with
the probe. DNA fragments of 14.5 and 24 kbp correspond to the wild-type and the Disc1 (D2-3) mutant alleles, respectively. (D) PCR genotyping of the
Disc1 (D2-3) mice. PCR products of 547 and 619 bp correspond to the wild-type and the Disc1 (D2-3) mutant alleles, respectively. (E) RT– PCR analysis
of the Disc1 (D2-3) mice. Total RNA prepared from P7 brain tissue was reverse transcribed and used for PCR amplification with primers specific for exons
2– 3 and exons 7 –10 of Disc1 and a housekeeping gene (Hprt). An amplification product for exons 2– 3 was not observed in the Disc1 D2-3/D2-3 mice.
However, exons 7– 10 were detected in both Disc1 +/+ and Disc1 D2-3/D2-3 mice. (F and G) Western blot analysis of the Disc1 (D2-3) mice. P7 brain lysates
were blotted with antibodies specific for the DISC1 N-terminal (F) and C-terminal (G) regions. The arrowhead indicates the position of full-length DISC1.
A 45 kDa protein band observed with the N-terminal antibody may be a nonspecific signal.

lysate but present in the Disc1 +/+ mouse brain lysate (Fig. 1G and chromatography tandem mass spectrometry analysis. Taken to-
Supplementary Material, Fig. S1B). To further examine whether gether, these results indicate that the Disc1 D2-3/D2-3 mice do not
the antibody recognized DISC1, DISC1 was immunoprecipitated express DISC1 isoforms containing exons 2 and 3. However,
from mouse brain extracts using both the N-terminal and C- we cannot dismiss the possibility that truncated forms of DISC1
terminal antibodies and the peptides derived from exons 5 and lacking exons 2 and 3 were produced from unidentified start
6 of Disc1 were detected in the immunoprecipitate by the liquid sites in Disc1 mRNA.
Human Molecular Genetics, 2011, Vol. 20, No. 23 4669

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Figure 2. No apparent morphological defects during brain development in the Disc1 D2-3/D2-3 mice. (A) Gross overview of brain tissue from P56 Disc1 +/+ and
Disc1 D2-3/D2-3 mice showed no apparent decrease in brain volume in the Disc1 D2-3/D2-3 mice. (B and C) Nissl-stained brain sections of P56 Disc1 +/+ and
Disc1 D2-3/D2-3 mice showed no deficit in the development of the prefrontal cortex (B), the neocortex, the ganglia and the hippocampus (C) in the Disc1 D2-3/
D2-3
mice. The regions within the boxes are shown at a higher magnification in adjacent panels. CC, corpus callosum; AC, anterior commissure; PC, posterior
commissure; CP, caudate putamen; TH, thalamus; DG, dentate gyrus. Asterisks indicate the pia mater.

Morphological defects in brain development were Evaluation of commercially available anti-DISC1


not observed in the Disc1 D2-3/D2-3 mice antibodies

Despite the results of the western blot analysis, which Several DISC1 antibodies are commercially available. During
showed abundant expression of DISC1 in the brain during the course of generating Disc1 (D2-3) mice, we noticed that
early development (Supplementary Material, Fig. S3B and none of the commercial antibodies recognized the 100 kDa
C), Disc1 D2-3/D2-3 embryos appeared normal upon examin- protein band that was absent in the Disc1 D2-3/D2-3 mice (Sup-
ation (data not shown), indicating that DISC1 is probably plementary Material, Fig. S1C). Of note, one antibody strong-
dispensable for embryogenesis. The Disc1 D2-3/D2-3 mice ly reacted with a protein band with a molecular mass of
develop and reproduce normally with no apparent morpho- 100 kDa. However, this immunoreactivity was not lost in
logical effects in the examined organs, including the brain, brain extracts from the Disc1 D2-3/D2-3 mice, although the
through postnatal (P) day 56, which may reflect the ability antigen recognized by this antibody was included in exon
of other proteins to compensate for the absence of DISC1. 2. Because the expression level of endogenous DISC1 is
No appreciable abnormalities in the volume of the cerebrum, low, even in the brain (Supplementary Material, Fig. S3B
the cerebellum or the olfactory bulb were observed in the and C), it was difficult to detect endogenous DISC1 using
brains of the Disc1 D2-3/D2-3 mice in comparison to their the commercially available antibodies under our conditions.
Disc1 +/+ littermates during the postnatal period or in adult-
hood (Fig. 2A). An examination of brain structure using
Nissl staining revealed a nearly normal cytoarchitecture of Mouse strains defective in DISC1 expression
the neocortex, the basal ganglia and the hippocampus in Gogos and associates identified a 25 bp deletion in exon 6 of
the Disc1 D2-3/D2-3 mice at P56 (Fig. 2B and C). Interestingly, Disc1 in the 129 strains of mice [Disc1 (D6) mice] (27,35).
the Disc1 D2-3/D2-3 mice exhibited no reduction in the size of These researchers claimed that this mutation interfered with
the white matter tracts, such as the corpus callosum and an- the production of full-length DISC1 (27,28). We determined
terior and posterior commissures (Fig. 2C). In addition, the whether full-length DISC1 was expressed in brain extracts
volume of the prefrontal cortex (Fig. 2C), which is a struc- from 129X1/SvJJmsSlc mice using our C-terminal antibodies
ture that has been implicated as a locus of dysfunction in (these mice, which are homozygous for the deletion of the
schizophrenia (33,34), did not appear to be reduced. allele, are hereafter termed Disc1 D6/D6 mice). The 100 kDa
4670 Human Molecular Genetics, 2011, Vol. 20, No. 23

protein band was not detected in the extract from Disc1 D6/D6 vicinity of the Golgi apparatus at the base of the apical den-
mice using western blot analysis using the exact conditions drites (Fig. 3E – G).
under which our antibody recognized DISC1 in extracts from In cultured hippocampal neurons and astrocytes, DISC1 was
wild-type C57BL/6J mice (Disc1 +/+ ) (Supplementary Mater- mainly localized to the vicinity of the Golgi apparatus
ial, Fig. S2), thereby indicating that the 129 strains do not (Fig. 4A – D). In the immunoelectron microscopic analysis of
express full-length DISC1. Furthermore, we examined hippocampal neurons labeled with the C-terminal antibody,
whether other outbred mice, such as ddY and ICR, harbor the the DISC1 immunoreactivity was associated with the mem-
same deletion as the 129 strains. Genomic PCR showed that brane structures of the Golgi apparatus (Fig. 4E).
77.5% of the ICR mice examined had homozygous deletion
alleles (Disc1 D6/D6), 22.5% of the ICR mice had one
non-deletion allele and one deletion allele (Disc1 +/D6) and Electrophysiological analyses in the Disc1 D2-3/D2-3 mice
none of the ICR mice had homozygous wild-type alleles Because Disc1 mRNA is highly expressed in the dentate gyrus
(Disc1 +/+ ). The C-terminal antibody recognized the 100 kDa of the hippocampus (14,36– 39), the effects of the disruption

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


band in the Disc1 +/+ mice but not in the Disc1 D6/D6 mice of of exons 2 and 3 of the Disc1 gene were investigated on the
the ddY and ICR strains (Supplementary Material, Fig. S2). synaptic properties of neurons in the dentate gyrus in mouse
hippocampal slices using an extracellular recording technique.
First, the strength of basal synaptic transmission was com-
Spatiotemporal expression pattern of DISC1 in mice
pared at medial perforant path-dentate granule cell synapses
In situ hybridization and northern blot analysis have shown between Disc1 +/+ and Disc1 D2-3/D2-3 mice. Basal synaptic
that Disc1 mRNA is chiefly expressed in the mouse cerebrum, transmission was estimated by measuring the amplitude of a
cerebellum and heart during development (3,14,36– 39). To presynaptic fiber volley (PSFV) and the initial slope of the
obtain an overview of the spatiotemporal expression pattern field excitatory postsynaptic potentials (fEPSPs) over a range
of the DISC1 protein in mice, a western blot analysis was per- of stimulus intensities. As seen in Supplementary Material,
formed using the C-terminal antibody. Expression of DISC1 Figure S4A, the stimulus – PSFV relationships in both
was detected in various tissues of prenatal mice (Supplemen- Disc1 +/+ and Disc1 D2-3/D2-3 mice were almost linear, and
tary Material, Fig. S3A). DISC1 was highly expressed in the the PSFV amplitude in both groups was nearly identical at
cerebellum and the lung and was moderately expressed in every stimulus intensity that was tested [two-way analysis of
the cerebrum, the brainstem, the heart and the kidney. The de- variance (ANOVA), F(1,126) ¼ 0.59, P . 0.1, n ¼ 10). In
velopmental profile of DISC1 expression in the cerebrum and the stimulus-fEPSP relationships, the mean value of the
the cerebellum was further analyzed (Supplementary Material, fEPSP slope in the Disc1 D2-3/D2-3 mice was smaller than in
Fig. S3B and C). The expression of DISC1 peaked during the the Disc1 +/+ mice over the stimulus range, but the difference
postnatal period and gradually declined thereafter. The amount was not significant [two-way ANOVA, F(1,126) ¼ 1.46,
of DISC1 protein in the cerebrum and the cerebellum was P . 0.1, n ¼ 10, Supplementary Material, Fig. S4B]. The
roughly calculated by quantitative western blot analysis mean value of the fEPSP slope evoked by test stimuli
using the recombinant DISC1 protein as a standard. The (50% of the maximum response) was 20.51 + 0.14 mV/ms
amount of DISC1 in the whole cerebrum and the whole cere- in the Disc1 +/+ mice (n ¼ 10) and 20.48 + 0.12 mV/ms in
bellum was estimated as 0.0002% and 0.0003% of the total the Disc1 D2-3/D2-3 mice (n ¼ 10).
protein, respectively, suggesting that DISC1 is a The synaptic responses to weaker high-frequency stimula-
low-abundance protein. tion (HFS, 50 Hz/100 pulses), standard HFS (100 Hz/100
pulses) and stronger HFS (200 Hz/100 pulses) were subse-
quently examined in the Disc1 +/+ and Disc1 D2-3/D2-3 mice.
Immunochemical analysis of DISC1 in hippocampal In hippocampal slices from the Disc1 +/+ mice, weaker
neurons and astrocytes HFS resulted in an enhancement of the fEPSP slope that per-
To further examine whether our antibodies recognized en- sisted for over 60 min, which was indicative of LTP
dogenous DISC1 in vivo and in vitro, immunofluorescent (123.3 + 2.0% of baseline 60 min after weaker HFS, n ¼ 4,
staining was performed with the C-terminal antibody in P , 0.001, paired t-test, Fig. 5A and D). However, in hippo-
brain sections from P2 mice and in cultured hippocampal campal slices from the Disc1 D2-3/D2-3 mice, the same proto-
neurons. In brain sections, DISC1 immunoreactivity was loca- col did not induce LTP but instead produced a phenomenon
lized in the pyramidal cell layer of the hippocampal CA1 and similar to short-term depression (109.7 + 8.1% of baseline
CA3 regions (Fig. 3A and B), which was consistent with after weaker HFS, n ¼ 5, P ¼ 0.056, paired t-test, Fig. 5A
the results of previous studies using in situ hybridization and D). In contrast, standard HFS-induced LTP in slices
(14,36 –39). Immunostaining with the DISC1 antibody and from the Disc1 +/+ and Disc1 D2-3/D2-3 mice with a signifi-
organelle-specific antibodies revealed that DISC1 immunor- cantly smaller magnitude of LTP in the Disc1 D2-3/D2-3
eactivity was mainly localized to the vicinity of the Golgi mice at 60 min after standard HFS (Disc1 +/+ , 130.8 +
apparatus (Fig. 3C and D). The immunoreactivity of DISC1 6.1% of baseline 60 min after standard HFS, n ¼ 4, P ¼
was diminished in the Disc1 D2-3/D2-3 mice (Fig. 3B and D). 0.002, paired t-test; Disc1 D2-3/D2-3, 117.7 + 7.0% of baseline
To analyze the subcellular distribution of DISC1 further, 60 min after standard HFS, n ¼ 5, P ¼ 0.005, paired t-test,
immunoelectron microscopic analysis was performed in hip- Fig. 5B and D). Notably, the form and magnitude of the
pocampal CA1 pyramidal cells stained with the C-terminal standard HFS-induced LTP in the Disc1 D2-3/D2-3 mice were
antibody. DISC1 immunoreactivity was detected in the similar to those of the weaker HFS-induced LTP in the
Human Molecular Genetics, 2011, Vol. 20, No. 23 4671

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Figure 3. Immunostaining of endogenous DISC1 in vivo. (A– D) Immunofluorescent analysis of DISC1 was performed with anti-DISC1 C-ter antibody (green)
in P2 mouse hippocampus prepared from Disc1 +/+ (A and C) and Disc1 D2-3/D2-3 (B and D) mice. The Golgi apparatus and nuclei were stained with anti-GM130
antibody (red) and Hoechst 33342 (blue), respectively. (A and B) DISC1 immunoreactivity was detected in pyramidal cell layer of the hippocampal CA1 and
CA3 region. (C and D) The boxed regions in (A) and (B) were shown at higher magnification. DISC1 immunoreactivity was mainly localized to the vicinity of
the Golgi apparatus in the hippocampal CA1 pyramidal cells. (E–G) Immunoelectron microscopic analysis of DISC1 was performed with anti-DISC1 C-ter
antibody in CA1 pyramidal cells of P2 mouse hippocampus prepared from the Disc1 +/+ mice. Golgi apparatus (arrow) was composed of stacks of membrane-
bound structures known as cisterna and localized at the base of apical dendrites (AD). DISC1 immunoreactivity (arrowhead) was detected in the vicinity of the
Golgi apparatus near the nucleus (N). The scale bar represents 100 mm (A and B), 10 mm (C and D), 3 mm (E), 500 nm (F) and 100 nm (G).

Disc1 +/+ mice. When a stronger HFS was used to induce HFS, n ¼ 5, P , 0.001, paired t-test; Disc1 D2-3/D2-3, 134.9 +
LTP, the magnitude of the LTP was significantly larger in 4.0% of baseline 60 min after a stronger HFS, n ¼ 4, P ,
the Disc1 D2-3/D2-3 mice than in the Disc1 +/+ mice 0.001, paired t-test, Fig. 5C and D). However, the magnitude
(Disc1 +/+ , 121.2 + 5.3% of baseline 60 min after a stronger of the stronger HFS-induced LTP in the Disc1 D2-3/D2-3 mice
4672 Human Molecular Genetics, 2011, Vol. 20, No. 23

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021

Figure 4. Immunostaining of endogenous DISC1 in vitro. (A– D) Immunofluorescent analysis of DISC1 was performed with anti-DISC1 C-ter antibody (green)
in cultured hippocampal neurons at DIV3 (A and B) and astrocytes at DIV8 (C and D) prepared from the Disc1 +/+ (A and C) and the Disc1 D2-3/D2-3 (B and D)
mice. DISC1 was localized to the vicinity of the Golgi apparatus, which was stained with anti-GM130 antibody (red). (E) Immunoelectron microscopic analysis
of DISC1 was performed with anti-DISC1 C-ter antibody in cultured hippocampal neurons at DIV5. DISC1 immunoreactivity (arrowhead) was detected in the
vicinity of the Golgi apparatus (arrow) near the nucleus (N). The broken lines represent the cell margins. The scale bar represents 5 mm (A– D) and 500 nm (E).

was similar to that of the standard HFS-induced LTP in the frequency of HFS was needed to produce an equivalent
Disc1 +/+ mice. The frequency dependencies of LTP LTP magnitude to that induced in the Disc1 +/+ mice.
magnitude in the Disc1 +/+ and Disc1 D2-3/D2-3 mice were These results suggest that LTP can be induced in the synap-
summarized by plotting the normalized fEPSP slope at ses of Disc1 D2-3/D2-3 mice but that the threshold for LTP in-
55– 60 min after HFS delivery against HFS frequency as duction is increased by the disruption of exons 2 and 3 of
shown in Figure 5D. In the Disc1 D2-3/D2-3 mice, a higher the Disc1 gene.
Human Molecular Genetics, 2011, Vol. 20, No. 23 4673

Behavioral analyses of the Disc1 D2-3/D2-3 mice

There were no apparent differences between juvenile


Disc1 +/+ and Disc1 D2-3/D2-3 mice (4 –8 weeks old) in most
of the behavioral tests conducted. In the open field test,
there was a marginal but significant increase in the locomotor
distance traveled within the inner circle of the open field in
the female (128 + 11%) but not the male (109 + 16%)
Disc1 D2-3/D2-3 mice compared with the activity of the
Disc1 +/+ mice (Table 1).
Abnormalities in emotional and social behavior were
observed in adult Disc1 D2-3/D2-3 mice (12 – 24 weeks old).
These behaviors were assessed using the elevated plus-maze

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


test, the cliff-avoidance test and the social interaction test
(Table 2). In the elevated plus-maze test, open arm time was
significantly increased, whereas closed arm time was signifi-
cantly decreased in the Disc1 D2-3/D2-3 mice compared with
the Disc1 +/+ mice (P , 0.01, Fig. 6A and B). The
Disc1 D2-3/D2-3 mice also exhibited a marked increase in open
and closed arm entries compared with the Disc1 +/+ mice
(P , 0.05, Supplementary Material, Fig. S5A and B). In the
cliff-avoidance test, the number of jumping events during a
10 min observation period was significantly increased in the
Disc1 D2-3/D2-3 mice compared with the Disc1 +/+ mice (P ,
0.05, Fig. 6C). Furthermore, the cumulative frequency of the
first jumping off of the platform during a 3 min observation
period was significantly increased in the female Disc1 D2-3/
D2-3
mice compared with the Disc1 +/+ mice (Supplementary
Material, Fig. S5C – E). In the social interaction test, a slight
but significant increase in social interaction time was evident
in the Disc1 D2-3/D2-3 mice compared with the Disc1 +/+ mice
[F(1,30) ¼ 4.54, P , 0.05] (Fig. 6D and Supplementary
Material, S5F –H). There was no difference in performance
in the open field or forced swim tests between the Disc1 D2-3/
D2-3
mice and the Disc1 +/+ mice (Table 2).
With respect to cognitive function, adult Disc1 D2-3/D2-3
mice, especially females, exhibited an impairment of sensori-
motor gating as indicated by PPI deficits (P , 0.05, Fig. 6E –
G) without changes in the acoustic startle amplitude (Supple-
mentary Material, Fig. S6I). Adult Disc1 D2-3/D2-3 mice were
generally normal with respect to various learning and
memory tests, including the Y-maze test, the novel object rec-
ognition test and the working memory test using the radial arm
maze (Table 2 and Supplementary Material, Fig. S5J – L).
Context-dependent conditioned fear freezing was significantly
enhanced in the Disc1 D2-3/D2-3 mice compared with the
Disc1 +/+ mice (P , 0.05, Fig. 6H), although no significant
difference in tone-dependent freezing time (Fig. 6I) or sensi-
Figure 5. Synaptic transmission and plasticity in the Disc1 D2-3/D2-3 mice.
(A) Induction of LTP by 100 pulses at 50 Hz in Disc1 +/+ and Disc1 D2-3/
tivity to an electric footshock was observed between the two
D2-3
mice. LTP was induced in the Disc1 +/+ mice, whereas was not in the groups (0.19 + 0.01 mA in the Disc1 +/+ mice, 0.18 +
Disc1 D2-3/D2-3 mice. (B) Induction of LTP by 100 pulses at 100 Hz in both 0.01 mA in the Disc1 D2-3/D2-3 mice). Latent inhibition of tone-
Disc1 +/+ and Disc1 D2-3/D2-3 mice. The magnitude of the LTP in the dependent freezing was also normal in adult Disc1 D2-3/D2-3
Disc1 D2-3/D2-3 mice was smaller than in the Disc1 +/+ mice. (C) Induction mice (Table 2).
of LTP by 100 pulses at 200 Hz in both Disc1 +/+ and Disc1 D2-3/D2-3 mice.
The magnitude of the LTP in the Disc1 D2-3/D2-3 mice was larger than in the Finally, sensitivities to METH, which is an indirect
Disc1 +/+ mice. (D) Summary of the LTP magnitude induced by 100 pulses dopaminergic agonist, and MK-801, a non-competitive N-
at 50, 100 and 200 Hz at 55– 60 min after HFS in both Disc1 +/+ and methyl-D-aspartate (NMDA) receptor antagonist, were deter-
Disc1 D2-3/D2-3 mice. In the Disc1 D2-3/D2-3 mice, the higher frequency of HFS mined in adult Disc1 D2-3/D2-3 mice. In the Disc1 D2-3/D2-3
than the Disc1 +/+ mice was needed to produce an equivalent LTP magnitude
to that in the Disc1 +/+ mice. Comparisons between Disc1 +/+ and Disc1 D2-3/
mice, especially the females, METH-induced hyperactivity
D2-3
mice were made using a two-tailed independent t-test. was significantly potentiated compared with the Disc1 +/+
4674 Human Molecular Genetics, 2011, Vol. 20, No. 23

Table 1. Summary of phenotypes of the Disc1 D2-3/D2-3 mice in juvenile group

Domain of function Test Parameter n Male+ n Male n Female


Female
+/+ D/D +/+ D/D +/+ D/D

Psychosis-related Locomotor Locomotor activity 12 15 ¼ 5 7 ¼ 7 8 ¼


behavior
Affective behavior Open field Total movement 12 15 ¼ 5 7 ¼ 7 8 ¼
Inner movement 12 15 ¼ 5 7 ¼ 7 8  (P ¼ 0.048)
Social interaction Interaction time 12 15 ¼ 5 7 ¼ 7 8 ¼
Cognition/learning and PPI Prepulse (69, 73, 77 18 20 ¼ 8 11 ¼ 10 9 ¼
memory and 81 dB)
Y-maze Alternation 18 20 ¼ 8 11 ¼ 10 9 ¼
Nobel object Exploratory preference 12 15 ¼ 5 7 ¼ 7 8 ¼

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


recognition Exploration time 12 15 ¼ 5 7 ¼ 7 8 ¼

n, number of animals; , higher than +/+; ¼, no difference.

mice [F(1,18) ¼ 9.83, P , 0.05] (Fig. 6J – L). There was no antibody detected a protein band with a molecular mass of
difference between the Disc1 D2-3/D2-3 and Disc1 +/+ mice in 100 kDa that was absent in brain lysates from the
their sensitivity to MK-801 (Table 2). Disc1 D2-3/D2-3 mice. Mass spectral analysis determined that
the N-terminal and C-terminal antibodies recognize the en-
dogenous mouse DISC1. We also confirmed that the full-
Effect of clozapine treatment on the Disc1 D2-3/D2-3 mice length DISC1 was not expressed in brain extracts of mice
Because the behavioral analysis of the Disc1 D2-3/D2-3 mice with a 25 bp deletion in exon 6 of Disc1 [Disc1 (D6) mice].
demonstrated an apparent impairment in performance in the These results indicate that the 100 kDa protein is the major
elevated plus-maze test, the effect of acute administration of isoform of DISC1. However, we cannot neglect the possibility
clozapine [1 mg/kg, intraperitoneally (i.p.)] was analyzed in of the existence of other isoforms, although the expression
the Disc1 D2-3/D2-3 mice. A two-way ANOVA revealed a levels of other isoforms, if present, are much lower than that
significant interaction between drug treatment and genotype of the 100 kDa DISC1.
with respect to the time spent in the open arms [Fig. 7A; DISC1 interacts with many proteins, including NDEL1,
drug treatment– genotype interaction F(1,33) ¼ 4.81, P , LIS1, FEZ1, PDE4B, KIF5B, GRB2, GSK3b, girdin, kalirin,
0.05] and the time spent in the closed arms [Fig. 7B; drug TNIK and DIXDC1 (8 – 19). A number of these proteins inter-
treatment – genotype interaction F(1,33) ¼ 5.84, P , 0.05]. A act with DISC1 through the region encoded by exons 2 and 3
multiple-comparison test with Tukey – Kramer post hoc tests (21). Therefore, the function of DISC1 as a scaffold protein,
indicated that the increase in the time spent in the open which requires interaction with these proteins, should be lost
arms by saline-treated Disc1 D2-3/D2-3 mice was significantly in Disc1 (D2-3) mice.
ameliorated by treatment with clozapine (P , 0.05, To understand the physiological significance of DISC1,
Fig. 7A). Treatment with clozapine also rescued the decrease many laboratories, including our own, have knocked down
in the time spent in the closed arms by the Disc1 D2-3/D2-3 mice the expression of DISC1 using small interfering RNA or
(P , 0.05, Fig. 7B). Clozapine treatment had no effect on the short hairpin RNA (8,10– 12,16– 19,40– 47). Although the
performance of Disc1 +/+ mice in the elevated plus-maze test acute knockdown of DISC1 impairs neuronal migration,
(Fig. 7 and Supplementary Material, S6). axon elongation, dendritic maturation and neurogenesis, the
Disc1 D2-3/D2-3 mice showed nearly normal cytoarchitecture
of the neocortex, the basal ganglia and the hippocampus
Monoamines and their metabolite levels in the brains and no reduction in the size of the white matter tracts or
of adult Disc1 D2-3/D2-3 mice the volume of the prefrontal cortex. Thus, a detailed analysis
will be required to examine the structural changes that may
In general, no differences between the Disc1 D2-3/D2-3 mice and occur in the Disc1 D2-3/D2-3 mice. Chronic depletion of
the Disc1 +/+ mice regarding monoamine levels or metabolite DISC1 may induce molecular compensation during develop-
contents were observed in any of the various brain regions that ment. To overcome this complicating factor, the generation
were examined. In one exception, a slight but significant re- of a conditional Disc1 knockout mouse is necessary.
duction in serotonin content was observed in the cerebellum Our immunohistochemical studies revealed that DISC1 is
of the Disc1 D2-3/D2-3 mice compared with the Disc1 +/+ mice localized to the vicinity of the Golgi apparatus in hippocam-
(Table 3). pal neurons and astrocytes. Previous studies have shown that
both NDEL1 and LIS1 are localized in the Golgi apparatus
(48,49). DISC1 can associate with ARFGEF2, PACS1 and
DISCUSSION
GM130 (17,50), all of which are involved in vesicle traffick-
We generated mice lacking exons 2 and 3 of Disc1 [Disc1 ing from the Golgi apparatus. Thus, it is possible that DISC1
(D2-3) mice] and made specific antibodies to the N- and localizes to the peripheral region of the Golgi apparatus by
C-termini of DISC1. Western blot analysis using either virtue of its association with these proteins. DISC1 may
Human Molecular Genetics, 2011, Vol. 20, No. 23 4675

Table 2. Summary of phenotypes of the Disc1 D2-3/D2-3 mice in adult group

n Male+Female n Male n Female


Domain of function Test Parameter +/+ D/D +/+ D/D +/+ D/D

Psychosis-related Locomotor Locomotor activity 26 26 ¼ 9 12 ¼ 17 14 ¼


behavior MK-801-induced Locomotor activity 17 19 ¼ 8 8 ¼ 9 11 ¼
hyperactivity
METH-induced Locomotor activity 18 20 ¼ 8 10 ¼ 10 10  (P ¼ 0.0057)
hyperactivity
Affective behavior Open field Total movement 14 15 ¼ 7 7 ¼ 7 8 ¼
Inner movement 14 15 ¼ 7 7 ¼ 7 8 ¼
Elevated Open arm entries 16 20  (P ¼ 0.00014) 8 10  (P ¼ 0.0031) 8 10  (P ¼ 0.024)
plus-maze Closed arm entries 16 20  (P ¼ 0.010) 8 10 ¼ 8 10  (P ¼ 0.037)
Time in open arms 16 20  (P ¼ 0.00013) 8 10  (P ¼ 0.0018) 8 10  (P ¼ 0.028)

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Time in closed arms 16 20  (P ¼ 0.00056) 8 10  (P ¼ 0.022) 8 10  (P ¼ 0.015)
Cliff-avoidance Frequency of 26 20  (P ¼ 0.037) 13 11 ¼ 13 9 ¼
jumping
Forced swim Immobility time 14 16 ¼ 8 10 ¼ 6 6 ¼
Social interaction Interaction time 12 20  (P ¼ 0.042) 6 8 ¼ 6 12 ¼
Cognition/learning PPI Prepulse (69, 73, 77 12 16  (P ¼ 0.027) 6 8 ¼ 6 8  (P ¼ 0.022)
and memory and 81 dB)
Latent inhibition Tone-dependent 15 17 ¼ 8 8 ¼ 7 9 ¼
freezing
Y-maze Alternation 14 16 ¼ 7 8 ¼ 7 8 ¼
Radial-maze Working memory 12 17 ¼ 7 12 ¼ 5 5 ¼
errors
Fear conditioning Context-dependent 14 16  (P ¼ 0.042) 7 8 ¼ 7 8 ¼
freezing
Tone-dependent 14 16 ¼ 7 8 ¼ 7 8 ¼
freezing
Novel object Exploratory 14 20 ¼ 7 10 ¼ 7 10 ¼
recognition preference
Exploration time 14 20 ¼ 7 10  (P ¼ 0.037) 7 10 ¼

n, number of animals; , higher than +/+; , lower than +/+; ¼, no difference.

participate in vesicle movement from the Golgi apparatus. hypothalamic supraoptic nucleus (54). Studies of the hippo-
DISC1 is also known to interact with KIF5B, NDELl and campus have revealed that pregnenolone sulfate, which is
LIS1 and to act as a cargo adapter linking kinesin and one of the most abundant neurosteroids, induces a modulatory
dynein to cargo proteins, such as NDEL1/LIS1 and GRB2 metaplasticity in an L-type voltage-gated calcium channel-
(16,17). Although DISC1 has been reported to be localized dependent manner (55) and that brain-derived neurotrophic
in the mitochondria (46,51,52), we did not detect immunor- factor (BDNF) is likely to alter the capacity for plastic
eactivity of DISC1 in association with these organelles. changes in synaptic efficacy (56). Interestingly, it has been
This discrepancy may be explained by the fact that the pre- reported that serum levels of D-serine, neurosteroids and
vious studies were performed using a commercial antibody. BDNF are altered in schizophrenia (57– 62). The relationship
However, we cannot dismiss the possibility that under the between these factors and DISC1 function represents an intri-
conditions used, our antibody did not detect mitochondria- guing subject for study that may lead to an understanding of
associated DISC1. the mechanisms through which DISC1 affects synaptic trans-
Electrophysiological analyses of the dentate gyrus of mission and higher order brain functions.
mouse hippocampal slices were conducted to examine the Adult, but not juvenile, Disc1 D2-3/D2-3 mice displayed
effect of the disruption of exons 2 and 3 of the Disc1 gene abnormal emotional behavior as assessed by the elevated
on synaptic functions. There was no difference in basal syn- plus-maze test and the cliff-avoidance test. This observation
aptic transmission at the medial perforant path-granule cell suggests that disruption of exons 2 and 3 of the Disc1 gene
synapses in the Disc1 +/+ and Disc1 D2-3/D2-3 mice. In con- may cause behavioral effects that are dependent upon neurode-
trast, though HFS-induced LTP could be observed at these velopment and that result in lower anxiety and/or higher
synapses in the Disc1 D2-3/D2-3 mice, the threshold for LTP in- impulsivity in adult mice. The Disc1 D2-3/D2-3 mice also exhib-
duction was significantly higher than in the Disc1 +/+ mice. ited increased social interaction with an unfamiliar intruder
Such a threshold shift in the LTP induction is observed in mouse during the social interaction test and increased explor-
metaplasticity, which has been proposed to relate to higher ation time of novel objects in the novel object recognition test;
order brain functions (53). The factors that contribute to the both of these behaviors may be associated with lower anxiety/
induction of metaplasticity are diverse. For example, glial- higher impulsivity. The higher impulsivity observed in the
derived D-serine affects NMDA receptor activity in such a Disc1 D2-3/D2-3 mice is consistent with the clinical features of
way that it shifts the threshold for LTP in the rat patients with schizophrenia (63,64).
4676 Human Molecular Genetics, 2011, Vol. 20, No. 23

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Figure 6. Behavioral analyses of the Disc1 D2-3/D2-3 mice. (A and B) Performance of adult Disc1 D2-3/D2-3 mice in the elevated plus-maze test. Time spent in the
open and closed arms as characterized by gender. Values indicate the mean + SE [Disc1 +/+ (+/+) (n ¼ 16) and Disc1 D2-3/D2-3 (D/D) (n ¼ 20) in female +
male, Disc1 +/+ (n ¼ 8) and Disc1 D2-3/D2-3 (n ¼ 10) in female, Disc1 +/+ (n ¼ 8) and Disc1 D2-3/D2-3 (n ¼ 10) in male]. (C) Performance of adult Disc1 D2-3/
D2-3
mice in the cliff-avoidance test. Number of jumping events during a 10 min observation categorized by gender. Values indicate the mean + SE
[Disc1 +/+ (n ¼ 26) and Disc1 D2-3/D2-3 (n ¼ 20) in female + male, Disc1 +/+ (n ¼ 13) and Disc1 D2-3/D2-3 (n ¼ 9) in female, Disc1 +/+ (n ¼ 13) and
Disc1 D2-3/D2-3 (n ¼ 11) in male]. (D) Performance of adult DISC1 Disc1 D2-3/D2-3 mice in the social interaction test. Accumulated time of social interaction cate-
gorized by gender over five trials. Values indicate the mean + SE [Disc1 +/+ (n ¼ 12) and Disc1 D2-3/D2-3 (n ¼ 20) in female + male, Disc1 +/+ (n ¼ 6) and
Disc1 D2-3/D2-3 (n ¼ 12) in female, Disc1 +/+ (n ¼ 6) and Disc1 D2-3/D2-3 (n ¼ 8) in male]. (E– G) Performance of adult Disc1 D2-3/D2-3 mice in the PPI test.
PPI (%) at four different prepulse intensities (69, 73, 77 and 81 dB) categorized by gender. Values indicate the mean + SE [Disc1 +/+ (n ¼ 12) and
Disc1 D2-3/D2-3 (n ¼ 16) in female + male, Disc1 +/+ (n ¼ 6) and Disc1 D2-3/D2-3 (n ¼ 8) in female, Disc1 +/+ (n ¼ 6) and Disc1 D2-3/D2-3 (n ¼ 8) in male].
(H and I) Performance of adult DISC1 Disc1 D2-3/D2-3 mice in the fear conditioning test. Context- and tone-dependent memory during the fear conditioning
test categorized by gender. Values indicate the mean + SE [Disc1 +/+ (n ¼ 14) and Disc1 D2-3/D2-3 (n ¼ 16) in female + male, Disc1 +/+ (n ¼ 7) and
Disc1 D2-3/D2-3 (n ¼ 8) in female, Disc1 +/+ (n ¼ 7) and Disc1 D2-3/D2-3 (n ¼ 8) in male]. (J–L) Performance of adult Disc1 D2-3/D2-3 mice in the METH-induced
hyperactivity test. Locomotor activity was measured for 180 min immediately after METH treatment categorized by gender. Single beam breaks were summar-
ized as ‘counts’. Data are shown as 5 min increments. Values indicate the mean + SE [Disc1 +/+ (n ¼ 18) and Disc1 D2-3/D2-3 (n ¼ 20) in female + male,
Disc1 +/+ (n ¼ 10) and Disc1 D2-3/D2-3 (n ¼ 10) in female, Disc1 +/+ (n ¼ 8) and Disc1 D2-3/D2-3 (n ¼ 10) in male]. ∗ P , 0.05, ∗∗ P , 0.01 versus Disc1 +/+ mice.

Clozapine is an atypical antipsychotic that has been shown treatment with clozapine. These results suggest that deficiency
to attenuate a broad range of symptoms in subjects with of DISC1 function can be rescued by treatment with anti-
schizophrenia (65). In our analysis of the effects of acute ad- psychotic drugs; this finding may reflect the therapeutic
ministration of clozapine to Disc1 D2-3/D2-3 mice on perform- effects of such drugs on psychotic symptoms in patients
ance in the elevated plus-maze test, clozapine-treated with schizophrenia.
Disc1 D2-3/D2-3 mice no longer exhibited deficits in the time Interestingly, the Disc1 D2-3/D2-3 female mice exhibited more
spent in the open arms and closed arms, which indicated severe deficits in some behavioral tests, such as the PPI test,
that the emotional deficits in these mice, such as lower the cliff-avoidance test and the METH-induced hyperactivity
anxiety and/or higher impulsivity, were attenuated by test than did the male Disc1 D2-3/D2-3 mice. Consistent with the
Human Molecular Genetics, 2011, Vol. 20, No. 23 4677

and their metabolites in the brain tissue of Disc1 D2-3/D2-3 and


wild-type mice. Therefore, it seems reasonable to infer that
the development of monoaminergic neurons is largely unaffect-
ed by the loss of DISC1 in mice. However, because
METH-induced hyperactivity and dopamine release in the
nucleus accumbens were potentiated in the Disc1 D2-3/D2-3
mice, further studies that examine potential alterations in dopa-
minergic neurotransmission in the Disc1 D2-3/D2-3 mice are war-
ranted.

MATERIALS AND METHODS

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Generation of Disc1 (D2-3) mice
Disc1 (D2-3) mice (Acc Nos CDB0583K and CDB0678K:
http://www.cdb.riken.jp/arg/mutant%20mice%20list.html)
were generated by gene targeting in TT2 ES cells (74) and
Figure 7. Effect of clozapine treatment on performance in the elevated
plus-maze test. (A and B) Effect of clozapine treatment on time spent in
HK3i ES cells (75) as described previously (http://www.cdb.
open and closed arms. Values indicate the mean + SE [saline-treated Disc1 +/+ riken.jp/arg/protocol.html). The targeting vector was designed
(n ¼ 11), saline-treated Disc1 D2-3/D2-3 (n ¼ 10), clozapine-treated Disc1 +/+ to replace most of exon 2 and all of exon 3 with a loxP/
(n ¼ 9) and clozapine-treated Disc1 D2-3/D2-3 (n ¼ 7)]. ∗ P , 0.05 versus saline- PGK-Neo-pA/loxP cassette (http://www.cdb.riken.jp/arg/ca
treated Disc1 +/+ mice, #P , 0.05 versus saline-treated Disc1 D2-3/D2-3 mice. ssette.html) and to insert an in-frame stop codon between
the remainder of exon 2 and the loxP site. Successful homolo-
gous recombination was confirmed in two clones of the TT2
ES cells (#5, #33) and two clones of the HK3i ES cells
observed gender-dependent changes in behavior, our in vivo (#24, #146) by PCR (forward primer, 5′ -GTA CTC GGA
dialysis study revealed gender-dependent alterations of TGG AAG CCG GTC TTG TC-3′ ; reverse primer, 5′ -GGT
METH-induced dopamine release in the nucleus accumbens TTC AGA GGA TCC TCC GTT CAC C-3′ ) and southern
of the Disc1 D2-3/D2-3 mice (data not shown). Although the blot analysis with a 5′ probe and a 3′ probe; the clones
reason for this gender difference remains unclear, sex hormones yielded highly chimeric mice. The heterozygous offspring
may play a role. For instance, it has been demonstrated that an were intercrossed to obtain homozygous mutants, which
increase in the plasma level of female hormones (e.g. estrogen were genotyped by southern blot analysis with a 5′ probe
and progesterone) impairs sensorimotor gating (66 –68). and PCR using a mixture of three primers (forward primer
In this study, we demonstrated that the Disc1 D2-3/D2-3 mice ‘Wt’, 5′ -GGT CAG AGC TCA GAG AAC TCC TTG
showed PPI deficits, which are a well-known phenomenon TGG-3′ , forward primer ‘Neo’, 5′ -CAT CGC CTT CTA
observed in patients with schizophrenia and other neuro- TCG CCT TCT TGA CG-3′ ; reverse primer ‘Rev’, 5′ -GCT
psychiatric disorders (69). Such deficits have been observed TCC TAC AGA GCA GAC TCC ACA CC-3′ ). Primers Wt
in most genetically engineered mouse models with mutated and Rev amplified the wild-type allele (547 bp), and primers
DISC1 (Table 4). In contrast, our newly developed Neo and Rev amplified the mutated allele (619 bp). Four
Disc1 D2-3/D2-3 mice displayed the potentiation of context- strains of Disc1 (D2-3) mice derived from four different
dependent fear memory, whereas other memory functions, in- clones of ES cells showed identical phenotypes. For the ana-
cluding cue-dependent fear memory and working memory, did lysis of the mutant mice, a marker-assisted selection congenic
not differ from those of wild-type mice. Therefore, it seems strategy was employed. Clone #33-derived mice, which had
unlikely that DISC1 plays an indispensable role in learning 75% C57BL/6 and 25% CBA background in the N1 gener-
and memory in mice. ation, were backcrossed with C57BL/6JJmsSlc more than
Previous reports have demonstrated that genetically engi- five times such that all 100 markers corresponded to C57BL/
neered mouse models with mutated DISC1 show decreased 6 alleles. Histological and behavioral analyses were performed
social interaction and memory impairment (22 – 25). The using littermates (both males and females) generated by cross-
present findings are somewhat inconsistent with these breeding of the backcrossed clone #33 mice. C57BL/
reports; this discrepancy may be due to the presence of differ- 6JJmsSlc, 129X1/SvJJmsSlc, Slc:ddY and Slc:ICR mice
ent amounts and/or functions of DISC1 in different genetically were obtained for breeding from SLC (Hamamatsu, Shizuoka,
engineered mice. The present study generated mice with tar- Japan). Mice were genotyped for the 25 bp deletion in Disc1
geted deletions in DISC1; in these mice, intact DISC1 was (D6) using PCR primers (forward primer, 5′ -GCT GTG
not expressed in the brain, while other previously reported ACC TGA TGG CAC T-3′ ; reverse primer, 5′ -GCA AAG
mouse models express intrinsic and/or extrinsic DISC1 TCA CCT CAA TAA CCA-3′ ) as described previously (35).
(Table 4) (22– 25,27,28,45,70,71). All of the animal protocols were approved by the Animal
Altered monoaminergic neurotransmission has been Care and Use Committee of the Nagoya University Graduate
reported in patients with schizophrenia (72), and these altera- School of Medicine, and the Principles for the Care and Use
tions may be associated with behavioral abnormalities (73). of Laboratory Animals, which were approved by the Japanese
There were few differences in the tissue content of monoamines Pharmacological Society and the National Institutes of Health
4678 Human Molecular Genetics, 2011, Vol. 20, No. 23

Table 3. Monoamines and their metabolite contents in each region of the Disc1 D2-3/D2-3 mouse brain

NE MHPG DA DOPAC HVA 5-HT 5-HIAA

Prefrontal cortex +/+ 618.9 + 41.5 204.9 + 12.4 83.3 + 5.9 34.1 + 3.6 87.6 + 7.2 1085.7 + 82.8 188.5 + 5.1
D/D 625.2 + 35.1 220.0 + 13.0 86.8 + 7.6 32.9 + 2.0 110.9 + 13.6 1038.5 + 59.1 181.1 + 12.7
Striatum +/+ 124.2 + 18.3 221.3 + 14.9 27141.5 + 952.8 1044.3 + 32.0 2522.2 + 142.5 1187.7 + 53.1 693.6 + 40.6
D/D 105.3 + 10.3 204.3 + 19.4 25055.4 + 1029.7 980.8 + 40.0 2464.6 + 180.7 1086.1 + 74.8 629.2 + 26.9
Hippocampus +/+ 433.4 + 39.0 160.9 + 16.6 13.7 + 2.6 18.5 + 1.3 36.1 + 3.8 1046.0 + 116.4 374.5 + 35.2
D/D 427.0 + 18.1 169.6 + 12.6 17.1 + 3.2 17.7 + 1.3 40.9 + 3.9 997.9 + 50.3 343.1 + 20.8
Midbrain +/+ 497.6 + 27.9 134.2 + 18.0 186.8 + 125.0 56.0 + 10.1 130.7 + 21.4 1045.0 + 71.6 308.3 + 26.3
D/D 503.9 + 18.9 154.5 + 4.8 343.2 + 19.9 67.6 + 2.9 157.8 + 8.2 1029.6 + 63.8 301.4 + 18.6
Cerebellum +/+ 403.1 + 22.5 171.5 + 8.7 15.7 + 4.0 18.0 + 2.5 18.7 + 2.2 227.5 + 14.7 98.1 + 8.2
D/D 363.0 + 8.6 186.3 + 13.2 11.3 + 2.2 17.3 + 1.9 48.3 + 18.0 191.2 + 7.6 ∗ 86.0 + 5.5

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Values are expressed as nanogram per gram of wet tissue (mean + SE). +/+ (n ¼ 7) and D/D (n ¼ 9). ∗ P , 0.05 versus +/+.
NE, norepinephrine; DA, dopamine; 5-HT, 5-hydroxytryptamine; MHPG, 3-methoxy-4-hydroxyphenylglycol; DOPAC, 3,4-dihydroxyphenylacetic acid; HVA,
homovanillic acid; 5-HIAA, 5-hydroxyindoleacetic acid.

Guide for the Care and Use of Laboratory Animals, were fol- (GE Healthcare). The MBP-mDISC1 C-terminal and
lowed. GST-mDISC1 C-terminal proteins were bound to a Mono Q
column (GE Healthcare) and eluted with a linear gradient
(0– 1000 mM NaCl in 20 column volume). A full-length
RNA extraction and RT – PCR GST-mDISC1 protein was produced in Rosetta-gami 2
Total RNA was extracted from P7 mouse brain tissue samples E. coli cells (Merck, Darmstadt, Germany) and purified with
using the RNeasy Lipid Tissue Mini Kit (QIAGEN, Hilden, glutathione-Sepharose 4B beads.
Germany). Reverse transcription was performed with 1 mg
of RNA using the Transcriptor First Strand cDNA Synthesis
Kit (Roche Applied Science, Mannheim, Germany). PCR Antibodies
amplification was performed using an Ex-Taq system Four guinea pig anti-DISC1 N-terminal antibodies were
(Takara Bio, Otsu, Shiga, Japan) with 35 cycles for Disc1 prepared by Medical & Biological Laboratories Co., Ltd.
exons 2 – 3 (forward primer, 5′ -GAA TGT GGC ACG GTC (MBL, Nagoya, Aichi, Japan) and purified using specific pep-
TCC TC-3′ ; reverse primer, 5′ -TAA TCG CCA TCC TCG tides (amino acid residues 131 – 170 of mDISC1) as antigens.
ACC AC-3′ ) and Disc1 exons 7– 10 (forward primer, Two rabbit and two guinea pig anti-DISC1 C-terminal anti-
5′ -TGG CTG TCA GAG AAC TCA CTG CTC AG-3′ ; bodies were prepared by MBL. To generate rabbit and
reverse primer, 5′ -CCA TGC ACT TCA CAG TGT TTG guinea pig polyclonal antibodies against the C-terminal
CCT TCA-3′ ). PCR amplification for Hprt was also performed region of DISC1, the MBP-mDISC1 C-terminal protein was
using PCR primers (forward primer, 5′ -CCT GCT GGA TTA used as the antigen. The antiserum was precleared using
CAT TAA AGC ACT G-3′ ; reverse primer, 5′ -GTC AAG GST-dnaK protein immobilized to CNBr-activated Sepharose
GGC ATA TCC AAC AAC AAA C-3′ ) as described previous- 4B beads (GE Healthcare). The anti-DISC1 C-terminal anti-
ly (26). body was affinity-purified from the precleared antiserum
using the GST-mDISC1 C-terminal protein immobilized on
CNBr-activated Sepharose 4B beads. Antibodies against
Plasmid construction and protein purification DISC1 (Mid and C-ter, rabbit polyclonal, Invitrogen, Carls-
cDNA encoding murine DISC1 (mDISC1) was generously bad, CA, USA and N-16, goat polyclonal, Santa Cruz Biotech-
provided by Dr Akira Sawa (Johns Hopkins University, Balti- nology, Santa Cruz, CA. USA), GM130 (mouse monoclonal,
more, MD, USA). The mDISC1 cDNA (amino acid residues BD, Franklin Lakes, NJ, USA) and glyceraldehyde-3-phos-
1 – 852 of mDISC1) was inserted into the pGEX-4T-2 vector phate dehydrogenase (mouse monoclonal, Invitrogen) were
(GE Healthcare, Uppsala, Sweden). An mDISC1 C-terminal purchased.
fragment (amino acid residues 658 – 852 of mDISC1) was
inserted into the pGEX-4T-2 and pMAL-c2 vectors (New
Tissue lysates
England Biolabs, Ipswich, MA, USA). The cDNA encoding
full-length dnaK was cloned from BL21 (DE3) Escherichia All of tissue lysates were extracted by the addition of RIPA
coli cells. Next, dnaK was amplified by PCR and subcloned buffer [20 mM Tris – HCl (pH 7.5), 1 mM EDTA, 150 mM
into the pGEX-6P-3 vector (GE Healthcare). The identity of NaCl, 1.0% NP-40, 0.1% sodium deoxycholate, 0.1%
each fragment was confirmed by DNA sequencing. The sodium dodecyl sulfate (SDS) and Complete Protease Inhibi-
MBP-mDISC1 C-terminal protein was produced in BL21 tor Cocktail (Roche Applied Science)]. Protein concentrations
(DE3) E. coli cells and was purified on an amylose resin were determined using the bicinchoninic acid Protein Assay
(New England Biolabs). The GST-mDISC1 C-terminal and Reagent Kit (Pierce, Rockford, IL, USA). The lysates were
GST-dnaK proteins were produced in BL21 (DE3) E. coli boiled with 3× SDS – polyacrylamide gel electrophoresis
cells and were purified with glutathione-Sepharose 4B beads (PAGE) sample buffer [187.5 mM Tris – HCl (pH 6.8), 9%
Human Molecular Genetics, 2011, Vol. 20, No. 23 4679

Table 4. Comparison with other Disc1 mutant mice

Hikida(23) Pletnikov and Li(24) Niwa(48) Clapcote and Koike and Present study
Ayhan (25,85) Lipina(22,86) Kvajo (27,28)
Human1-597 Inducible Inducible Knockdown Q31L L100P D6 (exon 6) D2-3 (exons
expression of DISC1 via in utero 2– 3)
Human1-597 C-terminal gene transfer
fragment
Test Parameter C57BL/6 B6;SJL;CBA C57BL/6N ICR C57BL/6J C57BL/6J C57BL/6J

Locomotor Locomotor activity  (Male)  (Male) ND ¼ ¼  ¼ ¼


MK-801-induced Locomotor activity ND  (Male) ND ND ND ND ND ¼
hyperactivity
METH-induced Locomotor activity ND ND ND  ND ND ND  (Female)
hyperactivity

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


AMP-induced Locomotor activity ND  (Male) ND ND ND  (Male) ND ND
hyperactivity
Open field Total movement  (Male) ¼ ND ND ND ND ND ¼
Inner movement ¼ (Male) ¼ ND ND ND ND ND ¼
Elevated plus-maze Open arm entries ¼ (Male) ND ND ND ND ND ND 
Closed arm entries ND ND ND ND ND ND ND 
Time in open arms ¼ (Male) ¼ ND ND ¼ ¼ ND 
Time in closed arms ND ¼ ND ND ND ND ND 
Cliff-avoidance test Frequency of jumping ND ND ND ND ND ND ND 
Forced swim test Immobility time  (Male)  (Female)  ¼  ¼ ND ¼
Social interaction Interaction time ¼ (Male)  (Male)  ND  ¼ ND 
PPI Prepulse (69–90 dB)  (Male) ¼ ND    ¼ 
Latent inhibition Suppression ratio ND ND ND ND   ND ¼
Y-maze Alternation ¼ (Male) ND ND ND ND ND ND ¼
Radial-maze Working memory ND ND  ND ND ND  (Male) ¼
Fear conditioning Context-dependent ND ND ND ND ND ND ¼ (Male) 
freezing
Tone-dependent ND ND ND ND ND ND ¼ (Male) ¼
freezing
Nobel object Exploratory preference ND ND ND  ND ND ¼ (Male) ¼
recognition test Exploration time ND ND ND ND ND ND ND  (Male)

METH, methamphetamine; AMP, amphetamine; , higher than WT; , lower than WT; ¼, no difference, ND, not determined.
(23) Hikida et al., 2007; (25) Pletnikov et al., 2008; (70) Ayhan et al., 2011; (24) Li et al., 2007; (45) Niwa et al., 2010; (22) Clapcote et al., 2007; (71) Lipina et al.,
2010; (27) Koike et al., 2006; (28) Kvajo et al., 2008.

SDS, 15% glycerol, 6% 2-mercaptoethanol and 0.06% bromo- (Thermo Fisher, San Jose, CA, USA) combined with a Para-
phenol blue]. digm MS4 HPLC system (Michrom BioResources Inc.,
Auburn, CA, USA).
Immunoprecipitation assay and mass spectral analysis
Cresyl violet (Nissl) staining
Lysates from P7 mouse brain tissue were extracted by the
addition of lysis buffer (20 mM Tris – HCl, 1 mM EDTA, Paraffin-embedded brain tissue sections were deparaffinized
50 mM NaCl, 1.0% NP-40 and Complete Protease Inhibitor and placed in 0.5% cresyl violet in distilled water at 378C
Cocktail) and were cleared by centrifugation at 100 000g for 10 min. The sections were briefly rinsed twice in 90%
for 60 min at 48C. The supernatants were incubated with N- ethanol and were dipped in 100% ethanol three times before
terminal or C-terminal anti-DISC1 antibodies for 1 h at 48C, being dehydrated in xylene three times for 5 min each. The
and the immunocomplexes were precipitated using protein sections were affixed to glass cover slips using Permount
A-Sepharose 4B beads (GE Healthcare). The immunocom- solution and were examined under a light microscope.
plexes were washed three times with lysis buffer
and were eluted by boiling in SDS –PAGE sample buffer.
The eluate was separated by SDS – PAGE, and the gel Immunostaining
was silver-stained using the SilverQuest Silver Staining Mice were transcardially perfused with 2% paraformaldehyde
Kit (Invitrogen). The protein band corresponding to the (PFA) in 100 mM phosphate buffer (PB) (pH 7.4) on day P2.
100 kDa DISC1 polypeptide was excised from the gel. The brains were removed, post-fixed with 2% PFA for 6 h
Gel pieces were destained, reduced, S-carboxymethylated and sectioned sagittally with a cryostat (Leica Microsystems,
and digested with trypsin. The resulting peptides were sub- Wetzlar, Germany) at 10 mm. The sections were blocked
jected to nano-electrospray tandem mass spectroscopic ana- using blocking buffer [100 mM PB (pH 7.4), 0.005%
lysis using a Finnigan LTQ Orbitrap XL mass spectrometer saponin, 1% Block Ace (DS Pharma Biomedical, Suita,
4680 Human Molecular Genetics, 2011, Vol. 20, No. 23

Osaka, Japan) and 5% normal goat serum (Vector Laboratories O2/5% CO2, and its pH was adjusted to 7.4. The slices were
Inc., Burlingame, CA, USA)]. The sections were incubated maintained for at least 1 h at RT (26 – 288C) in an incubation
with primary antibodies overnight at 48C in antibody dilution chamber containing gassed ACSF.
buffer [100 mM PB (pH 7.4), 0.005% saponin, and 1% Block
Ace], washed three times in wash buffer [100 mM PB (pH
7.4) and 0.005% saponin] and incubated with Alexa Fluor
488 goat anti-rabbit IgG, Alexa Fluor 555 goat anti-mouse
IgG (Invitrogen) and Hoechst 33342 (Nacalai, Kyoto, Japan) Electrophysiology
in the antibody dilution buffer for 1 h at room temperature
A single hippocampal slice was transferred to the recording
(RT). Finally, the sections were washed three times in wash
chamber and superfused continuously with gassed ACSF at
buffer.
a rate of 2 – 2.5 ml/min at RT. A stimulating electrode (mono-
Hippocampal neurons were prepared from mouse embryos
polar stimulation) was positioned in the molecular layer
at embryonic day 16 (E16) using papain (76). The neurons

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


30– 50 mm from the granule cell layer in the dentate gyrus
were seeded on poly-D-lysine- and laminin-coated cover
to activate the medial perforant path. Constant-current pulses
slips and were cultured in Neurobasal medium (Invitrogen)
(100 ms) were supplied by a stimulator (SEN-3301, Nihon
supplemented with the B-27 supplement (Invitrogen) and
Kohden, Tokyo, Japan) every 30 s. The intensity of the test
1 mM glutamine. The hippocampal neurons at day in vitro
stimuli was adjusted to evoke 50% of the maximum re-
(DIV) 3 and DIV8 were fixed with 4% PFA in PB for
sponse. Using the conventional extracellular recording tech-
10 min and were treated with blocking buffer for 10 min.
nique, fEPSPs were recorded from the middle molecular
The neurons were incubated with primary antibodies in
layer in the dentate gyrus with a glass pipette filled with
the antibody dilution buffer overnight at 48C, washed
2 M NaCl (2 – 3 MV). Baseline recordings were established
three times in wash buffer and incubated for 1 h with
by delivering test stimuli for more than 60 min before the ex-
Alexa Fluor 488 goat anti-rabbit IgG and Alexa Fluor 555
perimental recordings. LTP was induced by 100 pulses at 50,
goat anti-mouse IgG in the antibody dilution buffer for
100 and 200 Hz. The signals were amplified and filtered at
1 h at RT.
5 kHz with an amplifier (Axopatch 200B, Axon Instruments,
Immunofluorescence was performed using a laser scanning
CA, USA). Data acquisition and analysis were performed
confocal microscope (Model LSM 5 Pascal, Zeiss, Oberko-
using pCLAMP 9.0 software (Axon Instruments). The data
chen, Germany and Nikon-A1, Nikon, Tokyo, Japan).
are expressed as the mean + SD; N indicates the number of
slices tested, and one slice was prepared from each mouse.
Immunoelectron microscopy The synaptic transmission in the extracellular recordings was
estimated from the rising phase of the fEPSP slope using
Immunoelectron microscopy using the silver-enhanced
linear regression during the first 0.6 ms after the PSFV (78).
immunogold method was performed as previously described
The magnitude of LTP was expressed as the mean percentage
(77). P2 mouse brains and cultured hippocampal neurons at
change occurring 55 –60 min after HFS with respect to the
DIV5 were fixed with 2% PFA in 100 mM PB (pH 7.4)
final 5 min of the baseline period. Comparisons were made
and incubated at RT for 5 h with the primary anti-DISC1 C-
using a two-tailed t-test for paired or independent samples.
terminal antibody described above followed by incubation
P-values below 0.05 were considered to be statistically signifi-
with a secondary anti-rabbit antibody coupled to 1.4 nm
cant.
gold particles (Nanoprobes Inc., Yaphank, NY, USA). The
sample-bound gold particles were silver-enhanced using the
HQ-silver kit (Nanoprobes Inc.) at RT for 8 min. The
samples were post-fixed with 0.5% osmium oxide in a
buffer (pH 7.3) containing 100 mM cacodylate. The samples Behavioral analyses
were dehydrated by passage through a graded series of
ethanol (50, 70, 90 and 100%) and propylene oxide and em- Information about the open field test (79), the locomotor activ-
bedded in epoxy resin. From this sample, ultrathin sections ity test (79), the Y-maze test (80), the PPI test (79), the fear
were cut, stained with uranyl acetate and lead citrate and conditioning test (80), the elevated plus-maze test (81), the
observed with an electron microscope (JEM-1011 EX; novel object recognition test (82), the social interaction test
JEOL, Akishima, Tokyo, Japan). (79,80), the forced swim test (81), the latent inhibition
test (83), the cliff-avoidance test (84) and the radial-maze
test (85) is provided in the Supplementary Information
Hippocampal slice preparation section. Monoamine metabolism was analyzed as described
Electrophysiological analyses were conducted on 3-week-old previously (86). The data are presented as the mean + SE.
male mice. The mice were decapitated under deep anesthesia Differences between two groups were analyzed using a two-
with ethyl ether. The brains were quickly removed, and trans- tailed Student’s t-test. Differences in pre-pulse inhibition,
verse 400 mm thick slices were cut from the hippocampus locomotor activity and results of the social interaction test
using a vibratome in ice-cold artificial cerebrospinal fluid were analyzed by a repeated ANOVA. The time-course data
(ACSF) containing 128 mM NaCl, 5 mM KCl, 1.3 mM in the cliff-avoidance test were analyzed by the log-rank
MgSO4, 1.25 mM KH2PO4, 2.41 mM CaCl2, 26 mM NaHCO3 test. The effect of clozapine treatment was analyzed by a
and 10 mM D-glucose. The ACSF was gassed with 95% two-way ANOVA followed by Tukey – Kramer post hoc tests.
Human Molecular Genetics, 2011, Vol. 20, No. 23 4681

ACKNOWLEDGEMENTS Disrupted in Schizophrenia 1 interactome: evidence for the close


connectivity of risk genes and a potential synaptic basis for schizophrenia.
cDNA encoding mDISC1 was generously provided by Mol. Psychiatry, 12, 74– 86.
Dr Akira Sawa (Johns Hopkins University). We thank 10. Enomoto, A., Asai, N., Namba, T., Wang, Y., Kato, T., Tanaka, M.,
Dr Mutsuki Amano, Junko Uraguchi-Asaki, Yasutaka Tatsumi, H., Taya, S., Tsuboi, D., Kuroda, K. et al. (2009) Roles of
Disrupted-In-Schizophrenia 1-interacting protein girdin in postnatal
Fujino, Yasuhiro Funahashi, Yusuke Funahashi, Dr Takao development of the dentate gyrus. Neuron, 63, 774 –787.
Hikita, Dr Katsuhiro Kato, Takafumi Kinoshita, Ryo 11. Hayashi-Takagi, A., Takaki, M., Graziane, N., Seshadri, S., Murdoch, H.,
Kuwata, Shinichi Nakamuta, Dr Shinichiro Taya, Dr Shujie Dunlop, A.J., Makino, Y., Seshadri, A.J., Ishizuka, K., Srivastava, D.P.
Wang and Dr Takashi Watanabe for helpful discussions and et al. (2010) Disrupted-in-Schizophrenia 1 (DISC1) regulates spines of the
the preparation of some materials, Yuko Yamashita for her glutamate synapse via Rac1. Nat. Neurosci., 13, 327–332.
technical assistance and Takako Ishii and Naoko Hatamoto 12. Mao, Y., Ge, X., Frank, C.L., Madison, J.M., Koehler, A.N., Doud, M.K.,
Tassa, C., Berry, E.M., Soda, T., Singh, K.K. et al. (2009) Disrupted in
for secretarial assistance. We acknowledge the Division of Schizophrenia 1 regulates neuronal progenitor proliferation via
Medical Research Engineering of the Nagoya University modulation of GSK3beta/beta-catenin signaling. Cell, 136, 1017– 1031.

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


Graduate School of Medicine for the use of ImageQuant 13. Millar, J.K., Pickard, B.S., Mackie, S., James, R., Christie, S., Buchanan,
LAS 4010 (GE), a Leica CM3050 S cryostat and Nikon-A1 S.R., Malloy, M.P., Chubb, J.E., Huston, E., Baillie, G.S. et al. (2005)
microscopy, and we thank the Division for Research of DISC1 and PDE4B are interacting genetic factors in schizophrenia that
regulate cAMP signaling. Science, 310, 1187–1191.
Laboratory Animals, Center for Research of Laboratory 14. Miyoshi, K., Honda, A., Baba, K., Taniguchi, M., Oono, K., Fujita, T.,
Animals and Medical Research Engineering (Technical Kuroda, S., Katayama, T. and Tohyama, M. (2003)
Staff, Noboru Ogiso, Yasutaka Ohya and Kumiko Yano) for Disrupted-In-Schizophrenia 1, a candidate gene for schizophrenia,
animal care and use. participates in neurite outgrowth. Mol. Psychiatry, 8, 685–694.
15. Ozeki, Y., Tomoda, T., Kleiderlein, J., Kamiya, A., Bord, L., Fujii, K.,
Conflict of Interest statement. None declared. Okawa, M., Yamada, N., Hatten, M.E., Snyder, S.H. et al. (2003)
Disrupted-in-Schizophrenia-1 (DISC-1): mutant truncation prevents
binding to NudE-like (NUDEL) and inhibits neurite outgrowth. Proc. Natl
Acad. Sci. USA, 100, 289– 294.
FUNDING 16. Shinoda, T., Taya, S., Tsuboi, D., Hikita, T., Matsuzawa, R., Kuroda, S.,
Iwamatsu, A. and Kaibuchi, K. (2007) DISC1 regulates
This work was supported by JST, CREST, a Grant-in-Aid for
neurotrophin-induced axon elongation via interaction with Grb2.
Scientific Research (S) (20227006) from the Ministry of Edu- J. Neurosci., 27, 4 –14.
cation, Culture, Sports, Science and Technology of Japan 17. Taya, S., Shinoda, T., Tsuboi, D., Asaki, J., Nagai, K., Hikita, T., Kuroda,
(MEXT) and a Grant-in-Aid for GCOE research from MEXT. S., Kuroda, K., Shimizu, M., Hirotsune, S. et al. (2007) DISC1 regulates
the transport of the NUDEL/LIS1/14-3-3epsilon complex through
kinesin-1. J. Neurosci., 27, 15–26.
18. Kim, J.Y., Duan, X., Liu, C.Y., Jang, M.H., Guo, J.U., Pow-anpongkul,
REFERENCES N., Kang, E., Song, H. and Ming, G.L. (2009) DISC1 regulates new
1. Harrison, P.J. and Weinberger, D.R. (2005) Schizophrenia genes, gene neuron development in the adult brain via modulation of AKT-mTOR
expression, and neuropathology: on the matter of their convergence. Mol. signaling through KIAA1212. Neuron, 63, 761–773.
Psychiatry, 10, 40– 68 (image 45). 19. Singh, K.K., Ge, X., Mao, Y., Drane, L., Meletis, K., Samuels, B.A. and
2. Lewis, D.A. and Gonzalez-Burgos, G. (2006) Pathophysiologically based Tsai, L.H. (2010) Dixdc1 is a critical regulator of DISC1 and embryonic
treatment interventions in schizophrenia. Nat. Med., 12, 1016–1022. cortical development. Neuron, 67, 33–48.
3. Millar, J.K., Wilson-Annan, J.C., Anderson, S., Christie, S., Taylor, M.S., 20. Brandon, N.J., Millar, J.K., Korth, C., Sive, H., Singh, K.K. and Sawa, A.
Semple, C.A., Devon, R.S., St Clair, D.M., Muir, W.J., Blackwood, D.H. (2009) Understanding the role of DISC1 in psychiatric disease and during
et al. (2000) Disruption of two novel genes by a translocation normal development. J. Neurosci., 29, 12768–12775.
co-segregating with schizophrenia. Hum. Mol. Genet., 9, 1415–1423. 21. Chubb, J.E., Bradshaw, N.J., Soares, D.C., Porteous, D.J. and Millar, J.K.
4. St Clair, D., Blackwood, D., Muir, W., Carothers, A., Walker, M., (2008) The DISC locus in psychiatric illness. Mol. Psychiatry, 13, 36–64.
Spowart, G., Gosden, C. and Evans, H.J. (1990) Association within a 22. Clapcote, S.J., Lipina, T.V., Millar, J.K., Mackie, S., Christie, S., Ogawa,
family of a balanced autosomal translocation with major mental illness. F., Lerch, J.P., Trimble, K., Uchiyama, M., Sakuraba, Y. et al. (2007)
Lancet, 336, 13–16. Behavioral phenotypes of Disc1 missense mutations in mice. Neuron, 54,
5. Blackwood, D.H., Fordyce, A., Walker, M.T., St Clair, D.M., Porteous, 387– 402.
D.J. and Muir, W.J. (2001) Schizophrenia and affective disorders— 23. Hikida, T., Jaaro-Peled, H., Seshadri, S., Oishi, K., Hookway, C., Kong,
cosegregation with a translocation at chromosome 1q42 that directly
S., Wu, D., Xue, R., Andrade, M., Tankou, S. et al. (2007)
disrupts brain-expressed genes: clinical and P300 findings in a family.
Dominant-negative DISC1 transgenic mice display
Am. J. Hum. Genet., 69, 428–433.
schizophrenia-associated phenotypes detected by measures translatable to
6. Hodgkinson, C.A., Goldman, D., Jaeger, J., Persaud, S., Kane, J.M.,
humans. Proc. Natl Acad. Sci. USA, 104, 14501–14506.
Lipsky, R.H. and Malhotra, A.K. (2004) Disrupted in Schizophrenia 1
(DISC1): association with schizophrenia, schizoaffective disorder, and 24. Li, W., Zhou, Y., Jentsch, J.D., Brown, R.A., Tian, X., Ehninger, D.,
bipolar disorder. Am. J. Hum. Genet., 75, 862 –872. Hennah, W., Peltonen, L., Lonnqvist, J., Huttunen, M.O. et al. (2007)
7. Callicott, J.H., Straub, R.E., Pezawas, L., Egan, M.F., Mattay, V.S., Specific developmental disruption of Disrupted-In-Schizophrenia-1
Hariri, A.R., Verchinski, B.A., Meyer-Lindenberg, A., Balkissoon, R., function results in schizophrenia-related phenotypes in mice. Proc. Natl
Kolachana, B. et al. (2005) Variation in DISC1 affects hippocampal Acad. Sci. USA, 104, 18280–18285.
structure and function and increases risk for schizophrenia. Proc. Natl 25. Pletnikov, M.V., Ayhan, Y., Nikolskaia, O., Xu, Y., Ovanesov, M.V.,
Acad. Sci. USA, 102, 8627–8632. Huang, H., Mori, S., Moran, T.H. and Ross, C.A. (2008) Inducible
8. Wang, Q., Charych, E.I., Pulito, V.L., Lee, J.B., Graziane, N.M., Crozier, expression of mutant human DISC1 in mice is associated with brain and
R.A., Revilla-Sanchez, R., Kelly, M.P., Dunlop, A.J., Murdoch, H. et al. behavioral abnormalities reminiscent of schizophrenia. Mol. Psychiatry,
(2010) The psychiatric disease risk factors DISC1 and TNIK interact to 13, 173– 186, 115.
regulate synapse composition and function. Mol. Psychiatry., 10.1038/ 26. Shen, S., Lang, B., Nakamoto, C., Zhang, F., Pu, J., Kuan, S.L., Chatzi, C.,
mp.2010.87 He, S., Mackie, I., Brandon, N.J. et al. (2008) Schizophrenia-related
9. Camargo, L.M., Collura, V., Rain, J.C., Mizuguchi, K., Hermjakob, H., neural and behavioral phenotypes in transgenic mice expressing truncated
Kerrien, S., Bonnert, T.P., Whiting, P.J. and Brandon, N.J. (2007) Disc1. J. Neurosci., 28, 10893– 10904.
4682 Human Molecular Genetics, 2011, Vol. 20, No. 23

27. Koike, H., Arguello, P.A., Kvajo, M., Karayiorgou, M. and Gogos, J.A. maturation in the frontal cortex and leads to adult behavioral deficits.
(2006) Disc1 is mutated in the 129S6/SvEv strain and modulates working Neuron, 65, 480–489.
memory in mice. Proc. Natl Acad. Sci. USA, 103, 3693–3697. 46. Park, Y.U., Jeong, J., Lee, H., Mun, J.Y., Kim, J.H., Lee, J.S., Nguyen,
28. Kvajo, M., McKellar, H., Arguello, P.A., Drew, L.J., Moore, H., M.D., Han, S.S., Suh, P.G. and Park, S.K. (2010)
MacDermott, A.B., Karayiorgou, M. and Gogos, J.A. (2008) A mutation Disrupted-in-schizophrenia 1 (DISC1) plays essential roles in
in mouse Disc1 that models a schizophrenia risk allele leads to specific mitochondria in collaboration with Mitofilin. Proc. Natl Acad. Sci. USA,
alterations in neuronal architecture and cognition. Proc. Natl Acad. Sci. 107, 17785–17790.
USA, 105, 7076–7081. 47. Faulkner, R.L., Jang, M.H., Liu, X.B., Duan, X., Sailor, K.A., Kim, J.Y.,
29. Ishizuka, K., Chen, J., Taya, S., Li, W., Millar, J.K., Xu, Y., Clapcote, Ge, S., Jones, E.G., Ming, G.L., Song, H. et al. (2008) Development of
S.J., Hookway, C., Morita, M., Kamiya, A. et al. (2007) Evidence that hippocampal mossy fiber synaptic outputs by new neurons in the adult
many of the DISC1 isoforms in C57BL/6J mice are also expressed in brain. Proc. Natl Acad. Sci. USA, 105, 14157–14162.
129S6/SvEv mice. Mol. Psychiatry, 12, 897– 899. 48. Smith, D.S., Niethammer, M., Ayala, R., Zhou, Y., Gambello, M.J.,
30. Taylor, M.S., Devon, R.S., Millar, J.K. and Porteous, D.J. (2003) Wynshaw-Boris, A. and Tsai, L.H. (2000) Regulation of cytoplasmic
Evolutionary constraints on the Disrupted in Schizophrenia locus. dynein behaviour and microtubule organization by mammalian Lis1. Nat.
Genomics, 81, 67–77. Cell Biol., 2, 767 –775.

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


31. Bord, L., Wheeler, J., Paek, M., Saleh, M., Lyons-Warren, A., Ross, C.A., 49. Sasaki, S., Mori, D., Toyo-oka, K., Chen, A., Garrett-Beal, L.,
Sawamura, N. and Sawa, A. (2006) Primate Disrupted-In-Schizophrenia-1 Muramatsu, M., Miyagawa, S., Hiraiwa, N., Yoshiki, A., Wynshaw-Boris,
(DISC1): high divergence of a gene for major mental illnesses in recent A. et al. (2005) Complete loss of Ndel1 results in neuronal migration
evolutionary history. Neurosci. Res., 56, 286–293. defects and early embryonic lethality. Mol. Cell Biol., 25, 7812– 7827.
32. Nakata, K., Lipska, B.K., Hyde, T.M., Ye, T., Newburn, E.N., Morita, Y., 50. Millar, J.K., Christie, S. and Porteous, D.J. (2003) Yeast two-hybrid
Vakkalanka, R., Barenboim, M., Sei, Y., Weinberger, D.R. et al. (2009) screens implicate DISC1 in brain development and function. Biochem.
DISC1 splice variants are upregulated in schizophrenia and associated Biophys. Res. Commun., 311, 1019–1025.
with risk polymorphisms. Proc. Natl Acad. Sci. USA, 106, 15873–15878. 51. James, R., Adams, R.R., Christie, S., Buchanan, S.R., Porteous, D.J. and
33. Callicott, J.H., Bertolino, A., Mattay, V.S., Langheim, F.J., Duyn, J., Millar, J.K. (2004) Disrupted in Schizophrenia 1 (DISC1) is a
Coppola, R., Goldberg, T.E. and Weinberger, D.R. (2000) Physiological multicompartmentalized protein that predominantly localizes to
dysfunction of the dorsolateral prefrontal cortex in schizophrenia mitochondria. Mol. Cell Neurosci., 26, 112–122.
revisited. Cereb. Cortex, 10, 1078–1092. 52. Millar, J.K., James, R., Christie, S. and Porteous, D.J. (2005) Disrupted in
34. Meyer-Lindenberg, A. (2010) From maps to mechanisms through schizophrenia 1 (DISC1): subcellular targeting and induction of ring
neuroimaging of schizophrenia. Nature, 468, 194– 202. mitochondria. Mol. Cell Neurosci., 30, 477–484.
35. Clapcote, S.J. and Roder, J.C. (2006) Deletion polymorphism of Disc1 is 53. Abraham, W.C. and Bear, M.F. (1996) Metaplasticity: the plasticity of
common to all 129 mouse substrains: implications for gene-targeting synaptic plasticity. Trends Neurosci., 19, 126 –130.
studies of brain function. Genetics, 173, 2407–2410. 54. Panatier, A., Theodosis, D.T., Mothet, J.P., Touquet, B., Pollegioni, L.,
36. Austin, C.P., Ky, B., Ma, L., Morris, J.A. and Shughrue, P.J. (2004) Poulain, D.A. and Oliet, S.H. (2006) Glia-derived D-serine controls
Expression of Disrupted-In-Schizophrenia-1, a schizophrenia-associated NMDA receptor activity and synaptic memory. Cell, 125, 775 –784.
gene, is prominent in the mouse hippocampus throughout brain 55. Chen, L., Cai, W., Zhou, R., Furuya, K. and Sokabe, M. (2010)
development. Neuroscience, 124, 3 –10. Modulatory metaplasticity induced by pregnenolone sulfate in the rat
37. Austin, C.P., Ma, L., Ky, B., Morris, J.A. and Shughrue, P.J. (2003) hippocampus: a leftward shift in LTP/LTD-frequency curve.
DISC1 (Disrupted in Schizophrenia-1) is expressed in limbic regions of Hippocampus, 20, 499–512.
the primate brain. Neuroreport, 14, 951–954. 56. Ikegaya, Y., Ishizaka, Y. and Matsuki, N. (2002) BDNF attenuates
38. Honda, A., Miyoshi, K., Baba, K., Taniguchi, M., Koyama, Y., Kuroda, hippocampal LTD via activation of phospholipase C: implications for a
S., Katayama, T. and Tohyama, M. (2004) Expression of fasciculation and vertical shift in the frequency-response curve of synaptic plasticity.
elongation protein zeta-1 (FEZ1) in the developing rat brain. Brain Res. Eur. J. Neurosci., 16, 145–148.
Mol. Brain Res., 122, 89– 92. 57. Bendikov, I., Nadri, C., Amar, S., Panizzutti, R., De Miranda, J.,
39. Ma, L., Liu, Y., Ky, B., Shughrue, P.J., Austin, C.P. and Morris, J.A. Wolosker, H. and Agam, G. (2007) A CSF and postmortem brain study of
(2002) Cloning and characterization of Disc1, the mouse ortholog of D-serine metabolic parameters in schizophrenia. Schizophr. Res., 90,
DISC1 (Disrupted-in-Schizophrenia 1). Genomics, 80, 662– 672. 41– 51.
40. Kamiya, A., Kubo, K., Tomoda, T., Takaki, M., Youn, R., Ozeki, Y., 58. Yamada, K., Ohnishi, T., Hashimoto, K., Ohba, H., Iwayama-Shigeno, Y.,
Sawamura, N., Park, U., Kudo, C., Okawa, M. et al. (2005) A Toyoshima, M., Okuno, A., Takao, H., Toyota, T., Minabe, Y. et al.
schizophrenia-associated mutation of DISC1 perturbs cerebral cortex (2005) Identification of multiple serine racemase (SRR) mRNA isoforms
development. Nat. Cell Biol., 7, 1167–1178. and genetic analyses of SRR and DAO in schizophrenia and D-serine
41. Hashimoto, R., Numakawa, T., Ohnishi, T., Kumamaru, E., Yagasaki, Y., levels. Biol. Psychiatry, 57, 1493–1503.
Ishimoto, T., Mori, T., Nemoto, K., Adachi, N., Izumi, A. et al. (2006) 59. Ohnuma, T., Sakai, Y., Maeshima, H., Hatano, T., Hanzawa, R., Abe, S.,
Impact of the DISC1 Ser704Cys polymorphism on risk for major Kida, S., Shibata, N., Suzuki, T. and Arai, H. (2008) Changes in plasma
depression, brain morphology and ERK signaling. Hum. Mol. Genet., 15, glycine, L-serine, and D-serine levels in patients with schizophrenia as
3024– 3033. their clinical symptoms improve: results from the Juntendo University
42. Duan, X., Chang, J.H., Ge, S., Faulkner, R.L., Kim, J.Y., Kitabatake, Y., Schizophrenia Projects (JUSP). Prog. Neuropsychopharmacol. Biol.
Liu, X.B., Yang, C.H., Jordan, J.D., Ma, D.K. et al. (2007) Psychiatry, 32, 1905–1912.
Disrupted-In-Schizophrenia 1 regulates integration of newly generated 60. Thompson Ray, M., Weickert, C.S., Wyatt, E. and Webster, M.J. (2011)
neurons in the adult brain. Cell, 130, 1146– 1158. Decreased BDNF, trkB-TK+ and GAD(67) mRNA expression in the
43. Kamiya, A., Tan, P.L., Kubo, K., Engelhard, C., Ishizuka, K., Kubo, A., hippocampus of individuals with schizophrenia and mood disorders.
Tsukita, S., Pulver, A.E., Nakajima, K., Cascella, N.G. et al. (2008) J. Psychiatry Neurosci., 36, 100048.
Recruitment of PCM1 to the centrosome by the cooperative action of 61. Buckley, P.F., Pillai, A. and Howell, K.R. (2011) Brain-derived
DISC1 and BBS4: a candidate for psychiatric illnesses. Arch. Gen. neurotrophic factor: findings in schizophrenia. Curr. Opin. Psychiatry, 24,
Psychiatry, 65, 996–1006. 122–127.
44. Kubo, K., Tomita, K., Uto, A., Kuroda, K., Seshadri, S., Cohen, J., 62. Ritsner, M.S. (2010) Pregnenolone, dehydroepiandrosterone, and
Kaibuchi, K., Kamiya, A. and Nakajima, K. (2010) Migration defects by schizophrenia: alterations and clinical trials. CNS Neurosci. Ther., 16,
DISC1 knockdown in C57BL/6, 129X1/SvJ, and ICR strains via in utero 32– 44.
gene transfer and virus-mediated RNAi. Biochem. Biophys. Res. 63. Iancu, I., Bodner, E., Roitman, S., Piccone Sapir, A., Poreh, A. and Kotler,
Commun., 400, 631–637. M. (2010) Impulsivity, aggression and suicide risk among male
45. Niwa, M., Kamiya, A., Murai, R., Kubo, K., Gruber, A.J., Tomita, K., Lu, schizophrenia patients. Psychopathology, 43, 223–229.
L., Tomisato, S., Jaaro-Peled, H., Seshadri, S. et al. (2010) Knockdown of 64. Nestor, P.G. (2002) Mental disorder and violence: personality dimensions
DISC1 by in utero gene transfer disturbs postnatal dopaminergic and clinical features. Am. J. Psychiatry, 159, 1973–1978.
Human Molecular Genetics, 2011, Vol. 20, No. 23 4683

65. McEvoy, J.P., Lieberman, J.A., Stroup, T.S., Davis, S.M., Meltzer, H.Y., 76. Inagaki, N., Chihara, K., Arimura, N., Menager, C., Kawano, Y., Matsuo,
Rosenheck, R.A., Swartz, M.S., Perkins, D.O., Keefe, R.S., Davis, C.E. N., Nishimura, T., Amano, M. and Kaibuchi, K. (2001) CRMP-2 induces
et al. (2006) Effectiveness of clozapine versus olanzapine, quetiapine, and axons in cultured hippocampal neurons. Nat. Neurosci., 4, 781– 782.
risperidone in patients with chronic schizophrenia who did not respond to 77. Mizoguchi, A., Nakanishi, H., Kimura, K., Matsubara, K., Ozaki-Kuroda,
prior atypical antipsychotic treatment. Am. J. Psychiatry, 163, 600– 610. K., Katata, T., Honda, T., Kiyohara, Y., Heo, K., Higashi, M. et al. (2002)
66. Swerdlow, N.R., Hartman, P.L. and Auerbach, P.P. (1997) Changes in Nectin: an adhesion molecule involved in formation of synapses. J. Cell
sensorimotor inhibition across the menstrual cycle: implications for Biol., 156, 555–565.
neuropsychiatric disorders. Biol. Psychiatry, 41, 452– 460. 78. Hanse, E. and Gustafsson, B. (1992) Postsynaptic, but not presynaptic,
67. McEwen, B.S. and Alves, S.E. (1999) Estrogen actions in the central activity controls the early time course of long-term potentiation in the
nervous system. Endocr. Rev., 20, 279– 307. dentate gyrus. J. Neurosci., 12, 3226–3240.
68. Chavez, C., Hollaus, M., Scarr, E., Pavey, G., Gogos, A. and van den 79. Ibi, D., Nagai, T., Kitahara, Y., Mizoguchi, H., Koike, H., Shiraki, A.,
Buuse, M. (2010) The effect of estrogen on dopamine and serotonin Takuma, K., Kamei, H., Noda, Y., Nitta, A. et al. (2009) Neonatal polyI:C
receptor and transporter levels in the brain: an autoradiography study. treatment in mice results in schizophrenia-like behavioral and
Brain Res., 1321, 51–59. neurochemical abnormalities in adulthood. Neurosci. Res., 64, 297–305.
69. Braff, D.L., Geyer, M.A. and Swerdlow, N.R. (2001) Human studies of 80. Ibi, D., Nagai, T., Koike, H., Kitahara, Y., Mizoguchi, H., Niwa, M.,
Jaaro-Peled, H., Nitta, A., Yoneda, Y., Nabeshima, T. et al. (2010)

Downloaded from https://academic.oup.com/hmg/article/20/23/4666/559138 by guest on 09 August 2021


prepulse inhibition of startle: normal subjects, patient groups, and
pharmacological studies. Psychopharmacology (Berl.), 156, 234 –258. Combined effect of neonatal immune activation and mutant DISC1 on
70. Ayhan, Y., Abazyan, B., Nomura, J., Kim, R., Ladenheim, B., Krasnova, phenotypic changes in adulthood. Behav. Brain Res., 206, 32– 37.
81. Koike, H., Ibi, D., Mizoguchi, H., Nagai, T., Nitta, A., Takuma, K.,
I.N., Sawa, A., Margolis, R.L., Cadet, J.L., Mori, S. et al. (2011)
Nabeshima, T., Yoneda, Y. and Yamada, K. (2009) Behavioral
Differential effects of prenatal and postnatal expressions of mutant human
abnormality and pharmacologic response in social isolation-reared mice.
DISC1 on neurobehavioral phenotypes in transgenic mice: evidence for
Behav. Brain Res., 202, 114–121.
neurodevelopmental origin of major psychiatric disorders. Mol.
82. Nagai, T., Takuma, K., Kamei, H., Ito, Y., Nakamichi, N., Ibi, D.,
Psychiatry, 16, 293 –306.
Nakanishi, Y., Murai, M., Mizoguchi, H., Nabeshima, T. et al. (2007)
71. Lipina, T.V., Niwa, M., Jaaro-Peled, H., Fletcher, P.J., Seeman, P., Sawa, Dopamine D1 receptors regulate protein synthesis-dependent long-term
A. and Roder, J.C. (2010) Enhanced dopamine function in DISC1-L100P recognition memory via extracellular signal-regulated kinase 1/2 in the
mutant mice: implications for schizophrenia. Genes Brain Behav., 9, 777– prefrontal cortex. Learn Mem., 14, 117–125.
789. 83. Miyakawa, T., Leiter, L.M., Gerber, D.J., Gainetdinov, R.R., Sotnikova,
72. Remington, G. (2008) Alterations of dopamine and serotonin transmission T.D., Zeng, H., Caron, M.G. and Tonegawa, S. (2003) Conditional
in schizophrenia. Prog. Brain Res., 172, 117 –140. calcineurin knockout mice exhibit multiple abnormal behaviors related to
73. Hashimoto, H., Shintani, N., Tanaka, K., Mori, W., Hirose, M., Matsuda, schizophrenia. Proc. Natl Acad. Sci. USA, 100, 8987– 8992.
T., Sakaue, M., Miyazaki, J., Niwa, H., Tashiro, F. et al. (2001) Altered 84. Matsuoka, Y., Furuyashiki, T., Yamada, K., Nagai, T., Bito, H., Tanaka,
psychomotor behaviors in mice lacking pituitary adenylate Y., Kitaoka, S., Ushikubi, F., Nabeshima, T. and Narumiya, S. (2005)
cyclase-activating polypeptide (PACAP). Proc. Natl Acad. Sci. USA, 98, Prostaglandin E receptor EP1 controls impulsive behavior under stress.
13355– 13360. Proc. Natl Acad. Sci. USA, 102, 16066– 16071.
74. Yagi, T., Tokunaga, T., Furuta, Y., Nada, S., Yoshida, M., Tsukada, T., 85. Miyakawa, T., Yamada, M., Duttaroy, A. and Wess, J. (2001)
Saga, Y., Takeda, N., Ikawa, Y. and Aizawa, S. (1993) A novel ES cell Hyperactivity and intact hippocampus-dependent learning in mice
line, TT2, with high germline-differentiating potency. Anal. Biochem., lacking the M1 muscarinic acetylcholine receptor. J. Neurosci., 21,
214, 70–76. 5239–5250.
75. Kiyonari, H., Kaneko, M., Abe, S. and Aizawa, S. (2010) Three inhibitors 86. Miyamoto, Y., Yamada, K., Noda, Y., Mori, H., Mishina, M. and
of FGF receptor, ERK, and GSK3 establishes germline-competent Nabeshima, T. (2002) Lower sensitivity to stress and altered
embryonic stem cells of C57BL/6N mouse strain with high efficiency and monoaminergic neuronal function in mice lacking the NMDA receptor
stability. Genesis, 48, 317– 327. epsilon 4 subunit. J. Neurosci., 22, 2335– 2342.

You might also like