Mouse Models of Autism Spectrum Disorders: The Challenge for Behavioral Genetics

Page created by Debbie Cruz
 
CONTINUE READING
American Journal of Medical Genetics Part C (Semin. Med. Genet.) 142C:40 –51 (2006)

 A R T I C L E

Mouse Models of Autism Spectrum Disorders:
The Challenge for Behavioral Genetics
SHERYL S. MOY,* JESSICA J. NADLER, TERRY R. MAGNUSON, AND JACQUELINE N. CRAWLEY

               Autism is a severe neurodevelopmental disorder, which typically emerges early in childhood. The core symptoms
               of autism include deficits in social interaction, impaired communication, and aberrant repetitive behavior,
               including self-injury. Despite the strong genetic component for the disease, most cases of autism have not been
               linked to mutations in a specific gene, and the etiology of the disorder has yet to be established. At the present
               time, there is no generally accepted therapeutic strategy to treat the core symptoms of autism, and there remains
               a critical need for appropriate animal models and relevant behavioral assays to promote the understanding and
               treatment of the clinical syndrome. Challenges for the development of valid mouse models include complex
               genetic interactions underlying the high heritability of the disease in humans, diagnosis based on deficits in social
               interaction and communication, and the lack of confirmatory neuropathological markers to provide validation for
               genetic models of the disorder. Research focusing on genes that mediate social behavior in mice may help identify
               neural circuitry essential for normal social interaction, and lead to novel genetic animal models of the autism
               behavioral phenotype. ß 2006 Wiley-Liss, Inc.

KEY WORDS: autism; fragile X; mice; repetitive behavior; Rett syndrome; social interaction

INTRODUCTION                                       challenging. For example, the diagnostic          AUTISM CLINICAL
                                                   criteria for autism are based on social           DISORDER
Advances in the fields of genetics and             and communication deficits, rather than a
molecular biology have led to the                  general neurological pathology or neu-            Diagnostic and Neuropathological
creation of mouse models with genetic              rochemical marker that could be model-            Features
aberrations characteristic of human clin-          ed in an animal. In addition, the ASDs are
                                                                                                     Diagnosis of autism is based on beha-
ical disorders. These models can be                characterized by marked genetic and
                                                                                                     vioral symptomatology. The defining
tremendously beneficial for determining            phenotypic heterogeneity, suggesting
                                                                                                     characteristics include attenuated or
disease etiology, effects on organic and           that single-gene mouse mutants may not
                                                                                                     abnormal social interaction and com-
cellular function, and therapeutic efficacy        provide a model for the intrinsic com-            munication, as well as aberrant repetitive
of novel treatment strategies. However,            plexity of these diseases. This review
                                                                                                     behavior, with symptoms emerging
the development of mouse models for                describes current genetic mouse models
                                                                                                     early in childhood [American Psychia-
the autism spectrum disorders (ASDs),              for the ASDs, and discusses methodolog-
                                                                                                     tric Association, 1994]. In addition to
including autism, Asperger disorder, and           ical and theoretical issues in their beha-
                                                                                                     significant language deficits, autistic
Rett syndrome, has proven especially               vioral phenotyping and use.
                                                                                                     children demonstrate rigid adherence
                                                                                                     to routines and restricted interests, often
                                                                                                     forming obsessional preoccupations with
   Sheryl S. Moy, Ph.D., is the Associate Director of the Mouse Behavioral Phenotyping Laboratory    specific objects or topics. The autism
of the Neurodevelopmental Disorders Research Center, and an Associate Professor in the               phenotype also includes unusual motor-
Department of Psychiatry at UNC. Dr. Moy’s work focuses on the development of mouse models
relevant to human clinical disorders, including autism and schizophrenia.
                                                                                                     ic responses, such as tic-like stereotypies
   Jessica J. Nadler, Ph.D., is a Postdoctoral Fellow in the Department of Genetics at UNC.          and self-injury [Bodfish et al., 2000],
Dr. Nadler works on identifying the underlying gene expression changes associated with deficits      impaired motor function, and abnormal
in social and cognitive behaviors.
   Terry R. Magnuson, Ph.D., is the Chair of the UNC Department of Genetics and Director of the
                                                                                                     responses to sensory stimuli [Baranek,
Carolina Center for Genome Sciences.                                                                 2002]. Mental retardation is observed in
   Jacqueline N. Crawley, Ph.D., is Director of the Laboratory of Behavioral Neuroscience at NIMH    the majority of children diagnosed with
and the Mouse Behavioral Phenotyping Laboratory of the Neurodevelopmental Disorders
Research Center at UNC. Modeling neuropsychiatric disorders using mouse behavioral genetics
                                                                                                     autism, and seizures are present in an
represents a theme of Dr. Crawley’s research program.                                                estimated 30% of the autistic population
   Grant sponsor: STAART; Grant number: U54 MH66418; Grant sponsor: MRDDRC; Grant                    [Gillberg and Billstedt, 2000]. Other
number: P30 HD03110; Grant sponsor: NIMH Intramural Research Program.
   *Correspondence to: Sheryl S. Moy, Neurodevelopmental Disorders Research Center, CB no.
                                                                                                     symptoms reported in autism include
7146, University of North Carolina, Chapel Hill, NC 27599-7146. E-mail: ssmoy@med.unc.edu            anxiety [Muris et al., 1998], and sleep
   DOI 10.1002/ajmg.c.30081                                                                          disorders [Gillberg and Billstedt, 2000].

ß 2006 Wiley-Liss, Inc.
ARTICLE                             AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c            41

      Investigators have reported that        polymorphisms for the disease conferr-       protein. Mouse models based on tar-
autistic subjects may have various altera-    ing susceptibility in a non-deterministic    geted disruption of genes that may be
tions in brain size and neuroanatomy.         way [Folstein and Rosen-Sheidley,            involved in autism susceptibility include
These differences include increased head      2001; Keller and Persico, 2003].             the Fmr1-null mouse, a model for fragile
size and brain volume [Piven et al., 1996;                                                 X syndrome [Bakker et al., 1994],
Bailey et al., 1998; Courchesne et al.,                                                    Gabrb3- and Ube3a-null mice, models
                                              MOUSE MODELS FOR
2001], decreased volume of hippocam-                                                       for Angelman syndrome [DeLorey et al.,
                                              AUTISM SPECTRUM
pus, amygdala, and corpus callosum                                                         1998; Jiang et al., 1998], the DHCR7-
                                              DISORDERS
[Piven et al., 1997; Aylward et al.,                                                       null mouse, a model for Smith–Lemli–
1999; Saitoh et al., 2001], and decreased                                                  Optiz syndrome [Fitzky et al., 2001],
                                              Candidate Gene Approach
numbers of Purkinje cells in cerebellum                                                    and the Dvl1-null mouse [Lijam et al.,
[Bailey et al., 1998; Palmen et al., 2004].   With the advent of embryonic stem cell       1997] (Table I).
Neuroimaging assessment of regional           technology, mouse geneticists are able to         While a targeted disruption can pro-
brain function has demonstrated defic-        generate mouse models carrying the           vide clues on the role of the protein in
ient or abnormal activity in frontal and      targeted disruption or mutation of           normal brain development and func-
temporal cortex [Happé et al., 1996;         specific candidate genes. However, in        tion, a serious drawback of this approach
Schultz et al., 2000]. Unfortunately,         the case of the ASDs, determination of       is that the null allele may not be
many of the findings on neuropatholo-         which genes to target has proven             consistent with the allele present in the
gical changes in autism have not proven       problematic. Research in human popu-         ASD population. A second approach for
to be consistent across studies, or have      lations suggests that the number of loci     creating relevant animal models is to
unknown functional ramifications, such        associated with autism exceeds 15            engineer an allele known to be disease-
as the behavioral consequences of in-         [Risch et al., 1999]. Most mouse models      associated in humans. This modification
creased brain size.                           for the ASDs have been based on              may be more likely to mimic the
                                              manipulations of loci that mediate           molecular nature of the disease. How-
                                              single-gene human disorders character-       ever, there are often several disease-
Genetic Component of Autism
                                              ized by autistic symptoms, or that have      associated alleles for a given disorder, and
While the etiology of autism is not yet       been identified through association or       it can be unclear which allele to
known, support for a strong genetic           linkage studies in human genetics. Other     generate. The mouse models of ASDs
component is evident from the 70% to          mouse models involve mutations in path-      discussed below represent both targeted-
80% concordance between monozygo-             ways thought to be altered in autism.        disruption and disease-associated types
tic twins [Folstein and Rosen-Sheidley,                                                    of approaches and it is important to
2001]. Autism occurs about four times                                                      understand which type of allele is being
more frequently in males than in females,                                                  investigated when evaluating the validity
although this rate varies within subpop-         Most mouse models for the                 of the model.
ulations [Fombonne, 2002; Keller and             ASDs have been based on
Persico, 2003]. Association studies and
                                                 manipulations of loci that                Mouse Model for
familial linkage analyses have been
                                                mediate single-gene human                  Fragile X Syndrome
used to identify various candidate genes,
including WNT2 [Wassink et al., 2001],           disorders characterized by                Fragile X syndrome is characterized by
GABRB3 [Buxbaum et al., 2002],                                                             mental retardation, physical abnormal-
UBE3A [Nurmi et al., 2001], and                  autistic symptoms, or that                ities, and, in most cases, autistic-like
RELN [Persico et al., 2001]. The autism         have been identified through               behavior [Hagerman et al., 1986]. The
phenotype is also associated with muta-                                                    disease was mapped to a single locus on
tions in FMR1 (the underlying abnorm-
                                              association or linkage studies in            the X chromosome, Fragile X Mental
ality in fragile X syndrome; [Hagerman         human genetics. Other mouse                 Retardation 1 (FMR1). The most com-
et al., 1986]) and MECP2 (the genetic           models involve mutations in                mon allele found in fragile X patients is
basis for Rett disorder; [Shibayama et al.,                                                an expansion of a trinucleotide repeat
2004]). However, the percentage of            pathways thought to be altered               [Kremer et al., 1991]. In a normal FMR1
autism cases that can be linked to a                      in autism.                       allele, there are about 40 repeats of a
known genetic aberration is only around                                                    CCG nucleotide motif. In patients with
10% (e.g., Barton and Volkmar [1998]).                                                     fragile X, the number of these repeats is
Overall, studies suggest the autism           The most common strategy for genetic         dramatically increased, leading to the
phenotype cannot be attributed to a           modification is to generate a null allele,   severe reduction of FMR1 protein
single gene. Rather, the clinical syn-        or targeted disruption, which deletes a      (FMRP) expression. It is not known
drome results from multiple genes inter-      portion of the locus, and generally          how the loss of FMRP in brain leads to
acting together, with some genetic            prevents the production of functional        the deficits observed in the disease, but
42       AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c                                   ARTICLE

                                  TABLE I. Genes Linked to Autism and the Relevant Mouse Models

     Candidate gene                                                                   Genetic mouse model
     Genes for neurodevelopmental disorders associated with autism
       FMR1 (Fragile X syndrome)                                                      Fmr1-null mouse
       MECP2 (Rett syndrome)                                                          Mecp2-null mouse, Mecp2308/Y mouse
       GABRB3, UBE3A (Angelman’s syndrome)                                            Gabrb3-null mouse
                                                                                      Ube3a-null mouse
       NF1 (Neurofibromatosis type 1)                                                 Nf1-null, heterozygous, and NF123a/ mouse
       DHCR7 (Smith–Lemli–Opitz syndrome)                                             Dhcr7-null and heterozygous mouse
       TSC1, TSC2 (Tuberous sclerosis)                                                Tsc1-null mouse
     Other genes that may contribute to autism susceptibility
       WNT2                                                                           Wnt2-null mouse
                                                                                      Dvl1 (Dishevelled-1)-null mouse
       EN2                                                                            En1 and En2 (Engrailed) null mouse
       HOXA1, HOXA2                                                                   Hoxa1-null mouse
       SERT (SLC6A4)                                                                  Sert-null and heterozygous mouse
       MAOA                                                                           Maoa-null mouse
       RELN                                                                           Reln-null and heterozygous mouse

recent work suggests that normal FMRP                                                          normal performance is observed when
                                                     including altered dendritic
plays a role in RNA binding and                                                                the same mutation occurs on a C57BL/
regulating its translation [Oostra and                 spine morphology and                    6J background [Dobkin et al., 2000; see
Chiurazzi, 2001; Darnell et al., 2005].             macroorchidism, similar to                 also Paradee et al., 1999]. These differ-
     The Fmr1-null mouse is one of the                                                         ences suggest that there are multiple
best characterized animal models for               changes in fragile X patients.              genetic interactions, which can modu-
human disorders associated with autism.                                                        late complex behavioral phenotypes,
                                                                                               even in a defined, single-locus disease.
                                                  Comery et al., 1997]. The behavioral
 The Fmr1-null mouse is one                       phenotype of the fragile X-model mice
                                                                                               Mouse Models for Rett Syndrome
of the best characterized animal                  also reflects symptoms associated with
                                                  the human disorder. The null mice            Like fragile X syndrome, Rett syndrome
  models for human disorders                      show cognitive deficits, evidenced by        maps to the X chromosome. The disease
     associated with autism.                      impaired performance on reversal learn-      is observed primarily in females, with
                                                  ing in the Morris water maze [Bakker         symptoms including mental retardation,
                                                  et al., 1994; Kooy et al., 1996], and        slowed growth rate, hypoactivity, and
While the targeted disruption in this             altered sensitivity to acoustic stimuli      autistic-like behavior. Mutations in a
model is not the same as the common               [Chen and Toth, 2001; Nielson et al.,        single gene, methyl-CpG-binding pro-
human allele at the DNA level, the con-           2002; Frankland et al., 2004]. The mice      tein-2 (MECP2), account for the major-
sequence is likely similar: severe reduction      also demonstrate changes in social beha-     ity of Rett syndrome cases [Amir et al.,
or absence of the FMR1 gene product               vior, suggestive of enhanced social          1999]. As its name implies, MECP2
(see Yan et al. [2004]). FMRP-deficient           anxiety [Spencer et al., 2005]. However,     protein binds to methylated CpG islands
mice demonstrate cytoarchitectonic and            some studies have found normal beha-         in genomic DNA. MECP2 also associ-
physiological aberrations, including al-          vioral responses after disruption of the     ates with a protein complex containing
tered dendritic spine morphology and              Fmr1 gene [Fisch et al., 1999; Peier         histone deacetylase 1. Since DNA
macroorchidism, similar to changes in             et al., 2000; Nielson et al., 2002; Yan      methylation and histone deacetylation
fragile X patients [Bakker et al., 1994;          et al., 2004]. These differences may be      are two global methods of transcriptional
                                                  attributed, in part, to the effect of the    repression, the MECP2 protein con-
                                                  different genetic backgrounds of the         nects two major methods of down-
    FMRP-deficient mice                           mouse strains used for the Fmr1-null         regulating gene expression throughout
                                                  mice. In particular, the Fmr1-null allele    the mammalian genome [Nan et al.,
demonstrate cytoarchitectonic                     on a mixed FVB/129 background leads          1998]. Alleles found in the patient
and physiological aberrations,                    to deficits in spatial learning, while       population include missense mutations,
ARTICLE                            AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c           43

which cause amino acid changes in the        mice show increased anxiety-like beha-       gous for the Gabrb3 deletion have
MECP2 protein [Amir et al., 1999; Wan        vior, stereotypic forelimb movements         significantly reduced levels of the beta
et al., 1999], and nonsense mutations        [Shahbazian et al., 2002], and altered       3 subunit in brain, dependent upon both
that result in production of a truncated     home-cage and social behavior [Moretti       the parent of origin and gender, but do
protein [Zappella et al., 2001]. Early       et al., 2005]. In particular, Mecp2308/Y     not show seizures or hyperactivity [Lil-
truncation often leads to a classical Rett   mice spend less time investigating an        jelund et al., 2005]. Ube3a-null mice
syndrome phenotype, while later trun-        unfamiliar mouse, in comparison to           demonstrate poor rotarod performance,
cations are associated with the preserved    normal mice, suggesting that mutation        deficits in spatial learning in a Morris
speech variant of the disease [Zappella      of the Mecp2 locus can lead to deficits in   water maze task and in conditioned fear,
et al., 2001].                               social interaction [Moretti et al., 2005].   and enhanced susceptibility for seizures,
      Mouse models of Rett syndrome          Interestingly, a recent study has demon-     dependent upon maternal transmission
based on a null allele of the Mecp2          strated a link between deficient MECP2       of the mutation [Jiang et al., 1998; Miura
locus demonstrate a phenotype con-           expression in human brain or in mouse        et al., 2002].
sistent with the human disease. The          models, and expression deficits in                 Mortality early in development is an
mice exhibit normal development until        UBE3A and GABRB3, two genes                  issue for other mouse models relevant
about 5 weeks of age, followed by the        linked to Angelman syndrome [Samaco          to the ASDs. For example, gestational
emergence of motor impairment, tre-          et al., 2005].                               death is found with the Nf1-null mouse
mors, hypoactivity, aberrant hind-                                                        model for neurofibromatosis type 1
limb clasping, and respiratory symptoms                                                   (NF1), an autosomal dominant disorder
                                                                                          characterized by cognitive and language
                                             Mouse Models for Other
                                                                                          deficits, poor motor skills, and tumors of
                                             Disorders Associated
                                                                                          the peripheral nerves [Silva et al., 1997;
Mouse models of Rett syndrome                With Autism
                                                                                          North, 2000]. However, behavioral
  based on a null allele of the              Angelman syndrome is a severe neuro-         testing of heterozygotes has revealed that
                                             developmental disorder with clinical         Nf1þ/ mice demonstrate deficits in
  Mecp2 locus demonstrate a
                                             features of mental retardation, deficits     spatial learning in the Morris water
phenotype consistent with the                in language, movement disorder, and          maze, as well as an enhanced predisposi-
   human disease. The mice                   seizures [Williams, 2005]. The disease       tion for tumors [Silva et al., 1997; Costa
                                             has a complex genetic etiology, invol-       et al., 2002]. Further, mice carrying a
 exhibit normal development                  ving deletions in chromosome 15q11, an       specific allele of Nf1 show deficient
  until about 5 weeks of age,                area containing the GABAA receptor           social transmission of food preference,
                                             beta 3 subunit (GABRB3) gene and             impaired fear conditioning and spatial
 followed by the emergence of                the ubiquitin ligase gene, UBE3A.            learning, and retarded acquisition of
 motor impairment, tremors,                  GABRB3 was the first gene mapped to          motor performance on a rotarod, but
     hypoactivity, aberrant                  the Prader-Willi/Angelman syndrome           without the increased tumor incidence
                                             (PW/AS) region of 15q [Wagstaff et al.,      [Costa et al., 2001].
   hind-limb clasping, and                   1991]. Due to imprinting (differential             Unfortunately, early mortality has
     respiratory symptoms.                   expression of genes based on parent of       precluded behavioral phenotyping of
                                             origin), the phenotype of these deletions    some animal models, including the
                                             varies, depending upon which parent          model for Smith–Lemli–Opitz/RSH
[Chen et al., 2001; Guy et al., 2001].       transmitted the deletion. Transmission       syndrome (SLOS), an autosomal reces-
Female animals heterozygous for the          disequilibrium has been demonstrated         sive disorder arising from mutations in
targeted disruption have delayed onset of    with a marker of GABRB3, as well             DHCR7 [Fitzky et al., 1998]. The gene
the phenotype and evidence milder            as surrounding markers, in autism            encodes an enzyme crucial for choles-
symptoms. A conditional mutation of          [Buxbaum et al., 2002]. A targeted           terol biosynthesis, suggesting a possible
the locus, where the gene is disrupted       disruption of the mouse Gabrb3 locus         link between developmental cholesterol
only in the central nervous system,          reveals a severe phenotype, with high        deprivation and autistic spectrum beha-
shows a similar phenotype to the             mortality in neonatal homozygotes            viors. The targeted disruption of Dhcr7
Mecp2-null profile [Chen et al., 2001;       [Homanics et al., 1997]. The behavioral      in mice leads to severe respiratory failure
Guy et al., 2001], indicating that the       phenotype of surviving Gabrb3-null           and death in newborn homozygotes
protein’s role in the disease process can    mice includes seizures, hyperactivity,       [Fitzky et al., 2001; Wassif et al., 2001;
be attributed solely to its function in      learning and memory deficits, poor           see also Yu et al., 2005]. Similarly,
neural tissue, rather than in the periph-    motor performance on a rotarod task,         the Tsc1-null mouse, a model for
ery. Another allele, Mecp2308, is similar    and stereotyped behavior, such as repe-      tuberous sclerosis 1, is characterized by
to the truncation allele found in the        titive circling [Homanics et al., 1997;      an embryonic lethal phenotype [Wilson
human patient population. Mecp2308/Y         DeLorey et al., 1998]. Mice heterozy-        et al., 2005].
44     AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c                                      ARTICLE

Genes Implicated by Human                          The HOXA1 locus is located in the
                                                                                                including longer immobility
Genetic Studies of Autism                     region of human chromosome 7p14.2-
                                              15. A polymorphism changing a histi-                 in a forced swim task.
Multiple studies in human populations         dine residue to an arginine is enriched
have provided evidence that chromo-           in patients with ASD over both their
some 7q contains genes conferring             unaffected relatives and the rest of            [Holmes et al., 2002, 2003; Lira et al.,
susceptibility for autism [e.g., Collabo-     the population [Ingram et al., 2000].           2003]. Enhanced anxiety-like behavior
rative Linkage Study of Autism, 1999].        Hoxa1-null mice show aberrant prenatal          is not observed, however, when the null
This region includes several genes            development, with delayed hindbrain             allele for Sert is placed on a 129S6 strain
important for early brain development,        neural tube closure, absent cranial             background [Holmes et al., 2003].
including WNT2, Engrailed 2, and              nerves, and malformed inner ear struc-                Levels of serotonin are partially
HOXA1. WNT2 encodes a secreted                tures [Lufkin et al., 1991]. Neither the        mediated by monoamine oxidase A
growth factor, which triggers a canoni-       En2- nor the Hoxa1-null mice have been          (MAOA), an enzyme of the mitochon-
cal signal transduction cascade, utilizing    screened for autistic-like behaviors [see       drial outer membrane involved in the
the Disheveled 1 (Dvl1) protein. In           Murcia et al., 2004].                           metabolism of catecholamine and indo-
Drosophila, this signal is required for                                                       lamine neurotransmitters. In one study,
expression of the Engrailed locus [Vin-                                                       the alleles regulating levels of MAOA
cent and Lawrence, 1994]. The WNT2                                                            were found to correlate with autism
                                              Serotonergic Neurotransmission                  symptom severity, with the lowest-
locus was suggested to have a role in
autism due to its location within the         The strongest evidence for aberrant             expressing alleles associated with lower
7q31-3 chromosomal region of interest,        neurotransmission in autism involves            IQs and more severe autistic behavior
a premise supported by the identification     alterations in the serotonergic system,         [Cohen et al., 2003]. Mice homozygous
of an autistic patient with a chromoso-       with elevated levels of serotonin in blood      for the null allele for Maoa have increased
mal breakpoint in the WNT2 gene.              reported for up to 45% of autistic              serotonin and norepinephrine concen-
Additionally, a single nucleotide poly-       patients [Anderson et al., 1990; Cook           trations in the brain [Cases et al., 1995].
morphism associated with autism was           and Leventhal, 1996]. Clinical trials have      Pups exhibit trembling, difficulty right-
found in the locus [IMGSAC, 2001;             provided evidence that treatment with           ing themselves, and fearfulness, while
Wassink et al., 2001]. A null allele of the   fluoxetine, a selective inhibitor of the        adults show increased aggression [Cases
mouse Wnt2 locus results in a runting         serotonin transporter, can have benefi-         et al., 1995]. The MAOA-deficient mice
phenotype, with half of the mutant pups       cial effects in the treatment of autism         also demonstrate increased freezing in a
dying perinatally due to a placental          [Fatemi et al., 1998; Hollander et al.,         conditioned fear task [Kim et al., 1997],
defect potentially unrelated to autism        2005]. Human genetic studies have               hyper-responsivity to acoustic stimuli,
[Monkley et al., 1996]. A mouse model         found a link between a short variant of         and reduced exploration in a holeboard
with targeted disruption of another           the serotonin transporter allele (SERT)         test [Popova et al., 2000]. Overall, Sert-
member of the signaling pathway, the          and autism [Kim et al., 2002; Conroy            and Maoa-null mice demonstrate the
Dvl1-null mouse, shows behavioral and         et al., 2004].                                  significant behavioral effects of altera-
neuropathological defects, including a              Mice with targeted disruption of          tions in neurotransmission, and are
lack of homecage nesting and social           the Sert locus evidence marked decrea-          promising candidates for investigation
interaction deficits [Lijam et al., 1997;     ses in brain serotonin levels [Bengel et al.,   of behaviors relevant to the core symp-
Long et al., 2004]. Dvl1-null mice also       1998], as well as decreased aggre-              toms of autism.
evidence reduced dendritic arborization       ssion, increased anxiety, and increased
[Rosso et al., 2005].                         depression-related responses, including
                                                                                              Reeler Mouse
      The Engrailed 2 locus on human          longer immobility in a forced swim task
chromosome 7q36 has been implicated                                                           The RELN locus encodes a large protein
in ASD by an association study [Gharani                                                       that acts as a serine protease of the
et al., 2004]. A targeted disruption of the         Mice with targeted                        extracellular matrix [Fatemi, 2005]. The
mouse homolog was generated prior to                                                          locus was originally identified as a
the link with ASD. Homozygotes are
                                                   disruption of the Sert                     spontaneous mouse mutation, Reeler.
viable and fertile, but have abnormal             locus evidence marked                       Reeler animals showed impaired motor
foliation of the cerebellum [Joyner et al.,          decreases in brain                       coordination, tremors and ataxia
1991]. Further studies utilizing the En2                                                      [Falconer, 1951]. The gene was subse-
mutant mouse have suggested a role for          serotonin levels, as well as                  quently identified and the Reeler allele
the gene in regulating development of         decreased aggression, increased                 shown to be a deletion of about 150 kb,
serotonergic, noradrenergic, and dopa-                                                        generating a null allele. Examination of
minergic neurons [Alberi et al., 2004;
                                                  anxiety, and increased                      Reeler brains revealed that neurons failed
Simon et al., 2005].                           depression-related responses,                  to migrate to the correct location,
ARTICLE                             AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c              45

resulting in disorganization of lamina-       criteria. Therefore, validity of mouse         tained in one side chamber of the test
ted regions, such as the cerebellar and       models for this disorder is dependent          box, or remaining alone in the center or
cerebral cortices. Studies have reported      upon reflection of core behavioral             opposite side chambers. The unfamiliar
that specific human polymorphisms in          symptoms, including impairments in             stranger mouse is enclosed in a small
the RELN locus can be associated with         social interaction and deficiencies in         wire cage, which allows visual, auditory,
autism susceptibility [Persico et al.,        other functional domains. While the            olfactory, and some tactile contact, but
2001; Skaar et al., 2004], although this      use of a behavioral testing screen with        precludes aggressive or sexual interac-
association is not observed in all autism     measures relevant to the clinical syn-         tions. An identical wire cage is placed in
patient populations [Bonora et al.,           drome is crucial for the development and       the opposite side chamber to control for
2003].                                        evaluation of appropriate animal models        the effects of object novelty. Measures
      Due to overt neuroanatomical            [Rodier, 1996; Insel, 2001; Murcia et al.,     are taken of the amount of time spent in
pathology, the Reeler mouse has been          2004], few mouse tests have been               and entries into each side of the test
extensively investigated as an animal         targeted toward the constellation of           box, as well as time spent sniffing the
model for brain dysfunction. In addition      symptoms characteristic of the ASDs.           wire cages. Initial work with this test
to the association with autism, changes       Our research group at the University of        for social approach has shown that
in RELN have also been linked with            North Carolina, in consultation with           most, but not all, of selected inbred
many neuropsychiatric disorders, inclu-       clinicians, therapists, and other profes-      mouse strains choose to spend more time
ding schizophrenia, bipolar disorder, and     sionals working in the field of autism, has    near the unfamiliar stranger. This pre-
depression, as well as with lissencephaly     designed a set of mouse behavioral tasks       ference for social proximity can be
[Fatemi, 2001], suggesting that the Reeler    to model specific aspects of autistic-like     observed in juvenile and adult mice,
mouse does not provide a model for            behavior, such as tendencies for social        and in males and females [Brodkin et al.,
brain alterations specific to autism. Apart   avoidance [Moy et al., 2004; Nadler            2004; Moy et al., 2004; Nadler et al.,
from the overt motoric deficits, Reln         et al., 2004], repetitive responses, and       2004].
mutant mice show increased anxiety and        resistance to change. The testing screen             The second phase of the test assesses
stereotypy in an open field, increased        is composed of assays for sociability,         preference for social novelty. In this case,
social dominance in a tube test, and          preference for social novelty, and learn-      the mice are given a choice between
learning deficits in the Morris water         ing acquisition and reversal, as well as       the unfamiliar mouse from the first phase
maze task [Salinger et al., 2003; Lalonde     control measures for motor and sensory         (stranger 1), and a new unfamiliar mouse
et al., 2004]. Some studies have reported     function, including olfaction. Table II        (stranger 2) placed in a wire cage in
behavioral abnormalities in heterozy-         presents a summary of mouse behavioral         the opposite side of the test box. We have
gous Reeler mice, which have levels of        tasks that may be relevant to elements of      observed that most, but not all, of the
RELN protein in brain reduced by              the autism phenotype.                          inbred mouse strains we have tested
approximately 50% [Tuetling et al.,                                                          demonstrate a shift in preference, so that
1999]. Aberrant responses observed                                                           more time is spent with the novel
                                              Measures of Social Behavior
in Relnþ/ mice included increased                                                           stranger 2, rather than the more-familiar
anxiety, deficits in prepulse inhibition      Given that impairments in social inter-        stranger 1. This phase of the social
of acoustic startle, and learning delays      action and communication are the               preference test provides information on
in an olfactory discrimination task           primary diagnostic indicators in ASDs          the ability of the mice to distinguish
[Tuetling et al., 1999; Larson et al.,        [American Psychiatric Association,             between two unfamiliar conspecifics.
2003]. Heterozygous Reeler mice were          1994], we considered the assessment of               The social preference tests allow the
also found to have decreased dendritic        social behavior as an essential compo-         assessment of both social approach and
spine density [Liu et al., 2001] and          nent for our behavioral testing battery.       avoidance, using a relatively rapid, auto-
reduced levels of oxytocin receptors in       The first measure involves simple obser-       mated task [Nadler et al., 2004]. Further
brain [Liu et al., 2005]. However, other      vations of nest-building and huddling in       characterization of social behavior can
studies have failed to find differences       the home cage, which can provide early         utilize observations of social interaction
between Relnþ/ mice and normal               indications of aberrant social behavior        in an open field or neutral cage, the
controls, suggesting the lack of a clear      [Lijam et al., 1997; Mohn et al., 1999;        resident-intruder paradigm, or tube tests
gene-dosage relationship [Salinger et al.,    Moretti et al., 2005]. For a more quan-        for social dominance [Mineur et al.,
2003].                                        titative evaluation of social approach and     2002; Duncan et al., 2004; Long et al.,
                                              avoidance, we conduct a social investi-        2004; Spencer et al., 2005]. Measure-
                                              gation task in a three-chambered appa-         ment of ultrasonic vocalizations in
MOUSE BEHAVIORAL
                                              ratus [Moy et al., 2004; Nadler et al.,        mouse pups removed from the nesting
MEASURES RELEVANT TO
                                              2004; see also Brodkin et al., 2004]. In       area can provide information on respon-
THE AUTISM PHENOTYPE
                                              the first phase of this test, mice are given   ses to social isolation early in develop-
The diagnosis of autism is based on           a choice between spending time in the          ment [Holtzman et al., 1996; Moles
behavioral, rather than physiological,        proximity of an unfamiliar mouse, con-         et al., 2004].
46       AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c                                              ARTICLE

                                   TABLE II. Mouse Behavioral Tasks for Modeling the Autism Phenotype

     Symptom in autism                            Mouse behavioral task                      References
     Social interaction deficits                  Home cage observation                      Lijam et al. [1997]; Moretti et al. [2005]
                                                  Social approach tests                      Moy et al. [2004]; Nadler et al. [2004]
                                                  Resident-intruder interaction              Mohn et al. [1999]; Moretti et al. [2005]
     First symptoms early in development,         Ultrasonic vocalization in pups            Holtzman et al. [1996]; Moles et al. [2004]
       communication deficits
     Restricted interests, repetitive behavior;   Home cage observation                      Wu and Melton [1993]; Presti et al. [2003]
       stereotypies, self-injury                  Behavior in an open field                  DeLorey et al. [1998]; Turner et al. [2001]
                                                  Nose-poke, hole board test                 Popova et al. [2000]; Bell et al. [2003]
                                                  Novel object test                          Salinger et al. [2003]; Yan et al. [2004]
     Resistance to change                         Reversal learning                          Bakker et al. [1994]; Kooy et al. [1996]
     Anxiety                                      Elevated plus maze                         Griebel et al. [2000]; Ren-Patterson et al. [2005]
                                                  Light-dark transition test                 Bouwknecht and Paylor [2002]; Peier et al. [2000]
     Changes in sensory reactivity                Prepulse inhibition of startle responses   Tuetling et al. [1999]; Nielson et al. [2002]
                                                  Hot plate analgesia test                   Martinez-Cue et al. [1999]; Long et al. [2004]
     Motor deficits, clumsiness                   Balance beam, climbing pole tests          Lalonde et al. [2004]; Moretti et al. [2005]
                                                  Rotarod performance                        Gerlai et al. [1996]; Miura et al. [2002]
     Mental retardation                           Morris water maze, hidden platform task    Sago et al. [1998]; Lalonde et al. [2004]
                                                  Radial arm maze                            Mineur et al. [2002]; Yan et al. [2004]
                                                  Operant conditioning                       Driscoll et al. [2004]; Frankland et al. [2004]
                                                  Passive avoidance                          DeLorey et al. [1998]; Lira et al. [2003]
                                                  Conditioned fear                           Kim et al. [1997]; Costa et al. [2001]

Measures for Repetitive Behavior                   the task is measured by removal of the           forcement is switched, so that mice must
and Restricted Interests                           escape platform before a final probe trial.      now choose the opposite arm to earn
                                                   Normal animals will swim to and circle           rewards. One disadvantage of this task is
In mice, repetitive behavior can include           over the area where the platform had             that mice must be food-deprived before
perseverative sniffing, circling, digging,         been located. In the next phase of               acquisition, which can lead to issues
or continuous cage-running and jump-               testing, the escape platform is placed in        of motivational levels across strains.
ing [Cases et al., 1995; Homanics et al.,          a different region of the pool, and mice         Another procedure using the T-maze is
1997; Presti et al., 2003]. Repetitive             are re-assessed on learning the new              the spontaneous alternation test, which
grooming and scratching can lead to self-          location. Delayed or deficient acquisi-          relies on the nature tendency of mice to
injury, such as torn ears and skin lesions         tion during reversal learning could serve        alternate arm choices in a maze. Mice
[Wu and Melton, 1993]. Aberrant                    as a model for autistic-like cognitive           with the Relnrl-orl mutation demonstrate
stereotypies can be noted during                   rigidity. In line with this premise,             perseverative responses in this test
home-cage observations or during                   Fmr1-null mice show normal acquisi-              [Lalonde et al., 2004].
open-field testing.                                tion of spatial learning in the Morris                 Other ways to assess repetitive beha-
      The predisposition for repetitive            water maze task, but impaired learning           vior include measures of exploration on a
behavior in autism can also be seen                during the reversal phase [Bakker et al.,        hole-board, with counts taken of nose-
in an excessive adherence to routine.              1994; Kooy et al., 1996].                        pokes into a restricted set of holes, versus
Reversal learning in the Morris water                   Mice with visual or motor impair-           the entire board [Popova et al., 2000;
maze task can be used as a measure of              ments may not be able to perform in              Bell et al., 2003], and the investigation of
resistance to change a learned pattern of          the water maze test. In these cases, a           novel objects in an open field [Salinger
behavior. In this task, mice are trained to        simple T-maze task can be used to                et al., 2003; Yan et al., 2004].
locate a hidden escape platform, which is          evaluate learning acquisition and rever-
slightly submerged in a circular pool of           sal. Mice are trained to enter one
                                                                                                    Interpretation of Results From
opaque water. Over several days, mice              particular arm (left or right) of the maze
                                                                                                    Behavioral Tests
learn to swim directly to the escape               to obtain a food reward. When a high
platform from any release point in the             rate of correct choices has been                 While deficits in social interaction and
periphery of the pool. Spatial learning in         observed, the arm designated for rein-           aberrant repetitive behavior may be key
ARTICLE                             AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c              47

features of the autism behavioral phe-        mutation, a higher rate of inactivation       ches which can address the genetics of
notype, the human disorder is character-      for the mutation-bearing chromosome           these diverse pathways to achieve a more
ized by the variable inclusion of many        can result in a less severe phenotype.        global view of the etiology of autism.
other symptoms [e.g., Muris et al.,           However, the effect of X inactivation         Discovering genes important for normal
1998]. Table II lists several functional      can be controlled in Mecp2-mutant             social behavior in mice could lead to
domains of behavior, including anxiety,       mice by only using males as subjects          the identification of brain mechanisms
sensory reactivity, motor skills, and         [Shahbazian et al., 2002]. In Angelman        that play a role in the impaired inter-
cognitive abilities, that may or may not      syndrome, symptoms are dependent on           action and communication characteriz-
be changed in mouse models for ASDs.          whether the chromosome bearing the            ing ASDs. Gene expression profiles for
In addition, the behavioral battery           mutation was inherited from the father        mouse models characterized by aberrant
utilized by our laboratory includes con-      or the mother [for review, see Williams,      social behavior, such as oxytocin or
trol measures, such as tests for activity     2005]. Parent of origin imprinting            vasopressin deficient mice [Insel et al.,
level and olfactory ability, that can be      causes expression of a subset of genes        1999; Lim et al., 2005], 5-HT1A- and
crucial for the interpretation of observed    from only one of two copies, maternal         1B- knockouts [Zhuang et al., 1999;
deficits in social preference [Moy et al.,    and paternal, present in the genome.          see also Gingrich and Hen, 2001],
2004], as well as other tasks relevant to     The effect of imprinting can be con-          dopamine transporter knockout mice
autism.                                       trolled in mouse models by setting up         [Rodriguiz et al., 2004], or mice with
                                              crosses that transmit the mutation from       reduced levels of NMDA receptors in
                                              the appropriate parent [e.g., Jiang et al.,   brain [Mohn et al., 1999; Duncan et al.,
DISCUSSION
                                              1998; Miura et al., 2002; Liljelund et al.,   2004] could suggest new genetic targets
The complex interaction of genes that         2005].                                        for studies in human populations. Beha-
appears to underlie susceptibility for the         Possible interactions between the        vioral tasks relevant to the autism
autism phenotype is problematic for           disease mutation and the genetic back-        phenotype could also be revealing in
development of animal models of the           ground of the mouse model should also         the case of mouse models which reflect
ASDs. Many of the mouse models                be considered. Susceptibility for the         neuropathology relevant to autism,
described in this review involve targeted     effects of the targeted locus may differ      including changes in cerebellum [e.g.,
disruption of a candidate gene. One           across background strains, as observed in     the Engrailed mouse, Joyner et al., 1991;
disadvantage of this approach is that the     Fmr1-null mouse [Dobkin et al., 2000],        see also Murcia et al., 2004] or brain
complete removal of a protein in brain        Sert-null mice [Holmes et al., 2003],         overgrowth [e.g., IGF-overexpressing
can lead to severe impairments or early       and Tsc1 heterozygotes [Wilson et al.,        mice, Hodge et al., 2005]. Thorough
mortality, as observed with mouse             2005]. Mouse geneticists should also          behavioral phenotyping provides an
models for Angelman and Smith–                consider strain characteristics when gen-     advantageous first approach to deter-
Lemli–Optiz syndromes [Homanics               erating mouse models. For example, the        mine which mouse models for ASDs
et al., 1997; Fitzky et al., 2001]. In        129SvEv inbred mouse strain, a line           would offer the most valuable candidates
addition, the null allele might not           widely used in laboratory research, is        for detailed genetic characterization.
properly recapitulate the disease pheno-      characterized by reduction or loss of the
type in the animal model. In the case of      corpus callosum and, in some cases, poor
ASDs, many of the alleles identified in       performance in learning tasks [Balogh
candidate genes are not null alleles.         et al., 1999; Wahlsten et al., 2001].              Thorough behavioral
Rather than a complete loss of function,      In addition, several inbred mouse
                                                                                               phenotyping provides an
autism-susceptibility alleles may cause       strains, including FVB/NJ, C3H/HeJ,
alterations in the structure of the result-   and SJL/J, carry the gene for retinal         advantageous first approach to
ing proteins, affect the amount of protein    degeneration, with blindness occurring        determine which mouse models
produced, or protein stability within the     by weaning age [The Jackson Labora-
cell.                                         tory, 2002]. Intrinsic alterations in brain   for ASDs would offer the most
      Other biological processes contri-      morphology, sensory function, and             valuable candidates for detailed
bute to the variability seen in disease       learning ability are confounding factors
phenotype. Some genetic complexity,           in the use of these strains to model
                                                                                               genetic characterization.
such as X-inactivation or parent-of-          autistic-like behaviors.
origin imprinting, can be controlled               The genes involved in the regula-
for in mouse models. For example, the         tion of complex biological processes,
gene for Rett syndrome, MECP2, is             such as development, metabolism,              REFERENCES
carried on the X chromosome. In               imprinting, and X-inactivation, are all       Alberi L, Sgado P, Simon HH. 2004. Engrailed
affected females, in every cell, one X        likely to impinge upon the observable              genes are cell-autonomously required to
                                                                                                 prevent apoptosis in mesencephalic dopa-
chromosome is inactivated, or silenced.       autism phenotype in a human or a                   minergic neurons. Development 131:3229–
In cases where only one X carries a           mouse. Thus arises the need for approa-            3236.
48      AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c                                                        ARTICLE

American Psychiatric Association. 1994. Diag-             Fang Y, Song C-Y, Vitale R. 2002. Associa-             specific learning impairment. Neurosci 100:
      nostic and statistical manual of mental             tion between a GABRB3 polymorphism and                 423–429.
      disorders (DSM-IV). Washington, DC.                 autism. Mol Psychiatry 7: 311–316.               Driscoll LL, Carroll JC, Moon J, Crnic LS,
Amir RE, Van den Veyver IB, Wan M, Tran CQ,          Cases O, Seif I, Grimsby J, Gaspar P, Chen K,               Levitsky DA, Strupp BJ. 2004. Impaired
      Francke U, Zoghbi HY. 1999. Rett                    Pournin S, Muller U, Aguet M, Babinet C,               sustained attention and error-induced
      syndrome is caused by mutations in X-               Shih JC, De Maeyer E. 1995. Aggressive                 stereotypy in the aged Ts65Dn mouse: A
      linked MECP2, encoding methyl-CpG-                  behavior and altered amounts of brain                  mouse model of Down syndrome and
      binding protein 2. Nat Genet 23:185–188.            serotonin and norepinephrine in mice                   Alzheimer’s disease. Behav Neurosci 118:
Anderson GM, Horne WC, Chatterjee D, Cohen                lacking MAOA. Science 268:1763–1766.                   1196–1205.
      DJ. 1990. The hyperserotoninemia of aut-       Chen L, Toth M. 2001. Fragile X mice develop          Duncan GE, Moy SS, Perez A, Eddy DM, Zinow
      ism. Ann NY Acad Sci 600:331–340.                   sensory hyperreactivity to auditory stimuli.           W, Lieberman JA, Snouwaert JN, Koller
Aylward EH, Minshew NJ, Goldstein G, Honey-               Neuroscience 103:1043–1050.                            BH. 2004. Deficits in sensorimotor gating
      cutt NA, Augustine AM, Yates KO, Barta         Chen RZ, Akbarian S, Tudor M, Jaenisch R.                   and tests of social behavior in a genetic
      PE, Pearlson GD. 1999. MRI volumes of               2001. Deficiency of methyl-CpG binding                 model of reduced NMDA receptor func-
      amygdala and hippocampus in non-mentally            protein-2 in CNS neurons results in a Rett-            tion. Behav Brain Res 153:507–518.
      retarded autistic adolescents and adults.           like phenotype in mice. Nat Genet 27:327–        Falconer DS. 1951. Two new mutants. J Genet 50:
      Neurology 53:2145–2150.                             331.                                                   192–201.
Bailey A, Luthert P, Dean A, Harding B, Janota I,    Cohen IL, Liu X, Schutz C, White BN, Jenkins          Fatemi SH. 2001. Reelin mutations in mouse and
      Montgomery M, Rutter M, Lantos P. 1998.             EC, Brown WT, Holden JJA. 2003. Asso-                  man: From reeler mouse to schizophrenia,
      A clinicopathological study of autism. Brain        ciation of autism severity with a monoamine            mood disorders, autism and lissencephaly.
      121:889–905.                                        oxidase A functional polymorphism. Clin                Mol Psychiatry 6:129–133.
Bakker CE, Verheij C, Willemsen R, van der                Genet 64:190–197.                                Fatemi SH. 2005. Reelin glycoprotein: Structure,
      Helm R, Oerlemans F, Vermey M, Bygrave         Collaborative Linkage Study of Autism. 1999. An             biology and roles in health and disease. Mol
      A, Hoogeveen AT, Oostra BA, Reyniers E,             autosomal genomic screen for autism. Am J              Psychiatry 10:251–257.
      De Boulle K, D’Hooge R, Cras P, van                 Med Genet (Neuropsychiatr Genet) 88:             Fatemi SH, Realmuto GM, Khan L, Thuras P.
      Velzen D, Nagels G, Martin J-J, De Deyn             609–615.                                               1998. Fluoxetine in treatment of adoles-
      PP, Darby JK, Willems PJ. 1994. Fmr1           Comery TA, Harris JB, Willems PJ, Oostra BA,                cent patients with autism: A longitudinal
      knockout mice: A model to study fragile X           Irwin SA, Weiler IJ, Greenough WT. 1997.               open trial. J Aut Dev Disorders 28:303–
      mental retardation. Cell 78:23–33.                  Abnormal dendritic spines in fragile X                 307.
Balogh SA, McDowell CS, Stavnezer AJ, Denen-              knockout mice: Maturation and pruning            Fisch GS, Hao HK, Bakker C, Oostra BA. 1999.
      berg VH. 1999. A behavioral and neuroa-             deficits. Proc Natl Acad Sci 94:5401–5404.             Learning and memory in the FMR1 knock-
      natomical assessment of an inbred substrain    Conroy J, Meally E, Kearney G, Fitzgerald M, Gill           out mouse. Am J Med Genet 84:277–
      of 129 mice with behavioral comparisons to          M, Gallagher L. 2004. Serotonin transporter            282.
      C57BL/6J mice. Brain Res 836:38–48.                 gene and autism: A haplotype analysis in an      Fitzky BU, Witsch-Baumgartner M, Erdel M, Lee
Baranek GT. 2002. Efficacy of sensory and motor           Irish autistic population. Mol Psychiatry              J-N, Paik Y-K, Glossmann H, Utermann G,
      interventions for children with autism. J           9:587–593.                                             Moebius FF. 1998. Mutations in the 7-sterol
      Autism Dev Disord 32:397–422.                  Cook EH, Leventhal BL. 1996. The serotonin sys-             reductase gene in patients with the Smith-
Barton M, Volkmar F. 1998. How commonly are               tem in autism. Curr Opin Pediatr 8:348–                Lemli-Opitz syndrome. Proc Natl Acad Sci
      known medical conditions associated with            354.                                                   USA 95:8181–8186.
      autism? J Aut Dev Dis 28:273–278.              Costa RM, Yang T, Huynh DP, Pulst SM,                 Fitzky BU, Moebius FF, Asaoka H, Waage-Baudet
Bell K, Shokrian D, Potenzieri C, Whitaker-               Viskochil DH, Silva AJ, Brannan CI. 2001.              H, Xu L, Xu G, Maeda N, Kluckman K,
      Azmitia PM. 2003. Harm avoidance, anxi-             Learning deficits, but normal development              Hiller S, Yu H, Batta AK, Shefer S, Chen
      ety, and response to novelty in the adoles-         and tumor predisposition, in mice lacking              T, Salen G, Sulik K, Simoni RD, Ness GC,
      cent S-100B transgenic mouse: Role of               exon 23a of Nf1. Nat Genet 27:399–405.                 Glossmann H, Patel SB, Tint GS. 2001.
      serotonin and relevance to Down syndrome.      Costa RM, Federov NB, Kogan JH, Murphy GG,                  7-dehydrocholesterol-dependent proteolysis
      Neuropsychopharmacol 28:1810–1816.                  Stern J, Ohno M, Kucherlapati R, Jacks T,              of HMG-CoA reductase suppresses sterol
Bengel D, Murphy DL, Andrews AM, Wichems                  Silva AJ. 2002. Mechanism for the learning             biosynthesis in a mouse model of Smith–
      CH, Feltner D, Heils A, Mossner R,                  deficits in a mouse model of neurofibroma-             Lemli–Opitz/RSH syndrome. J Clin Invest
      Westphal H, Lesch KP. 1998. Altered brain           tosis type 1. Nature 415:526–530.                      108:905–915.
      serotonin homeostasis and locomotor insen-     Courchesne E, Karns CM, Davis HR, Ziccardi R,         Folstein SE, Rosen-Sheidley B. 2001. Genetics of
      sitivity to 3,4-methylenedioxymethampheta-          Carper RA, Tigue ZD, Chisum HJ, Moses                  autism: Complex aetiology for a hetero-
      mine (‘‘Ecstasy’’) in serotonin transporter-        P, Pierce K, Lord C, Lincoln AJ, Pizzo S,              geneous disorder. Nat Rev Genet 2:943–
      deficient mice. Mol Pharmacol 53:649–655.           Schreibman L, Haas RH, Akshoomoff NA,                  955.
Bodfish JW, Symons FJ, Parker DE, Lewis M.                Courchesne RY. 2001. Unusual brain               Fombonne E. 2002. Epidemiological trends in
      2000. Varieties of repetitive behavior in           growth patterns in early life in patients with         rates of autism. Mol Psychiatry 7:S4–S6.
      autism: Comparisons to mental retardation.          autistic disorder. An MRI study. Neurology       Frankland PW, Wang Y, Rosner B, Shimizu T,
      J Aut Dev Disorders 30:237–243.                     57:245–254.                                            Balleine BW, Dykens EM, Ornitz EM, Silva
Bonora E, Beyer KS, Lamb JA, Parr JR, Klauck         Darnell JC, Fraser CE, Mostovetsky O, Stefani G,            AJ. 2004. Sensorimotor gating abnormalities
      SM, Benner A, Paolucci M, Abbott A,                 Jones TA, Eddy SR, Darnell RB. 2005.                   in young males with fragile X syndrome and
      Ragoussis I, Poustka A, Bailey AJ, Monaco           Kissing complex RNAs mediate interaction               Fmr1-knockout mice. Mol Psychiatry 9:
      AP, International Molecular Genetic Study           between the Fragile-X mental retardation               417–425.
      of Autism (IMGSAC). 2003. Analysis of               protein KH2 domain and brain polyribo-           Gerlai R, Millen KJ, Herrup K, Fabien K, Joyner
      reelin as a candidate gene for autism. Mol          somes. Genes Dev 19:903–918.                           AL, Roder J. 1996. Impaired motor learning
      Psychiatry 8:885–892.                          DeLorey TM, Handforth A, Anagnostaras SG,                   performance in cerebellar En-2 mutant
Bouwknecht JA, Paylor R. 2002. Behavioral and             Homanics GE, Minassian BA, Asatourian A,               mice. Behav Neurosci 110:126–133.
      physiological mouse assays for anxiety: A           Fanselow MS, Delgado-Escueta A, Ellison          Gharani N, Benayed R, Mancuso V, Brzustowicz
      survey in nine mouse strains. Behav Brain           GD, Olsen RW. 1998. Mice lacking the b3                LM, Millonig JH. 2004. Association of the
      Res 136:489–501.                                    subunit of the GABAA receptor have the                 homeobox transcription factor, ENGRAILED
Brodkin ES, Hagemann A, Nemetski SM, Silver               epilepsy phenotype and many of the beha-               2, 3, with autism spectrum disorder. Mol
      LM. 2004. Social approach-avoidance beha-           vioral characteristics of Angelman syn-                Psychiatry 9(5):474–484.
      vior of inbred mouse strains towards DBA/2          drome. J Neurosci 18:8505–8514.                  Gillberg C, Billstedt E. 2000. Autism and
      mice. Brain Res 1002:151–157.                  Dobkin C, Rabe A, Dumas R, El Idrissi A,                    Asperger syndrome: Coexistence with other
Buxbaum JD, Silverman JM, Smith CJ, Greenberg             Haubenstock H, Brown WT. 2000. Fmr1                    clinical disorders. Acta Psychiatr Scand 102:
      DA, Kilifarski M, Reichert J, Cook EH Jr,           knockout mouse has a distinctive strain-               321–330.
ARTICLE                                      AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMIN. MED. GENET.): DOI 10.1002/ajmg.c                             49

Gingrich JA, Hen R. 2001. Dissecting the role of        Insel TR, O’Brien DJ, Leckman JF. 1999.                Liu WS, Pesold C, Rodriguez MA, Carboni G,
      the serotonin system in neuropsychiatric                 Oxytocin, vasopressin, and autism: Is there          Auta J, Lacor P, Larson J, Condie BG,
      disorders using knockout mice. Psychophar-               a connection? Biol Psychiatry 45:145–157.            Guidotti A, Costa E. 2001. Down-regula-
      macology 155:1–10.                                Jiang Y, Armstrong D, Albrecht U, Atkins CM,                tion of dendritic spine and glutamic acid
Griebel G, Belzung C, Perrault G, Sanger DJ.                   Noebels JL, Eichele G, Sweatt JD, Beaudet            decarboxylase 67 expressions in the reelin
      2000. Differences in anxiety-related beha-               AL. 1998. Mutation of the Angelman                   haploinsufficient heterozygous reeler mouse.
      viours and in sensitivity to diazepam in                 ubiquitin ligase in mice causes increased            Proc Natl Acad Sci 98:3477–3482.
      inbred and outbred strains of mice. Psycho-              cytoplasmic p53 and deficits of contextual      Liu W, Pappas GD, Carter CS. 2005. Oxytocin
      pharmacology 148:164–170.                                learning and long-term potentiation. Neu-            receptors in brain cortical regions are
Guy J, Hendrich B, Holmes M, Martin JE, Bird                   ron 21:799–811.                                      reduced in haploinsufficient (þ/) reeler
      A. 2001. A mouse Mecp2-null mutation              Joyner AL, Herrup K, Auerbach BA, Davis CA,                 mice. Neurol Res 27(4):339–345.
      causes neurological symptoms that mimic                  Rossant J. 1991. Subtle cerebellar pheno-       Long JM, LaPorte P, Paylor R, Wynshaw-Boris A.
      Rett syndrome. Nat Genet 27:322–326.                     type in mice homozygous for a targeted               2004. Expanded characterization of the
Hagerman RJ, Jackson AWIII, Levitas A, Rim-                    deletion of the En-2 homeobox. Science               social interaction abnormalities in mice
      land B, Braden M. 1986. An analysis of                   251:1239–1243.                                       lacking Dvl1. Genes Brain Behav 3:51–62.
      autism in fifty males with the fragile X          Keller F, Persico AM. 2003. The neurobiological        Lufkin T, Dierich A, LeMeur M, Mark M,
      syndrome. Am J Med Genet 23:359–374.                     context of autism. Mol Neurobiol 28:1–22.            Chambon P. 1991. Disruption of the Hox-
Happé F, Ehlers S, Fletcher P, Frith U, Johansson M,   Kim JJ, Shih JC, Chen K, Chen L, Bao S, Maren               1.6 homeobox gene results in defects in a
      Gillberg C, Dolan R, Frackowiak R, Frith C.              S, Anagnostaras SG, Fanselow MS, Maeyer              region corresponding to its rostral domain of
      1996.‘‘Theoryofmind’’ inthebrain. Evidence               ED, Seif I, Thompson RF. 1997. Selective             expression. Cell 66:1105–1119.
      from a PET scan study of Asperger syndrome.              enhancement of emotional, but not motor,        Martinez-Cue C, Baamonde C, Lumbreras MA,
      Neuro Report 8(1): 197–201.                              learning in monoamine oxidase A-deficient            Vallina IF, Dierssen M, Florez J. 1999. A
Hodge RD, D’Ercole AJ, O’Kusky JR. 2005.                       mice. Proc Natl Acad Sci 94:5929–5933.               murine model for Down syndrome shows
      Increased expression of insulin-like growth       Kim S-J, Cox N, Courchesne R, Lord C, Corsello              reduced responsiveness to pain. Neurore-
      factor-I (IGF-I) during embryonic develop-               C, Akshoomoff N, Guter S, Leventhal BL,              port 10:1119–1122.
      ment produces neocortical overgrowth with                Courchesne E, Cook EHJr. 2002. Transmis-        Mineur YS, Sluyter F, de Wit S, Oostra BA, Crusio
      differentially greater effects on specific               sion disequilibrium mapping at the seroto-           WE. 2002. Behavioral and neuroanatomical
      cytoarchitectonic areas and cortical layers.             nin transporter gene (SLC6A4) region in              characterization of the Fmr1 knockout
      Dev Brain Res 154:227–237.                               autistic disorder. Mol Psychiatry 7:278–288.         mouse. Hippocampus 12:39–46.
Hollander E, Phillips A, Chaplin W, Zagursky K,         Kooy RF, D’Hooge R, Reyniers E, Bakker CE,             Miura K, Kishino T, Li E, Webber H, Dikkes P,
      Novotny S, Wasserman S, Iyengar R. 2005.                 Nagels G, De Boulle K, Storm K, Clincke              Holmes GL, Wagstaff J. 2002. Neurobeha-
      A placebo controlled crossover trial of liquid           G, De Deyn PP, Oostra BA, Willems PJ.                vioral and electroencephalographic abnor-
      fluoxetine on repetitive behaviors in child-             1996. Transgenic mouse model for the                 malities in Ube3a maternal-deficient mice.
      hood and adolescent autism. Neuropsycho-                 fragile X syndrome. American J Med Gen               Neurobio Dis 9:149–159.
      pharmacology 30:582–589.                                 64:241–245.                                     Mohn AR, Gainetdinov RR, Caron MG, Koller
Holmes A, Yang RJ, Murphy DL, Crawley JN.               Kremer EJ, Pritchard M, Lynch M, Yu S, Holman               BH. 1999. Mice with reduced NMDA
      2002. Evaluation of antidepressant-related               K, Baker E, Warren ST, Schlessinger D,               receptor expression display behaviors related
      behavioral responses in mice lacking the                 Sutherland GR, Richards RI. 1991. Map-               to schizophrenia. Cell 98:427–436.
      serotonin transporter. Neuropsychophar-                  ping of DNA instability at the fragile X to a   Moles A, Kieffer BL, D’Amato F. 2004. Deficit
      macology 27(6):914–923.                                  trinucleotide repeat sequence p(CCG)n.               in attachment behavior in mice lacking the
Holmes A, Li Q, Murphy DL, Gold E, Crawley                     Science 252:1711–1714.                               m-opioid receptor gene. Science 304:1983–
      JN. 2003. Abnormal anxiety-related beha-          Lalonde R, Hayzoun K, Derer M, Mariani J,                   1986.
      vior in serotonin transporter null mutant                Strazielle C. 2004. Neurobehavioral evalua-     Monkley SJ, Delaney SJ, Pennisi DJ, Christiansen
      mice: The influence of genetic background.               tion of Relnrl-orl mutant mice and correla-          JH, Wainwright BJ. 1996. Targeted disrup-
      Genes Brain Behav 2(6):365–380.                          tions with cytochrome oxidase activity.              tion of the Wnt2 gene results in placentation
Holtzman DM, Santucci D, Kilbridge J, Chua-                    Neurosci Res 49:297–305.                             defects. Development 122:3343–3353.
      Couzens J, Fontana DJ, Daniels SE, Johnson        Larson J, Hoffman JS, Guidotti A, Costa E. 2003.       Moretti P, Bouwknecht JA, Teague R, Paylor R,
      RM, Chen K, Sun Y, Carlson E, Alleva E,                  Olfactory discrimination learning deficit in         Zoghbi HY. 2005. Abnormalities of social
      Epstein CJ, Mobley WC. 1996. Develop-                    heterozygous reeler mice. Brain Res 971:             interactions and home-cage behavior in a
      mental abnormalities and age-related neu-                40–46.                                               mouse model of Rett syndrome. Hum Mol
      rodegeneration in a mouse model of Down           Lijam N, Paylor R, McDonald MP, Crawley JN,                 Gen 14:205–220.
      syndrome. Proc Natl Acad Sci 93:13333–                   Deng C-X, Herrup K, Stevens KE, Macca-          Moy SS, Nadler JJ, Perez A, Barbaro RP, Johns
      13338.                                                   ferri G, McBain CJ, Sussman DJ, Wynshaw-             JM, Magnuson TR, Piven J, Crawley JN.
Homanics GE, DeLorey TM, Firestone LL,                         Boris A. 1997. Social interaction and                2004. Sociability and preference for social
      Quinlan JJ, Handforth A, Harrison NL,                    sensorimotor gating abnormalities in mice            novelty in five inbred strains: An approach to
      Krasowski MD, Rick CEM, Korpi ER,                        lacking Dv/1. Cell 90:895–905.                       assess autistic-like behavior in mice. Genes
      Makela R, Brilliant MH, Hagiwara N,               Liljelund P, Handforth A, Homanics GE, Olsen                Brain Behav 3:287–302.
      Ferguson C, Synder K, Olsen RW. 1997.                    RW. 2005. GABAA receptor b3 subunit             Murcia CL, Gulden F, Herrup K. 2004. A
      Mice devoid of a-aminobutyrate type A                    gene-deficient heterozygous mice show                question of balance: A proposal for new
      receptor b3 subunit have epilepsy, cleft                 parent-of-origin and gender-related differ-          mouse models of autism. Int J Devl
      palate, and hypersensitive behavior. Proc                ences in b3 subunit levels, EEG, and                 Neurosci 23:265–275.
      Natl Acad Sci USA 94:4143–4148.                          behavior. Dev Brain Res 157:150–161.            Muris P, Steerneman P, Merckelbach H, Holdri-
IMGSAC. 2001. A genomewide screen for                   Lim MM, Bielsky IF, Young LJ. 2005. Neuropep-               net I, Meesters C. 1998. Comorbid anxiety
      autism: Strong evidence for linkage to                   tides and the social brain: Potential rodent         symptoms in children with pervasive devel-
      chromosomes 2q, 7q, and 16p. Am J Hum                    models of autism. Int J Devl Neurosci 23:            opmental disorders. J Anxiety Dis 12:387–
      Genet 69:570–581.                                        235–243.                                             393.
Ingram JL, Stodgell CJ, Hyman SL, Figlewicz DA,         Lira A, Zhou M, Castanon N, Ansorge MS,                Nadler JJ, Moy SS, Dold G, Trang D, Simmons N,
      Weitkamp LR, Rodier PM. 2000. Discov-                    Gordon JA, Francis JH, Bradley-Moore M,              Perez A, Young NB, Barbaro RP, Piven J,
      ery of allelic variants of HOXA1 and                     Lira J, Underwood MD, Arango V, Kung                 Magnuson T, Crawley JN. 2004. Automated
      HOXB1: Genetic susceptibility to autism                  HF, Hofer MA, Hen R, Gingrich JA. 2003.              apparatus for rapid quantitation of autism-
      spectrum disorders. Teratology 62:393–405.               Altered depression-related behaviors and             like social deficits in mice. Genes Brain
Insel TR. 2001. Mouse models for autism: Report                functional changes in the dorsal raphe               Behav 3:303–314.
      from a meeting. Mamm Genome 12:755–                      nucleus of serotonin transporter-deficient      Nan X, Ng H-H, Johnson CA, Laherty CD,
      757.                                                     mice. Bio Psychiatry 54:960–971.                     Turner BM, Eisenman RN, Bird A. 1998.
You can also read