Baker. The Social Skills Picture Book
Baker. The Social Skills Picture Book
Baker. The Social Skills Picture Book
REVIEW OF
NEUROBIOLOGY
VOLUME 113
SERIES EDITORS
R. ADRON HARRIS
Waggoner Center for Alcohol and Drug Addiction Research
The University of Texas at Austin
Austin, Texas, USA
PETER JENNER
Division of Pharmacology and Therapeutics
GKT School of Biomedical Sciences
King's College, London, UK
EDITORIAL BOARD
ERIC AAMODT HUDA AKIL
PHILIPPE ASCHER MATTHEW J. DURING
DONARD S. DWYER DAVID FINK
MARTIN GIURFA BARRY HALLIWELL
PAUL GREENGARD JON KAAS
NOBU HATTORI LEAH KRUBITZER
DARCY KELLEY KEVIN MCNAUGHT
BEAU LOTTO JOSÉ A. OBESO
MICAELA MORELLI CATHY J. PRICE
JUDITH PRATT SOLOMON H. SNYDER
EVAN SNYDER STEPHEN G. WAXMAN
JOHN WADDINGTON
Academic Press is an imprint of Elsevier
32 Jamestown Road, London NW1 7BY, UK
Radarweg 29, PO Box 211, 1000 AE Amsterdam, The Netherlands
The Boulevard, Langford Lane, Kidlington, Oxford, OX5 1GB, UK
225 Wyman Street, Waltham, MA 02451, USA
525 B Street, Suite 1800, San Diego, CA 92101-4495, USA
Permissions may be sought directly from Elsevier’s Science & Technology Rights
Department in Oxford, UK: phone (þ44) (0) 1865 843830; fax (þ44) (0) 1865 853333;
email: permissions@elsevier.com. Alternatively you can submit your request online
by visiting the Elsevier web site at http://elsevier.com/locate/permissions, and selecting
Obtaining permission to use Elsevier material
Notice
No responsibility is assumed by the publisher for any injury and/or damage to persons
or property as a matter of products liability, negligence or otherwise, or from any use
or operation of any methods, products, instructions or ideas contained in the material
herein. Because of rapid advances in the medical sciences, in particular, independent
verification of diagnoses and drug dosages should be made
ISBN: 978-0-12-418700-9
ISSN: 0074-7742
Ashley Anderson
Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
Esther B.E. Becker
MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics,
University of Oxford, Oxford, United Kingdom
Brent R. Bill
Department of Psychiatry, David Geffen School of Medicine, Center for Autism Research
and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience
and Human Behavior, University of California, Los Angeles, California, USA
Daniel B. Campbell
Zilkha Neurogenetic Institute, and Department of Psychiatry and the Behavioral Sciences,
University of Southern California, Los Angeles, California, USA
Leanne Chukoskie
Institute for Neural Computation, University of California, San Diego, California, USA
Joseph D. Dougherty
Department of Genetics, and Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri, USA
Christina T. DyBuncio
Department of Psychiatry, David Geffen School of Medicine, Center for Autism Research
and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience
and Human Behavior, and Program in Neurogenetics, Department of Neurology, David
Geffen School of Medicine, University of California, Los Angeles, California, USA
Brent L. Fogel
Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine,
University of California, Los Angeles, California, USA
Elaine Y. Hsiao
Division of Biology and Biological Engineering, Division of Chemistry and Chemical
Engineering, California Institute of Technology, Pasadena, California, USA
Matthew Huentelman
Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona,
USA
Genevieve Konopka
Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
Kenneth Y. Kwan
Department of Human Genetics, Molecular & Behavioral Neuroscience Institute (MBNI),
University of Michigan, Ann Arbor, Michigan, USA
ix
x Contributors
Stephanie Lepp
Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
Jennifer K. Lowe
Department of Psychiatry, David Geffen School of Medicine, Center for Autism Research
and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience
and Human Behavior, and Program in Neurogenetics, Department of Neurology, David
Geffen School of Medicine, University of California, Los Angeles, California, USA
Susan E. Maloney
Department of Genetics, and Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri, USA
Shingo Miyauchi
School of Biotechnology and Biomolecular Sciences, University of New South Wales,
Sydney, Australia
Yun Peng
Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix,
Arizona, USA
Shenfeng Qiu
Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix,
Arizona, USA
Michael A. Rieger
Department of Genetics, and Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri, USA
Christopher Smith
Southwest Autism Research Center, Phoenix, Arizona, USA
Catherine J. Stoodley
Department of Psychology, American University, Washington, District of Columbia, USA
Jeanne Townsend
Department of Neurosciences, University of California, San Diego, California, USA
Irina Voineagu
School of Biotechnology and Biomolecular Sciences, University of New South Wales,
Sydney, Australia
Marissa Westerfield
Institute for Neural Computation, University of California, San Diego, California, USA
Brent Wilkinson
Program in Biological and Biomedical Sciences, and Zilkha Neurogenetic Institute,
University of Southern California, Los Angeles, California, USA
PREFACE
The Neurobiology of Autism: Integrating
Genetics, Brain Development, Behavior,
and the Environment
The definition and diagnosis of autism spectrum disorder (ASD) has remained
somewhat constant: impairment in social and communicative behavior and
the manifestation of repetitive and restrictive behaviors. However, our
understanding of the mechanisms underlying ASD is continuously evolving
as both more researchers across broad disciplines enter the field and as
techniques improve for greater resolution into genetics, genomics, brain
connectivity, and behavior. Together, these advances have determined that
ASD is not caused by a single source, but rather these collective studies have
demonstrated that there are a multitude of genetic and environmental mech-
anisms at play that all ultimately manifest (in a variety of combinations)
in ASD.
The genetic component of ASD has been well documented as a central
contributor to etiology. However, this genetic component rarely takes the
form of a monogenetic disorder, and it is unlikely that genetics can
completely explain all incidences of ASD. Thus, the genetics of ASD are
complex and how this genetic component interacts with environmental
components, for example, at the epigenetic level, remains a topic of great
interest. In this volume, Campbell and colleagues discuss the role of noncod-
ing RNAs in ASD. This is yet another level of genetic regulation that is just
beginning to be explored but certainly plays an important function in ASD.
Specific genes are also discussed in this volume as windows into understand-
ing basic brain mechanisms at risk in ASD: the chapter by Qiu and colleagues
focuses on MET and its role in brain connectivity, the chapter by Lepp and
colleagues highlights the role of the FOXP genes in language and commu-
nication, and the chapter by Fogel and colleagues outlines how a splicing
factor, RBFOX1, may have a pivotal role in ASD gene networks.
How such an altered genetic architecture may lead to a manifestation in
changes in gene expression in the brain is a relatively underexplored topic in
ASD research due to the paucity of tissue for such studies. However, the
chapter by Miyauchi and Voineagu summarizes this field and presents
new insights into these data including the expression pattern of ASD genes
xi
xii Preface
GENEVIEVE KONOPKA
Department of Neuroscience, UT Southwestern Medical Center,
Dallas, Texas, USA
CHAPTER ONE
Contents
1. Introduction 2
2. Cerebellar Organization 3
3. ASD Symptoms in Patients with Cerebellar Disorders 5
4. Motor Impairment in Autistic Individuals 6
5. Cerebellar Pathology in Autism 8
6. Cerebellar Differences in Autism: Structural Neuroimaging 9
7. Abnormal Cerebellar Activation in Autism 11
8. Autoimmune Studies in ASD Implicating the Cerebellum 13
9. Autism Genes in Mouse Cerebellar Development 14
10. Cerebellar Phenotypes in Rodent Models of Autism 17
11. Functional Evidence from Mouse Genetics 20
12. Conclusions 21
Acknowledgment 21
References 22
Abstract
The cerebellum has been long known for its importance in motor learning and coor-
dination. Recently, anatomical, clinical, and neuroimaging studies strongly suggest that
the cerebellum supports cognitive functions, including language and executive func-
tions, as well as affective regulation. Furthermore, the cerebellum has emerged as
one of the key brain regions affected in autism. Here, we discuss our current understand-
ing of the role of the cerebellum in autism, including evidence from genetic, molecular,
clinical, behavioral, and neuroimaging studies. Cerebellar findings in autism suggest
developmental differences at multiple levels of neural structure and function, indicating
that the cerebellum is an important player in the complex neural underpinnings of
autism spectrum disorder, with behavioral implications beyond the motor domain.
1. INTRODUCTION
Autism spectrum disorder (ASD) comprises a collection of neu-
rodevelopmental diseases defined by deficits in communication and social
interaction, and repetitive and restrictive behaviors (American Psychiatric
Association, 2013). The etiology of autism is complex. The past decade
has seen revolutionary advances in our understanding of the genetics of
ASD and several hundreds of genetic variants have been identified
(Berg & Geschwind, 2012; Betancur, 2011; Devlin & Scherer, 2012). In
addition to its intricate genetic landscape, various environmental factors
and specific gene–environment interactions are thought to contribute to
the pathogenesis of ASD (Hallmayer et al., 2011; Herbert, 2010).
Despite the recent advances in autism research, the molecular underpin-
nings and neural and circuit substrates of autism remain incompletely under-
stood. ASD is widely regarded as a disorder of connectivity between
different parts of the brain. A number of different brain areas have been
implicated in autism (Amaral, Schumann, & Nordahl, 2008; Courchesne,
Campbell, & Solso, 2011; Di Martino et al., 2013), including the cerebellum
(Fatemi et al., 2012; Rogers, McKimm, et al., 2013).
Termed the “little brain,” the cerebellum comprises 10% of total brain
volume but contains more neurons than the rest of the brain and has the
highest cell density of any brain area, approximately four times that of the
neocortex (Herculano-Houzel, 2010). Its unique geometric arrangement,
relatively simple structure, and sophisticated circuitry have been the subject
of intense scrutiny for over two centuries. The cerebellum is at the cross-
roads between the sensory and motor systems and is essential for coordinat-
ing communications between these two systems. Importantly, the
cerebellum is not necessary for basic elements of perception or movement,
but rather controls the spatial accuracy and temporal coordination of move-
ment. In addition, the cerebellum has long been implicated in motor skill
learning. More recently, driven by increasingly sophisticated imaging tech-
niques and advances in genetic studies, mounting evidence points to a role
for the cerebellum in cognition and emotion.
In this chapter, we will give a brief introduction to the cerebellum and
discuss the different lines of evidence that link the cerebellum to autism.
Autism Spectrum Disorder and the Cerebellum 3
2. CEREBELLAR ORGANIZATION
The cerebellum lies behind the pons and is connected to the brain stem
by three pairs of peduncles. Structurally, the cerebellum is composed of an
outer mantle of gray matter (GM) (cerebellar cortex), which surrounds the
internal white matter (WM), with three pairs of embedded deep nuclei (from
medial to lateral, the fastigial, interposed, and dentate nuclei). Morphologi-
cally, the cerebellum is subdivided into a central vermis flanked by two hemi-
spheres. The hemispheres are evolutionarily more recent and their volume
increases progressively from lower vertebrates to higher mammals; this dra-
matic increase parallels the expansion of the neocortex in higher mammalian
orders (Balsters et al., 2010). Notably, this region (cerebrocerebellum)
receives input exclusively from the cerebral cortex. The cerebellum of higher
vertebrates is highly folded into a series of parallel folia and subdivided into 3
lobes and 10 lobules (I–X). In humans, lobule VII is subdivided into Crus I,
Crus II, and VIIB, and lobule VIII is divided into VIIIA and VIIIB (Fig. 1.1A).
Figure 1.1 Cerebellar anatomy. (A) Illustration of the lobes and lobules of the cerebel-
lum, with the lobules color-coded (Spatially Unbiased Infratentorial (SUIT) Atlas;
Diedrichsen et al., 2006, 2009). (B) Schematic representation of the basic cerebellar cell
types and circuitry. The cerebellar neurons receive input from the climbing fibers (CF)
originating in the inferior olive (IO) and the mossy fibers (MF) coming from the
precerebellar nuclei (PCN). Purkinje cells (PC) form the sole output of the cerebellar cor-
tex. Excitatory (þ) and inhibitory () inputs are indicated. BC, basket cell; DCN, deep
cerebellar nuclei; GC, granule cell; GL, granular layer; GoC, Golgi cell; ML, molecular layer;
PCL, Purkinje cell layer; PF, parallel fiber, SC, stellate cell; WM, white matter.
4 Esther B.E. Becker and Catherine J. Stoodley
Figure 1.2 Cerebellar functional topography. Task-based functional MRI activation pat-
terns reflect contralateral connections with cerebral cortex and ipsilateral connections
with the spinal cord. Language tasks engage right-lateralized cerebellar lobule VII, spa-
tial task activation is left-lateralized, and right-handed finger-tapping activates the right
cerebellar anterior lobe and lobule VIII. Adapted and reproduced with permission from
Stoodley and Schmahmann (2009).
Autism Spectrum Disorder and the Cerebellum 5
suggested that adults and adolescents with ASD are impaired in calibrating
the relationship between their body and environment, and this is strongly
correlated with their social and communication impairments
(Linkenauger, Lerner, Ramenzoni, & Proffitt, 2012).
Motor impairments are among the earliest signs of an autistic phenotype
(Esposito et al., 2011; Teitelbaum, Teitelbaum, Nye, Fryman, & Maurer,
1998; Zwaigenbaum, Bryson, & Garon, 2013). Prospective studies of
at-risk infants have shown that children who are later diagnosed with
ASD show poorer fine and gross motor skills than typically developing
(TD) and language-impaired children (Landa & Garrett-Mayer, 2006),
and motor impairments are predictive of ASD outcome (Zwaigenbaum
et al., 2013). Greater head lag during pull-to-sit was more frequently
observed in infants at high-risk for ASD and was associated with ASD status
upon follow-up (Flanagan, Landa, Bhat, & Bauman, 2012). Similarly, oral
and manual motor skills in infancy and toddlerhood differentiated ASD indi-
viduals and predicted later speech fluency (Gernsbacher, Sauer, Geye,
Schweigert, & Hill Goldsmith, 2008), and early motor delays are more com-
mon in infants at risk for ASD and are related to later communication delays
(Bhat, Galloway, & Landa, 2012).
Performance on motor tasks also correlates with ASD symptoms,
including emotional/behavioral disturbance and communication disorder
(Papadopoulos et al., 2012). ASD children that were significantly impaired dur-
ing quiet stance had a higher number of restricted and repetitive behaviors
(Radonovich et al., 2013). It has been suggested that the lack of gesture and imi-
tation in ASD might be related to motor dysfunction, providing a mechanism
by which cerebellar dysfunction could impact the core social communication
symptoms of ASD (Gidley Larson & Mostofsky, 2006; Jones & Prior, 1985).
Consistent with this, ASD children are impaired on both the recognition
and imitation of gestures (Stieglitz Ham et al., 2011), and imitation impairment
was associated with increased ASD symptoms and poorer joint attention
(Rogers et al., 2003).
2008) and likely related to cerebellar WM (Allen, 2005; Amaral et al., 2008;
Courchesne, Webb, & Schumann, 2012). By adulthood, smaller cerebellar
volume has been reported (Hallahan et al., 2009).
Numerous imaging studies have reported cerebellar hypoplasia in autism,
specifically smaller cerebellar vermal lobules VI and VII (e.g., Allen, 2005;
Courchesne et al., 2011, 2012; Courchesne, Yeung-Courchesne,
Hesselink, & Jernigan, 1988). Reduced vermal VI–VII is associated with
ASD symptoms (Kaufmann et al., 2003), including reduced exploration
and increased stereotyped and repetitive movements (Pierce &
Courchesne, 2001). The imaging findings in other vermal lobules are incon-
sistent (e.g., Courchesne et al., 2012; Stanfield et al., 2008; Webb et al.,
2009). Reduced size of the cerebellar hemispheres has also been observed
and is correlated with vermal hypoplasia (Murakami, Courchesne, Press,
Yeung-Courchesne, & Hesselink, 1989). Differences in the volume of
the vermis and anterior lobe and abnormal left-lateralization in lobule
VIIIA have been associated with language impairment in ASD (Hodge
et al., 2010).
Differences in cerebellar WM tracts have also been reported. Increased
diffusivity of the superior cerebellar peduncles suggests abnormal connectiv-
ity between the cerebellum and its rostral connections (Sivaswamy et al.,
2010), and Catani et al. (2008) found a correlation between the degree of
social impairment and the integrity of the superior cerebellar peduncle. In
ASD children, cerebellar WM abnormalities were associated with repetitive
behaviors (Cheung et al., 2009).
Voxel-based morphometry studies have reported both increases and
decreases in cerebellar GM and WM (Fig. 1.3). Decreased GM is consis-
tently found in midline IX, right Crus I, and lobule VIII in ASD, and in
some studies lobule IX was the most significant cluster in the entire brain;
increased GM was reported in lobule VI (Cauda et al., 2011; Duerden,
Mak-Fan, Taylor, & Roberts, 2012; Nickl-Jockschat et al., 2012; Yu,
Cheung, Chua, & McAlonan, 2011). Resting-state activity in lobule IX
and right Crus I most strongly correlates with the default mode network
(Buckner et al., 2011), and healthy males engage Crus I bilaterally during
both theory of mind and empathy tasks (Vollm et al., 2006). In ASD chil-
dren, GM reductions bilaterally in Crus II and in vermal lobules VIII–IX
correlated with communication scores (Riva et al., 2013), and lower GM
in Crus I was associated with increased repetitive and stereotyped behaviors
(Rojas et al., 2006). Although increases in right Crus I have been reported,
these were in a different region of right Crus I that is thought to be part of
Autism Spectrum Disorder and the Cerebellum 11
it is unclear whether the latter result specifically from loss of En2 in the
cerebellum, as En2 is also expressed, albeit at lower levels, in other parts of
the brain, including the cortex and the thalamus (Brielmaier et al., 2012).
RAR-related orphan receptor alpha (ROR-alpha) is another transcrip-
tion factor crucial for cerebellar development that recently has been associ-
ated with autism (SFARI Gene). It has long been known that ROR-alpha is
vital for early PC development (Boukhtouche et al., 2006; Gold, Gent, &
Hamilton, 2007). Both the staggerer mouse, harboring a spontaneous intra-
genic Rora deletion, and targeted Rora knockout mice display abnormal PC
development, followed by progressive loss of PCs and secondary loss of GCs
(Sidman, Lane, & Dickie, 1962) (see also Mouse Genome Database (MGD);
Eppig et al., 2012). Behaviorally, Rora-deficient mice are ataxic and exhibit
impaired motor coordination and learning phenotypes (MGD).
PC development is also known to be under control of the transcription
factor forkhead box 2 (Foxp2). Human mutations in FOXP2 cause devel-
opmental speech and language deficits and are associated with autism
(SFARI Gene, Autism KB). Mice deficient for the Foxp2 gene show impair-
ments in PC development, cerebellar radial glia morphology, and GC
migration (Shu, 2005). Furthermore, mice with a language disorder-
implicated missense mutation (R522H) in Foxp2 also show cerebellar hypo-
plasia, abnormal PC development, cerebellar synaptic deficits, and impaired
motor learning (Fujita et al., 2008; Groszer et al., 2008).
The extracellular matrix protein reelin is important for neuronal migra-
tion in both the cortex and the cerebellum (Rice & Curran, 2001). Several
studies have found a genetic association between the RELN gene and
autism, and rare mutations in RELN have been identified in individuals with
ASD (SFARI Gene). Furthermore, reelin signaling was impaired in the
frontal cortex and cerebellum of autistic postmortem brains (Fatemi et al.,
2005). In the cerebellum, reelin is highly expressed in GCs. Reln mouse
mutants, including the classic spontaneous reeler mouse, display cerebellar
hypoplasia with severely reduced GC numbers and secondary PC migration
deficits (Mariani, Crepel, Mikoshiba, Changeux, & Sotelo, 1977) (MGD).
Behaviorally, Reln mouse mutants display ataxia, motor coordination, and
balance deficits (MGD). Consistent with the findings in mice, human RELN
mutations have also been associated with profound cerebellar hypoplasia
(Hong et al., 2000). The adaptor proteins Dab1 and CrkL are downstream
effectors in the reelin-signaling pathway (Ballif et al., 2004; Park & Curran,
2008). Similar to the reeler mice, mice deficient for Dab1 (scrambler mouse
mutant) or Crkl knockout mice display cerebellar hypofoliation and failure
16 Esther B.E. Becker and Catherine J. Stoodley
treatment of the mice with the mTOR inhibitor rapamycin (Reith et al.,
2011, 2013; Tsai et al., 2012), identifying the mTOR signaling pathway
as a key molecular mechanism in the cerebellum contributing to autistic-like
behavior.
12. CONCLUSIONS
There is ample evidence at multiple levels of inquiry that link differ-
ences in cerebellar structure and function to autism. From mouse models of
autism-related genes to human studies of cerebellar malformations, cerebel-
lar dysfunction is related to the core behaviors that comprise the autism spec-
trum. Vice versa, differences in cerebellar structure and function, and
behavioral evidence of cerebellar-type motor impairments, have been
clearly documented in autistic populations. Given the evidence presented
here, it seems unlikely that changes in cerebellar structure and function in
autism are a mere anatomical beacon of dysfunction elsewhere (Ziats &
Rennert, 2013). Instead, the cerebellum appears to be part of extensive neu-
ral networks that together govern the social, communication, and repeti-
tive/restrictive behaviors impaired in autism. Future research will
undoubtedly extend our current understanding of the link between the cer-
ebellum and autism. It will be important to further characterize the lobular
localization of cellular, molecular, and structural differences in the cerebel-
lum, their relevance to specific motor and nonmotor autistic symptoms, and
the effects of these differences on downstream cortical targets. As extensive
exome and whole genome sequencing studies of autistic patients are now
underway, these discoveries should be linked to specific human gene muta-
tions. This will give important insight into the specific molecular and neural
pathways that underlie distinct autistic traits. The generation of temporally
and spatially conditional mouse models will help to clarify which cell types
and autism symptoms are affected by specific human genetic mutations.
Ultimately, our increasing knowledge of the specific role of the cerebellum
in ASD should lead to better diagnosis and promising targets for more effec-
tive clinical interventions.
ACKNOWLEDGMENT
E. B. is a Royal Society Research Fellow. We thank Friederike Winter for critical reading of
the chapter.
22 Esther B.E. Becker and Catherine J. Stoodley
REFERENCES
Albrecht, U., Sutcliffe, J. S., Cattanach, B. M., Beechey, C. V., Armstrong, D., Eichele, G.,
et al. (1997). Imprinted expression of the murine Angelman syndrome gene, Ube3a, in
hippocampal and Purkinje neurons. Nature Genetics, 17(1), 75–78.
Aldinger, K. A., Kogan, J., Kimonis, V., Fernandez, B., Horn, D., Klopocki, E., et al. (2013).
Cerebellar and posterior fossa malformations in patients with autism-associated chromo-
some 22q13 terminal deletion. American Journal of Medical Genetics A, 161A(1), 131–136.
Allen, G. (2005). The cerebellum in autism. Clinical Neuropsychiatry, 2(6), 321–337.
Allen, G., & Courchesne, E. (2003). Differential effects of developmental cerebellar abnor-
mality on cognitive and motor functions in the cerebellum: An fMRI study of autism.
American Journal of Psychiatry, 160(2), 262–273.
Amaral, D. G., Schumann, C. M., & Nordahl, C. W. (2008). Neuroanatomy of autism.
Trends in Neurosciences, 31(3), 137–145.
American Psychiatric Association (2013). Diagnostic and statistical manual of mental disorders
(DSM-5) (5th ed.). Washington, DC: American Psychiatric Association.
Backman, S. A., Stambolic, V., Suzuki, A., Haight, J., Elia, A., Pretorius, J., et al. (2001).
Deletion of Pten in mouse brain causes seizures, ataxia and defects in soma size resem-
bling Lhermitte-Duclos disease. Nature Genetics, 29(4), 396–403.
Bailey, A., Luthert, P., Dean, A., Harding, B., Janota, I., Montgomery, M., et al. (1998).
A clinicopathological study of autism. Brain, 121(5), 889–905.
Ballif, B. A., Arnaud, L., Arthur, W. T., Guris, D., Imamoto, A., & Cooper, J. A. (2004).
Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Current
Biology, 14(7), 606–610.
Balsters, J. H., Cussans, E., Diedrichsen, J., Phillips, K. A., Preuss, T. M., Rilling, J. K., et al.
(2010). Evolution of the cerebellar cortex: The selective expansion of prefrontal-
projecting cerebellar lobules. NeuroImage, 49(3), 2045–2052.
Basu, S. N., Kollu, R., & Banerjee-Basu, S. (2009). AutDB: A gene reference resource for
autism research. Nucleic Acids Research, 37, D832–D836.
Baudouin, S. J., Gaudias, J., Gerharz, S., Hatstatt, L., Zhou, K., Punnakkal, P., et al. (2012).
Shared synaptic pathophysiology in syndromic and nonsyndromic rodent models of
autism. Science, 338(6103), 128–132.
Bauman, M. L., & Kemper, T. L. (2005). Neuroanatomic observations of the brain in autism:
A review and future directions. International Journal of Developmental Neuroscience, 23(2–3),
183–187.
Bauman, M. L., Kemper, T. L., & Arin, D. M. (1995). Pervasive neuroanatomic abnormal-
ities of the brain in three cases of Rett’s syndrome. Neurology, 45(8), 1581–1586.
Belichenko, N. P., Belichenko, P. V., Li, H. H., Mobley, W. C., & Francke, U. (2008).
Comparative study of brain morphology in Mecp2mutant mouse models of Rett syn-
drome. The Journal of Comparative Neurology, 508(1), 184–195.
Belmonte, M. K., Gomot, M., & Baron-Cohen, S. (2010). Visual attention in autism fam-
ilies: ‘Unaffected’ sibs share atypical frontal activation. Journal of Child Psychology and Psy-
chiatry, 51(3), 259–276.
Ben-Shachar, S., Chahrour, M., Thaller, C., Shaw, C. A., & Zoghbi, H. Y. (2009). Mouse
models of MeCP2 disorders share gene expression changes in the cerebellum and hypo-
thalamus. Human Molecular Genetics, 18(13), 2431–2442.
Berg, J. M., & Geschwind, D. H. (2012). Autism genetics: Searching for specificity and con-
vergence. Genome Biology, 13(7), 247.
Betancur, C. (2011). Etiological heterogeneity in autism spectrum disorders: More than 100
genetic and genomic disorders and still counting. Brain Research, 1380, 42–77.
Betancur, C., Sakurai, T., & Buxbaum, J. D. (2009). The emerging role of synaptic
cell-adhesion pathways in the pathogenesis of autism spectrum disorders. Trends in
Neurosciences, 32(7), 402–412.
Autism Spectrum Disorder and the Cerebellum 23
Bhat, A. N., Galloway, J. C., & Landa, R. J. (2012). Relation between early motor delay and
later communication delay in infants at risk for autism. Infant Behavior and Development,
35(4), 838–846.
Böckers, T. M., Mameza, M. G., Kreutz, M. R., Bockmann, J., Weise, C., Buck, F., et al.
(2001). Synaptic scaffolding proteins in rat brain. The Journal of Biological Chemistry,
276(43), 40104–40112.
Boer, K., Troost, D., Jansen, F., Nellist, M., van den Ouweland, A. M. W., Geurts, J. J. G.,
et al. (2008). Clinicopathological and immunohistochemical findings in an autopsy case
of tuberous sclerosis complex. Neuropathology, 28(6), 577–590.
Bolduc, M. E., du Plessis, A. J., Sullivan, N., Guizard, N., Zhang, X., Robertson, R. L., et al.
(2012). Regional cerebellar volumes predict functional outcome in children with cere-
bellar malformations. Cerebellum, 11(2), 531–542.
Bolduc, M. E., Du Plessis, A. J., Sullivan, N., Khwaja, O. S., Zhang, X., Barnes, K., et al.
(2011). Spectrum of neurodevelopmental disabilities in children with cerebellar mal-
formations. Developmental Medicine and Child Neurology, 53(5), 409–416.
Bolduc, M. E., & Limperopoulos, C. (2009). Neurodevelopmental outcomes in children
with cerebellar malformations: A systematic review. Developmental Medicine and Child
Neurology, 51(4), 256–267.
Boukhtouche, F., Janmaat, S., Vodjdani, G., Gautheron, V., Mallet, J., Dusart, I., et al.
(2006). Retinoid-related orphan receptor alpha controls the early steps of Purkinje cell
dendritic differentiation. The Journal of Neuroscience, 26(5), 1531–1538.
Bozdagi, O., Sakurai, T., Papapetrou, D., Wang, X., Dickstein, D. L., Takahashi, N., et al.
(2010). Haploinsufficiency of the autism-associated Shank3 gene leads to deficits
in synaptic function, social interaction, and social communication. Molecular Autism,
1(1), 15.
Braunschweig, D., Ashwood, P., Krakowiak, P., Hertz-Picciotto, I., Hansen, R.,
Croen, L. A., et al. (2008). Autism: Maternally derived antibodies specific for fetal brain
proteins. Neurotoxicology, 29(2), 226–231.
Braunschweig, D., Duncanson, P., Boyce, R., Hansen, R., Ashwood, P., Pessah, I. N., et al.
(2012). Behavioral correlates of maternal antibody status among children with autism.
Journal of Autism and Developmental Disorders, 42(7), 1435–1445.
Braunschweig, D., & Van de Water, J. (2012). Maternal autoantibodies in autism. Archives of
Neurology, 69(6), 693–699.
Brielmaier, J., Matteson, P. G., Silverman, J. L., Senerth, J. M., Kelly, S., Genestine, M., et al.
(2012). Autism-relevant social abnormalities and cognitive deficits in engrailed-2 knock-
out mice. PLoS One, 7(7), e40914.
Buckner, R. L., Krienen, F. M., Castellanos, A., Diaz, J. C., & Yeo, B. T. (2011). The orga-
nization of the human cerebellum estimated by intrinsic functional connectivity. Journal
of Neurophysiology, 106(5), 2322–2345.
Caria, A., Venuti, P., & de Falco, S. (2011). Functional and dysfunctional brain circuits
underlying emotional processing of music in autism spectrum disorders. Cerebral Cortex,
21(12), 2838–2849.
Catani, M., Jones, D. K., Daly, E., Embiricos, N., Deeley, Q., Pugliese, L., et al. (2008).
Altered cerebellar feedback projections in Asperger syndrome. NeuroImage, 41,
1184–1191.
Cauda, F., Geda, E., Sacco, K., D’Agata, F., Duca, S., Geminiani, G., et al. (2011).
Grey matter abnormality in autism spectrum disorder: An activation likelihood estima-
tion meta-analysis study. Journal of Neurology, Neurosurgery, and Psychiatry, 82(12),
1304–1313.
Chang, C. H., Wade, M. G., Stoffregen, T. A., Hsu, C. Y., & Pan, C. Y. (2010). Visual tasks
and postural sway in children with and without autism spectrum disorders. Research in
Developmental Disabilities, 31(6), 1536–1542.
24 Esther B.E. Becker and Catherine J. Stoodley
Cheh, M. A., Millonig, J. H., Roselli, L. M., Ming, X., Jacobsen, E., Kamdar, S., et al.
(2006). En2 knockout mice display neurobehavioral and neurochemical alterations rel-
evant to autism spectrum disorder. Brain Research, 1116(1), 166–176.
Chen, R. Z., Akbarian, S., Tudor, M., & Jaenisch, R. (2001). Deficiency of methyl-CpG
binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nature
Genetics, 27(3), 327–331.
Cheron, G., Servais, L., Wagstaff, J., & Dan, B. (2005). Fast cerebellar oscillation asso-
ciated with ataxia in a mouse model of Angelman syndrome. Neuroscience, 130(3),
631–637.
Cheung, C., Chua, S. E., Cheung, V., Khong, P. L., Tai, K. S., Wong, T. K., et al. (2009).
White matter fractional anisotrophy differences and correlates of diagnostic symptoms in
autism. Journal of Child Psychology and Psychiatry, 50(9), 1102–1112.
Cisternas, F. A., Vincent, J. B., Scherer, S. W., & Ray, P. N. (2003). Cloning and charac-
terization of human CADPS and CADPS2, new members of the Ca2þ-dependent acti-
vator for secretion protein family. Genomics, 81(3), 279–291.
Corbett, B. A., Carmean, V., Ravizza, S., Wendelken, C., Henry, M. L., Carter, C., et al.
(2009). A functional and structural study of emotion and face processing in children with
autism. Psychiatry Research, 173(3), 196–205.
Cornish, K., Kogan, C., Turk, J., Manly, T., James, N., Mills, A., et al. (2005). The emerging
fragile X premutation phenotype: Evidence from the domain of social cognition. Brain
and Cognition, 57(1), 53–60.
Courchesne, E., Campbell, K., & Solso, S. (2011). Brain growth across the life span in autism:
Age-specific changes in anatomical pathology. Brain Research, 1380, 138–145.
Courchesne, E., Karns, C. M., Davis, H. R., Ziccardi, R., Carper, R. A., Tigue, Z. D., et al.
(2001). Unusual brain growth patterns in early life in patients with autistic disorder.
Neurology, 57(2), 245–254.
Courchesne, E., Webb, S. J., & Schumann, C. M. (2012). From toddlers to adults: The
changing landscape of the brain in autism. In D. Amaral, D. Geschwind, &
G. Dawson (Eds.), Autism spectrum disorders. Oxford: Oxford University Press.
Courchesne, E., Yeung-Courchesne, R., Hesselink, J. R., & Jernigan, T. L. (1988). Hypo-
plasia of cerebellar vermal lobules VI and VII in autism. The New England Journal of Med-
icine, 318(21), 1349–1354.
Critchley, H., Daly, E., Phillips, M., Brammer, M., Bullmore, E., Williams, S., et al. (2000).
Explicit and implicit neural mechanisms for processing of social information from facial
expressions: A functional magnetic resonance imaging study. Human Brain Mapping, 9(2),
93–105.
Croen, L. A., Braunschweig, D., Haapanen, L., Yoshida, C. K., Fireman, B., Grether, J. K.,
et al. (2008). Maternal mid-pregnancy autoantibodies to fetal brain protein: The early
markers for autism study. Biological Psychiatry, 64(7), 583–588.
Dalton, P., Deacon, R., Blamire, A., Pike, M., McKinlay, I., Stein, J., et al. (2003). Maternal
neuronal antibodies associated with autism and a language disorder. Annals of Neurology,
53(4), 533–537.
DeLorey, T. M., Handforth, A., Anagnostaras, S. G., Homanics, G. E., Minassian, B. A.,
Asatourian, A., et al. (1998). Mice lacking the beta3 subunit of the GABAA receptor
have the epilepsy phenotype and many of the behavioral characteristics of Angelman syn-
drome. The Journal of Neuroscience, 18(20), 8505–8514.
DeLorey, T. M., Sahbaie, P., Hashemi, E., Homanics, G. E., & Clark, J. D. (2008). Gabrb3
gene deficient mice exhibit impaired social and exploratory behaviors, deficits in non-
selective attention and hypoplasia of cerebellar vermal lobules: A potential model of
autism spectrum disorder. Behavioural Brain Research, 187(2), 207–220.
Devlin, B., & Scherer, S. W. (2012). Genetic architecture in autism spectrum disorder.
Current Opinion in Genetics and Development, 22(3), 229–237.
Autism Spectrum Disorder and the Cerebellum 25
Diedrichsen, J., Balsters, J. H., Flavell, J., Cussans, E., & Ramnani, N. (2009). A probabilistic
MR atlas of the human cerebellum. Neuroimage, 46, 39–46.
Diedrichsen, J. (2006). A spatially unbiased atlas template of the human cerebellum.
Neuroimage, 33, 127–138.
Di Martino, A., Yan, C.-G., Li, Q., Denio, E., Castellanos, F. X., Alaerts, K., et al. (2013).
The autism brain imaging data exchange: Towards a large-scale evaluation of the intrinsic
brain architecture in autism. Molecular Psychiatry, http://dx.doi.org/10.1038/
mp.2013.78. (Epub ahead of Print).
Dindot, S. V., Antalffy, B. A., Bhattacharjee, M. B., & Beaudet, A. L. (2008). The Angelman
syndrome ubiquitin ligase localizes to the synapse and nucleus, and maternal deficiency
results in abnormal dendritic spine morphology. Human Molecular Genetics, 17(1),
111–118.
Duerden, E. G., Mak-Fan, K. M., Taylor, M. J., & Roberts, S. W. (2012). Regional differ-
ences in grey and white matter in children and adults with autism spectrum disorders: An
activation likelihood estimate (ALE) meta-analysis. Autism Research, 5(1), 49–66.
Dziuk, M. A., Gidley Larson, J. C., Apostu, A., Mahone, E. M., Denckla, M. B., &
Mostofsky, S. H. (2007). Dyspraxia in autism: Association with motor, social, and com-
municative deficits. Developmental Medicine and Child Neurology, 49(10), 734–739.
Ecker, C., Rocha-Rego, V., Johnston, P., Mourao-Miranda, J., Marquand, A., Daly, E. M.,
et al. (2010). Investigating the predictive value of whole-brain structural MR scans in
autism: A pattern classification approach. NeuroImage, 49(1), 44–56.
Egawa, K., Kitagawa, K., Inoue, K., Takayama, M., Takayama, C., Saitoh, S., et al. (2012).
Decreased tonic inhibition in cerebellar granule cells causes motor dysfunction in a
mouse model of Angelman Syndrome. Science Translational medicine, 4(163), 163ra157.
Ellegood, J., Pacey, L. K., Hampson, D. R., Lerch, J. P., & Henkelman, R. M. (2010). Ana-
tomical phenotyping in a mouse model of fragile X syndrome with magnetic resonance
imaging. NeuroImage, 53(3), 1023–1029.
Eluvathingal, T. J., Behen, M. E., Chugani, H. T., Janisse, J., Bernardi, B., Chakraborty, P.,
et al. (2006). Cerebellar lesions in tuberous sclerosis complex. Journal of Child Neurology,
21(10), 846–851.
Eppig, J. T., Blake, J. A., Bult, C. J., Kadin, J. A., Richardson, J. E., & Mouse Genome
Database Group (2012). The Mouse Genome Database (MGD): Comprehensive
resource for genetics and genomics of the laboratory mouse. Nucleic Acids Research,
40, D881–D886.
Esposito, G., Venuti, P., Apicella, F., & Muratori, F. (2011). Analysis of unsupported gait in
toddlers with autism. Brain and Development, 33(5), 367–373.
Fatemi, S. H., Aldinger, K. A., Ashwood, P., Bauman, M. L., Blaha, C. D., Blatt, G. J., et al.
(2012). Consensus paper: Pathological role of the cerebellum in autism. Cerebellum,
11(3), 777–807.
Fatemi, S. H., Halt, A. R., Realmuto, G., Earle, J., Kist, D. A., Thuras, P., et al. (2002).
Purkinje cell size is reduced in cerebellum of patients with autism. Cellular and Molecular
Neurobiology, 22(2), 171–175.
Fatemi, S. H., Reutiman, T. J., Folsom, T. D., & Thuras, P. D. (2008). GABAA receptor
downregulation in brains of subjects with autism. Journal of Autism and Developmental
Disorders, 39(2), 223–230.
Fatemi, S. H., Snow, A. V., Stary, J. M., Araghi-Niknam, M., Reutiman, T. J., Lee, S., et al.
(2005). Reelin signaling is impaired in autism. Biological Psychiatry, 57(7), 777–787.
Flanagan, J. E., Landa, R., Bhat, A., & Bauman, M. (2012). Head lag in infants at risk for
autism: A preliminary study. American Journal of Occupational Therapy, 66(5), 577–585.
Fournier, K. A., Hass, C. J., Naik, S. K., Lodha, N., & Cauraugh, J. H. (2010). Motor coor-
dination in autism spectrum disorders: A synthesis and meta-analysis. Journal of Autism and
Developmental Disorders, 40(10), 1227–1240.
26 Esther B.E. Becker and Catherine J. Stoodley
Fournier, K. A., Kimberg, C. I., Radonovich, K. J., Tillman, M. D., Chow, J. W.,
Lewis, M. H., et al. (2010). Decreased static and dynamic postural control in children
with autism spectrum disorders. Gait and Posture, 32(1), 6–9.
Freitag, C. M., Kleser, C., Schneider, M., & von Gontard, A. (2007). Quantitative assessment
of neuromotor function in adolescents with high functioning autism and Asperger syn-
drome. Journal of Autism and Developmental Disorders, 37(5), 948–959.
Frith, U. (1991). Autism and Asperger syndrome. Cambridge: Cambridge University Press.
Fujita, E., Tanabe, Y., Shiota, A., Ueda, M., Suwa, K., Momoi, M. Y., et al. (2008). Ultra-
sonic vocalization impairment of Foxp2 (R552H) knockin mice related to speech-
language disorder and abnormality of Purkinje cells. Proceedings of the National Academy
of Sciences of the United States of America, 105(8), 3117–3122.
Furge, K. A., Zhang, Y. W., & Van de Woude, G. F. (2000). Met receptor tyrosine kinase:
Enhanced signaling through adapter proteins. Oncogene, 19(49), 5582–5589.
Gaffrey, M. S., Kleinhans, N. M., Haist, F., Akshoomoff, N., Campbell, A., Courchesne, E.,
et al. (2007). Atypical participation of visual cortex during word processing in autism: An
fMRI study of semantic decision. Neuropsychologia, 45(8), 1672–1684.
Gepner, B., & Mestre, D. (2002). Rapid visual-motion integration deficit in autism. Trends in
Cognitive Sciences, 6(11), 455.
Gernsbacher, M. A., Sauer, E. A., Geye, H. M., Schweigert, E. K., & Hill Goldsmith, H.
(2008). Infant and toddler oral- and manual-motor skills predict later speech fluency
in autism. Journal of Child Psychology and Psychiatry, 49(1), 43–50.
Gidley Larson, J. C., & Mostofsky, S. H. (2006). Motor deficits in autism. In R. Tuchman &
I. Rapin (Eds.), Autism: A neurological disorder of early brain development (pp. 231–247).
London: MacKeith Press.
Gilbert, S. J., Bird, G., Brindley, R., Frith, C. D., & Burgess, P. W. (2008). Atypical recruit-
ment of medial prefrontal cortex in autism spectrum disorders: An fMRI study of two
executive function tasks. Neuropsychologia, 46(9), 2281–2291.
Goffin, D., Allen, M., Zhang, L., Amorim, M., Wang, I.-T. J., Reyes, A.-R. S., et al. (2011).
Rett syndrome mutation MeCP2 T158A disrupts DNA binding, protein stability and
ERP responses. Nature Neuroscience, 15(2), 274–283.
Goines, P., Haapanen, L., Boyce, R., Duncanson, P., Braunschweig, D., Delwiche, L., et al.
(2011). Autoantibodies to cerebellum in children with autism associate with behavior.
Brain, Behavior, and Immunity, 25(3), 514–523.
Gold, D. A., Gent, P. M., & Hamilton, B. A. (2007). ROR alpha in genetic control of cer-
ebellum development: 50 staggering years. Brain Research, 1140, 19–25.
Gomot, M., Bernard, F. A., Davis, M. H., Belmonte, M. K., Ashwin, C., Bullmore, E. T.,
et al. (2006). Change detection in children with autism: An auditory event-related fMRI
study. NeuroImage, 29(2), 475–484.
Gowen, E., & Hamilton, A. (2013). Motor abilities in autism: a review using a computational
context. Journal of Autism and Developmental Disorders, 43, 323–344.
Greco, C. M., Navarro, C. S., Hunsaker, M. R., Maezawa, I., Shuler, J. F., Tassone, F., et al.
(2011). Neuropathologic features in the hippocampus and cerebellum of three older men
with fragile X syndrome. Molecular Autism, 2(1), 2.
Green, D., Baird, G., Barnett, A. L., Henderson, L., Huber, J., & Henderson, S. E. (2002).
The severity and nature of motor impairment in Asperger’s syndrome: A comparison
with specific developmental disorder of motor function. Journal of Child Psychology and
Psychiatry, 43(5), 655–668.
Green, D., Charman, T., Pickles, A., Chandler, S., Loucas, T., Simonoff, E., et al. (2009).
Impairment in movement skills of children with autistic spectrum disorders. Developmen-
tal Medicine and Child Neurology, 51(4), 311–316.
Greffou, S., Bertone, A., Hahler, E. M., Hanssens, J. M., Mottron, L., & Faubert, J. (2012).
Postural hypo-reactivity in autism is contingent on development and visual
Autism Spectrum Disorder and the Cerebellum 27
environment: A fully immersive virtual reality study. Journal of Autism and Developmental
Disorders, 42(6), 961–970.
Groen, W. B., Tesink, C., Petersson, K. M., van Berkum, J., van der Gaag, R. J.,
Hagoort, P., et al. (2010). Semantic, factual, and social language comprehension in ado-
lescents with autism: An fMRI study. Cerebral Cortex, 20(8), 1937–1945.
Groszer, M., Keays, D. A., Deacon, R. M. J., de Bono, J. P., Prasad-Mulcare, S., Gaub, S.,
et al. (2008). Impaired synaptic plasticity and motor learning in mice with a point muta-
tion implicated in human speech deficits. Current Biology, 18(5), 354–362.
Gutmann, D. H., Zhang, Y., Hasbani, M. J., Goldberg, M. P., Plank, T. L., & Petri
Henske, E. (2000). Expression of the tuberous sclerosis complex gene products, hamartin
and tuberin, in central nervous system tissues. Acta Neuropathologica, 99(3), 223–230.
Guy, J., Hendrich, B., Holmes, M., Martin, J. E., & Bird, A. (2001). A mouse Mecp2-null
mutation causes neurological symptoms that mimic Rett syndrome. Nature Genetics,
27(3), 322–326.
Haist, F., Adamo, M., Westerfield, M., Courchesne, E., & Townsend, J. (2005). The func-
tional neuroanatomy of spatial attention in autism spectrum disorder. Developmental
Neuropsychology, 27(3), 425–458.
Hallahan, B., Daly, E. M., McAlonan, G., Loth, E., Toal, F., O’Brien, F., et al. (2009). Brain
morphometry volume in autistic spectrum disorder: A magnetic resonance imaging study
of adults. Psychological Medicine, 39(2), 337–346.
Hallett, M., Lebiedowska, M. K., Thomas, S. L., Stanhope, S. J., Denckla, M. B., &
Rumsey, J. (1993). Locomotion of autistic adults. Archives of Neurology, 50(12),
1304–1308.
Hallmayer, J., Cleveland, S., Torres, A., Phillips, J., Cohen, B., Torigoe, T., et al. (2011).
Genetic heritability and shared environmental factors among twin pairs with autism.
Archives of General Psychiatry, 68(11), 1095–1102.
Harris, G. J., Chabris, C. F., Clark, J., Urban, T., Aharon, I., Steele, S., et al. (2006). Brain
activation during semantic processing in autism spectrum disorders via functional mag-
netic resonance imaging. Brain and Cognition, 61(1), 54–68.
Heath, R. G. (1977). Modulation of emotion with a brain pacemamer. Treatment for intrac-
table psychiatric illness. Journal of Nervous and Mental Disease, 165(5), 300–317.
Heck, D. H., Zhao, Y., Roy, S., LeDoux, M. S., & Reiter, L. T. (2008). Analysis of cere-
bellar function in Ube3a-deficient mice reveals novel genotype-specific behaviors.
Human Molecular Genetics, 17(14), 2181–2189.
Herbert, M. R. (2010). Contributions of the environment and environmentally vulnerable
physiology to autism spectrum disorders. Current Opinion in Neurology, 23(2), 103–110.
Herculano-Houzel, S. (2010). Coordinated scaling of cortical and cerebellar numbers of neu-
rons. Frontiers in Neuroanatomy, 4, 12.
Hesling, I., Dilharreguy, B., Peppe, S., Amirault, M., Bouvard, M., & Allard, M. (2010). The
integration of prosodic speech in high functioning autism: A preliminary fMRI study.
PLoS One, 5(7), e11571.
Heuer, L., Braunschweig, D., Ashwood, P., Van de Water, J., & Campbell, D. B. (2011).
Association of a MET genetic variant with autism-associated maternal autoantibodies
to fetal brain proteins and cytokine expression. Translational Psychiatry, 1(10), e48.
Hilton, C. L., Zhang, Y., Whilte, M. R., Klohr, C. L., & Constantino, J. (2012). Motor
impairment in sibling pairs concordant and discordant for autism spectrum disorders.
Autism, 16(4), 430–441.
Hodge, S. M., Makris, N., Kennedy, D. N., Caviness, V. S., Jr., Howard, J., McGrath, L.,
et al. (2010). Cerebellum, language, and cognition in autism and specific language
impairment. Journal of Autism and Developmental Disorders, 40(3), 300–316.
Honda, S., Kagoshima, M., Wanaka, A., Tohyama, M., Matsumoto, K., & Nakamura, T.
(1995). Localization and functional coupling of HGF and c-Met/HGF receptor in rat
28 Esther B.E. Becker and Catherine J. Stoodley
brain: Implication as neurotrophic factor. Brain Research Molecular Brain Research, 32(2),
197–210.
Hong, S. E., Shugart, Y. Y., Huang, D. T., Shahwan, S. A., Grant, P. E., Hourihane, J. O.,
et al. (2000). Autosomal recessive lissencephaly with cerebellar hypoplasia is associated
with human RELN mutations. Nature Genetics, 26(1), 93–96.
Ieraci, A., Forni, P. E., & Ponzetto, C. (2002). Viable hypomorphic signaling mutant of the
Met receptor reveals a role for hepatocyte growth factor in postnatal cerebellar develop-
ment. Proceedings of the National Academy of Sciences, 99(23), 15200–15205.
Ingram, J. L., Peckham, S. M., Tisdale, B., & Rodier, P. M. (2000). Prenatal exposure of rats
to valproic acid reproduces the cerebellar anomalies associated with autism. Neu-
rotoxicology and Teratology, 22(3), 319–324.
Jeste, S. S. (2011). The neurology of autism spectrum disorders. Current Opinion Neurology,
24(2), 132–139.
Jiang, Y., Armstrong, D., Albrecht, U., Atkins, C., Noebels, J., Eichele, G., et al. (1998).
Mutation of the angelman ubiquitin ligase in mice causes increased cytoplasmic p53
and deficits of contextual learning and long-term potentiation. Neuron, 21(4), 799–811.
Jiang, Y.-H., & Ehlers, M. D. (2013). Modeling autism by SHANK gene mutations in mice.
Neuron, 78(1), 8–27.
Jones, V., & Prior, M. (1985). Motor imitation abilities and neurological signs in autistic chil-
dren. Journal of Autism and Developmental Disorders, 15(1), 37–46.
Jordan, C., Li, H. H., Kwan, H. C., & Francke, U. (2007). Cerebellar gene expression pro-
files of mouse models for Rett syndrome reveal novel MeCP2 targets. BMC Medical
Genetics, 8(1), 36.
Kaufmann, W. E., Cooper, K. L., Mostofsky, S. H., Capone, G. T., Kates, W. R.,
Newschaffer, C. J., et al. (2003). Specificity of cerebellar vermian abnormalities in
autism: A quantitative magnetic resonance imaging study. Journal of Child Neurology,
18(7), 463–470.
Kemper, T. L., & Bauman, M. (1998). Neuropathology of infantile autism. Journal of
Neuropathology and Experimental Neurology, 57(7), 645–652.
Kleinhans, N. M., Schweinsburg, B. C., Cohen, D. N., Muller, R. A., & Courchesne, E.
(2007). N-acetyl aspartate in autism spectrum disorders: Regional effects and relationship
to fMRI activation. Brain Research, 1162, 85–97.
Knaus, T. A., Silver, A. M., Lindgren, K. A., Hadjikhani, N., & Tager-Flusberg, H. (2008).
FMRI activation during a language task in adolescents with ASD. Journal of the Interna-
tional Neuropsychological Society, 14(6), 967–979.
Koekkoek, S. K. E., Yamaguchi, K., Milojkovic, B. A., Dortland, B. R., Ruigrok, T. J. H.,
Maex, R., et al. (2005). Deletion of FMR1 in Purkinje cells enhances parallel fiber LTD,
enlarges spines, and attenuates cerebellar eyelid conditioning in fragile X syndrome. Neu-
ron, 47(3), 339–352.
Kuemerle, B., Zanjani, H., Joyner, A., & Herrup, K. (1997). Pattern deformities and cell loss
in Engrailed-2 mutant mice suggest two separate patterning events during cerebellar
development. The Journal of Neuroscience, 17(20), 7881–7889.
Kwon, C.-H., Zhu, X., Zhang, J., Knoop, L. L., Tharp, R., Smeyne, R. J., et al. (2001).
Pten regulates neuronal soma size: A mouse model of Lhermitte-Duclos disease. Nature
Genetics, 29(4), 404–411.
Landa, R., & Garrett-Mayer, E. (2006). Development in infants with autism spectrum dis-
orders: A prospective study. Journal of Child Psychology and Psychiatry, 47(6), 629–638.
Laurie, D. J., Wisden, W., & Seeburg, P. H. (1992). The distribution of thirteen GABAA
receptor subunit mRNAs in the rat brain. III. Embryonic and postnatal development.
The Journal of Neuroscience, 12(11), 4151–4172.
Limperopoulos, C., Bassan, H., Gauvreau, K., Jr., Robertson, R. L., Sullivan, N. R.,
Benson, C. B., et al. (2007). Does cerebellar injury in premature infants contribute to
Autism Spectrum Disorder and the Cerebellum 29
the high prevalence of long-term cognitive, learning, and behavioral disability in survi-
vors? Pediatrics, 120, 584–593.
Limperopoulos, C., Bassan, H., Sullivan, N. R., Soul, J. S., Robertson, R. L., Jr., Moore, M.,
et al. (2008). Positive screening for autism in ex-preterm infants: Prevalence and risk fac-
tors. Pediatrics, 121(4), 758–765.
Limperopoulos, C., Chilingaryan, G., Guizard, N., Robertson, R. L., & Du Plessis, A. J.
(2010). Cerebellar injury in the premature infant is associated with impaired growth
of specific cerebral regions. Pediatric Research, 68(2), 145–150.
Linkenauger, S. A., Lerner, M. D., Ramenzoni, V. C., & Proffitt, D. R. (2012). A perceptual-
motor deficit predicts social and communicative impairments in individuals with autism
spectrum disorders. Autism Research, 5(5), 352–362.
MacNeil, L. K., & Mostofsky, S. H. (2012). Specificity of dyspraxia in children with autism.
Neuropsychology, 26(2), 165–171.
Mariani, J., Crepel, F., Mikoshiba, K., Changeux, J. P., & Sotelo, C. (1977). Anatomical,
physiological and biochemical studies of the cerebellum from Reeler mutant mouse.
Philosophical Transactions of the Royal Society B: Biological Sciences, 281(978), 1–28.
Marino, S., Krimpenfort, P., Leung, C., van der Korput, H. A. G. M., Trapman, J.,
Camenisch, I., et al. (2002). PTEN is essential for cell migration but not for fate
determination and tumourigenesis in the cerebellum. Development, 129(14), 3513–3522.
Millen, K. J., Wurst, W., Herrup, K., & Joyner, A. L. (1994). Abnormal embryonic cerebellar
development and patterning of postnatal foliation in two mouse Engrailed-2 mutants.
Development, 120, 695–706.
Minshew, N. J., Sung, K., Jones, B. L., & Furman, J. M. (2004). Underdevelopment of the
postural control system in autism. Neurology, 63(11), 2056–2061.
Miura, K., Kishino, T., Li, E., Webber, H., Dikkes, P., Holmes, G. L., et al. (2002). Neu-
robehavioral and electroencephalographic abnormalities in Ube3a maternal-deficient
mice. Neurobiology of Disease, 9(2), 149–159.
Molloy, C. A., Dietrich, K. N., & Bhattacharya, A. (2003). Postural stability in children
with autism spectrum disorder. Journal of Autism and Developmental Disorders, 33(6),
643–652.
Moran, M. F., Foley, J. T., Parker, M. E., & Weiss, M. J. (2013). Two-legged hopping in
autism spectrum disorders. Frontiers in Integrative Neuroscience, 7, 14.
Mosconi, M. W., Kay, M., D’Cruz, A. M., Guter, S., Kapur, K., Macmillan, C., et al. (2010).
Neurobehavioral abnormalities in first-degree relatives of individuals with autism.
Archives of General Psychiatry, 67(8), 830–840.
Mosconi, M. W., Luna, B., Kay-Stacey, M., Nowinski, C. V., Rubin, L. H., Scudder, C.,
et al. (2013). Saccade adaptation abnormalities implicate dysfunction of cerebellar-
dependent learning mechanisms in autism spectrum disorders (ASD). PLoS One, 8(5),
e63709.
Mostofsky, S. H., Goldberg, M. C., Landa, R. J., & Denckla, M. B. (2000). Evidence for a
deficit in procedural learning in children and adolescents with autism: Implications for
cerebellar contribution. Journal of the International Neuropsychological Society, 6(7),
752–759.
Mostofsky, S. H., Mazzocco, M. M., Aakalu, G., Warsofsky, I. S., Denckla, M. B., &
Reiss, A. L. (1998). Decreased cerebellar posterior vermis size in fragile X syndrome:
Correlation with neurocognitive performance. Neurology, 50(1), 121–130.
Mostofsky, S. H., Powell, S. K., Simmonds, D. J., Goldberg, M. C., Caffo, B., & Pekar, J. J.
(2009). Decreased connectivity and cerebellar activity in autism during motor task per-
formance. Brain, 132(Pt. 9), 2413–2425.
Mullaney, B. C., Johnston, M. V., & Blue, M. E. (2004). Developmental expression of
methyl-CpG binding protein 2 is dynamically regulated in the rodent brain. Neuroscience,
123(4), 939–949.
30 Esther B.E. Becker and Catherine J. Stoodley
Muller, R. A., Kleinhans, N., Kemmotsu, N., Pierce, K., & Courchesne, E. (2003). Abnor-
mal variability and distribution of functional maps in autism: An fMRI study of
visuomotor learning. American Journal of Psychiatry, 160(10), 1847–1862.
Murakami, J. W., Courchesne, E., Haas, R. H., Press, G. A., & Yeung-Courchesne, R.
(1992). Cerebellar and cerebral abnormalities in Rett syndrome: A quantitative MR
analysis. American Journal of Roentgenology, 159(1), 177–183.
Murakami, J. W., Courchesne, E., Press, G. A., Yeung-Courchesne, R., & Hesselink, J. R.
(1989). Reduced cerebellar hemisphere size and its relationship to vermal hypoplasia in
autism. Archives of Neurology, 46(6), 689–694.
Mychasiuk, R., Richards, S., Nakahashi, A., Kolb, B., & Gibb, R. (2012). Effects of rat pre-
natal exposure to valproic acid on behaviour and neuro-anatomy. Developmental Neuro-
science, 34(2–3), 268–276.
Nickl-Jockschat, T., Habel, U., Michel, T. M., Manning, J., Laird, A. R., Fox, P. T., et al.
(2012). Brain structure anomalies in autism spectrum disorder—A meta-analysis of VBM
studies using anatomic likelihood estimation. Human Brain Mapping, 33(6), 1470–1489.
Noterdaeme, M., Mildenberger, K., Minow, F., & Amorosa, H. (2002). Evaluation of
neuromotor deficits in children with autism and children with a specific speech and lan-
guage disorder. European Child and Adolescent Psychiatry, 11(5), 219–225.
Oldfors, A., Sourander, P., Armstrong, D. L., Percy, A. K., Witt-Engerström, I., &
Hagberg, B. A. (1990). Rett syndrome: Cerebellar pathology. Pediatric Neurology,
6(5), 310–314.
Ozonoff, S., Williams, B. J., Gale, S., & Miller, J. N. (1999). Autism and autistic behavior in
Joubert syndrome. Journal of Child Neurology, 14(10), 636–641.
Paakki, J. J., Rahko, J., Long, X., Moilanen, I., Tervonen, O., Nikkinen, J., et al. (2010).
Alterations in regional homogeneity of resting-state brain activity in autism spectrum dis-
orders. Brain Research, 1321, 169–179.
Palmen, S. J. M. C. (2004). Neuropathological findings in autism. Brain, 127(12),
2572–2583.
Papadopoulos, N., McGinley, J., Tonge, B., Bradshaw, J., Saunders, K., Murphy, A., et al.
(2012). Motor proficiency and emotional/behavioural disturbance in autism and
Asperger’s disorder: Another piece of the neurological puzzle? Autism, 16(6), 627–640.
Park, T. J., & Curran, T. (2008). Crk and Crk-like play essential overlapping roles down-
stream of disabled-1 in the reelin pathway. Journal of Neuroscience, 28(50), 13551–13562.
Peça, J., Feliciano, C., Ting, J. T., Wang, W., Wells, M. F., Venkatraman, T. N., et al.
(2011). Shank3 mutant mice display autistic-like behaviours and striatal dysfunction.
Nature, 472(7344), 437–442.
Peça, J., & Feng, G. (2012). Cellular and synaptic network defects in autism. Current Opinion
in Neurobiology, 22(5), 866–872.
Pierce, K., & Courchesne, E. (2001). Evidence for a cerebellar role in reduced exploration
and stereotyped behavior in autism. Biological Psychiatry, 49(8), 655–664.
Pitskel, N. B., Bolling, D. Z., Hudac, C. M., Lantz, S. D., Minshew, N. J., Vander
Wyk, B. C., et al. (2011). Brain mechanisms for processing direct and averted gaze in
individuals with autism. Journal of Autism and Developmental Disorders, 41(12), 1686–1693.
Radonovich, K. J., Fournier, K. A., & Hass, C. J. (2013). Relationship between postural
control and restricted, repetitive behaviors in autism spectrum disorders. Frontiers in
Integrative Neuroscience, 7, 28.
Radyushkin, K., Hammerschmidt, K., Boretius, S., Varoqueaux, F., El-Kordi, A.,
Ronnenberg, A., et al. (2009). Neuroligin-3-deficient mice: Model of a monogenic
heritable form of autism with an olfactory deficit. Genes, Brain, and Behavior, 8(4),
416–425.
Autism Spectrum Disorder and the Cerebellum 31
Reith, R. M., McKenna, J., Wu, H., Hashmi, S. S., Cho, S.-H., Dash, P. K., et al. (2013).
Loss of Tsc2 in Purkinje cells is associated with autistic-like behavior in a mouse model of
tuberous sclerosis complex. Neurobiology of Disease, 51, 93–103.
Reith, R. M., Way, S., McKenna, J., III., Haines, K., & Gambello, M. J. (2011). Loss
of the tuberous sclerosis complex protein tuberin causes Purkinje cell degeneration.
Neurobiology of Disease, 43(1), 113–122.
Reynolds, S., Millette, A., & Devine, P. D. (2012). Sensory and motor characterization in the
postnatal valproate rat model of autism. Developmental Neuroscience, 34(2–3), 258–267.
Rice, D. S., & Curran, T. (2001). Role of the reelin signaling pathway in central nervous
system development. Annual Review of Neuroscience, 24, 1005–1039.
Rinehart, N. J., Bellgrove, M. A., Tonge, B. J., Brereton, A. V., Howells-Rankin, D., &
Bradshaw, J. L. (2006). An examination of movement kinematics in young people with
high-functioning autism and Asperger’s disorder: Further evidence for a motor planning
deficit. Journal of Autism and Developmental Disorders, 36(6), 757–767.
Rinehart, N. J., Tonge, B. J., Iansek, R., McGinley, J., Brereton, A. V., Enticott, P. G., et al.
(2006). Gait function in newly diagnosed children with autism: Cerebellar and basal
ganglia related motor disorder. Developmental Medicine and Child Neurology, 48(10),
819–824.
Riva, D., Annunziata, S., Contarino, V., Erbetta, A., Aquino, D., & Bulgheroni, S. (2013).
Gray matter reduction in the VERMIS and CRUS-II is associated with social and inter-
action deficits in low-functioning children with autistic spectrum disorders: A VBM-
DARTEL study. Cerebellum, 12(5), 676–685.
Riva, D., & Giorgi, C. (2000). The cerebellum contributes to higher functions during devel-
opment: Evidence from a series of children surgically treated for posterior fossa tumours.
Brain, 123(Pt. 5), 1051–1061.
Rodier, P. M. (1996). Animal model of autism based on developmental data. Mental Retar-
dation and Developmental Disabilities Research Reviews, 2(4), 249–256.
Rogers, T. D., Dickson, P. E., McKimm, E., Heck, D. H., Goldowitz, D., Blaha, C. D.,
et al. (2013). Reorganization of circuits underlying cerebellar modulation of prefrontal
cortical dopamine in mouse models of autism spectrum disorder. Cerebellum, 12(4),
547–556.
Rogers, S. J., Hepburn, S. L., Stackhouse, T., & Wehner, E. (2003). Imitation performance
in toddlers with autism and those with other developmental disorders. Journal of Child
Psychology and Psychiatry, 44(5), 763–781.
Rogers, T. D., McKimm, E., Dickson, P. E., Goldowitz, D., Blaha, C. D., & Mittleman, G.
(2013). Is autism a disease of the cerebellum? An integration of clinical and pre-clinical
research. Frontiers in Systems Neuroscience, 7, 15.
Rojas, D. C., Peterson, E., Winterrowd, E., Reite, M. L., Rogers, S. J., & Tregellas, J. R.
(2006). Regional gray matter volumetric changes in autism associated with social and
repetitive behavior symptoms. BMC Psychiatry, 6, 56.
Roullet, F. I., Wollaston, L., deCatanzaro, D., & Foster, J. A. (2010). Behavioral and molec-
ular changes in the mouse in response to prenatal exposure to the anti-epileptic drug
valproic acid. Neuroscience, 170(2), 514–522.
Roy, S., Zhao, Y., Allensworth, M., Farook, M. F., LeDoux, M. S., Reiter, L. T., et al.
(2011). Comprehensive motor testing in Fmr1-KO mice exposes temporal defects in
oromotor coordination. Behavioral Neuroscience, 125(6), 962–969.
Sabaratnam, M. (2000). Pathological and neuropathological findings in two males with
fragile-X syndrome. Journal of Intellectual Disability Research, 44, 81–85.
Sadakata, T. (2004). The secretory granule-associated protein CAPS2 regulates neurotrophin
release and cell survival. Journal of Neuroscience, 24(1), 43–52.
Sadakata, T., Kakegawa, W., Mizoguchi, A., Washida, M., Katoh-Semba, R.,
Shutoh, F., et al. (2007). Impaired cerebellar development and function in mice
32 Esther B.E. Becker and Catherine J. Stoodley
Stoodley, C. J., & Schmahmann, J. D. (2009). Functional topography in the human cerebel-
lum: A meta-analysis of neuroimaging studies. NeuroImage, 44(2), 489–501.
Stoodley, C. J., & Schmahmann, J. D. (2010). Evidence for topographic organization in the cer-
ebellum of motor control versus cognitive and affective processing. Cortex, 46(7), 831–844.
Sweet, H. O., Bronson, R. T., Johnson, K. R., Cook, S. A., & Davisson, M. T. (1996).
Scrambler, a new neurological mutation of the mouse with abnormalities of neuronal
migration. Mammalian Genome, 7(11), 798–802.
Tabuchi, K., Blundell, J., Etherton, M. R., Hammer, R. E., Liu, X., Powell, C. M., et al.
(2007). A neuroligin-3 mutation implicated in autism increases inhibitory synaptic trans-
mission in mice. Science, 318(5847), 71–76.
Takarae, Y., Minshew, N. J., Luna, B., Krisky, C. M., & Sweeney, J. A. (2004). Pursuit eye
movement deficits in autism. Brain, 127(Pt. 12), 2584–2594.
Takarae, Y., Minshew, N. J., Luna, B., & Sweeney, J. A. (2004). Oculomotor abnormal-
ities parallel cerebellar histopathology in autism. Journal of Neurology, Neurosurgery, and
Psychiatry, 75(9), 1359–1361.
Takarae, Y., Minshew, N. J., Luna, B., & Sweeney, J. A. (2007). Atypical involvement of
frontostriatal systems during sensorimotor control in autism. Psychiatry Research,
156(2), 117–127.
Tavano, A., Grasso, R., Gagliardi, C., Triulzi, F., Bresolin, N., Fabbro, F., et al. (2007). Dis-
orders of cognitive and affective development in cerebellar malformations. Brain, 130,
2646–2660.
Teitelbaum, P., Teitelbaum, O., Nye, J., Fryman, J., & Maurer, R. G. (1998). Movement
analysis in infancy may be useful for early diagnosis of autism. Proceedings of the National
Academy of Sciences of the United States of America, 95(23), 13982–13987.
Tesink, C. M., Buitelaar, J. K., Petersson, K. M., van der Gaag, R. J., Teunisse, J. P., &
Hagoort, P. (2011). Neural correlates of language comprehension in autism spectrum
disorders: When language conflicts with world knowledge. Neuropsychologia, 49(5),
1095–1104.
Tobia, M. J., & Woodruff-Pak, D. S. (2009). Delay eyeblink classical conditioning is
impaired in Fragile X syndrome. Behavioral Neuroscience, 123(3), 665–676.
Travers, B. G., Adluru, N., Ennis, C., Tromp do, P. M., Destiche, D., Doran, S., et al.
(2012). Diffusion tensor imaging in autism spectrum disorder: A review. Autism Research,
5(5), 289–313.
Tsai, P. T., Hull, C., Chu, Y., Greene-Colozzi, E., Sadowski, A. R., Leech, J. M., et al.
(2012). Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant
mice. Nature, 488(7413), 647–651.
Vaughn, J., Hagiwara, M., Katz, J., Roth, J., Devinsky, O., Weiner, H., et al. (2013). MRI
characterization and longitudinal study of focal cerebellar lesions in a young tuberous
sclerosis cohort. American Journal of Neuroradiology, 34(3), 655–659.
Villalobos, M. E., Mizuno, A., Dahl, B. C., Kemmotsu, N., & Muller, R. A. (2005).
Reduced functional connectivity between V1 and inferior frontal cortex associated with
visuomotor performance in autism. NeuroImage, 25(3), 916–925.
Vollm, B. A., Taylor, A. N., Richardson, P., Corcoran, R., Stirling, J., McKie, S., et al.
(2006). Neuronal correlates of theory of mind and empathy: A functional magnetic res-
onance imaging study in a nonverbal task. NeuroImage, 29(1), 90–98.
Wagner, G. C., Reuhl, K. R., Cheh, M., McRae, P., & Halladay, A. K. (2006). A new neu-
robehavioral model of autism in mice: Pre- and postnatal exposure to sodium valproate.
Journal of Autism and Developmental Disorders, 36(6), 779–793.
Wang, A. T., Lee, S. S., Sigman, M., & Dapretto, M. (2007). Reading affect in the face and
voice: Neural correlates of interpreting communicative intent in children and adoles-
cents with autism spectrum disorders. Archives of General Psychiatry, 64(6), 698–708.
34 Esther B.E. Becker and Catherine J. Stoodley
Webb, S. J., Sparks, B. F., Friedman, S. D., Shaw, D. W., Giedd, J., Dawson, G., et al. (2009).
Cerebellar vermal volumes and behavioral correlates in children with autism spectrum
disorder. Psychiatry Research, 172(1), 61–67.
Weber, A. M., Egelhoff, J. C., McKellop, J. M., & Franz, D. N. (2000). Autism and the cer-
ebellum: Evidence from tuberous sclerosis. Journal of Autism and Developmental Disorders,
30(6), 511–517.
Wegiel, J., Kuchna, I., Nowicki, K., Imaki, H., Wegiel, J., Marchi, E., et al. (2010). The
neuropathology of autism: Defects of neurogenesis and neuronal migration, and dysplas-
tic changes. Neuropsychology Review, 119(6), 755–770.
Weisenfeld, N. I., Peters, J. M., Tsai, P. T., Prabhu, S. P., Dies, K. A., Sahin, M., et al. (2013).
A magnetic resonance imaging study of cerebellar volume in tuberous sclerosis complex.
Pediatric Neurology, 48(2), 105–110.
Whitney, E. R., Kemper, T. L., Bauman, M. L., Rosene, D. L., & Blatt, G. J. (2008). Cer-
ebellar Purkinje cells are reduced in a subpopulation of autistic brains: A stereological
experiment using calbindin-D28k. Cerebellum, 7(3), 406–416.
Whitney, E. R., Kemper, T. L., Rosene, D. L., Bauman, M. L., & Blatt, G. J. (2009). Density
of cerebellar basket and stellate cells in autism: Evidence for a late developmental loss of
Purkinje cells. Journal of Neuroscience Research, 87(10), 2245–2254.
Whyatt, C. P., & Craig, C. M. (2012). Motor skills in children aged 7-10 years, diagnosed
with autism spectrum disorder. Journal of Autism and Developmental Disorders, 42(9),
1799–1809.
Wills, S., Cabanlit, M., Bennett, J., Ashwood, P., Amaral, D. G., & Van de Water, J. (2009).
Detection of autoantibodies to neural cells of the cerebellum in the plasma of subjects
with autism spectrum disorders. Brain, Behavior, and Immunity, 23(1), 64–74.
Xu, L.-M., Li, J.-R., Huang, Y., Zhao, M., Tang, X., & Wei, L. (2012). AutismKB: An
evidence-based knowledgebase of autism genetics. Nucleic Acids Research, 40,
D1016–D1022.
Yang, M., Bozdagi, O., Scattoni, M. L., Wöhr, M., Roullet, F. I., Katz, A. M., et al. (2011).
Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adoles-
cent Shank3 null mutant mice. The Journal of Neuroscience, 32(19), 6525–6541.
Yochum, C. L., Dowling, P., Reuhl, K. R., Wagner, G. C., & Ming, X. (2008). VPA-
induced apoptosis and behavioral deficits in neonatal mice. Brain Research, 1203,
126–132.
Yu, K. K., Cheung, C., Chua, S. E., & McAlonan, G. M. (2011). Can Asperger syndrome be
distinguished from autism? An anatomic likelihood meta-analysis of MRI studies. Journal
of Psychiatry and Neuroscience, 36(6), 412–421.
Ziats, M. N., & Rennert, O. M. (2013). The cerebellum in autism: Pathogenic or an ana-
tomical beacon? Cerebellum, 12(5), 776–777.
Zimmerman, A. W., Connors, S. L., Matteson, K. J., Lee, L.-C., Singer, H. S.,
Castaneda, J. A., et al. (2007). Maternal antibrain antibodies in autism. Brain, Behavior,
and Immunity, 21(3), 351–357.
Zwaigenbaum, L., Bryson, S., & Garon, N. (2013). Early identification of autism spectrum
disorders. Behavioural Brain Research, 251, 133–146.
CHAPTER TWO
Contents
1. Small ncRNA 36
2. Long ncRNA 38
3. LncRNA in Fundamental Genetic Mechanisms 38
4. LncRNAs in Cancer 40
5. LncRNAs in the Brain 41
5.1 Brain development and function 41
5.2 Neurodegenerative diseases 44
5.3 Neurodevelopmental disorders 45
6. LncRNAs Contribute to ASD 47
7. Conclusions 48
References 49
Abstract
Accumulating evidence indicates that long noncoding RNAs (lncRNAs) contribute to
autism spectrum disorder (ASD) risk. Although a few lncRNAs have long been recog-
nized to have important functions, the vast majority of this class of molecules remains
uncharacterized. Because lncRNAs are more abundant in human brain than protein-
coding RNAs, it is likely that they contribute to brain disorders, including ASD. We review
here the known functions of lncRNAs and the potential contributions of lncRNAs to ASD.
1. SMALL ncRNA
The classification, small ncRNA, encompasses a wide variety of sub-
classes including microRNAs (miRNAs), short interfering RNAs
(siRNAs), small nucleolar RNAs (snoRNAs), and PIWI-interacting RNAs
(piRNAs). These can be derived from a variety of sources within the
LncRNAs in Autism 37
genome (review in Mattick & Makunin, 2005) and it has been suggested that
one potential source is also from the processing of long noncoding RNAs
(lncRNAs) (Jalali, Jayaraj, & Scaria, 2012; Keniry et al., 2012). In general,
one of the most prominent forms of genetic regulation carried out by small
ncRNAs is by binding to their specific target transcripts via complementary
nucleotide sequences and subsequently influencing the translation of the tar-
get through a diverse set of downstream events. For an in-depth explanation
of small ncRNA, including subclasses, biogenesis, and forms of genetic reg-
ulation, we refer the reader to these reviews: Carthew and Sontheimer
(2009), Kim, Han, and Siomi (2009), Luteijn and Ketting (2013), Matera,
Terns, and Terns (2007), Mattick and Makunin (2005), and Winter,
Jung, Keller, Gregory, and Diederichs (2009).
One of the most thoroughly studied classes of small ncRNAs, miRNAs,
was first discovered in Caenorhabditis elegans in 1993 (Lee, Feinbaum, &
Ambros, 1993) and since then has been postulated to be relatively conserved
among vertebrate species (Altuvia et al., 2005; Berezikov et al., 2005;
Ibáñez-Ventoso, Vora, & Driscoll, 2008). MiRNAs can participate in post-
transcriptional gene regulation by binding to either the 30 -UTR (Lai, 2002)
or the 50 -UTR (Lytle, Yario, & Steitz, 2007) of a target transcript which
subsequently inhibits its translation. A single miRNA has the potential to
target multiple transcripts and similarly, a given transcript may be regulated
by more than one miRNA (Krek et al., 2005). Both the importance and
prevalence of miRNAs in the human genome is underscored by the fact that
greater than 60% of human protein-coding genes are predicted targets of
miRNAs (Friedman, Farh, Burge, & Bartel, 2009). The regulation per-
formed by miRNAs is a critical process in the development of mammals
and like many other ncRNAs, their expression can be specific in both
particular tissue types and developmental stages as reviewed in Alvarez-
Garcia and Miska (2005), Sayed and Abdellatif (2011), and Wienholds
and Plasterk (2005).
Like miRNA, siRNA also regulates gene expression by binding to com-
plementary sequences on target transcripts, but with more stringent require-
ments as they have to have close to perfect sequence complementation
(Elbashir, Martinez, Patkaniowska, Lendeckel, & Tuschl, 2001). In general,
this difference results in two separate mechanisms where targets with close to
perfect complementation undergo direct cleavage (as with siRNA) and
those with relatively loose complementation are destabilized or transcrip-
tionally repressed (as with miRNA) (Guo, Ingolia, Weissman, & Bartel,
2010; Hutvágner & Zamore, 2002; Martinez, Patkaniowska, Urlaub,
38 Brent Wilkinson and Daniel B. Campbell
2. LONG ncRNA
LncRNA is a broad category which encompasses transcripts of diverse
structural features and mechanisms of action. They can be derived from
sense or antisense strands overlapping with protein-coding genes, within
intergenic regions (lincRNAs), or within pseudogenes. Once transcribed
they may undergo splicing and be processed to include a 50 -
methyl-guanosine cap and 30 -poly (A) tail. Within the cell, lncRNAs can
be localized to the nucleus or the cytoplasm (Ponting, Oliver, & Reik,
2009). They have been found to perform a wide variety of functions includ-
ing participating in the recruitment of chromatin-modifying complexes,
acting as competing endogenous RNAs (ceRNAs), providing a scaffold
for the assembly of protein complexes, modulating alternative splicing,
and employing enhancer-like functions (Nagano & Fraser, 2011; Rinn &
Chang, 2012; Wang & Chang, 2011). This diverse set of functions further
emphasizes the notion that lncRNAs are key components of numerous cel-
lular processes (Table 2.1).
with Down’s syndrome. In the iPSC model, this corrected for the trisomy of
chromosome 21 involved in the pathogenesis of Down’s syndrome and
improved deficiencies in proliferation and neural rosette formation (Jiang
et al., 2013). In addition, it was recently discovered that there is also a human
specific lncRNA, XACT, that coats the active X chromosome in pluripo-
tent cells (Vallot et al., 2013). Xist displays a common trait among lncRNAs
as they can associate with chromatin and recruit proteins with epigenetic
functions in order to regulate the transcription of multiple genes.
LncRNAs have also been shown to be involved in genomic imprinting,
the process by which genes are expressed in a parent-specific manner. The
paternally expressed lncRNA, Antisense Igf 2r (Air), silences three protein-
coding genes (Igf2r, Slc22a2, and Slc22a3) located within the Igf2r cluster on
the same allele (Sleutels, Zwart, & Barlow, 2002). Like Xist, Air is conserved
between mice and humans (Yotova et al., 2008) and is involved in epige-
netic regulation as it inactivates the Slc22a3 gene by recruiting G9a (a histone
methyltransferase) (Nagano et al., 2008). Factors influencing genomic
imprinting are of extreme importance as dysregulation of this process is hall-
mark of a number of conditions, including the neurodevelopmental disor-
ders, Angelman Syndrome (AS), and Prader-Willi Syndrome (PWS)
(Horsthemke & Wagstaff, 2008).
4. LncRNAs IN CANCER
Downregulation of the tumor suppressor, PTEN, has been associated
with numerous types of cancers (Cairns et al., 1997; Li et al., 1997; Vlietstra,
van Alewijk, Hermans, van Steenbrugge, & Trapman, 1998). The lncRNA,
PTENpg1 (also known as PTENP1), is selectively lost in cancer, has similar
expression levels to that of PTEN, and functions as an miRNA decoy by
intercepting miRNA species targeting PTEN (Poliseno et al., 2010).
Two PTENpg1 antisense (as) isoforms, a and b, regulate the expression
levels of PTEN by diverse mechanisms, illustrating the complex networks
of regulation lncRNAs can take part in. PTENpg1 asRNA a is able to
decrease PTEN expression levels through epigenetic mechanisms involving
DNMT3A and EZH2, while PTENpg1 asRNA b interacts with PTENpg1
in order to positively influence its stability and decoy function ( Johnsson
et al., 2013). Interestingly, abnormalities in PTEN have also been implicated
in ASD (Butler et al., 2005; O’Roak et al., 2012).
The HOX genes are essential for specifying patterning during the devel-
opment of bilateral animals (Pearson, Lemons, & McGinnis, 2005).
LncRNAs in Autism 41
HOTAIR is an lncRNA transcribed from the HOXC locus that acts in trans
to regulate transcription at the HOXD locus. Here, HOTAIR associates
with Polycomb Repressive Complex 2 (PRC2) in order to influence His-
tone H3 Lysine-27 trimethylation which results in transcriptional silencing
(Rinn et al., 2007). Increased expression of HOTAIR has been associated
with a variety of cancer types including breast (Gupta et al., 2010), colon
(Kogo et al., 2011), and liver (Ishibashi et al., 2013) cancers. This may be
due to massive epigenetic changes in which increased HOTAIR reverts
the epigenetic profile of cancerous cells to something resembling that of
embryonic fibroblasts through retargeting of PRC2 (Gupta et al., 2010).
Maternally expressed gene 3 (MEG3) is a conserved, imprinted gene
encoding an lncRNA. In meningiomas, there is a strong association between
loss of MEG3 expression and increase in tumor grade (Zhang et al., 2010).
This can be attributed to the activation of the p53 pathway (Zhao, Dahle,
Zhou, Zhang, & Klibanski, 2005) and increased methylation of MEG3 reg-
ulatory region. The effect of decreased expression of MEG3 has also been
implicated in pituitary adenomas (Zhang et al., 2003), gliomas (Wang,
Ren, & Sun, 2012), cervical cancer (Qin et al., 2013), and bladder cancer
(Ying et al., 2013).
7. CONCLUSIONS
The Central Dogma of Molecular Biology posits that DNA is tran-
scribed into RNA, which is translated into protein. Genetic information
is stored in protein-coding genes, while RNA is merely an intermediary
between genes and functional proteins. Prior to the completion of the
Human Genome Project, the prevailing hypothesis was that the human
genome would produce approximately 100,000 protein-coding genes. This
seemed like a reasonable estimate based on the size of the human genome
and the complexity of human anatomy. However, the human genome con-
tains only approximately 21,000 protein-coding genes, slightly more than a
mouse and slightly less than a grape. Over the last decade, RNA has become
increasingly recognized as a functional entity. The whole genome and trans-
criptome sequencing suggests that the complexity of an organism may be
regulated by noncoding portions of the genome rather than by proteins.
LncRNAs in Autism 49
B rs7704909 rs4307059
C MSNP1: Chr 5p14.1 pseudogene with 94% sequence identity to Chr X gene MSN
+ Strand
– Strand
Figure 2.1 MSNP1AS maps within the chromosome 5p14.1 GWAS-significant ASD-
association peak and is the only significantly expressed transcript within 500 kb of
the GWAS peak. (A) The GWAS results from Wang et al. (2009), indicating ASD-
associated markers on chromosome 5p14.1. (B) Genome-wide RNA-Seq data from a
variety of tissue sources indicate that a single major transcript of 4 kb is expressed
within 500 kb of the GWAS peak. (C) The þ strand of this 4 kb chromosome 5p14.1
region is the pseudogene moesin-like 1 (MSNP1), which has 94% sequence identity
to the X chromosome gene-encoding moesin (MSN) but does not appear to be tran-
scribed. Instead, our data indicate that a 3.9 kb RNA is transcribed from the strand,
producing a non-protein-coding RNA that is antisense to the X chromosome gene-
encoding moesin (MSN). Because the chromosome 5p14.1 transcript represents the
antisense of the pseudogene, we designate it MSNP1AS (moesin pseudogene 1, anti-
sense). Data from the UCSC Genome Browser.
Among long RNAs produced in the human brain, the majority do not code
for proteins but are instead lncRNAs. Increasing evidence suggests that these
lncRNAs may contribute to brain disorders.
REFERENCES
Altuvia, Y., Landgraf, P., Lithwick, G., Elefant, N., Pfeffer, S., Aravin, A., et al. (2005). Clus-
tering and conservation patterns of human microRNAs. Nucleic Acids Research, 33,
2697–2706.
Alvarez-Garcia, I., & Miska, E. A. (2005). MicroRNA functions in animal development and
human disease. Development, 132, 4653–4662.
50 Brent Wilkinson and Daniel B. Campbell
Amaral, P. P., Neyt, C., Wilkins, S. J., Askarian-Amiri, M. E., Sunkin, S. M., Perkins, A. C.,
et al. (2009). Complex architecture and regulated expression of the Sox2ot locus during
vertebrate development. RNA, 15, 2013–2027.
Amiri, A., Cho, W., Zhou, J., Birnbaum, S. G., Sinton, C. M., McKay, R. M., et al. (2012).
Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnor-
malities and alters neurogenesis. Journal of Neuroscience, 32, 5880–5890.
Anderson, S. A., Eisenstat, D. D., Shi, L., & Rubenstein, J. L. (1997). Interneuron migration
from basal forebrain to neocortex: Dependence on Dlx genes. Science, 278, 474–476.
Ashley-Koch, A. E., Jaworski, J., Ma, de, Q., Mei, H., Ritchie, M. D., Skaar, D. A., et al.
(2007). Investigation of potential gene-gene interactions between APOE and RELN
contributing to autism risk. Psychiatric Genetics, 17, 221–226.
Atwal, J. K., Chen, Y., Chiu, C., Mortensen, D. L., Meilandt, W. J., Liu, Y., et al. (2011).
A therapeutic antibody targeting BACE1 inhibits amyloid-b production in vivo. Science
Translational Medicine, 3, 84ra43.
Babiarz, J. E., Ruby, J. G., Wang, Y., Bartel, D. P., & Blelloch, R. (2008). Mouse ES cells
express endogenous shRNAs, siRNAs, and other Microprocessor-independent, Dicer-
dependent small RNAs. Genes and Development, 22, 2773–2785.
Barry, G., Briggs, J. A., Vanichkina, D. P., Poth, E. M., Beveridge, N. J., Ratnu, V. S., et al.
(2013). The long non-coding RNA Gomafu is acutely regulated in response to neuronal
activation and involved in schizophrenia-associated alternative splicing. Molecular Psychiatry,
http://dx.doi.org/10.1038/mp.2013.45.
Berezikov, E., Guryev, V., van de Belt, J., Wienholds, E., Plasterk, R. H., & Cuppen, E.
(2005). Phylogenetic shadowing and computational identification of human microRNA
genes. Cell, 120, 21–24.
Bernard, D., Prasanth, K. V., Tripathi, V., Colasse, S., Nakamura, T., Xuan, Z., et al. (2010).
A long nuclear-retained non-coding RNA regulates synaptogenesis by modulating gene
expression. The EMBO Journal, 29, 3082–3093.
Bertone, P., Stolc, V., Royce, T. E., Rozowsky, J. S., Urban, A. E., Zhu, X., et al. (2004).
Global identification of human transcribed sequences with genome tiling arrays. Science,
306, 2242–2246.
Birney, E., Stamatoyannopoulos, J. A., Dutta, A., Guigó, R., Gingeras, T. R.,
Margulies, E. H., et al. (2007). Identification and analysis of functional elements in
1% of the human genome by the ENCODE pilot project. Nature, 447, 799–816.
Bond, A. M., Vangompel, M. J., Sametsky, E. A., Clark, M. F., Savage, J. C., Disterhoft, J. F.,
et al. (2009). Balanced gene regulation by an embryonic brain ncRNA is critical for adult
hippocampal GABA circuitry. Nature Neuroscience, 12, 1020–1027.
Bourgeron, T. (2009). A synaptic trek to autism. Current Opinion in Neurobiology, 19,
231–234.
Brown, S. D. (1991). XIST and the mapping of the X chromosome inactivation centre. Bio-
essays, 13, 607–612.
Brown, C. J., Hendrich, B. D., Rupert, J. L., Lafrenière, R. G., Xing, Y., Lawrence, J., et al.
(1992). The human XIST gene: Analysis of a 17 kb inactive X-specific RNA that con-
tains conserved repeats and is highly localized within the nucleus. Cell, 71, 527–542.
Burnett, J. C., & Rossi, J. J. (2012). RNA-based therapeutics: Current progress and future
prospects. Chemistry and Biology, 19, 60–71.
Butler, M. G., Dasouki, M. J., Zhou, X. P., Talebizadeh, Z., Brown, M., Takahashi, T. N.,
et al. (2005). Subset of individuals with autism spectrum disorders and extreme
macrocephaly associated with germline PTEN tumour suppressor gene mutations. Jour-
nal of Medical Genetics, 42, 318–321.
Cai, H., Wang, Y., McCarthy, D., Wen, H., Borchelt, D. R., Price, D. L., et al. (2001).
BACE1 is the major beta-secretase for generation of Abeta peptides by neurons. Nature
Neuroscience, 4, 233–234.
LncRNAs in Autism 51
Cairns, P., Okami, K., Halachmi, S., Halachmi, N., Esteller, M., Herman, J. G., et al. (1997).
Frequent inactivation of PTEN/MMAC1 in primary prostate cancer. Cancer Research,
57, 4997–5000.
Carninci, P., Kasukawa, T., Katayama, S., Gough, J., Frith, M. C., Maeda, N., et al. (2005).
The transcriptional landscape of the mammalian genome. Science, 309, 1559–1563.
Carninci, P., Sandelin, A., Lenhard, B., Katayama, S., Shimokawa, K., Ponjavic, J., et al.
(2006). Genome-wide analysis of mammalian promoter architecture and evolution.
Nature Genetics, 38, 626–635.
Carthew, R. W., & Sontheimer, E. J. (2009). Origins and mechanisms of miRNAs and
siRNAs. Cell, 136, 642–655.
Chan, A. S., Thorner, P. S., Squire, J. A., & Zielenska, M. (2002). Identification of a novel
gene NCRMS on chromosome 12q21 with differential expression between rhabdo-
myosarcoma subtypes. Oncogene, 21, 3029–3037.
Chan, W. L., Yuo, C. Y., Yang, W. K., Hung, S. Y., Chang, Y. S., Chiu, C. C., et al. (2013).
Transcribed pseudogene cPPM1K generates endogenous siRNA to suppress oncogenic
cell growth in hepatocellular carcinoma. Nucleic Acids Research, 41, 3734–3747.
Cheng, J., Kapranov, P., Drenkow, J., Dike, S., Brubaker, S., Patel, S., et al. (2005). Transcrip-
tional maps of 10 human chromosomes at 5-nucleotide resolution. Science, 308, 1149–1154.
Chodroff, R. A., Goodstadt, L., Sirey, T. M., Oliver, P. L., Davies, K. E., Green, E. D., et al.
(2010). Long noncoding RNA genes: Conservation of sequence and brain expression
among diverse amniotes. Genome Biology, 11, R72.
Chung, W. J., Okamura, K., Martin, R., & Lai, E. C. (2008). Endogenous RNA interference
provides a somatic defense against Drosophila transposons. Current Biology, 18, 795–802.
Core, L. J., Waterfall, J. J., & Lis, J. T. (2008). Nascent RNA sequencing reveals widespread
pausing and divergent initiation at human promoters. Science, 322, 1845–1848.
de la Grange, P., Gratadou, L., Delord, M., Dutertre, M., & Auboeuf, D. (2010). Splicing
factor and exon profiling across human tissues. Nucleic Acids Research, 38, 2825–2838.
Djebali, S., Davis, C. A., Merkel, A., Dobin, A., Lassmann, T., Mortazavi, A., et al. (2012).
Landscape of transcription in human cells. Nature, 489, 101–108.
Eastwood, S. L., & Harrison, P. J. (2006). Cellular basis of reduced cortical reelin expression
in schizophrenia. The American Journal of Psychiatry, 163, 540–542.
Egan, M. F., Kojima, M., Callicott, J. H., Goldberg, T. E., Kolachana, B. S., Bertolino, A.,
et al. (2003). The BDNF val66met polymorphism affects activity-dependent secretion of
BDNF and human memory and hippocampal function. Cell, 112, 257–269.
Elbashir, S. M., Martinez, J., Patkaniowska, A., Lendeckel, W., & Tuschl, T. (2001). Func-
tional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster
embryo lysate. The EMBO Journal, 20, 6877–6888.
Ellis, P., Fagan, B. M., Magness, S. T., Hutton, S., Taranova, O., Hayashi, S., et al. (2004).
SOX2, a persistent marker for multipotential neural stem cells derived from embryonic
stem cells, the embryo or the adult. Developmental Neuroscience, 26, 148–165.
Faghihi, M. A., Modarresi, F., Khalil, A. M., Wood, D. E., Sahagan, B. G., Morgan, T. E.,
et al. (2008). Expression of a noncoding RNA is elevated in Alzheimer’s disease and
drives rapid feed-forward regulation of beta-secretase. Nature Medicine, 14, 723–730.
Fantes, J., Ragge, N. K., Lynch, S. A., McGill, N. I., Collin, J. R., Howard-Peebles, P. N.,
et al. (2003). Mutations in SOX2 cause anophthalmia. Nature Genetics, 33, 461–463.
Fatemi, S. H., Folsom, T. D., Kneeland, R. E., & Liesch, S. B. (2011). Metabotropic glu-
tamate receptor 5 upregulation in children with autism is associated with under-
expression of both Fragile X mental retardation protein and GABAA receptor beta 3
in adults with autism. Anatomical Record, 294, 1635–1645.
Favaro, R., Valotta, M., Ferri, A. L., Latorre, E., Mariani, J., Giachino, C., et al. (2009). Hip-
pocampal development and neural stem cell maintenance require Sox2-dependent reg-
ulation of Shh. Nature Neuroscience, 12, 1248–1256.
52 Brent Wilkinson and Daniel B. Campbell
Feng, J., Bi, C., Clark, B. S., Mady, R., Shah, P., & Kohtz, J. D. (2006). The Evf-2 non-
coding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as
a Dlx-2 transcriptional coactivator. Genes and Development, 20, 1470–1484.
Ferri, A. L., Cavallaro, M., Braida, D., Di Cristofano, A., Canta, A., Vezzani, A., et al. (2004).
Sox2 deficiency causes neurodegeneration and impaired neurogenesis in the adult mouse
brain. Development, 131, 3805–3819.
Friedman, R. C., Farh, K. K., Burge, C. B., & Bartel, D. P. (2009). Most mammalian
mRNAs are conserved targets of microRNAs. Genome Research, 19, 92–105.
Gadow, K. D., Roohi, J., DeVincent, C. J., Kirsch, S., & Hatchwell, E. (2009). Association
of COMT (Val158Met) and BDNF (Val66Met) gene polymorphisms with anxiety,
ADHD and tics in children with autism spectrum disorder. Journal of Autism and Devel-
opmental Disorders, 39, 1542–1551.
Garber, K. B., Visootsak, J., & Warren, S. T. (2008). Fragile X syndrome. European Journal of
Human Genetics, 16, 666–672.
Grant, S. G. (2012). Synaptopathies: Diseases of the synaptome. Current Opinion in Neuro-
biology, 22, 522–529.
Grosso, A. R., Gomes, A. Q., Barbosa-Morais, N. L., Caldeira, S., Thorne, N. P., Grech, G.,
et al. (2008). Tissue-specific splicing factor gene expression signatures. Nucleic Acids
Research, 36, 4823–4832.
Guidi, S., Bonasoni, P., Ceccarelli, C., Santini, D., Gualtieri, F., Ciani, E., et al. (2008). Neu-
rogenesis impairment and increased cell death reduce total neuron number in the hip-
pocampal region of fetuses with Down syndrome. Brain Pathology, 18, 180–197.
Guo, H., Ingolia, N. T., Weissman, J. S., & Bartel, D. P. (2010). Mammalian microRNAs
predominantly act to decrease target mRNA levels. Nature, 466, 835–840.
Gupta, R. A., Shah, N., Wang, K. C., Kim, J., Horlings, H. M., Wong, D. J., et al. (2010).
Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer
metastasis. Nature, 464, 1071–1076.
Hock, C., Heese, K., Hulette, C., Rosenberg, C., & Otten, U. (2000). Region-specific neu-
rotrophin imbalances in Alzheimer disease: Decreased levels of brain-derived neuro-
trophic factor and increased levels of nerve growth factor in hippocampus and cortical
areas. Archives of Neurology, 57, 846–851.
Hogart, A., Nagarajan, R. P., Patzel, K. A., Yasui, D. H., & Lasalle, J. M. (2007). 15q11-13
GABAA receptor genes are normally biallelically expressed in brain yet are subject to
epigenetic dysregulation in autism-spectrum disorders. Human Molecular Genetics, 16,
691–703.
Horch, H. W., & Katz, L. C. (2002). BDNF release from single cells elicits local dendritic
growth in nearby neurons. Nature Neuroscience, 5, 1177–1184.
Horder, J., Lavender, T., Mendez, M. A., O’Gorman, R., Daly, E., Craig, M. C., et al.
(2013). Reduced subcortical glutamate/glutamine in adults with autism spectrum disor-
ders: A [(1)H]MRS study. Translational Psychiatry, 3, e279.
Horsthemke, B., & Wagstaff, J. (2008). Mechanisms of imprinting of the Prader-Willi/
Angelman region. American Journal of Medical Genetics Part A, 146A, 2041–2052.
Howells, D. W., Porritt, M. J., Wong, J. Y., Batchelor, P. E., Kalnins, R., Hughes, A. J., et al.
(2000). Reduced BDNF mRNA expression in the Parkinson’s disease substantia nigra.
Experimental Neurology, 166, 127–135.
Hsieh, J., & Eisch, A. J. (2010). Epigenetics, hippocampal neurogenesis, and neuropsychiatric
disorders: Unraveling the genome to understand the mind. Neurobiology of Disease, 39,
73–84.
Hu, V. W. (2013). The expanding genomic landscape of autism: Discovering the ‘forest’
beyond the ‘trees’. Future Neurology, 8, 29–42.
Hu, V. W., Sarachana, T., Kim, K. S., Nguyen, A., Kulkarni, S., Steinberg, M. E., et al.
(2009). Gene expression profiling differentiates autism case-controls and phenotypic
LncRNAs in Autism 53
modification and is associated with poor prognosis in colorectal cancers. Cancer Research,
71, 6320–6326.
Krebs, M. O., Guillin, O., Bourdell, M. C., Schwartz, J. C., Olie, J. P., Poirier, M. F., et al.
(2000). Brain derived neurotrophic factor (BDNF) gene variants association with age at
onset and therapeutic response in schizophrenia. Molecular Psychiatry, 5, 558–562.
Krek, A., Grün, D., Poy, M. N., Wolf, R., Rosenberg, L., Epstein, E. J., et al. (2005). Com-
binatorial microRNA target predictions. Nature Genetics, 37, 495–500.
Kuwajima, T., Nishimura, I., & Yoshikawa, K. (2006). Necdin promotes GABAergic neu-
ron differentiation in cooperation with Dlx homeodomain proteins. Journal of Neuro-
science, 26, 5383–5392.
Ladd, P. D., Smith, L. E., Rabaia, N. A., Moore, J. M., Georges, S. A., Hansen, R. S., et al.
(2007). An antisense transcript spanning the CGG repeat region of FMR1 is upregulated in
premutation carriers but silenced in full mutation individuals. Human Molecular Genetics, 16,
3174–3187.
Lai, E. C. (2002). Micro RNAs are complementary to 3’ UTR sequence motifs that mediate
negative post-transcriptional regulation. Nature Genetics, 30, 363–364.
Lander, E. S., Linton, L. M., Birren, B., Nusbaum, C., Zody, M. C., Baldwin, J., et al.
(2001). Initial sequencing and analysis of the human genome. Nature, 409, 860–921.
Law, A. J., Kleinman, J. E., Weinberger, D. R., & Weickert, C. S. (2007). Disease-associated
intronic variants in the ErbB4 gene are related to altered ErbB4 splice-variant expression
in the brain in schizophrenia. Human Molecular Genetics, 16, 129–141.
Lee, R. C., Feinbaum, R. L., & Ambros, V. (1993). The C. elegans heterochronic gene lin-4
encodes small RNAs with antisense complementarity to lin-14. Cell, 75, 843–854.
Lewis, D. A., Hashimoto, T., & Volk, D. W. (2005). Cortical inhibitory neurons and schizo-
phrenia. Nature Reviews Neuroscience, 6, 312–324.
Li, H., Li, W. X., & Ding, S. W. (2002). Induction and suppression of RNA silencing by an
animal virus. Science, 296, 1319–1321.
Li, J., Yen, C., Liaw, D., Podsypanina, K., Bose, S., Wang, S. I., et al. (1997). PTEN, a puta-
tive protein tyrosine phosphatase gene mutated in human brain, breast, and prostate can-
cer. Science, 275, 1943–1947.
Lin, M., Pedrosa, E., Shah, A., Hrabovsky, A., Maqbool, S., Zheng, D., et al. (2011).
RNA-Seq of human neurons derived from iPS cells reveals candidate long non-
coding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS One,
6, e23356.
Lipovich, L., Dachet, F., Cai, J., Bagla, S., Balan, K., Jia, H., et al. (2012). Activity-
dependent human brain coding/noncoding gene regulatory networks. Genetics, 192,
1133–1148.
Lipovich, L., Tarca, A. L., Cai, J., Jia, H., Chugani, H. T., Sterner, K. N., et al. (2013).
Developmental changes in the transcriptome of human cerebral cortex tissue: Long non-
coding RNA transcripts. Cerebral Cortex, http://dx.doi.org/10.1093/cercor/bhs414.
Liu, Q. R., Lu, L., Zhu, X. G., Gong, J. P., Shaham, Y., & Uhl, G. R. (2006). Rodent
BDNF genes, novel promoters, novel splice variants, and regulation by cocaine. Brain
Research, 1067, 1–12.
Loesch, D. Z., Godler, D. E., Evans, A., Bui, Q. M., Gehling, F., Kotschet, K. E., et al.
(2011). Evidence for the toxicity of bidirectional transcripts and mitochondrial dysfunc-
tion in blood associated with small CGG expansions in the FMR1 gene in patients with
parkinsonism. Genetics in Medicine, 13, 392–399.
Luteijn, M. J., & Ketting, R. F. (2013). PIWI-interacting RNAs: From generation to trans-
generational epigenetics. Nature Reviews Genetics, 14, 523–534.
Lytle, J. R., Yario, T. A., & Steitz, J. A. (2007). Target mRNAs are repressed as efficiently by
microRNA-binding sites in the 5’ UTR as in the 3’ UTR. Proceedings of the National
Academy of Sciences of the United States of America, 104, 9667–9672.
LncRNAs in Autism 55
Martignetti, J. A., & Brosius, J. (1993). BC200 RNA: A neural RNA polymerase III product
encoded by a monomeric Alu element. Proceedings of the National Academy of Sciences of the
United States of America, 90, 11563–11567.
Martinez, J., Patkaniowska, A., Urlaub, H., Lührmann, R., & Tuschl, T. (2002).
Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell, 110,
563–574.
Matera, A. G., Terns, R. M., & Terns, M. P. (2007). Non-coding RNAs: Lessons from the
small nuclear and small nucleolar RNAs. Nature Reviews Molecular Cell Biology, 8, 209–220.
Mattick, J. S., & Makunin, I. V. (2005). Small regulatory RNAs in mammals. Human Molec-
ular Genetics, 14(Spec. No. 1), R121–R132.
Meng, L., Person, R. E., & Beaudet, A. L. (2012). Ube3a-ATS is an atypical RNA polymer-
ase II transcript that represses the paternal expression of Ube3a. Human Molecular Genetics,
21, 3001–3012.
Mercer, T. R., Dinger, M. E., & Mattick, J. S. (2009). Long non-coding RNAs: Insights into
functions. Nature Reviews Genetics, 10, 155–159.
Mercer, T. R., Dinger, M. E., Sunkin, S. M., Mehler, M. F., & Mattick, J. S. (2008). Specific
expression of long noncoding RNAs in the mouse brain. Proceedings of the National Acad-
emy of Sciences of the United States of America, 105, 716–721.
Millar, J. K., Wilson-Annan, J. C., Anderson, S., Christie, S., Taylor, M. S., Semple, C. A.,
et al. (2000). Disruption of two novel genes by a translocation co-segregating with
schizophrenia. Human Molecular Genetics, 9, 1415–1423.
Ming, G. L., & Song, H. (2005). Adult neurogenesis in the mammalian central nervous sys-
tem. Annual Review of Neuroscience, 28, 223–250.
Morikawa, T., & Manabe, T. (2010). Aberrant regulation of alternative pre-mRNA splicing
in schizophrenia. Neurochemistry International, 57, 691–704.
Moseley, M. L., Zu, T., Ikeda, Y., Gao, W., Mosemiller, A. K., Daughters, R. S., et al. (2006).
Bidirectional expression of CUG and CAG expansion transcripts and intranuclear poly-
glutamine inclusions in spinocerebellar ataxia type 8. Nature Genetics, 38, 758–769.
Muddashetty, R., Khanam, T., Kondrashov, A., Bundman, M., Iacoangeli, A.,
Kremerskothen, J., et al. (2002). Poly(A)-binding protein is associated with neuronal
BC1 and BC200 ribonucleoprotein particles. Journal of Molecular Biology, 321, 433–445.
Mus, E., Hof, P. R., & Tiedge, H. (2007). Dendritic BC200 RNA in aging and in
Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of
America, 104, 10679–10684.
Nagano, T., & Fraser, P. (2011). No-nonsense functions for long noncoding RNAs. Cell,
145, 178–181.
Nagano, T., Mitchell, J. A., Sanz, L. A., Pauler, F. M., Ferguson-Smith, A. C., Feil, R., et al.
(2008). The Air noncoding RNA epigenetically silences transcription by targeting G9a
to chromatin. Science, 322, 1717–1720.
Nakata, K., Lipska, B. K., Hyde, T. M., Ye, T., Newburn, E. N., Morita, Y., et al. (2009).
DISC1 splice variants are upregulated in schizophrenia and associated with risk polymor-
phisms. Proceedings of the National Academy of Sciences of the United States of America, 106,
15873–15878.
Neves-Pereira, M., Cheung, J. K., Pasdar, A., Zhang, F., Breen, G., Yates, P., et al. (2005).
BDNF gene is a risk factor for schizophrenia in a Scottish population. Molecular Psychiatry,
10, 208–212.
Ng, S. Y., Johnson, R., & Stanton, L. W. (2012). Human long non-coding RNAs promote
pluripotency and neuronal differentiation by association with chromatin modifiers and
transcription factors. The EMBO Journal, 31, 522–533.
Nicholls, R. D., & Knepper, J. L. (2001). Genome organization, function, and imprinting in
Prader-Willi and Angelman syndromes. Annual Review of Genomics and Human Genetics,
2, 153–175.
56 Brent Wilkinson and Daniel B. Campbell
Nieto, R., Kukuljan, M., & Silva, H. (2013). BDNF and schizophrenia: From
neurodevelopment to neuronal plasticity, learning, and memory. Frontiers in Psychiatry,
4, 45.
O’Roak, B. J., Vives, L., Fu, W., Egertson, J. D., Stanaway, I. B., Phelps, I. G., et al. (2012).
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum
disorders. Science, 338, 1619–1622.
Pang, K. C., Frith, M. C., & Mattick, J. S. (2006). Rapid evolution of noncoding RNAs:
Lack of conservation does not mean lack of function. Trends in Genetics, 22, 1–5.
Pearson, J. C., Lemons, D., & McGinnis, W. (2005). Modulating Hox gene functions during
animal body patterning. Nature Reviews Genetics, 6, 893–904.
Penny, G. D., Kay, G. F., Sheardown, S. A., Rastan, S., & Brockdorff, N. (1996). Require-
ment for Xist in X chromosome inactivation. Nature, 379, 131–137.
Persico, A. M., D’Agruma, L., Maiorano, N., Totaro, A., Militerni, R., Bravaccio, C., et al.
(2001). Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder.
Molecular Psychiatry, 6, 150–159.
Petanjek, Z., Judaš, M., Šimic, G., Rasin, M. R., Uylings, H. B., Rakic, P., et al. (2011).
Extraordinary neoteny of synaptic spines in the human prefrontal cortex. Proceedings of
the National Academy of Sciences of the United States of America, 108, 13281–13286.
Plath, K., Fang, J., Mlynarczyk-Evans, S. K., Cao, R., Worringer, K. A., Wang, H., et al.
(2003). Role of histone H3 lysine 27 methylation in X inactivation. Science, 300,
131–135.
Poliseno, L., Salmena, L., Zhang, J., Carver, B., Haveman, W. J., & Pandolfi, P. P. (2010).
A coding-independent function of gene and pseudogene mRNAs regulates tumour biol-
ogy. Nature, 465, 1033–1038.
Pollard, K. S., Salama, S. R., Lambert, N., Lambot, M. A., Coppens, S., Pedersen, J. S., et al.
(2006). An RNA gene expressed during cortical development evolved rapidly in
humans. Nature, 443, 167–172.
Ponjavic, J., Oliver, P. L., Lunter, G., & Ponting, C. P. (2009). Genomic and transcriptional
co-localization of protein-coding and long non-coding RNA pairs in the developing
brain. PLoS Genetics, 5, e1000617.
Ponting, C. P., Oliver, P. L., & Reik, W. (2009). Evolution and functions of long noncoding
RNAs. Cell, 136, 629–641.
Pruunsild, P., Kazantseva, A., Aid, T., Palm, K., & Timmusk, T. (2007). Dissecting the
human BDNF locus: Bidirectional transcription, complex splicing, and multiple pro-
moters. Genomics, 90, 397–406.
Qin, R., Chen, Z., Ding, Y., Hao, J., Hu, J., & Guo, F. (2013). Long non-coding RNA
MEG3 inhibits the proliferation of cervical carcinoma cells through the induction of cell
cycle arrest and apoptosis. Neoplasma, 60, 486–492.
Reif, A., Fritzen, S., Finger, M., Strobel, A., Lauer, M., Schmitt, A., et al. (2006). Neural
stem cell proliferation is decreased in schizophrenia, but not in depression. Molecular Psy-
chiatry, 11, 514–522.
Ricci, S., Businaro, R., Ippoliti, F., Lo Vasco, V. R., Massoni, F., Onofri, E., et al. (2013).
Altered cytokine and BDNF levels in autism spectrum disorder. Neurotoxicity Research,
24, 491–501.
Rinn, J. L., & Chang, H. Y. (2012). Genome regulation by long noncoding RNAs. Annual
Review of Biochemistry, 81, 145–166.
Rinn, J. L., Kertesz, M., Wang, J. K., Squazzo, S. L., Xu, X., Brugmann, S. A., et al. (2007).
Functional demarcation of active and silent chromatin domains in human HOX loci by
noncoding RNAs. Cell, 129, 1311–1323.
Ross-Inta, C., Omanska-Klusek, A., Wong, S., Barrow, C., Garcia-Arocena, D.,
Iwahashi, C., et al. (2010). Evidence of mitochondrial dysfunction in fragile
X-associated tremor/ataxia syndrome. The Biochemical Journal, 429, 545–552.
LncRNAs in Autism 57
Rougeulle, C., Cardoso, C., Fontés, M., Colleaux, L., & Lalande, M. (1998). An imprinted
antisense RNA overlaps UBE3A and a second maternally expressed transcript. Nature
Genetics, 19, 15–16.
Samaco, R. C., Hogart, A., & LaSalle, J. M. (2005). Epigenetic overlap in autism-spectrum
neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A
and GABRB3. Human Molecular Genetics, 14, 483–492.
Sayed, D., & Abdellatif, M. (2011). MicroRNAs in development and disease. Physiological
Reviews, 91, 827–887.
Seila, A. C., Calabrese, J. M., Levine, S. S., Yeo, G. W., Rahl, P. B., Flynn, R. A., et al.
(2008). Divergent transcription from active promoters. Science, 322, 1849–1851.
Singer, O., Marr, R. A., Rockenstein, E., Crews, L., Coufal, N. G., Gage, F. H., et al.
(2005). Targeting BACE1 with siRNAs ameliorates Alzheimer disease neuropathology
in a transgenic model. Nature Neuroscience, 8, 1343–1349.
Sleutels, F., Zwart, R., & Barlow, D. P. (2002). The non-coding Air RNA is required for
silencing autosomal imprinted genes. Nature, 415, 810–813.
Song, R., Hennig, G. W., Wu, Q., Jose, C., Zheng, H., & Yan, W. (2011). Male germ cells
express abundant endogenous siRNAs. Proceedings of the National Academy of Sciences of the
United States of America, 108, 13159–13164.
St Pourcain, B., Wang, K., Glessner, J. T., Golding, J., Steer, C., Ring, S. M., et al. (2010).
Association between a high-risk autism locus on 5p14 and social communication spec-
trum phenotypes in the general population. The American Journal of Psychiatry, 167,
1364–1372.
Sullivan, P. F., Kendler, K. S., & Neale, M. C. (2003). Schizophrenia as a complex trait:
Evidence from a meta-analysis of twin studies. Archives of General Psychiatry, 60,
1187–1192.
Taft, R. J., Pheasant, M., & Mattick, J. S. (2007). The relationship between non-protein-
coding DNA and eukaryotic complexity. Bioessays, 29, 288–299.
Tam, O. H., Aravin, A. A., Stein, P., Girard, A., Murchison, E. P., Cheloufi, S., et al. (2008).
Pseudogene-derived small interfering RNAs regulate gene expression in mouse oocytes.
Nature, 453, 534–538.
Tanaka, J., Horiike, Y., Matsuzaki, M., Miyazaki, T., Ellis-Davies, G. C., & Kasai, H. (2008).
Protein synthesis and neurotrophin-dependent structural plasticity of single dendritic
spines. Science, 319, 1683–1687.
Taylor, M. S., Devon, R. S., Millar, J. K., & Porteous, D. J. (2003). Evolutionary constraints
on the disrupted in schizophrenia locus. Genomics, 81, 67–77.
Tiedge, H., Chen, W., & Brosius, J. (1993). Primary structure, neural-specific expression,
and dendritic location of human BC200 RNA. Journal of Neuroscience, 13, 2382–2390.
Tripathi, V., Ellis, J. D., Shen, Z., Song, D. Y., Pan, Q., Watt, A. T., et al. (2010). The
nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulat-
ing SR splicing factor phosphorylation. Molecular Cell, 39, 925–938.
Uhde, C. W., Vives, J., Jaeger, I., & Li, M. (2010). Rmst is a novel marker for the mouse
ventral mesencephalic floor plate and the anterior dorsal midline cells. PLoS One, 5,
e8641.
Vallot, C., Huret, C., Lesecque, Y., Resch, A., Oudrhiri, N., Bennaceur-Griscelli, A., et al.
(2013). XACT, a long noncoding transcript coating the active X chromosome in human
pluripotent cells. Nature Genetics, 45, 239–241.
Vlietstra, R. J., van Alewijk, D. C., Hermans, K. G., van Steenbrugge, G. J., & Trapman, J.
(1998). Frequent inactivation of PTEN in prostate cancer cell lines and xenografts. Cancer
Research, 58, 2720–2723.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature, 474,
380–384.
58 Brent Wilkinson and Daniel B. Campbell
Wang, X. H., Aliyari, R., Li, W. X., Li, H. W., Kim, K., Carthew, R., et al. (2006). RNA
interference directs innate immunity against viruses in adult Drosophila. Science, 312,
452–454.
Wang, K. C., & Chang, H. Y. (2011). Molecular mechanisms of long noncoding RNAs.
Molecular Cell, 43, 904–914.
Wang, P., Ren, Z., & Sun, P. (2012). Overexpression of the long non-coding RNA
MEG3 impairs in vitro glioma cell proliferation. Journal of Cellular Biochemistry, 113,
1868–1874.
Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner, J. T., Abrahams, B. S., et al. (2009).
Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature,
459, 528–533.
Watanabe, T., Totoki, Y., Toyoda, A., Kaneda, M., Kuramochi-Miyagawa, S., Obata, Y.,
et al. (2008). Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in
mouse oocytes. Nature, 453, 539–543.
Watson, P., Black, G., Ramsden, S., Barrow, M., Super, M., Kerr, B., et al. (2001). Angel-
man syndrome phenotype associated with mutations in MECP2, a gene encoding a
methyl CpG binding protein. Journal of Medical Genetics, 38, 224–228.
Wegiel, J., Kuchna, I., Nowicki, K., Imaki, H., Marchi, E., Ma, S. Y., et al. (2010). The
neuropathology of autism: Defects of neurogenesis and neuronal migration, and dysplas-
tic changes. Acta Neuropathologica, 119, 755–770.
Wheeler, E., Huang, N., Bochukova, E. G., Keogh, J. M., Lindsay, S., Garg, S., et al. (2013).
Genome-wide SNP and CNV analysis identifies common and low-frequency variants
associated with severe early-onset obesity. Nature Genetics, 45, 513–517.
Wienholds, E., & Plasterk, R. H. (2005). MicroRNA function in animal development.
FEBS Letters, 579, 5911–5922.
Winter, J., Jung, S., Keller, S., Gregory, R. I., & Diederichs, S. (2009). Many roads to matu-
rity: MicroRNA biogenesis pathways and their regulation. Nature Cell Biology, 11,
228–234.
Wonders, C. P., & Anderson, S. A. (2006). The origin and specification of cortical interneu-
rons. Nature Reviews Neuroscience, 7, 687–696.
Xia, J., Joyce, C. E., Bowcock, A. M., & Zhang, W. (2013). Noncanonical microRNAs and
endogenous siRNAs in normal and psoriatic human skin. Human Molecular Genetics, 22,
737–748.
Yamasaki, K., Joh, K., Ohta, T., Masuzaki, H., Ishimaru, T., Mukai, T., et al. (2003). Neu-
rons but not glial cells show reciprocal imprinting of sense and antisense transcripts of
Ube3a. Human Molecular Genetics, 12, 837–847.
Yang, N., & Kazazian, H. H. (2006). L1 retrotransposition is suppressed by endogenously
encoded small interfering RNAs in human cultured cells. Nature Structural & Molecular
Biology, 13, 763–771.
Ying, L., Huang, Y., Chen, H., Wang, Y., Xia, L., Chen, Y., et al. (2013). Downregulated
MEG3 activates autophagy and increases cell proliferation in bladder cancer. Molecular
BioSystems, 9, 407–411.
Yotova, I. Y., Vlatkovic, I. M., Pauler, F. M., Warczok, K. E., Ambros, P. F., Oshimura, M.,
et al. (2008). Identification of the human homolog of the imprinted mouse Air non-
coding RNA. Genomics, 92, 464–473.
Zhang, X., Gejman, R., Mahta, A., Zhong, Y., Rice, K. A., Zhou, Y., et al. (2010). Mater-
nally expressed gene 3, an imprinted noncoding RNA gene, is associated with menin-
gioma pathogenesis and progression. Cancer Research, 70, 2350–2358.
Zhang, X., Zhou, Y., Mehta, K. R., Danila, D. C., Scolavino, S., Johnson, S. R., et al.
(2003). A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor
cells. Journal of Clinical Endocrinology and Metabolism, 88, 5119–5126.
LncRNAs in Autism 59
Zhao, J., Dahle, D., Zhou, Y., Zhang, X., & Klibanski, A. (2005). Hypermethylation of the
promoter region is associated with the loss of MEG3 gene expression in human pituitary
tumors. Journal of Clinical Endocrinology and Metabolism, 90, 2179–2186.
Ziats, M. N., & Rennert, O. M. (2013). Aberrant expression of long noncoding RNAs in
autistic brain. Journal of Molecular Neuroscience, 49, 589–593.
Zoghbi, H. Y. (2003). Postnatal neurodevelopmental disorders: Meeting at the synapse?
Science, 302, 826–830.
CHAPTER THREE
Contents
1. Introduction to Cell Types and Autism Spectrum Disorder 62
2. Genetics of ASD 63
3. Brief Review of Rodent Behavioral Assays Relevant to ASD Symptoms 66
4. ASD Models Involving Serotonergic Neurons 69
5. ASD Models Involving GABAergic Interneurons 74
6. ASD Models Involving the Cerebellum 78
7. ASD Models Involving the Striatum 81
8. Other Regions and Cell Types 83
9. Conclusions 84
Acknowledgments 85
References 85
Abstract
Autism spectrum disorder (ASD) is highly genetic in its etiology, with potentially hun-
dreds of genes contributing to risk. Despite this heterogeneity, these disparate genetic
lesions may result in the disruption of a limited number of key cell types or circuits—
information which could be leveraged for the design of therapeutic interventions. While
hypotheses for cellular disruptions can be identified by postmortem anatomical analysis
and expression studies of ASD risk genes, testing these hypotheses requires the use of
animal models. In this review, we explore the existing evidence supporting the contri-
bution of different cell types to ASD, specifically focusing on rodent studies disrupting
serotonergic, GABAergic, cerebellar, and striatal cell types, with particular attention to
studies of the sufficiency of specific cellular disruptions to generate ASD-related behav-
ioral abnormalities. This evidence suggests multiple cellular routes can create features of
the disorder, though it is currently unclear if these cell types converge on a final com-
mon circuit. We hope that in the future, systematic studies of cellular sufficiency and
genetic interaction will help to classify patients into groups by type of cellular disrup-
tions which suggest tractable therapeutic targets.
How can one identify these cellular disruptions? There are several com-
plimentary approaches. Most directly, human brain imaging of patient
populations and postmortem anatomical experiments allow for identifica-
tion of regional and/or cellular abnormalities from human patients, respec-
tively. More indirectly, one can examine the expression patterns of known
ASD risk genes to identify the cells and regions most likely to be impacted by
their disruption. For example, if an ASD risk gene is only expressed in one
population of cells in the brain, then these cells must almost certainly medi-
ate the effect of the gene’s mutation. These approaches allow for the gen-
eration of hypotheses regarding cellular deficits, but testing these
hypotheses requires the use of animal models. Therefore, an essential com-
plement to patient-oriented approaches is the use of animal models to test
the sufficiency of disruptions in particular cell types to recreate key symp-
toms of the disorder. This can be done genetically in the mouse either by
the deletion of a gene only employed by a single cell type in the brain, or
by creating conditional knockouts using the Cre–Lox system (Nagy, 2000).
Here, we are going to focus on reviewing the existing data, particularly
from conditional mouse models, regarding the sufficiency of certain cell
types for the generation of ASD-like behavioral features. We will briefly
overview the genetics of the disorder with a focus on using the expression
of ASD-associated genes to guide us toward circuits that may be most
disrupted by their loss. We will then briefly discuss behavioral methods of
assessment of the ASD-like symptoms in rodents, as a preview to presenting
the current knowledge regarding the cell types that may mediate ASD-like
behaviors. We will focus on the four regions or cell types that have received
substantial attention in ASD thus far: serotonin-producing neurons,
GABAergic interneurons, the cerebellum, and the striatum.
2. GENETICS OF ASD
The genetics of ASD are thoroughly reviewed in recent publications
(Berg & Geschwind, 2012; Geschwind, 2011; Persico & Napolioni, 2013).
Briefly, the genetic variation contributing to risk in ASD has been investi-
gated in one of several ways. These have included identification of genes for
classic ASD-associated syndromes, common variant association studies, and
rare variant analyses. ASD-associated syndromes, such as Fragile
X syndrome, Rett syndrome, and tuberous sclerosis are typically caused
by highly penetrant, loss-of-function mutations in single genes (FMR1,
64 Susan E. Maloney et al.
Table 3.1 Genes identified by recent whole-exome sequencing (WES) studies with
regionalized patterns of expression
Gene
symbol Region, cell type References
FOXP1 Striatum, medium spiny neurons Ferland, Cherry, Preware,
Morrisey, and Walsh (2003),
Tamura, Morikawa, Iwanishi,
Hisaoka, and Senba (2004)
GRIN2B Striatum, all cell types Kuppenbender, Standaert,
Feuerstein, Penney, and Young
(2000)
NRXN1 Cortex, Ntsr1 þ and Cck þ cells; Dougherty, Schmidt, Nakajima,
cerebellum, granule cells and Heintz (2010)
PLXNB1 Cerebellum, purkinje cells Fazzari, Penachioni, Gianola,
Rossi, and Eickholt (2007)
RELN Cortex, GABA interneurons; Pesold, Impagnatiello, Pisu,
hippocampus, GABA Uzunov, and Costa (1998)
interneurons; cerebellum, granule
cells
TBR1 Cortex Bulfone, Smiga, Shimamura,
Peterson, and Puelles (1995),
Englund et al. (2005)
A minority of genes recently identified by WES studies have patterns of expression which have been
previously studied in the brain. Relevant references are listed alongside the regions and cell types
implicated.
more we learn about the spatial and temporal dynamics of ASD risk genes,
the better poised we are to make testable hypotheses about underlying cel-
lular and molecular mechanisms in the disorder. Currently, lacking is a
method akin to GO, which provides statistical analysis for the enrichment
of candidate genes in particular regions or cell types. We have recently
developed such a method, cell-type-specific expression analysis, and appli-
cation of this method to lists of previously implicated genes in ASD (Basu,
Kollu, & Banerjee-Basu, 2009) suggests a modest enrichment in genes found
in cortical interneurons and the striatum (not shown). These analyses will
likely be more informative as the number of candidate genes increases.
Furthermore, with advances in genome editing technologies ( Joung &
Sander, 2013; Wang et al., 2013), it will become easier to model the effects
of targeted mutations, in single and multiple genes, and study the effects of
these on behavior. While the relevance to ASD risk has been explored in
mouse knockout models for many genes, targeted knockout studies in
66 Susan E. Maloney et al.
specific cell populations have not been performed for the overwhelming
majority of candidate genes, with the exception of genes for some ASD-
related syndromes such as Tsc1 (Reith et al., 2013) and Mecp2 (Adachi,
Autry, Covington, & Monteggia, 2009; Alvarez-Saavedra, Saez, Kang,
Zoghbi, & Young, 2007; Chao et al., 2010; Fyffe et al., 2008;
Michaelson et al., 2012; Samaco et al., 2009).
Mouse models of ASD risk genes provide powerful tools for both explor-
ing the cellular phenotype that results when these genes are disrupted, as well
as the larger phenotype of the behaving animal, often mirroring the symp-
toms observed in humans. In the subsequent sections, we will first review
how ASD-like behaviors are identified and interpreted in mice. Then we
will proceed to discuss the existing evidence in several mouse models, with
a focus on conditional deletions and ASD genes that implicate certain cir-
cuits by their spatially or temporally restricted patterns of expression.
Wohr & Scattoni, 2013). Nonetheless, the assays discussed are the most
widely used in the studies highlighted below characterizing the cellular
mediators of ASD behaviors.
syndrome phenotype as these mice also did not show motor deficits, breath-
ing irregularities, or heightened anxiety (Samaco et al., 2009). In contrast to
this conditional deletion, there are a fair number of deletions of genes spe-
cific to serotonin cells, such as Tph2 and Slc6a4 (the serotonin transporter),
which can also serve to more broadly elucidate the sufficiency of serotoner-
gic disruption in generating ASD-like behaviors.
Many mutations of 5-HT-cell-specific genes result in ASD-like behav-
iors (see Table 3.2). Complete depletion of brain 5-HT by deletion of the
gene encoding Tph2, the rate-limiting enzyme in the synthesis of CNS
5-HT, results in abnormal social behaviors, communication deficits, and
repetitive behaviors (Alenina et al., 2009; Angoa-Perez et al., 2012; Kane
et al., 2012; Mosienko et al., 2012). A knock-in mouse model expressing
a mutant, low-activity form of Tph2, equivalent to a rare human variant,
also exhibits abnormal social behavior and an approximate 80% reduction
in brain 5-HT (Beaulieu et al., 2008). Likewise, mice null for Slc6a4, exhibit
abnormal social behaviors and repetitive behaviors as well as a loss of about
half of the serotonin-expressing neurons and reduced overall brain 5-HT
levels (Kalueff, Fox, Gallagher, & Murphy, 2007; Moy et al., 2009). While
an increase in 5-HT concentration has been reported in specific brain areas
like the striatum of Slc6a4/ mice (Mossner, Simantov, Marx, Lesch, &
Seif, 2006), it is likely due to compensatory 5-HT uptake by the dopamine
transporter in these areas (Zhou, Lesch, & Murphy, 2002) and not reflective
of an overall increase in brain 5-HT. Heterozygous Slc6a4 mutants display
ASD-like behaviors to a lesser degree than Slc6a4/ mice; however, 5-HT
levels were not reported in these mice (Kyzar et al., 2012; Moy et al., 2009).
Mice expressing a high activity Slc6a4 variant, Ala56, have unchanged over-
all 5-HT levels but do exhibit increased 5-HT clearance rates (Veenstra-
VanderWeele et al., 2012). Disrupted social and communicative behaviors
as well as increased stereotyped behaviors are demonstrated by these mice.
Interestingly, both mice lacking Slc6a4 and those expressing a high activity
Slc6a4 variant exhibit ASD-like phenotypes. Both models would be
predicted to have a decrease of synaptic serotonin overall—the Ala56 variant
due to more rapid clearance and the knockout due to long-term depletion of
serotonin from the presynapse in the absence of the ability to efficiently
recycle the transmitter. Finally, mice mutant for the Itgb3 gene, which
encodes a protein that interacts with Slc6a4, show slight social behavior def-
icits and repetitive behaviors as well as a reduced volume of the serotonergic-
expressing neurons of the dorsal raphe nucleus (Carter et al., 2011; Ellegood,
Henkelman, & Lerch, 2012).
Table 3.2 ASD-related phenotypes of genetic mouse models of the serotonin system
Stereotyped/repetitive
5-HT Abnormal social Communication behaviors, resistance to
Mutation 5-HT levels neurons behaviors deficits change References
/
Celf6 Reduced – Normal sociability Decreased pup Trend toward failed Dougherty et al. (2013)
(30%) USVs reversal performance;
failure to change hole-
poking behavior
Itgb3/ – Reduced Normal sociability; lack – Increased self-grooming Carter et al. (2011),
of preference for social in novel environment Ellegood et al. (2012)
novelty
Itgb3þ/ – – Normal sociability and – Slightly increased self- Carter et al. (2011)
preference for social grooming in novel
novelty environment
Slc6a4/ Reduced Reduced Decreased sociability; – Increased self-grooming Kalueff et al. (2007),
(50%) increased sensitivity to in homecage; normal Moy et al. (2009)
social stress; reduced self-grooming in novel
aggression; increased environment; normal
sociopositive behaviors nest building
Slc6a4þ/ – – Normal sociability in – Increased self-grooming Page et al. (2009), Kyzar
males; decreased in homecage; normal et al. (2012), Moy et al.
sociability in females; self-grooming in novel (2009)
normal preference for environment
social novelty; slightly
reduced aggression
Slc6a4 Unchanged – Decreased sociability; Decreased pup Repetitive homecage Veenstra-VanderWeele
Ala56 with increased submission to USVs wire hanging; normal et al. (2012)
(high- increased social dominance marble burying and
activity 5-HT self-grooming in
variant) clearance homecage
Tph2/ Absent Intact Postnatal lack of Decreased Increased nestlet Angoa-Perez et al.
preference for maternal urinary scent shredding, marbling, (2012), Kane et al.
scent; social memory marking burying, and digging in (2012), Mosienko et al.
deficits; social odorant episodes and mixed C57BL/6J-129Sv (2012), Alenina et al.
disinterest; decreased investigation background; decreased (2009)
social interaction time; marble burying with
lack of preference for increased activity in
social novelty; increased C57BL/6J background;
aggression increased motor
impulsivity; normal
reversal performance
Tph2 Reduced – Increased aggression – – Beaulieu et al. (2008)
R441H (80%)
(low-
activity
variant)
The impact of serotonin-related genetic mutation on 5-HT levels, 5-HT-expressing neurons, and behavioral phenotypes relevant to the core ASD symptoms.
Dash (–) indicates behavior was not assessed.
74 Susan E. Maloney et al.
be due to acute loss of Mecp2, and not to abnormal circuit formation during
development; inducible deletion of Mecp2 in the adult mouse was sufficient
to recapitulate some of the behavioral features of the germline mutation
(although social behaviors were not assessed) (McGraw, Samaco, &
Zoghbi, 2011). This suggests the imbalance of inhibitory control over exci-
tation induced by the absence of Mecp2 in the brain may be reversible.
In Mecp2, more than in any other model, multiple groups have
attempted postnatal “rescue” experiments by variously expressing Mecp2
under the control of various cell-specific and ubiquitous promoters. These
are an important complement to the cell-specific deletion experiments.
Deletions indicate which cell types are sufficient to disrupt the behavior.
Cell-specific rescue experiments indicate which cell types are sufficient
for normal behavior, and also provide some indication if the deficits are
due to acute loss of the protein, or permanent abnormalities that are a con-
sequence of the absence of Mecp2 during development. They also serve as a
model for potential treatment strategies. However, the interpretation of
these studies is complicated by the potentially nonphysiological levels of
expression of Mecp2 from exogenous promoters, and may account for
the differences seen across studies (Giacometti, Luikenhuis, Beard, &
Jaenisch, 2007; Guy, Gan, Selfridge, Cobb, & Bird, 2007; Jugloff et al.,
2008; Luikenhuis, Giacometti, Beard, & Jaenisch, 2004). Thus far, an
interneuron-specific Mecp2 rescue has not been demonstrated.
Other mutant models have provided evidence that disruption of
GABAergic inhibitory neurotransmission can result in an ASD-like pheno-
type. The mouse model of the ASD-related syndrome, Dravet’s syndrome,
which is caused in humans by heterozygous loss-of-function mutations in
the SCN1A gene, exhibits a 20–50% reduction in the a-subunit of the brain
voltage-gated Naþ channels. This is the primary Naþ channel in GABAergic
interneurons and thus is critical for action potentials in these neurons (Han
et al., 2012). GABAergic-specific deletion of Scn1a using the Dlx1/2 Cre
revealed that ASD-relevant behaviors in Dlx1/2–Scn1aþ/ mice, particu-
larly abnormal social behaviors, are due to decreased GABAergic neuro-
transmission specifically in the forebrain. These behaviors were reversed
with benzodiazepine administration. This study not only strongly implicates
the sufficiency of the loss of inhibitory control in the forebrain for abnormal
social behaviors but also further suggests abnormal social behaviors in some
ASD patients may not be irreversible consequences of neural development
and may, in fact, be treated in some manner with anticonvulsants or anxi-
olytics. Benzodiazepines are often prescribed to individuals with ASD
78 Susan E. Maloney et al.
These studies have mostly employed the Purkinje cell protein 2 (Pcp2)
sequence as a promoter driving Cre recombinase for cerebellar-specific dele-
tion of ASD-relevant genes. Pcp2–Fmr1/ mice exhibit altered dendritic
morphology in Purkinje cells and recapitulate the attenuated eyeblink con-
ditioning observed in global Fmr1/ mice and Fragile X patients, who have
a mutation in the FMR1 gene (Koekkoek et al., 2005). The Pcp2–Fmr1/
mice also show impaired sensorimotor gating, but other ASD-relevant
behavior testing was not reported. Cerebellar-specific deletion of either
Tsc1 or Tsc2, genes inactivated in the ASD-related syndrome tuberous scle-
rosis, results in a progressive loss of Purkinje cells due to apoptosis possibly
induced by neuronal stress (Reith et al., 2013; Tsai et al., 2012). Surprisingly,
these mice also display decreased social behaviors and increased repetitive
behaviors and USVs. The loss of Purkinje cells and the abnormal behaviors
were prevented with postnatal-onset of rapamycin treatment, which rec-
tifies the dysregulation of mTOR signaling downstream of Tsc1 or Tsc2.
These cerebellar-specific genetic deletions suggest a role for Purkinje cells
in ASD-relevant behaviors, likely resulting from the influence of these cells
on the excitation/inhibition balance in other brain areas.
The loss of Purkinje cells may alter the functioning of the frontal cortex.
An association was reported between early signs of ASD and dorsolateral
prefrontal cortex volume in premature infants with cerebellar injury
(Limperopoulos et al., 2012). The Purkinje cells receive excitatory input
from glutamatergic granule cells and provide GABAergic inhibition to
other areas of the cerebellum, particularly deep cerebellar nuclei. These
nuclei then send projections to the thalamus and cerebral cortex
(Gonzalo-Ruiz & Leichnetz, 1990; Middleton & Strick, 2001; Saab &
Willis, 2003; Sarna & Hawkes, 2003; Yamamoto, Yoshida, Yoshikawa,
Kishimoto, & Oka, 1992). Therefore, disruption of GABAergic inhibition
in the Purkinje cells can influence functioning in thalamocortical circuits.
Reduced Purkinje cell function has been suggested to ultimately produce
reduced cerebellar modulation of dopamine release in the medial prefrontal
cortex (Rogers et al., 2013). It is possible that loss of Purkinje cells ultimately
leads to an imbalance of the excitation/inhibition ratio in the cortex, which,
as discussed above, is hypothesized as an underlying mechanism of ASD.
However, there is also evidence suggesting that the Purkinje cells are a
particularly vulnerable population. For example, neonatal exposure to toxins
like alcohol or nicotine can reduce Purkinje cell numbers (Chen, Parnell, &
West, 1998). Thus, it is possible that the cerebellar abnormalities seen in
individuals with ASD are simply indicators of a broader developmental def-
icit influencing many systems.
Essential Cell Types and Circuits in ASD 81
factors FOXP1 (Ferland et al., 2003; Tamura et al., 2004) and FOXP2
(Takahashi, Liu, Hirokawa, & Takahashi, 2003), the dopamine receptor
DRD3 (Staal, de Krom, & de Jonge, 2012), and the postsynaptic density
scaffolding protein SHANK3 (Peca et al., 2011). While several disruptions
in FOXP1 are linked to ASD (Hamdan et al., 2010; O’Roak et al., 2011;
Talkowski et al., 2012), Foxp1 null mice have not yet been assessed for
behaviors relevant to ASD-like symptoms. FOXP2 is considered a potential
risk gene for ASD primarily due to its apparent role in speech and language
(Newbury & Monaco, 2010), as well as its regulation of downstream genes
MET and CNTNAP2 which have been associated previously with ASD risk
(Arking et al., 2008; Bakkaloglu et al., 2008; Mukamel et al., 2011; Vernes
et al., 2011). Reports on FOXP2 have focused on motor function and pro-
duction of USV (Fisher & Scharff, 2009), the latter of which has had some
conflicting evidence—either reporting a deficit in amount of vocalization
(Shu et al., 2005) or lack thereof, with a subtler phenotype in amplitude
of vocalization (Gaub, Groszer, Fisher, & Ehret, 2010). Mice deficient in
Foxp2 protein have not yet been assessed on other ASD-like measures, such
as the three-chambered test of sociability.
Among genes important to striatal function modeled in mice, perhaps
one of the most well documented in relation to ASD risk is SHANK3.
SH3 and multiple ankyrin repeat domains 3 (Shank3) is a scaffolding protein
associated with the postsynaptic density, which links receptors and ion chan-
nels at the postsynaptic terminus to the cytoskeleton and downstream
molecular signaling pathways (Sheng & Kim, 2000). Mice null for Shank3
protein show ASD-like behaviors in a number of behavioral assays as well as
disrupted corticostriatal neuronal transmission (Bozdagi et al., 2010;
Folstein, Dowd, Mankoski, & Tadevosyan, 2003; Verpelli et al., 2011;
Wang et al., 2011; Yang et al., 2012). Shank3 mutant mice display stereo-
typed motor behaviors, which has been proposed as correlated to deficits in
striatal function (Peca et al., 2011). Specifically, they show excessive
grooming (but not allogrooming) which leads to facial lesions (Peca et al.,
2011). In the same report, Shank3/ mice were found to have striatal
hypertrophy—both in the surface area and dendritic length of MSNs—a
finding which the authors suggest may mirror human reports of increased
volume in the caudate nucleus in autistic patients. Whole-cell patch-clamp
recording of Shank3/ mice showed reduced frequency and amplitude to
MSN AMPAR-mediated mEPSCs. Peca and colleagues in that report argue
that this dysfunction is restricted to the striatum, based upon lack of such
deficits in transmission in the hippocampus, as well as normal reversal
Essential Cell Types and Circuits in ASD 83
learning in the Morris water maze task (Peca et al., 2011). However, because
Shank3 is also expressed in the cerebellum (Welch, Wang, & Feng, 2004),
and because the cerebellum may also have a role in the expression of autistic-
like phenotypes, it is not clear that there is not also cerebellar dysfunction in
the Shank3 null mouse. Furthermore, more recently, Yang et al. (2012) have
shown reduced glutamatergic synaptic transmission in the hippocampus and
a deficit in long-term potentiation in Shank3/ mice.
Overall, in the models described, it is difficult to assess the contribution
of striatal dysfunction to the observed phenotype, as genes such as SHANK3
are not exclusive to the striatum. Furthermore, many other ASD risk genes,
which have more global expression, may have a particularly crucial role to
play in the striatum that has yet been undiscovered. To address these prob-
lems, it will be useful to look at specific striatal disruption of these genes.
There are a number of transgenic mice, expressing Cre recombinase under
the control of different gene promoters, which can be used to mediate dis-
ruption in the striatum. The promoters driving Cre expression are as
described in Gong et al. (2007) (and on gensat.org) and their genes are sum-
marized in Table 3.3. Novel methodologies, such as translational profiling of
cell populations (Doyle et al., 2008; Heiman et al., 2008), have the potential
to uncover highly specific markers of different cell types, which can be used
to benefit future genetic manipulations.
Table 3.3 Promoters used to drive Cre recombination in the striatum (from GENSAT)
Gene
symbol Gene Pattern of Cre expression
Adora2a Adenosine A2A receptor Drd2 þ (striatopallidal) projection neurons
Dlx5 Distal-less homeobox 5 Projection neurons (also expressed in GABA
interneurons of the cortex and in the reticular
nucleus of the thalamus)
Drd1a Dopamine receptor D1A Drd1a þ (striatonigral) projection neurons
(some limited expression in cortex and
hypothalamus)
Drd2 Dopamine receptor D2 Drd2 þ (striatopallidal) projection neurons
(some limited expression in limbic cortex and
hypothalamus)
Drd3 Dopamine receptor D3 Drd3 þ (ventral striatum) neurons
(expression in layers 2 and 3 of cortex and in
the EC of the hippocampus)
Gng7 Guanine nucleotide Both Drd1a þ and Drd2 þ projection
binding protein, gamma 7 neurons (scattered expression in cortex and
hippocampus as well)
Vipr2 Vasoactive intestinal Both Drd1a þ and Drd2 þ (also cortex,
peptide receptor 2 layer 5)
A list of available Cre recombinase-expressing transgenic mouse lines available with expression in the
striatum, varying in specificity of expression.
LaSalle, & Jin, 2009) and neural stem cells (Amiri et al., 2012). In-depth ana-
lyses of the sufficiency of these cell types to create ASD-like behavior dis-
ruptions are certainly needed. Determination of sufficiency in conditional
deletion experiments must take into account that drivers of recombination
have varying levels of specificity (Gofflot et al., 2011). Ultimately, converg-
ing lines of evidence, from multiple mouse models and human neuroanat-
omy, will help to define the cell types and circuits that form the basis of ASD
symptoms.
9. CONCLUSIONS
From the current review of the consequences of conditional deletions
and deletions of genes enriched in certain cell types, it is clear there are mul-
tiple cellular disruptions that are sufficient to recreate some ASD-like
Essential Cell Types and Circuits in ASD 85
symptoms in the mouse. What does this suggest to us about the likely cellular
mechanisms of human ASD? Some things are becoming clearer.
First, there are mutations that lead to broad deficits in the early organi-
zation of the brain such as in the gene RELN (Goffinet, 1984; Neale et al.,
2012) or CNTNAP2 (Penagarikano et al., 2011). These mutations disrupt
many different circuits and lead to multiple deficits including intellectual dis-
ability, epilepsy, motor coordination difficulties, and finally ASD. These
deficits may be more difficult to treat with a single strategy, and may repre-
sent a class of developmental disorders that need to be considered differently
than other diagnoses of ASD (Gillberg, 2010).
Second, even among those individuals without broad cellular disorgani-
zation of CNS development, it seems likely that, much like the heteroge-
neity of ASD genetics, there is likely to be some heterogeneity of cellular
mechanisms as well. Thus, compared to Parkinson’s disease, it seems
unlikely that all ASD patients will share a single common cellular pathology.
Yet, it may still be the case that there are a limited number of distinct cellular
pathologies leading to the disorder. For example, it is possible that a subset of
patients develop ASD as a consequence of serotonergic abnormalities, while
another subset as a consequence of disrupted social reward processing in the
striatum. If the ASD cases can be clustered by cellular deficits, then at
least within these clusters, patients with distinct genetic causes may still
respond to a single treatment strategy. Genetic interaction experiments as
well as conditional deletion of a variety of ASD risk genes across different
cell types, in conjunction with careful and consistent phenotyping, are going
to be key to understanding whether such a clustering of cellular mechanisms
indeed exists.
ACKNOWLEDGMENTS
This work was supported by NINDS (4R00NS067239-03) to J. D. D., and an NIMH ACE
Network Grant (9R01MH100027-06). M. A. R. was supported by Kirschtein-NRSA
(5T32GM007067-38).
REFERENCES
Abrahams, B. S., & Geschwind, D. H. (2008). Advances in autism genetics: On the threshold
of a new neurobiology. Nature Reviews Genetics, 9(5), 341–355.
Adachi, M., Autry, A. E., Covington, H. E., 3rd., & Monteggia, L. M. (2009). MeCP2-
mediated transcription repression in the basolateral amygdala may underlie heightened
anxiety in a mouse model of Rett syndrome. Journal of Neuroscience, 29(13), 4218–4227.
Alenina, N., Kikic, D., Todiras, M., Mosienko, V., Qadri, F., Plehm, R., et al. (2009).
Growth retardation and altered autonomic control in mice lacking brain serotonin.
86 Susan E. Maloney et al.
Proceedings of the National Academy of Sciences of the United States of America, 106(25),
10332–10337.
Allen, G., Muller, R. A., & Courchesne, E. (2004). Cerebellar function in autism: Functional
magnetic resonance image activation during a simple motor task. Biological Psychiatry,
56(4), 269–278.
Alvarez-Saavedra, M., Saez, M. A., Kang, D., Zoghbi, H. Y., & Young, J. I. (2007). Cell-
specific expression of wild-type MeCP2 in mouse models of Rett syndrome yields
insight about pathogenesis. Human Molecular Genetics, 16(19), 2315–2325.
Amiri, A., Cho, W., Zhou, J., Birnbaum, S. G., Sinton, C. M., McKay, R. M., et al. (2012).
Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnor-
malities and alters neurogenesis. The Journal of Neuroscience: The official journal of the Society
for Neuroscience, 32(17), 5880–5890.
Anderson, G. M., Freedman, D. X., Cohen, D. J., Volkmar, F. R., Hoder, E. L.,
McPhedran, P., et al. (1987). Whole blood serotonin in autistic and normal subjects. Jour-
nal of Child Psychology and Psychiatry, 28(6), 885–900.
Angoa-Perez, M., Kane, M. J., Briggs, D. I., Sykes, C. E., Shah, M. M., Francescutti, D. M.,
et al. (2012). Genetic depletion of brain 5HT reveals a common molecular pathway
mediating compulsivity and impulsivity. Journal of Neurochemistry, 121(6), 974–984.
Anney, R., Klei, L., Pinto, D., Almeida, J., Bacchelli, E., Baird, G., et al. (2012). Individual
common variants exert weak effects on the risk for autism spectrum disorders. Human
Molecular Genetics, 21(21), 4781–4792.
Arakawa, H., Blanchard, D. C., Arakawa, K., Dunlap, C., & Blanchard, R. J. (2008). Scent
marking behavior as an odorant communication in mice. Neuroscience and Biobehavioral
Reviews, 32(7), 1236–1248.
Arking, D. E., Cutler, D. J., Brune, C. W., Teslovich, T. M., West, K., Ikeda, M., et al.
(2008). A common genetic variant in the neurexin superfamily member CNTNAP2
increases familial risk of autism. American Journal of Human Genetics, 82(1), 160–164.
Azmitia, E. C., Singh, J. S., & Whitaker-Azmitia, P. M. (2011). Increased serotonin axons
(immunoreactive to 5-HT transporter) in postmortem brains from young autism donors.
Neuropharmacology, 60(7–8), 1347–1354.
Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E., et al. (1995).
Autism as a strongly genetic disorder: Evidence from a British twin study. Psychological
Medicine, 25(1), 63–77.
Bailey, A., Luthert, P., Dean, A., Harding, B., Janota, I., Montgomery, M., et al. (1998).
A clinicopathological study of autism. Brain, 121(Pt. 5), 889–905.
Bakkaloglu, B., O’Roak, B. J., Louvi, A., Gupta, A. R., Abelson, J. F., Morgan, T. M., et al.
(2008). Molecular cytogenetic analysis and resequencing of contactin associated protein-
like 2 in autism spectrum disorders. American Journal of Human Genetics, 82(1), 165–173.
Ballas, N., Lioy, D. T., Grunseich, C., & Mandel, G. (2009). Non-cell autonomous influence
of MeCP2-deficient glia on neuronal dendritic morphology. Nature Neuroscience, 12(3),
311–317.
Basu, S. N., Kollu, R., & Banerjee-Basu, S. (2009). AutDB: A gene reference resource for
autism research. Nucleic Acids Research, 37, D832–D836.
Beaulieu, J. M., Zhang, X., Rodriguiz, R. M., Sotnikova, T. D., Cools, M. J.,
Wetsel, W. C., et al. (2008). Role of GSK3 beta in behavioral abnormalities induced
by serotonin deficiency. Proceedings of the National Academy of Sciences of the United States
of America, 105(4), 1333–1338.
Berg, J. M., & Geschwind, D. H. (2012). Autism genetics: Searching for specificity and con-
vergence. Genome Biology, 13(7), 247.
Betancur, C., Corbex, M., Spielewoy, C., Philippe, A., Laplanche, J. L., Launay, J. M., et al.
(2002). Serotonin transporter gene polymorphisms and hyperserotonemia in autistic dis-
order. Molecular Psychiatry, 7(1), 67–71.
Essential Cell Types and Circuits in ASD 87
Bozdagi, O., Sakurai, T., Papapetrou, D., Wang, X., Dickstein, D. L., Takahashi, N., et al.
(2010). Haploinsufficiency of the autism-associated Shank3 gene leads to deficits in
synaptic function, social interaction, and social communication. Molecular Autism,
1(1), 15.
Brielmaier, J., Matteson, P. G., Silverman, J. L., Senerth, J. M., Kelly, S., Genestine, M., et al.
(2012). Autism-relevant social abnormalities and cognitive deficits in engrailed-2 knock-
out mice. PLoS One, 7(7), e40914.
Bulfone, A., Smiga, S. M., Shimamura, K., Peterson, A., Puelles, L., et al. (1995). T-brain-1:
a homolog of Brachyury whose expression defines molecularly distinct domains within
the cerebral cortex. Neuron, 15, 63–78.
Buxbaum, J. D., Silverman, J. M., Smith, C. J., Greenberg, D. A., Kilifarski, M., Reichert, J.,
et al. (2002). Association between a GABRB3 polymorphism and autism. Molecular Psy-
chiatry, 7(3), 311–316.
Canitano, R., & Scandurra, V. (2011). Psychopharmacology in autism: An update. Progress in
Neuro-Psychopharmacology & Biological Psychiatry, 35(1), 18–28.
Carrasco, M., Volkmar, F. R., & Bloch, M. H. (2012). Pharmacologic treatment of repetitive
behaviors in autism spectrum disorders: Evidence of publication bias. Pediatrics, 129(5),
e1301–e1310.
Carter, M. D., Shah, C. R., Muller, C. L., Crawley, J. N., Carneiro, A. M., & Veenstra-
VanderWeele, J. (2011). Absence of preference for social novelty and increased
grooming in integrin beta3 knockout mice: Initial studies and future directions. Autism
Research, 4(1), 57–67.
Chahrour, M. H., Yu, T. W., Lim, E. T., Ataman, B., Coulter, M. E., Hill, R. S., et al.
(2012). Whole-exome sequencing and homozygosity analysis implicate
depolarization-regulated neuronal genes in autism. PLoS Genetics, 8(4), e1002635.
Chao, H. T., Chen, H., Samaco, R. C., Xue, M., Chahrour, M., Yoo, J., et al. (2010). Dys-
function in GABA signalling mediates autism-like stereotypies and Rett syndrome phe-
notypes. Nature, 468(7321), 263–269.
Cheh, M. A., Millonig, J. H., Roselli, L. M., Ming, X., Jacobsen, E., Kamdar, S., et al.
(2006). En2 knockout mice display neurobehavioral and neurochemical alterations rel-
evant to autism spectrum disorder. Brain Research, 1116(1), 166–176.
Chen, R. Z., Akbarian, S., Tudor, M., & Jaenisch, R. (2001). Deficiency of methyl-CpG
binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nature
Genetics, 27(3), 327–331.
Chen, W. J., Parnell, S. E., & West, J. R. (1998). Neonatal alcohol and nicotine exposure
limits brain growth and depletes cerebellar Purkinje cells. Alcohol, 15(1), 33–41.
Ching, H., & Pringsheim, T. (2012). Aripiprazole for autism spectrum disorders (ASD).
Cochrane Database of Systematic Reviews, 5, CD009043.
Cook, E. H., Jr., Courchesne, R. Y., Cox, N. J., Lord, C., Gonen, D., Guter, S. J., et al.
(1998). Linkage-disequilibrium mapping of autistic disorder, with 15q11-13 markers.
American Journal of Human Genetics, 62(5), 1077–1083.
Cook, E. H., & Leventhal, B. L. (1996). The serotonin system in autism. Current Opinion in
Pediatrics, 8(4), 348–354.
Courchesne, E., Yeung-Courchesne, R., Press, G. A., Hesselink, J. R., & Jernigan, T. L.
(1988). Hypoplasia of cerebellar vermal lobules VI and VII in autism. New England Journal
of Medicine, 318(21), 1349–1354.
Crawley, J. N. (2012). Translational animal models of autism and neurodevelopmental dis-
orders. Dialogues in Clinical Neuroscience, 14(3), 293–305.
Dani, V. S., Chang, Q., Maffei, A., Turrigiano, G. G., Jaenisch, R., & Nelson, S. B. (2005).
Reduced cortical activity due to a shift in the balance between excitation and inhibition
in a mouse model of Rett syndrome. Proceedings of the National Academy of Sciences of the
United States of America, 102(35), 12560–12565.
88 Susan E. Maloney et al.
Deane, C. M., Salwinski, L., Xenarios, I., & Eisenberg, D. (2002). Protein interactions: Two
methods for assessment of the reliability of high throughput observations. Molecular &
Cellular Proteomics, 1(5), 349–356.
DeLorey, T. M., Handforth, A., Anagnostaras, S. G., Homanics, G. E., Minassian, B. A.,
Asatourian, A., et al. (1998). Mice lacking the beta3 subunit of the GABAA receptor
have the epilepsy phenotype and many of the behavioral characteristics of Angelman syn-
drome. Journal of Neuroscience, 18(20), 8505–8514.
DeLorey, T. M., Sahbaie, P., Hashemi, E., Homanics, G. E., & Clark, J. D. (2008). Gabrb3
gene deficient mice exhibit impaired social and exploratory behaviors, deficits in non-
selective attention and hypoplasia of cerebellar vermal lobules: A potential model of
autism spectrum disorder. Behavioural Brain Research, 187(2), 207–220.
Di Martino, A., Kelly, C., Grzadzinski, R., Zuo, X. N., Mennes, M., Mairena, M. A., et al.
(2011). Aberrant striatal functional connectivity in children with autism. Biological Psy-
chiatry, 69(9), 847–856.
Dougherty, J. D., Schmidt, E. F., Nakajima, M., & Heintz, N. (2010). Analytical approaches
to RNA profiling data for the identification of genes enriched in specific cells. Nucleic
Acids Res, 38, 4218–4230.
Dougherty, J. D., Maloney, S. E., Wozniak, D. F., Rieger, M. A., Sonnenblick, L.,
Coppola, G., et al. (2013). The disruption of Celf6, a gene identified by translational
profiling of serotonergic neurons, results in autism-related behaviors. Journal of Neurosci-
ence, 33(7), 2732–2753.
Doyle, J. P., Dougherty, J. D., Heiman, M., Schmidt, E. F., Stevens, T. R., Ma, G., et al.
(2008). Application of a translational profiling approach for the comparative analysis of
CNS cell types. Cell, 135(4), 749–762.
du Plessis, L., Skunca, N., & Dessimoz, C. (2011). The what, where, how and why of
gene ontology—A primer for bioinformaticians. Briefings in Bioinformatics, 12(6), 723–735.
Ellegood, J., Henkelman, R. M., & Lerch, J. P. (2012). Neuroanatomical assessment of the
integrin beta3 mouse model related to autism and the serotonin system using high res-
olution MRI. Front Psychiatry, 3, 37.
Englund, C., et al. (2005). Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia,
intermediate progenitor cells, and postmitotic neurons in developing neocortex. J Neu-
rosci, 25, 247–251.
Fazzari, P., Penachioni, J., Gianola, S., Rossi, F., Eickholt, B. J., et al. (2007). Plexin-B1 plays
a redundant role during mouse development and in tumour angiogenesis. BMC Dev Biol,
7, 55.
Ferland, R. J., Cherry, T. J., Preware, P. O., Morrisey, E. E., & Walsh, C. A. (2003).
Characterization of Foxp2 and Foxp1 mRNA and protein in the developing and mature
brain. J Comp Neurol, 460(2), 266–279.
Fisher, S. E., & Scharff, C. (2009). FOXP2 as a molecular window into speech and language.
Trends in Genetics, 25(4), 166–177.
Folstein, S. E., Dowd, M., Mankoski, R., & Tadevosyan, O. (2003). How might genetic
mechanisms operate in autism? Novartis Foundation Symposium, 251, 70–80, discussion
80–73, 109–111, 281–297.
Fyffe, S. L., Neul, J. L., Samaco, R. C., Chao, H. T., Ben-Shachar, S., Moretti, P., et al.
(2008). Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2
in feeding behavior, aggression, and the response to stress. Neuron, 59(6), 947–958.
Gaub, S., Groszer, M., Fisher, S. E., & Ehret, G. (2010). The structure of innate vocalizations
in Foxp2-deficient mouse pups. Genes, Brain, and Behavior, 9(4), 390–401.
Gerfen, C. R. (1992). The neostriatal mosaic: Multiple levels of compartmental organization.
Trends in Neurosciences, 15(4), 133–139.
Geschwind, D. H. (2011). Genetics of autism spectrum disorders. Trends in Cognitive Sciences,
15(9), 409–416.
Essential Cell Types and Circuits in ASD 89
Gharani, N., Benayed, R., Mancuso, V., Brzustowicz, L. M., & Millonig, J. H. (2004). Asso-
ciation of the homeobox transcription factor, ENGRAILED 2, 3, with autism spectrum
disorder. Molecular Psychiatry, 9(5), 474–484.
Giacometti, E., Luikenhuis, S., Beard, C., & Jaenisch, R. (2007). Partial rescue of MeCP2
deficiency by postnatal activation of MeCP2. Proceedings of the National Academy of Sciences
of the United States of America, 104(6), 1931–1936.
Gillberg, C. (2010). The ESSENCE in child psychiatry: Early symptomatic syndromes
eliciting neurodevelopmental clinical examinations. Research in Developmental Disabilities,
31(6), 1543–1551.
Goffinet, A. M. (1984). Events governing organization of postmigratory neurons: Studies on
brain development in normal and reeler mice. Brain Research, 319(3), 261–296.
Gofflot, F., Wendling, O., Chartoire, N., Birling, M.-C., Warot, X., & Auwerx, J. (2011).
Characterization and validation of Cre-driver mouse lines. Current Protocols in Mouse
Biology, 1, 1–15.
Gong, S., Doughty, M., Harbaugh, C. R., Cummins, A., Hatten, M. E., Heintz, N., et al.
(2007). Targeting Cre recombinase to specific neuron populations with bacterial artificial
chromosome constructs. Journal of Neuroscience, 27(37), 9817–9823.
Gonzalo-Ruiz, A., & Leichnetz, G. R. (1990). Connections of the caudal cerebellar inter-
positus complex in a new world monkey (Cebus apella). Brain Research Bulletin, 25(6),
919–927.
Gregorios-Pippas, L., Tobler, P. N., & Schultz, W. (2009). Short-term temporal discounting
of reward value in human ventral striatum. Journal of Neurophysiology, 101(3), 1507–1523.
Guy, J., Gan, J., Selfridge, J., Cobb, S., & Bird, A. (2007). Reversal of neurological defects in
a mouse model of Rett syndrome. Science, 315(5815), 1143–1147.
Guy, J., Hendrich, B., Holmes, M., Martin, J. E., & Bird, A. (2001). A mouse Mecp2-null
mutation causes neurological symptoms that mimic Rett syndrome. Nature Genetics,
27(3), 322–326.
Hallmayer, J., Cleveland, S., Torres, A., Phillips, J., Cohen, B., Torigoe, T., et al. (2011).
Genetic heritability and shared environmental factors among twin pairs with autism.
Archives of General Psychiatry, 68(11), 1095–1102.
Hamdan, F. F., Daoud, H., Rochefort, D., Piton, A., Gauthier, J., Langlois, M., et al. (2010).
De novo mutations in FOXP1 in cases with intellectual disability, autism, and language
impairment. American Journal of Human Genetics, 87(5), 671–678.
Han, S., Tai, C., Westenbroek, R. E., Yu, F. H., Cheah, C. S., Potter, G. B., et al. (2012).
Autistic-like behaviour in Scn1a þ/- mice and rescue by enhanced GABA-mediated
neurotransmission. Nature, 489(7416), 385–390.
Heiman, M., Schaefer, A., Gong, S., Peterson, J. D., Day, M., Ramsey, K. E., et al. (2008).
A translational profiling approach for the molecular characterization of CNS cell types.
Cell, 135(4), 738–748.
Hendricks, T. J., Fyodorov, D. V., Wegman, L. J., Lelutiu, N. B., Pehek, E. A.,
Yamamoto, B., et al. (2003). Pet-1 ETS gene plays a critical role in 5-HT neuron devel-
opment and is required for normal anxiety-like and aggressive behavior. Neuron, 37(2),
233–247.
Hofer, M. A., Shair, H. N., & Brunelli, S. A. (2002). Ultrasonic vocalizations in rat and
mouse pups. Current Protocols in Neuroscience, (Chapter 8), Unit 8.14.
Hollander, E., Anagnostou, E., Chaplin, W., Esposito, K., Haznedar, M. M., Licalzi, E., et al.
(2005). Striatal volume on magnetic resonance imaging and repetitive behaviors in
autism. Biological Psychiatry, 58(3), 226–232.
Iossifov, I., Ronemus, M., Levy, D., Wang, Z., Hakker, I., Rosenbaum, J., et al. (2012). De
novo gene disruptions in children on the autistic spectrum. Neuron, 74(2), 285–299.
Joung, J. K., & Sander, J. D. (2013). TALENs: A widely applicable technology for targeted
genome editing. Nature Reviews Molecular Cell Biology, 14(1), 49–55.
90 Susan E. Maloney et al.
Joyner, A. L., Herrup, K., Auerbach, B. A., Davis, C. A., & Rossant, J. (1991). Subtle cer-
ebellar phenotype in mice homozygous for a targeted deletion of the En-2 homeobox.
Science, 251(4998), 1239–1243.
Jugloff, D. G., Vandamme, K., Logan, R., Visanji, N. P., Brotchie, J. M., & Eubanks, J. H.
(2008). Targeted delivery of an Mecp2 transgene to forebrain neurons improves the
behavior of female Mecp2-deficient mice. Human Molecular Genetics, 17(10), 1386–1396.
Kalueff, A. V., Fox, M. A., Gallagher, P. S., & Murphy, D. L. (2007). Hypolocomotion,
anxiety and serotonin syndrome-like behavior contribute to the complex phenotype
of serotonin transporter knockout mice. Genes, Brain, and Behavior, 6(4), 389–400.
Kane, M. J., Angoa-Perez, M., Briggs, D. I., Sykes, C. E., Francescutti, D. M.,
Rosenberg, D. R., et al. (2012). Mice genetically depleted of brain serotonin display
social impairments, communication deficits and repetitive behaviors: Possible relevance
to autism. PLoS One, 7(11), e48975.
Kemp, J. M., & Powell, T. P. (1970). The cortico-striate projection in the monkey. Brain,
93(3), 525–546.
Kirov, G., Pocklington, A. J., Holmans, P., Ivanov, D., Ikeda, M., Ruderfer, D., et al.
(2012). De novo CNV analysis implicates specific abnormalities of postsynaptic signalling
complexes in the pathogenesis of schizophrenia. Molecular Psychiatry, 17(2), 142–153.
Kirsten, T. B., Chaves-Kirsten, G. P., Chaible, L. M., Silva, A. C., Martins, D. O.,
Britto, L. R., et al. (2012). Hypoactivity of the central dopaminergic system and
autistic-like behavior induced by a single early prenatal exposure to lipopolysaccharide.
Journal of Neuroscience Research, 90(10), 1903–1912.
Klei, L., Sanders, S. J., Murtha, M. T., Hus, V., Lowe, J. K., Willsey, A. J., et al. (2012).
Common genetic variants, acting additively, are a major source of risk for autism. Molec-
ular Autism, 3(1), 9.
Koekkoek, S. K., Yamaguchi, K., Milojkovic, B. A., Dortland, B. R., Ruigrok, T. J.,
Maex, R., et al. (2005). Deletion of FMR1 in Purkinje cells enhances parallel fiber
LTD, enlarges spines, and attenuates cerebellar eyelid conditioning in Fragile
X syndrome. Neuron, 47(3), 339–352.
Kron, M., Howell, C. J., Adams, I. T., Ransbottom, M., Christian, D., Ogier, M., et al.
(2012). Brain activity mapping in Mecp2 mutant mice reveals functional deficits in fore-
brain circuits, including key nodes in the default mode network, that are reversed with
ketamine treatment. Journal of Neuroscience, 32(40), 13860–13872.
Kuppenbender, K. D., Standaert, D. G., Feuerstein, T. J., Penney, J. B., Jr., Young, A. B.,
et al. (2000). Expression of NMDA receptor subunit mRNAs in neurochemically iden-
tified projection and interneurons in the human striatum. J Comp Neurol, 419, 407–421.
Kyzar, E. J., Pham, M., Roth, A., Cachat, J., Green, J., Gaikwad, S., et al. (2012). Alterations
in grooming activity and syntax in heterozygous SERT and BDNF knockout mice: The
utility of behavior-recognition tools to characterize mutant mouse phenotypes. Brain
Research Bulletin, 89(5–6), 168–176.
Langen, M., Schnack, H. G., Nederveen, H., Bos, D., Lahuis, B. E., de Jonge, M. V., et al.
(2009). Changes in the developmental trajectories of striatum in autism. Biological Psychi-
atry, 66(4), 327–333.
Lesch, K. P., Wolozin, B. L., Murphy, D. L., & Reiderer, P. (1993). Primary structure of the
human platelet serotonin uptake site: Identity with the brain serotonin transporter. Jour-
nal of Neurochemistry, 60(6), 2319–2322.
Liljelund, P., Handforth, A., Homanics, G. E., & Olsen, R. W. (2005). GABAA receptor
beta3 subunit gene-deficient heterozygous mice show parent-of-origin and gender-
related differences in beta3 subunit levels, EEG, and behavior. Brain Research Developmen-
tal Brain Research, 157(2), 150–161.
Limperopoulos, C., Chilingaryan, G., Sullivan, N., Guizard, N., Robertson, R. L., & du
Plessis, A. J. (2012). Injury to the premature cerebellum: Outcome is related to remote
cortical development. Cerebral Cortex. [E-pub ahead of print].
Essential Cell Types and Circuits in ASD 91
Lord, C., Risi, S., Lambrecht, L., Cook, E. H., Jr., Leventhal, B. L., DiLavore, P. C., et al.
(2000). The autism diagnostic observation schedule-generic: A standard measure of social
and communication deficits associated with the spectrum of autism. Journal of Autism and
Developmental Disorders, 30(3), 205–223.
Lord, C., Rutter, M., & Le Couteur, A. (1994). Autism Diagnostic Interview-Revised:
A revised version of a diagnostic interview for caregivers of individuals with possible
pervasive developmental disorders. Journal of Autism and Developmental Disorders, 24(5),
659–685.
Luikenhuis, S., Giacometti, E., Beard, C. F., & Jaenisch, R. (2004). Expression of MeCP2 in
postmitotic neurons rescues Rett syndrome in mice. Proceedings of the National Academy of
Sciences of the United States of America, 101(16), 6033–6038.
Maezawa, I., Swanberg, S., Harvey, D., LaSalle, J. M., & Jin, L. W. (2009). Rett syndrome
astrocytes are abnormal and spread MeCP2 deficiency through gap junctions. The Journal
of Neuroscience: The official journal of the Society for Neuroscience, 29(16), 5051–5061.
Markram, H., Toledo-Rodriguez, M., Wang, Y., Gupta, A., Silberberg, G., & Wu, C.
(2004). Interneurons of the neocortical inhibitory system. Nature Reviews Neuroscience,
5(10), 793–807.
McDougle, C. J., Naylor, S. T., Goodman, W. K., Volkmar, F. R., Cohen, D. J., &
Price, L. H. (1993). Acute tryptophan depletion in autistic disorder: A controlled case
study. Biological Psychiatry, 33(7), 547–550.
McFarlane, H. G., Kusek, G. K., Yang, M., Phoenix, J. L., Bolivar, V. J., & Crawley, J. N.
(2008). Autism-like behavioral phenotypes in BTBR T þ tf/J mice. Genes, Brain, and
Behavior, 7(2), 152–163.
McGraw, C. M., Samaco, R. C., & Zoghbi, H. Y. (2011). Adult neural function requires
MeCP2. Science, 333(6039), 186.
Michaelson, J. J., Shi, Y., Gujral, M., Zheng, H., Malhotra, D., Jin, X., et al. (2012). Whole-
genome sequencing in autism identifies hot spots for de novo germline mutation. Cell,
151(7), 1431–1442.
Miczek, K. A., Faccidomo, S., De Almeida, R. M., Bannai, M., Fish, E. W., & Debold, J. F.
(2004). Escalated aggressive behavior: New pharmacotherapeutic approaches and oppor-
tunities. Annals of the New York Academy of Sciences, 1036, 336–355.
Middleton, F. A., & Strick, P. L. (2000). Basal ganglia output and cognition: Evidence from
anatomical, behavioral, and clinical studies. Brain and Cognition, 42(2), 183–200.
Middleton, F. A., & Strick, P. L. (2001). Cerebellar projections to the prefrontal cortex of the
primate. Journal of Neuroscience, 21(2), 700–712.
Millen, K. J., Wurst, W., Herrup, K., & Joyner, A. L. (1994). Abnormal embryonic cerebellar
development and patterning of postnatal foliation in two mouse Engrailed-2 mutants.
Development, 120(3), 695–706.
Mosienko, V., Bert, B., Beis, D., Matthes, S., Fink, H., Bader, M., et al. (2012). Exaggerated
aggression and decreased anxiety in mice deficient in brain serotonin. Translational Psychiatry,
2, e122.
Mossner, R., Simantov, R., Marx, A., Lesch, K. P., & Seif, I. (2006). Aberrant accumulation
of serotonin in dopaminergic neurons. Neuroscience Letters, 401(1–2), 49–54.
Moy, S. S., Nadler, J. J., Magnuson, T. R., & Crawley, J. N. (2006). Mouse models of autism
spectrum disorders: The challenge for behavioral genetics. American Journal of Medical
Genetics Part C, Seminars in Medical Genetics, 142C(1), 40–51.
Moy, S. S., Nadler, J. J., Perez, A., Barbaro, R. P., Johns, J. M., Magnuson, T. R., et al.
(2004). Sociability and preference for social novelty in five inbred strains: An
approach to assess autistic-like behavior in mice. Genes, Brain, and Behavior, 3(5),
287–302.
Moy, S. S., Nadler, J. J., Poe, M. D., Nonneman, R. J., Young, N. B., Koller, B. H., et al.
(2008). Development of a mouse test for repetitive, restricted behaviors: Relevance to
autism. Behavioural Brain Research, 188(1), 178–194.
92 Susan E. Maloney et al.
Moy, S. S., Nadler, J. J., Young, N. B., Nonneman, R. J., Grossman, A. W., Murphy, D. L.,
et al. (2009). Social approach in genetically engineered mouse lines relevant to autism.
Genes, Brain, and Behavior, 8(2), 129–142.
Moy, S. S., Nadler, J. J., Young, N. B., Nonneman, R. J., Segall, S. K., Andrade, G. M., et al.
(2008). Social approach and repetitive behavior in eleven inbred mouse strains. Behav-
ioural Brain Research, 191(1), 118–129.
Muhle, R., Trentacoste, S. V., & Rapin, I. (2004). The genetics of autism. Pediatrics, 113(5),
e472–e486.
Mukamel, Z., Konopka, G., Wexler, E., Osborn, G. E., Dong, H., Bergman, M. Y., et al.
(2011). Regulation of MET by FOXP2, genes implicated in higher cognitive dysfunc-
tion and autism risk. Journal of Neuroscience, 31(32), 11437–11442.
Mullen, B. R., Khialeeva, E., Hoffman, D. B., Ghiani, C. A., & Carpenter, E. M. (2013).
Decreased reelin expression and organophosphate pesticide exposure alters mouse
behaviour and brain morphology. ASN Neuro, 5(1), e00106.
Nagy, A. (2000). Cre recombinase: The universal reagent for genome tailoring. Genesis,
26(2), 99–109.
Nakao, T., Nakagawa, A., Yoshiura, T., Nakatani, E., Nabeyama, M., Yoshizato, C., et al.
(2005). A functional MRI comparison of patients with obsessive-compulsive disorder
and normal controls during a Chinese character Stroop task. Psychiatry Research,
139(2), 101–114.
Neale, B. M., Kou, Y., Liu, L., Ma’ayan, A., Samocha, K. E., Sabo, A., et al. (2012). Patterns
and rates of exonic de novo mutations in autism spectrum disorders. Nature, 485(7397),
242–245.
Newbury, D. F., & Monaco, A. P. (2010). Genetic advances in the study of speech and lan-
guage disorders. Neuron, 68(2), 309–320.
O’Roak, B. J., Deriziotis, P., Lee, C., Vives, L., Schwartz, J. J., Girirajan, S., et al. (2011).
Exome sequencing in sporadic autism spectrum disorders identifies severe de novo muta-
tions. Nature Genetics, 43(6), 585–589.
O’Roak, B. J., Vives, L., Girirajan, S., Karakoc, E., Krumm, N., Coe, B. P., et al. (2012).
Sporadic autism exomes reveal a highly interconnected protein network of de novo
mutations. Nature, 485(7397), 246–250.
Oswald, D. P., & Sonenklar, N. A. (2007). Medication use among children with autism spec-
trum disorders. Journal of Child and Adolescent Psychopharmacology, 17(3), 348–355.
Page, L. A., Rubia, K., Deeley, Q., Daly, E., Toal, F., Mataix-Cols, D., et al. (2009).
A functional magnetic resonance imaging study of inhibitory control in obsessive-
compulsive disorder. Psychiatry Research, 174(3), 202–209.
Panksepp, J. B., Jochman, K. A., Kim, J. U., Koy, J. J., Wilson, E. D., Chen, Q., et al. (2007).
Affiliative behavior, ultrasonic communication and social reward are influenced by
genetic variation in adolescent mice. PLoS One, 2(4), e351.
Peca, J., Feliciano, C., Ting, J. T., Wang, W., Wells, M. F., Venkatraman, T. N., et al.
(2011). Shank3 mutant mice display autistic-like behaviours and striatal dysfunction.
Nature, 472(7344), 437–442.
Pellis, S. M., & Pasztor, T. J. (1999). The developmental onset of a rudimentary form of play
fighting in C57 mice. Developmental Psychobiology, 34, 175–182.
Penagarikano, O., Abrahams, B. S., Herman, E. I., Winden, K. D., Gdalyahu, A., Dong, H.,
et al. (2011). Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities,
and core autism-related deficits. Cell, 147(1), 235–246.
Persico, A. M., D’Agruma, L., Maiorano, N., Totaro, A., Militerni, R., Bravaccio, C., et al.
(2001). Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder.
Molecular Psychiatry, 6(2), 150–159.
Persico, A. M., & Napolioni, V. (2013). Autism genetics. Behavioural Brain Research, 251,
95–112.
Essential Cell Types and Circuits in ASD 93
Pesold, C., Impagnatiello, F., Pisu, M. G., Uzunov, D. P., Costa, E., et al. (1998). Reelin is
preferentially expressed in neurons synthesizing gamma-aminobutyric acid in cortex and
hippocampus of adult rats. Proc Natl Acad Sci U S A, 95, 3221–3226.
Peterson, T. C., Villatoro, L., Arneson, T., Ahuja, B., Voss, S., & Swain, R. A. (2012).
Behavior modification after inactivation of cerebellar dentate nuclei. Behavioral Neurosci-
ence, 126(4), 551–562.
Peyron, C., Faraco, J., Rogers, W., Ripley, B., Overeem, S., Charnay, Y., et al. (2000).
A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin
peptides in human narcoleptic brains. Nature Medicine, 6(9), 991–997.
Rauch, S. L., Savage, C. R., Alpert, N. M., Dougherty, D., Kendrick, A., Curran, T., et al.
(1997). Probing striatal function in obsessive-compulsive disorder: A PET study of implicit
sequence learning. Journal of Neuropsychiatry and Clinical Neurosciences, 9(4), 568–573.
Reith, R. M., McKenna, J., Wu, H., Hashmi, S. S., Cho, S. H., Dash, P. K., et al. (2013).
Loss of Tsc2 in Purkinje cells is associated with autistic-like behavior in a mouse model of
tuberous sclerosis complex. Neurobiology of Disease, 51, 93–103.
Ritvo, E. R., Freeman, B. J., Scheibel, A. B., Duong, T., Robinson, H., Guthrie, D., et al.
(1986). Lower Purkinje cell counts in the cerebella of four autistic subjects: Initial find-
ings of the UCLA-NSAC autopsy research report. The American Journal of Psychiatry,
143(7), 862–866.
Riva, D., Annunziata, S., Contarino, V., Erbetta, A., Aquino, D., & Bulgheroni, S. (2013).
Gray matter reduction in the Vermis and CRUS-II is associated with social and inter-
action deficits in low-functioning children with autistic spectrum disorders: A VBM-
DARTEL study. Cerebellum, 12(5), 676–685.
Rogers, T. D., Dickson, P. E., McKimm, E., Heck, D. H., Goldowitz, D., Blaha, C. D., et al.
(2013). Reorganization of circuits underlying cerebellar modulation of prefrontal cortical
dopamine in mouse models of autism spectrum disorder. Cerebellum, 12(4), 547–556.
Rogers, J. T., Zhao, L., Trotter, J. H., Rusiana, I., Peters, M. M., Li, Q., et al. (2013).
Reelin supplementation recovers sensorimotor gating, synaptic plasticity and associa-
tive learning deficits in the heterozygous reeler mouse. Journal of Psychopharmacology,
27(4), 386–395.
Rojas, D. C., Peterson, E., Winterrowd, E., Reite, M. L., Rogers, S. J., & Tregellas, J. R.
(2006). Regional gray matter volumetric changes in autism associated with social and
repetitive behavior symptoms. BMC Psychiatry, 6, 56.
Rubenstein, J. L., & Merzenich, M. M. (2003). Model of autism: Increased ratio of excita-
tion/inhibition in key neural systems. Genes, Brain, and Behavior, 2(5), 255–267.
Saab, C. Y., & Willis, W. D. (2003). The cerebellum: Organization, functions and its role in
nociception. Brain Research Brain Research Reviews, 42(1), 85–95.
Salinger, W. L., Ladrow, P., & Wheeler, C. (2003). Behavioral phenotype of the reeler
mutant mouse: Effects of RELN gene dosage and social isolation. Behavioral Neuroscience,
117(6), 1257–1275.
Samaco, R. C., Mandel-Brehm, C., Chao, H. T., Ward, C. S., Fyffe-Maricich, S. L., Ren, J.,
et al. (2009). Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in
neurotransmitter synthesis and specific behavioral abnormalities. Proceedings of the
National Academy of Sciences of the United States of America, 106(51), 21966–21971.
Sanders, S. J., Murtha, M. T., Gupta, A. R., Murdoch, J. D., Raubeson, M. J., Willsey, A. J.,
et al. (2012). De novo mutations revealed by whole-exome sequencing are strongly asso-
ciated with autism. Nature, 485(7397), 237–241.
Sarna, J. R., & Hawkes, R. (2003). Patterned Purkinje cell death in the cerebellum. Progress in
Neurobiology, 70(6), 473–507.
Scattoni, M. L., Martire, A., Cartocci, G., Ferrante, A., & Ricceri, L. (2013). Reduced social
interaction, behavioural flexibility and BDNF signalling in the BTBR T þ tf/J strain, a
mouse model of autism. Behavioural Brain Research, 251, 35–40.
94 Susan E. Maloney et al.
Scattoni, M. L., McFarlane, H. G., Zhodzishsky, V., Caldwell, H. K., Young, W. S.,
Ricceri, L., et al. (2008). Reduced ultrasonic vocalizations in vasopressin 1b knockout
mice. Behavioural Brain Research, 187(2), 371–378.
Scattoni, M. L., Ricceri, L., & Crawley, J. N. (2011). Unusual repertoire of vocalizations in
adult BTBR T þ tf/J mice during three types of social encounters. Genes, Brain, and
Behavior, 10(1), 44–56.
Schaevitz, L. R., Moriuchi, J. M., Nag, N., Mellot, T. J., & Berger-Sweeney, J. (2010). Cog-
nitive and social functions and growth factors in a mouse model of Rett syndrome. Phys-
iology & Behavior, 100(3), 255–263.
Schain, R. J., & Freedman, D. X. (1961). Studies on 5-hydroxyindole metabolism in autistic
and other mentally retarded children. The Journal of Pediatrics, 58, 315–320.
Schmahmann, J. D., & Sherman, J. C. (1998). The cerebellar cognitive affective syndrome.
Brain, 121(Pt. 4), 561–579.
Sears, L. L., Vest, C., Mohamed, S., Bailey, J., Ranson, B. J., & Piven, J. (1999). An MRI
study of the basal ganglia in autism. Progress in Neuro-Psychopharmacology and Biological Psy-
chiatry, 23(4), 613–624.
Sen, B., Singh, A. S., Sinha, S., Chatterjee, A., Ahmed, S., Ghosh, S., et al. (2010). Family-
based studies indicate association of Engrailed 2 gene with autism in an Indian popula-
tion. Genes, Brain, and Behavior, 9(2), 248–255.
Shahbazian, M., Young, J., Yuva-Paylor, L., Spencer, C., Antalffy, B., Noebels, J., et al.
(2002). Mice with truncated MeCP2 recapitulate many Rett syndrome features and dis-
play hyperacetylation of histone H3. Neuron, 35(2), 243–254.
Sheng, M., & Kim, E. (2000). The Shank family of scaffold proteins. Journal of Cell Science,
113(Pt. 11), 1851–1856.
Shu, W., Cho, J. Y., Jiang, Y., Zhang, M., Weisz, D., Elder, G. A., et al. (2005). Altered
ultrasonic vocalization in mice with a disruption in the Foxp2 gene. Proceedings of the
National Academy of Sciences of the United States of America, 102(27), 9643–9648.
Sillitoe, R. V., Stephen, D., Lao, Z., & Joyner, A. L. (2008). Engrailed homeobox genes
determine the organization of Purkinje cell sagittal stripe gene expression in the adult
cerebellum. Journal of Neuroscience, 28(47), 12150–12162.
Silverman, J. L., Yang, M., Lord, C., & Crawley, J. N. (2010). Behavioural phenotyping
assays for mouse models of autism. Nature Reviews Neuroscience, 11(7), 490–502.
Staal, W. G., de Krom, M., & de Jonge, M. V. (2012). Brief report: The dopamine-3-
receptor gene (DRD3) is associated with specific repetitive behavior in autism spectrum
disorder (ASD). Journal of Autism and Developmental Disorders, 42(5), 885–888.
Stocco, A., Lebiere, C., & Anderson, J. R. (2010). Conditional routing of information to the
cortex: A model of the basal ganglia’s role in cognitive coordination. Psychological Review,
117(2), 541–574.
Takahashi, K., Liu, F. C., Hirokawa, K., & Takahashi, H. (2003). Expression of Foxp2, a
gene involved in speech and language, in the developing and adult striatum. Journal of
Neuroscience Research, 73(1), 61–72.
Talkowski, M. E., Rosenfeld, J. A., Blumenthal, I., Pillalamarri, V., Chiang, C., Heilbut, A.,
et al. (2012). Sequencing chromosomal abnormalities reveals neurodevelopmental loci
that confer risk across diagnostic boundaries. Cell, 149(3), 525–537.
Tamura, S., Morikawa, Y., Iwanishi, H., Hisaoka, T., & Senba, E. (2004). Foxp1 gene
expression in projection neurons of the mouse striatum. Neuroscience, 124(2), 261–267.
Taniguchi, H., He, M., Wu, P., Kim, S., Paik, R., Sugino, K., et al. (2011). A resource of Cre
driver lines for genetic targeting of GABAergic neurons in cerebral cortex. Neuron, 71(6),
995–1013.
The Dutch-Belgian Fragile X Consortium (1994). Fmr1 knockout mice: A model to study
fragile X mental retardation. Cell, 78(1), 23–33.
Essential Cell Types and Circuits in ASD 95
Thomas, A., Burant, A., Bui, N., Graham, D., Yuva-Paylor, L. A., & Paylor, R. (2009). Mar-
ble burying reflects a repetitive and perseverative behavior more than novelty-induced
anxiety. Psychopharmacology, 204(2), 361–373.
Trouillas, P., Takayanagi, T., Hallett, M., Currier, R. D., Subramony, S. H., Wessel, K.,
et al. (1997). International Cooperative Ataxia Rating Scale for pharmacological assess-
ment of the cerebellar syndrome. The Ataxia Neuropharmacology Committee of the
World Federation of Neurology. Journal of the Neurological Sciences, 145(2), 205–211.
Tsai, P. T., Hull, C., Chu, Y., Greene-Colozzi, E., Sadowski, A. R., Leech, J. M., et al.
(2012). Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant
mice. Nature, 488(7413), 647–651.
Ubeda-Banon, I., Novejarque, A., Mohedano-Moriano, A., Pro-Sistiaga, P., de la Rosa-
Prieto, C., Insausti, R., et al. (2007). Projections from the posterolateral olfactory amyg-
dala to the ventral striatum: Neural basis for reinforcing properties of chemical stimuli.
BMC Neuroscience, 8, 103.
Ugarte, S. D., Homanics, G. E., Firestone, L. L., & Hammond, D. L. (2000). Sensory thresh-
olds and the antinociceptive effects of GABA receptor agonists in mice lacking the beta3
subunit of the GABA(A) receptor. Neuroscience, 95(3), 795–806.
van den Heuvel, O. A., Veltman, D. J., Groenewegen, H. J., Cath, D. C., van Balkom, A. J.,
van Hartskamp, J., et al. (2005). Frontal-striatal dysfunction during planning in obsessive-
compulsive disorder. Archives of General Psychiatry, 62(3), 301–309.
Veenstra-VanderWeele, J., Muller, C. L., Iwamoto, H., Sauer, J. E., Owens, W. A.,
Shah, C. R., et al. (2012). Autism gene variant causes hyperserotonemia, serotonin
receptor hypersensitivity, social impairment and repetitive behavior. Proceedings of the
National Academy of Sciences of the United States of America, 109(14), 5469–5474.
Vernes, S. C., Oliver, P. L., Spiteri, E., Lockstone, H. E., Puliyadi, R., Taylor, J. M., et al.
(2011). Foxp2 regulates gene networks implicated in neurite outgrowth in the develop-
ing brain. PLoS Genetics, 7(7), e1002145.
Verpelli, C., Dvoretskova, E., Vicidomini, C., Rossi, F., Chiappalone, M., Schoen, M., et al.
(2011). Importance of Shank3 protein in regulating metabotropic glutamate receptor 5
(mGluR5) expression and signaling at synapses. Journal of Biological Chemistry, 286(40),
34839–34850.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature,
474(7351), 380–384.
Wagstaff, J., Knoll, J. H., Fleming, J., Kirkness, E. F., Martin-Gallardo, A., Greenberg, F.,
et al. (1991). Localization of the gene encoding the GABAA receptor beta 3 subunit to
the Angelman/Prader-Willi region of human chromosome 15. American Journal of Human
Genetics, 49(2), 330–337.
Waider, J., Proft, F., Langlhofer, G., Asan, E., Lesch, K. P., & Gutknecht, L. (2013). GABA
concentration and GABAergic neuron populations in limbic areas are differentially
altered by brain serotonin deficiency in Tph2 knockout mice. Histochemistry and Cell Biol-
ogy, 139(2), 267–281.
Wang, X., McCoy, P. A., Rodriguiz, R. M., Pan, Y., Je, H. S., Roberts, A. C., et al. (2011).
Synaptic dysfunction and abnormal behaviors in mice lacking major isoforms of Shank3.
Human Molecular Genetics, 20(15), 3093–3108.
Wang, H., Yang, H., Shivalila, C. S., Dawlaty, M. M., Cheng, A. W., Zhang, F., et al.
(2013). One-step generation of mice carrying mutations in multiple genes by
CRISPR/Cas-mediated genome engineering. Cell, 153(4), 910–918.
Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner, J. T., Abrahams, B. S., et al. (2009).
Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature,
459(7246), 528–533.
96 Susan E. Maloney et al.
Wegiel, J., Kuchna, I., Nowicki, K., Imaki, H., Yong Ma, S., Azmitia, E. C., et al. (2013).
Contribution of olivofloccular circuitry developmental defects to atypical gaze in autism.
Brain Research, 1512, 106–122.
Weiss, L. A., Arking, D. E., Daly, M. J., & Chakravarti, A. (2009). A genome-wide linkage
and association scan reveals novel loci for autism. Nature, 461(7265), 802–808.
Welch, J. M., Wang, D., & Feng, G. (2004). Differential mRNA expression and protein
localization of the SAP90/PSD-95-associated proteins (SAPAPs) in the nervous system
of the mouse. Journal of Comparative Neurology, 472(1), 24–39.
Whiteside, S. P., Port, J. D., & Abramowitz, J. S. (2004). A meta-analysis of functional neu-
roimaging in obsessive-compulsive disorder. Psychiatry Research, 132(1), 69–79.
Willins, D. L., Deutch, A. Y., & Roth, B. L. (1997). Serotonin 5-HT2A receptors are
expressed on pyramidal cells and interneurons in the rat cortex. Synapse, 27(1), 79–82.
Wohr, M., & Scattoni, M. L. (2013). Behavioural methods used in rodent models of autism
spectrum disorders: Current standards and new developments. Behavioural Brain Research,
251, 5–17.
Yamamoto, T., Yoshida, K., Yoshikawa, H., Kishimoto, Y., & Oka, H. (1992). The medial
dorsal nucleus is one of the thalamic relays of the cerebellocerebral responses to the fron-
tal association cortex in the monkey: Horseradish peroxidase and fluorescent dye double
staining study. Brain Research, 579(2), 315–320.
Yang, M., Bozdagi, O., Scattoni, M. L., Wohr, M., Roullet, F. I., Katz, A. M., et al. (2012).
Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adoles-
cent Shank3 null mutant mice. Journal of Neuroscience, 32(19), 6525–6541.
Zhang, L., He, J., Jugloff, D. G., & Eubanks, J. H. (2008). The MeCP2-null mouse hippo-
campus displays altered basal inhibitory rhythms and is prone to hyperexcitability. Hip-
pocampus, 18(3), 294–309.
Zhang, S., Lin, L., Kaur, S., Thankachan, S., Blanco-Centurion, C., Yanagisawa, M., et al.
(2007). The development of hypocretin (orexin) deficiency in hypocretin/ataxin-3
transgenic rats. Neuroscience, 148(1), 34–43.
Zhou, F. C., Lesch, K. P., & Murphy, D. L. (2002). Serotonin uptake into dopamine neurons
via dopamine transporters: A compensatory alternative. Brain Research, 942(1–2),
109–119.
CHAPTER FOUR
Contents
1. Introduction 98
2. Neuroanatomy of Communication Deficits in ASD 99
3. Genes Linking Language to ASD 102
3.1 FOXP2 102
3.2 CNTNAP2 107
3.3 FOXP1 109
4. Modeling Communication Phenotypes Using Animal Models 116
4.1 Ultrasonic vocalizations in rodents 116
4.2 Songbird models 119
5. Evolutionary Comparisons 119
6. Conclusions and Future Directions 122
Acknowledgments 124
References 124
Abstract
Language is a human-specific trait that likely facilitated the rapid increase in higher cog-
nitive function in our species. A consequence of the selective pressures that have per-
mitted language and cognition to flourish in humans is the unique vulnerability of
humans to developing cognitive disorders such as autism. Therefore, progress in under-
standing the genetic and molecular mechanisms of language evolution should provide
insight into such disorders. Here, we discuss the few genes that have been identified in
both autism-related pathways and language. We also detail the use of animal models to
uncover the function of these genes at a mechanistic and circuit level. Finally, we pre-
sent the use of comparative genomics to identify novel genes and gene networks
involved in autism. Together, all of these approaches will allow for a broader and deeper
view of the molecular brain mechanisms involved in the evolution of language and the
gene disruptions associated with autism.
1. INTRODUCTION
Deficits in language or communication are key features of autism
spectrum disorder (ASD). Such deficits can span a wide range of phenotypes
from complete lack of verbal communication to inappropriate use of lan-
guage in social situations. The fact that language plays such an integral role
in ASD lends credence to the idea that the evolution of higher cognitive
functions, such as language, has led to humans having increased susceptibil-
ities to disorders of cognition such as ASD or schizophrenia (Crespi,
Summers, & Dorus, 2007; Crow, 1997). This also ties into the prevailing,
albeit controversial, notion that language is a human-specific trait as some
of the cognitive processes underlying language are not human-specific
(Hauser, Chomsky, & Fitch, 2002; Penn, Holyoak, & Povinelli, 2008;
Pinker & Jackendoff, 2005; Premack, 2007). Furthermore, most cognitive
disorders are also thought to be human-specific. Thus, understanding such
human-specific traits (and language is debatably the only human-specific
trait) can provide a unique perspective into both human brain evolution
and the pathophysiology of human diseases such as ASD.
One of the challenging aspects of studying language phenotypes as they
relate to ASD is that language deficits are prevalent across many neu-
rodevelopmental disorders such as schizophrenia and reactive attachment
disorder (Gillberg, 2010; King & Lord, 2011; Li, Branch, & DeLisi,
2009). In addition, children with early language impairments are often diag-
nosed with specific language impairment, or SLI, and longitudinal studies
have demonstrated that these children often develop other disorders such
as ASD (Gillberg, 2010). Consequently, phenotypes observed early in devel-
opment can be red flags that there are other broader pathological processes at
play that should be investigated and potentially treated through early behav-
ioral interventional processes. Due to this high comorbidity of phenotypic
features across neurodevelopmental disorders, it is very difficult to predict
how early language disturbances will correlate with later behavioral out-
comes and diagnoses. Therefore, in the absence of any other phenotypic
or genetic biomarkers, language dysfunction is not a strong predictor of
ASD. However, in the context of other ASD phenotypes such as social
impairments and repetitive or restricted behaviors, the presence of language
or communication deficits can strengthen and verify the diagnosis of ASD.
Another complicating factor in deciphering the genetic and molec-
ular mechanisms of language as they relate to ASD is that other
Language and Autism 99
While the cerebral cortex has been a primary focus of research related to
both ASD and language, there are several other key players: the cerebellum,
striatum, and thalamus. The cerebellum plays a role in ASD (Rogers et al.,
2013) and will be discussed in detail in Chapter 1. The cortex, thalamus, and
striatum all form a complex circuit that is likely important for many cogni-
tive functions (Fig. 4.1A). The striatum has been a brain region of intense
focus in the study of ASD and language (Di Martino et al., 2011;
Lieberman, 2002) since spoken language involves complex regulation of
motor output and frontal cortical–striatal circuitry is frequently disrupted
in numerous cognitive disorders (Shepherd, 2013). In addition, the thala-
mus, often referred to as the “gateway to the neocortex,” plays an important
role in parsing complex input sensory data and incoming data from the neo-
cortex that may be important in sensory feedback loops and dysfunctional in
ASD. For example, imaging studies have demonstrated ASD patients
exhibiting excessive cortical–striatal connectivity (Di Martino et al.,
A B
Visual Somatosensory
cortex cortex
Thalamus
direct ties between brain circuit abnormalities and specific behavioral abnor-
malities is still needed to more fully appreciate how mutations in specific
genes in ASD patients result in atypical behaviors. In summary, given the
robust circuitry linking the cortex, striatum, and thalamus, directed
approaches to finding pathways underlying language can be uncovered by
studying how these regions form connections and overlaying imaging data
of these connections with genetic studies of patients. Together, this overlap
will inform our understanding of how disorders affecting language, like
ASD, disrupt these connections.
3.1. FOXP2
The gene encoding FOXP2 ( forkhead box P2) was the first gene directly
implicated in a language disorder (Lai, Fisher, Hurst, Vargha-Khadem, &
Monaco, 2001). The affected individuals of a large, intergenerational family
(termed the KE family) had a dominantly inherited verbal dyspraxia with
additional cognitive impairments (Vargha-Khadem, Gadian, Copp, &
Mishkin, 2005; Vargha-Khadem, Watkins, Alcock, Fletcher, &
Passingham, 1995; Watkins, Dronkers, & Vargha-Khadem, 2002). FOXP2
is a member of the family of forkhead transcription factors expressed in areas
of the brain including the neocortex, striatum, thalamus, and cerebellum,
which are thought to be important for language and the coordination of
sequential motor output required for speech (Ferland, Cherry, Preware,
Morrisey, & Walsh, 2003; Teramitsu et al., 2004; Vargha-Khadem et al.,
2005). Moreover, the molecular evolution of FOXP2 on the human lineage
supports the idea that FOXP2 modification (at both the coding and noncod-
ing levels) may have been important for the emergence of language in
humans (Enard et al., 2002, 2009; Konopka et al., 2009; Maricic et al.,
2013; Zhang, Webb, & Podlaha, 2002). Thus, a deeper understanding of
Language and Autism 103
studies using both human cells and mouse tissue have identified additional
FoxP2 target genes that are associated with ASD (Enard et al., 2009;
Konopka et al., 2009; Mukamel et al., 2011; Vernes et al., 2007, 2008,
2011). In Table 4.1, we summarize the current knowledge of FOXP2 target
genes involved in ASD. However, such a table is a moving target. The list of
ASD genes is constantly evolving, and a recent estimate based on ASD
patient exome sequencing studies puts the projected number of genes likely
to have a genetic link to ASD at over 800 genes (O’Roak, Vives, Girirajan,
et al., 2012). For the analysis presented in Table 4.1, we used two curated
databases, the SFARI (Simons Foundation Autism Research Initiative)
Gene database (https://gene.sfari.org/autdb/; Basu, Kollu, & Banerjee-
Basu, 2009) containing over 500 genes and the AutismKB database from
Peking University, which has over 3000 ASD-associated genes including
a list of over 400 “core” ASD genes (Xu et al., 2011).
The second list that is not completely solidified is the list of direct
FOXP2 target genes. Although, we and others previously identified pro-
moters bound by FOXP2 in human fetal brain tissue and cells using
ChIP-chip (Spiteri et al., 2007; Vernes et al., 2007), these studies had tech-
nical limitations due to the fact that only the proximal promoter regions of
6000 genes were included on the promoter arrays. FOXP2 was one of the
transcription factors recently assessed using ChIP-seq (chromatin immuno-
precipitation followed by DNA sequencing) as part of the ENCODE
(Encyclopedia of DNA Elements) project to determine the function and
regulation of all “nongenic” regions of the genome (Dunham et al.,
2012). These FOXP2 ChIP-seq data have recently been analyzed for deter-
mining the optimal FOXP2 DNA binding motif (Nelson et al., 2013).
However, two recent studies have shown that the antibody used for the
ChIP portion of the experiment in the ENCODE dataset (Abcam antibody
ab16046) also recognizes FOXP1 (Campbell et al., 2010; Tsui, Vessey,
Tomita, Kaplan, & Miller, 2013). Therefore, the ENCODE FOXP2
ChIP-seq data likely contain numerous false-positives. Moreover, the
ENCODE experiments were conducted in transformed human cell lines
(PFSK-1 and SK-N-MC), which are not as optimal as using primary human
neuronal cells or tissue. In summary, we are still lacking a true genome-wide
assessment (using ChIP-seq) of FOXP2 binding in human neurons due to
technical limitations.
Even if ChIP-seq studies for FOXP2 could be reliably carried out in
human neurons, it is estimated that less than 50% of the identified genes will
actually be transcriptionally regulated by FOXP2 (Marson et al., 2007; Wei
Language and Autism 105
et al., 2006), due to spatial and temporal factors necessary for activation or
repression. Thus, it is important to confirm that genes physically bound by
FOXP2 exhibit a change in expression upon manipulation of FOXP2 levels.
To this end, we have identified gene expression changes downstream of
human FOXP2 in human cell lines and primary human neuronal progeni-
tors using whole genome microarrays (Konopka et al., 2009; Konopka,
Friedrich, et al., 2012). Gene expression microarrays using Foxp2 knockout
mice and expression microarray studies in “humanized” Foxp2 mice have
also been conducted (Enard et al., 2009; Vernes et al., 2011). Expression
microarrays have also been utilized to identify networks of gene
coexpression in the songbird striatum and uncovered one network in which
FoxP2 had high connectivity (Hilliard, Miller, Fraley, Horvath, & White,
Language and Autism 107
2012). Thus, once ChIP-seq for FOXP2 is successfully carried out in one or
more species, several published gene expression studies can be combined
across species to identify conserved and species-specific transcriptional tar-
gets of FoxP2.
A handful of direct FOXP2 target genes associated with ASD have been
identified and studied in detail. These include CNTNAP2 (Vernes et al.,
2008), DISC1 (disrupted in schizophrenia 1) (Walker et al., 2012), the receptor
tyrosine kinase MET (Mukamel et al., 2011), and the genes for SRPX2/
uPAR (sushi-repeat-containing protein, X-linked 2; urokinase plasminogen activator
receptor/urokinase receptor) (Roll et al., 2010). One of the interesting defining
features of even just these few genes is the strong interrelatedness of the genes
with phenotypes and the connection not only to disease but also to language.
As discussed in detail in the succeeding text, CNTNAP2 is associated with
language and epilepsy. DISC1 has been studied extensively within the con-
text of schizophrenia (Crespi et al., 2007), another developmental disorder
that is characterized by disruptions to language and communication (Li et al.,
2009; Morice & Igram, 1983; Rapoport, Addington, Frangou, & Psych,
2005; van Os & Kapur, 2009). Using data from the Allen Brain Atlas
(Hawrylycz et al., 2012), MET is the most enriched gene in the human tem-
poral lobe, an important cortical area involved in language. A number of
studies have suggested that MET may have genetic or protein interactions
with uPAR (also known as PLAUR) to mediate signaling through MET
or coordinate oppositional excitatory versus inhibitory signaling in the cor-
tex (Campbell, Li, Sutcliffe, Persico, & Levitt, 2008; Eagleson, Campbell,
Thompson, Bergman, & Levitt, 2011), an important component of normal
brain functioning that is often disrupted in ASD (LeBlanc & Fagiolini, 2011;
Penzes et al., 2013; Yizhar et al., 2011). (For a more detailed discussion of
the role of MET in ASD, see Chapter 5.) Together, these data suggest an
integral role for FOXP2-mediated signaling pathways in both language
and ASD.
3.2. CNTNAP2
The gene encoding CNTNAP2 was first identified as a candidate ASD gene
in several studies conducting fine mapping of a genetic locus significantly
associated with a particular endophenotype of autism, age at first word
(Alarcon, Cantor, Liu, Gilliam, & Geschwind, 2002; Alarcon et al., 2008;
Arking et al., 2008; Bakkaloglu et al., 2008). Additional studies have found
genetic variation or association in CNTNAP2 in individuals with ASD,
108 Stephanie Lepp et al.
3.3. FOXP1
FOXP1 is also a member of the forkhead family of transcription factors, is
highly homologous to FOXP2, and can physically interact with FOXP2
(heterodimerize) to regulate transcription (Li, Weidenfeld, & Morrisey,
2004; Shu, Yang, Zhang, Lu, & Morrisey, 2001). FOXP1 is also expressed
in areas of the brain associated with language such as the neocortex and stri-
atum; however, unlike FOXP2, it has high expression in the hippocampus
and low expression in the cerebellum (Ferland et al., 2003; Teramitsu et al.,
2004). The ability to heterodimerize and the somewhat overlapping expres-
sion patterns, especially in the striatum, support the idea that FOXP1 and
FOXP2 function in a coordinated manner to regulate signaling pathways
in the brain.
110 Stephanie Lepp et al.
ASD (Gibney & Drexhage, 2013; Michel, Schmidt, & Mirnics, 2012). One
of the largest gene expression studies to date using brain tissue from ASD and
control brains found an enrichment of immune-related genes that did not
appear to have a genetic basis (i.e., no enrichment in genome-wide associ-
ation studies) (Voineagu et al., 2011). Thus, the coexpression and regulation
of these immune-related genes may possibly be coordinated through a tran-
scriptional network regulated by a factor such as FOXP1. In fact, the
immune-related genes in this dataset are coexpressed with the RNA binding
and splicing factor A2BP1 (also known as RBFOX1) and there are data for
dysregulation of A2BP1-mediated splicing in the ASD brains (Voineagu
et al., 2011). In addition, previous work has shown that A2BP1 is a target
of FOXP2 in both humans (Spiteri et al., 2007) and mice (Vernes et al.,
2011). More interestingly, A2BP1 was identified as a target of Foxp1 in
Language and Autism 115
mouse striatum with forced expression (Tang et al., 2012). (For a more
detailed discussion of the role of A2BP1 in ASD, see Chapter 8.) Thus, it
is possible that while FOXP1 was not identified as differentially expressed
or coexpressed in the ASD brain dataset, it could be playing an upstream role
in the dysregulated transcriptional networks identified in ASD brain. Overall
though, there is a need for further study of both the transcriptional targets of
FOXP1 and its role in normal brain development. In Table 4.2, we include
our unpublished RNA-seq data from primary human neural progenitors
(Konopka, Wexler, et al., 2012) with forced expression of human FOXP1
with known ASD genes. What is interesting from Tables 4.1 and 4.2 is that
there are twice as many FOXP1 target genes that are associated with ASD
than FOXP2 target genes, again suggesting a more direct role for FOXP1 in
ASD pathophysiology. These and other future studies will begin to elucidate
the role of FOXP1 in ASD and how FOXP1 heterodimerization with
FOXP2 may be mediating signaling pathways important in language. What
has become quite clear though is that both FOXP1 and FOXP2 regulate
convergent and divergent signaling pathways that are likely important for
a myriad of neurodevelopmentally regulated processes that are frequently
disrupted in disorders like ASD (Fig. 4.2).
FOXP1 FOXP2
Figure 4.2 Shared molecular pathways downstream of FOXP2 and FOXP1. Individuals
with mutations in FOXP1 or FOXP2 develop unique and overlapping phenotypes affect-
ing aspects of language, motor control, and cognition. This flowchart shows FOXP1- and
FOXP2-regulating molecular pathways underlying these phenotypes through activation
of shared and distinct direct and indirect gene targets, of which many have been linked
to neurological disorders, such as ASD.
116 Stephanie Lepp et al.
Figure 4.3 Experimental set up for collecting USVs using Avisoft Bioacoustics recording
equipment and software. (A) A mouse pup is placed in a plastic container (a) inside a
polystyrene recording chamber (b). An Avisoft ultrasound microphone is mounted
on the lid of the recording chamber (c) and connected to the Avisoft-UltraSoundGate.
The UltraSoundGate is connected via USB to a PC with Avisoft-RECORDER USGH soft-
ware installed (e). (B) A photograph of the equipment setup.
Figure 4.4 Examples of mouse isolation calls: (A) Simple calls made by PN4 animals.
(B) More complex calls made by PN7 animals. (C) Calls are often preceded by clicks (den-
oted here with a yellow arrow). Clicks can also appear independently of calls (data not
shown).
mutants with Foxp2 knocked down in specific brain regions or at later time
points would be of interest assuming that they survive until adulthood
(French et al., 2007).
5. EVOLUTIONARY COMPARISONS
A different approach to identify the genes and molecular pathways
involved in ASD is to use evolutionary comparisons to identify human-
specific gene expression patterns. As the susceptibility to cognitive disorders,
such as ASD, may have coevolved with our increased capacity for social
communication, uncovering the molecular mechanisms driving human
brain evolution will likely provide insights into the pathophysiology of cog-
nitive disorders.
120 Stephanie Lepp et al.
being coupled to brain imaging studies so that we can begin to have better
connections between genotypes and phenotypes. These studies will likely
push the ongoing molecular and cellular studies into new frontiers.
Such genetic studies in humans are, however, limited to observational
and correlative reports due to obvious ethical issues with altering gene
expression in humans. While these correlative studies of genotype and func-
tion can obviously be extremely valuable, they are also limited by the avail-
ability of large cohorts of patients with known genetic alterations (although
this is rapidly changing as exome and whole genome sequencing costs fall
and analytic methods improve). Therefore, the study of specific genetic
alterations in vivo in animal models can provide groundbreaking insights into
normal brain development and function relevant to diseases such as ASD.
We have discussed using standard transgenic techniques or viral approaches
for candidate genes in the preceding text. However, new techniques have
revolutionized the field of transgenic animals. These techniques include
nucleases such as zinc finger nucleases or transcription activator-like effector
nucleases and the even newer clustered regularly interspaced short palin-
dromic repeats system (Cong et al., 2013; Gaj, Gersbach, & Barbas, 2013;
Li et al., 2011; Mali et al., 2013; Moehle et al., 2007). This burgeoning field
of alternative genome-editing techniques has opened up the possibility of
genetically manipulating model organisms that have been challenging to
study using standard transgenic technology (e.g., rats or birds) (Geurts
et al., 2009; Huang et al., 2011). The use of animal models beyond the stan-
dard laboratory mouse will be critical for the study of the molecular under-
pinnings of language. Having genetically modified vocal learners, such as
songbirds, will uncover conserved neural circuitry that is necessary for vocal-
izations. It will also be intriguing to see how ASD genes alter the social
aspects of these learned vocalizations and whether there are gender-relevant
differences to these social behaviors upon ASD gene manipulations.
The genetic studies have been leading the way in ASD research, and the
pace of these studies has made it challenging for the confirmatory, functional
studies to keep up (State & Levitt, 2011). In addition, the molecular study of
language has primarily focused on FOXP2 and its target genes, whereas
there are likely to be many more equally relevant genes to follow in such
detail. All of these avenues of research have been instrumental for furthering
our understanding of ASD and how language-relevant neurobiology is at
play during brain development and learning. The future of this field will
require delving deeper into the genetic studies and connecting them to func-
tional studies. This will encompass not only imaging studies in humans but
124 Stephanie Lepp et al.
also new animal models that display ASD and language-relevant behaviors
and phenotypes. Moreover, meta-analyses of all of these studies will be
required to fully flesh out the relevant brain circuitry in finer detail. Overall,
though, tremendous advances have already been made and the road to the
development of pharmacotherapeutics based on these studies is becoming
more tangible and achievable.
ACKNOWLEDGMENTS
G. K. is a Jon Heighten Scholar in Autism Research at UT Southwestern. This work was
supported by the NIMH (R00MH090238), a March of Dimes Basil O’Connor Starter
Scholar Research Award, and CREW Dallas to G. K. S. L. is supported by NIDA
(T32DA07290, Basic Science Training Program in Drug Abuse, Amelia J. Eisch, PhD PI).
REFERENCES
Abrahams, B. S., Tentler, D., Perederiy, J. V., Oldham, M. C., Coppola, G., &
Geschwind, D. H. (2007). Genome-wide analyses of human perisylvian cerebral cortical
patterning. Proceedings of the National Academy of Sciences of the United States of America,
104(45), 17849–17854.
Alarcon, M., Abrahams, B. S., Stone, J. L., Duvall, J. A., Perederiy, J. V., Bomar, J. M., et al.
(2008). Linkage, association, and gene-expression analyses identify CNTNAP2 as an
autism-susceptibility gene. American Journal of Human Genetics, 82(1), 150–159.
Alarcon, M., Cantor, R. M., Liu, J., Gilliam, T. C., & Geschwind, D. H. (2002). Evidence
for a language quantitative trait locus on chromosome 7q in multiplex autism families.
American Journal of Human Genetics, 70(1), 60–71.
Al-Murrani, A., Ashton, F., Aftimos, S., George, A. M., & Love, D. R. (2012). Amino-
terminal microdeletion within the CNTNAP2 gene associated with variable expressivity
of speech delay. Case Reports in Genetics, 2012, 172408.
Anderson, G. R., Galfin, T., Xu, W., Aoto, J., Malenka, R. C., & Sudhof, T. C. (2012).
Candidate autism gene screen identifies critical role for cell-adhesion molecule CASPR2
in dendritic arborization and spine development. Proceedings of the National Academy of
Sciences of the United States of America, 109(44), 18120–18125.
Arking, D. E., Cutler, D. J., Brune, C. W., Teslovich, T. M., West, K., Ikeda, M., et al.
(2008). A common genetic variant in the neurexin superfamily member CNTNAP2
increases familial risk of autism. American Journal of Human Genetics, 82(1), 160–164.
Arriaga, G., & Jarvis, E. D. (2013). Mouse vocal communication system: Are ultrasounds
learned or innate? Brain and Language, 124(1), 96–116.
Arriaga, G., Zhou, E. P., & Jarvis, E. D. (2012). Of mice, birds, and men: The mouse ultra-
sonic song system has some features similar to humans and song-learning birds. PLoS
One, 7(10), e46610.
Bacon, C., & Rappold, G. A. (2012). The distinct and overlapping phenotypic spectra of
FOXP1 and FOXP2 in cognitive disorders. Human Genetics, 131(11), 1687–1698.
Bakkaloglu, B., O’Roak, B. J., Louvi, A., Gupta, A. R., Abelson, J. F., Morgan, T. M., et al.
(2008). Molecular cytogenetic analysis and resequencing of contactin associated protein-
like 2 in autism spectrum disorders. American Journal of Human Genetics, 82(1), 165–173.
Language and Autism 125
Barttfeld, P., Wicker, B., Cukier, S., Navarta, S., Lew, S., & Sigman, M. (2011). A big-world
network in ASD: Dynamical connectivity analysis reflects a deficit in long-range connec-
tions and an excess of short-range connections. Neuropsychologia, 49(2), 254–263.
Basu, S. N., Kollu, R., & Banerjee-Basu, S. (2009). AutDB: A gene reference resource for
autism research. Nucleic Acids Research, 37(Database issue), D832–D836.
Benavides-Piccione, R., Ballesteros-Yanez, I., DeFelipe, J., & Yuste, R. (2002). Cortical
area and species differences in dendritic spine morphology. Journal of Neurocytology,
31(3–5), 337–346.
Berg, J. M., & Geschwind, D. H. (2012). Autism genetics: Searching for specificity and con-
vergence. Genome Biology, 13(7), 247.
Berkel, S., Marshall, C. R., Weiss, B., Howe, J., Roeth, R., Moog, U., et al. (2010). Muta-
tions in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental
retardation. Nature Genetics, 42(6), 489–491.
Bowers, J. M., & Konopka, G. (2012a). ASD-relevant animal models of the Foxp family of
transcription factors. Autism Open Access, S1(10). http://dx.doi.org/10.4172/2165-7890.
S1-010.
Bowers, J. M., & Konopka, G. (2012b). The role of the FOXP family of transcription factors
in ASD. Disease Markers, 33(5), 251–260.
Bowers, J. M., Perez-Pouchoulen, M., Edwards, N. S., & McCarthy, M. M. (2013). Foxp2
mediates sex differences in ultrasonic vocalization by rat pups and directs order of mater-
nal retrieval. Journal of Neuroscience, 33(8), 3276–3283.
Campbell, D. B., Li, C., Sutcliffe, J. S., Persico, A. M., & Levitt, P. (2008). Genetic evidence
implicating multiple genes in the MET receptor tyrosine kinase pathway in autism spec-
trum disorder. Autism Research, 1(3), 159–168.
Campbell, A. J., Lyne, L., Brown, P. J., Launchbury, R. J., Bignone, P., Chi, J., et al. (2010).
Aberrant expression of the neuronal transcription factor FOXP2 in neoplastic plasma
cells. British Journal of Haematology, 149(2), 221–230.
Carr, C. W., Moreno-De-Luca, D., Parker, C., Zimmerman, H. H., Ledbetter, N.,
Martin, C. L., et al. (2010). Chiari I malformation, delayed gross motor skills, severe
speech delay, and epileptiform discharges in a child with FOXP1 haploinsufficiency.
European Journal of Human Genetics, 18(11), 1216–1220.
Casey, J. P., Magalhaes, T., Conroy, J. M., Regan, R., Shah, N., Anney, R., et al. (2012).
A novel approach of homozygous haplotype sharing identifies candidate genes in autism
spectrum disorder. Human Genetics, 131(4), 565–579.
Chien, Y. L., Wu, Y. Y., Chiu, Y. N., Liu, S. K., Tsai, W. C., Lin, P. I., et al. (2011). Asso-
ciation study of the CNS patterning genes and autism in Han Chinese in Taiwan. Progress
in Neuro-Psychopharmacology and Biological Psychiatry, 35(6), 1512–1517.
Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto, R., Habib, N., et al. (2013). Multiplex
genome engineering using CRISPR/Cas systems. Science, 339(6121), 819–823.
Cook, E. H., Jr., & Scherer, S. W. (2008). Copy-number variations associated with neuro-
psychiatric conditions. Nature, 455(7215), 919–923.
Crespi, B. (2008). Genomic imprinting in the development and evolution of psychotic
spectrum conditions. Biological Reviews of the Cambridge Philosophical Society, 83(4),
441–493.
Crespi, B., & Badcock, C. (2008). Psychosis and autism as diametrical disorders of the social
brain. Behavioral and Brain Sciences, 31(3), 241–261, discussion 261–320.
Crespi, B., Summers, K., & Dorus, S. (2007). Adaptive evolution of genes underlying schizo-
phrenia. Proceedings of the Biological Sciences, 274(1627), 2801–2810.
Crosson, B. (2013). Thalamic mechanisms in language: A reconsideration based on recent
findings and concepts. Brain and Language, 126(1), 73–88.
Crow, T. J. (1997). Is schizophrenia the price that Homo sapiens pays for language?
Schizophrenia Research, 28(2–3), 127–141.
126 Stephanie Lepp et al.
Dasen, J. S., De Camilli, A., Wang, B., Tucker, P. W., & Jessell, T. M. (2008). Hox reper-
toires for motor neuron diversity and connectivity gated by a single accessory factor,
FoxP1. Cell, 134(2), 304–316.
Di Martino, A., Kelly, C., Grzadzinski, R., Zuo, X. N., Mennes, M., Mairena, M. A., et al.
(2011). Aberrant striatal functional connectivity in children with autism. Biological
Psychiatry, 69(9), 847–856.
Dumontheil, I., Burgess, P. W., & Blakemore, S. J. (2008). Development of rostral prefron-
tal cortex and cognitive and behavioural disorders. Developmental Medicine and Child
Neurology, 50(3), 168–181.
Dunham, I., Kundaje, A., Aldred, S. F., Collins, P. J., Davis, C. A., Doyle, F., et al. (2012).
An integrated encyclopedia of DNA elements in the human genome. Nature, 489(7414),
57–74.
Eagleson, K. L., Campbell, D. B., Thompson, B. L., Bergman, M. Y., & Levitt, P. (2011).
The autism risk genes MET and PLAUR differentially impact cortical development.
Autism Research, 4(1), 68–83.
Ehret, G. (2005). Infant rodent ultrasounds—A gate to the understanding of sound commu-
nication. Behavior Genetics, 35(1), 19–29.
Elston, G. N., Benavides-Piccione, R., & DeFelipe, J. (2001). The pyramidal cell in cogni-
tion: A comparative study in human and monkey. Journal of Neuroscience, 21(17), RC163.
Enard, W., Gehre, S., Hammerschmidt, K., Holter, S. M., Blass, T., Somel, M., et al. (2009).
A humanized version of Foxp2 affects cortico-basal ganglia circuits in mice. Cell, 137(5),
961–971.
Enard, W., Przeworski, M., Fisher, S. E., Lai, C. S., Wiebe, V., Kitano, T., et al. (2002).
Molecular evolution of FOXP2, a gene involved in speech and language. Nature,
418(6900), 869–872.
Feng, X., Ippolito, G. C., Tian, L., Wiehagen, K., Oh, S., Sambandam, A., et al. (2010).
Foxp1 is an essential transcriptional regulator for the generation of quiescent naive
T cells during thymocyte development. Blood, 115(3), 510–518.
Ferland, R. J., Cherry, T. J., Preware, P. O., Morrisey, E. E., & Walsh, C. A. (2003). Char-
acterization of Foxp2 and Foxp1 mRNA and protein in the developing and mature
brain. Journal of Comparative Neurology, 460(2), 266–279.
Feuk, L., Kalervo, A., Lipsanen-Nyman, M., Skaug, J., Nakabayashi, K., Finucane, B., et al.
(2006). Absence of a paternally inherited FOXP2 gene in developmental verbal
dyspraxia. American Journal of Human Genetics, 79(5), 965–972.
Ford, A. A., Triplett, W., Sudhyadhom, A., Gullett, J., McGregor, K., Fitzgerald, D. B., et al.
(2013). Broca’s area and its striatal and thalamic connections: A diffusion-MRI
tractography study. Frontiers in Neuroanatomy, 7, 8.
French, C. A., Groszer, M., Preece, C., Coupe, A. M., Rajewsky, K., & Fisher, S. E. (2007).
Generation of mice with a conditional Foxp2 null allele. Genesis, 45(7), 440–446.
Gaj, T., Gersbach, C. A., & Barbas, C. F., 3rd. (2013). ZFN, TALEN, and CRISPR/Cas-
based methods for genome engineering. Trends in Biotechnology, 31(7), 397–405.
Gaub, S., Groszer, M., Fisher, S. E., & Ehret, G. (2010). The structure of innate vocalizations
in Foxp2-deficient mouse pups. Genes, Brain and Behavior, 9(4), 390–401.
Gauthier, J., Joober, R., Mottron, L., Laurent, S., Fuchs, M., De Kimpe, V., et al. (2003).
Mutation screening of FOXP2 in individuals diagnosed with autistic disorder. American
Journal of Medical Genetics Part A, 118A(2), 172–175.
Geurts, A. M., Cost, G. J., Freyvert, Y., Zeitler, B., Miller, J. C., Choi, V. M., et al.
(2009). Knockout rats via embryo microinjection of zinc-finger nucleases. Science,
325(5939), 433.
Gibney, S. M., & Drexhage, H. A. (2013). Evidence for a dysregulated immune system
in the etiology of psychiatric disorders. Journal of NeuroImmune Pharmacology, 8(4),
900–920.
Language and Autism 127
King, B. H., & Lord, C. (2011). Is schizophrenia on the autism spectrum? Brain Research,
1380, 34–41.
Konopka, G., Bomar, J. M., Winden, K., Coppola, G., Jonsson, Z. O., Gao, F., et al. (2009).
Human-specific transcriptional regulation of CNS development genes by FOXP2.
Nature, 462(7270), 213–217.
Konopka, G., Friedrich, T., Davis-Turak, J., Winden, K., Oldham, M. C., Gao, F., et al.
(2012). Human-specific transcriptional networks in the brain. Neuron, 75(4), 601–617.
Konopka, G., Wexler, E., Rosen, E., Mukamel, Z., Osborn, G. E., Chen, L., et al. (2012).
Modeling the functional genomics of autism using human neurons. Molecular Psychiatry,
17(2), 202–214.
Kos, M., van den Brink, D., Snijders, T. M., Rijpkema, M., Franke, B., Fernandez, G., et al.
(2012). CNTNAP2 and language processing in healthy individuals as measured with
ERPs. PLoS One, 7(10), e46995.
Kotz, S. A., Schwartze, M., & Schmidt-Kassow, M. (2009). Non-motor basal ganglia func-
tions: A review and proposal for a model of sensory predictability in auditory language
perception. Cortex, 45(8), 982–990.
Lai, C. S., Fisher, S. E., Hurst, J. A., Vargha-Khadem, F., & Monaco, A. P. (2001).
A forkhead-domain gene is mutated in a severe speech and language disorder. Nature,
413(6855), 519–523.
Langfelder, P., & Horvath, S. (2008). WGCNA: An R package for weighted correlation net-
work analysis. BMC Bioinformatics, 9, 559.
Laroche, F., Ramoz, N., Leroy, S., Fortin, C., Rousselot-Paillet, B., Philippe, A., et al.
(2008). Polymorphisms of coding trinucleotide repeats of homeogenes in neu-
rodevelopmental psychiatric disorders. Psychiatric Genetics, 18(6), 295–301.
LeBlanc, J. J., & Fagiolini, M. (2011). Autism: A “critical period” disorder? Neural Plasticity,
2011, 921680.
Li, X., Branch, C. A., & DeLisi, L. E. (2009). Language pathway abnormalities in schizophre-
nia: A review of fMRI and other imaging studies. Current Opinion in Psychiatry, 22(2),
131–139.
Li, X., Hu, Z., He, Y., Xiong, Z., Long, Z., Peng, Y., et al. (2010). Association analysis of
CNTNAP2 polymorphisms with autism in the Chinese Han population. Psychiatric
Genetics, 20(3), 113–117.
Li, T., Huang, S., Zhao, X., Wright, D. A., Carpenter, S., Spalding, M. H., et al. (2011).
Modularly assembled designer TAL effector nucleases for targeted gene knockout and
gene replacement in eukaryotes. Nucleic Acids Research, 39(14), 6315–6325.
Li, S., Weidenfeld, J., & Morrisey, E. E. (2004). Transcriptional and DNA binding activity of
the Foxp1/2/4 family is modulated by heterotypic and homotypic protein interactions.
Molecular and Cellular Biology, 24(2), 809–822.
Li, H., Yamagata, T., Mori, M., & Momoi, M. Y. (2005). Absence of causative mutations and
presence of autism-related allele in FOXP2 in Japanese autistic patients. Brain Dev, 27(3),
207–210.
Lieberman, P. (2002). On the nature and evolution of the neural bases of human language.
The American Journal of Physical Anthropology, 35(Suppl.), 36–62.
Lin, P. I., Chien, Y. L., Wu, Y. Y., Chen, C. H., Gau, S. S., Huang, Y. S., et al. (2012). The
WNT2 gene polymorphism associated with speech delay inherent to autism. Research in
Developmental Disabilities, 33(5), 1533–1540.
Lipkind, D., Marcus, G. F., Bemis, D. K., Sasahara, K., Jacoby, N., Takahasi, M., et al.
(2013). Stepwise acquisition of vocal combinatorial capacity in songbirds and human
infants. Nature, 498(7452), 104–108.
Liu, X., Somel, M., Tang, L., Yan, Z., Jiang, X., Guo, S., et al. (2012). Extension of cortical
synaptic development distinguishes humans from chimpanzees and macaques. Genome
Research, 22(4), 611–622.
Language and Autism 129
Mali, P., Yang, L., Esvelt, K. M., Aach, J., Guell, M., DiCarlo, J. E., et al. (2013). RNA-
guided human genome engineering via Cas9. Science, 339(6121), 823–826.
Maricic, T., Gunther, V., Georgiev, O., Gehre, S., Curlin, M., Schreiweis, C., et al. (2013).
A recent evolutionary change affects a regulatory element in the human FOXP2 gene.
Molecular Biology and Evolution, 30(4), 844–852.
Marson, A., Kretschmer, K., Frampton, G. M., Jacobsen, E. S., Polansky, J. K.,
MacIsaac, K. D., et al. (2007). Foxp3 occupancy and regulation of key target genes dur-
ing T-cell stimulation. Nature, 445(7130), 931–935.
Marui, T., Koishi, S., Funatogawa, I., Yamamoto, K., Matsumoto, H., Hashimoto, O., et al.
(2005). No association of FOXP2 and PTPRZ1 on 7q31 with autism from the Japanese
population. Neuroscience Research, 53(1), 91–94.
Mefford, H. C., Muhle, H., Ostertag, P., von Spiczak, S., Buysse, K., Baker, C., et al. (2010).
Genome-wide copy number variation in epilepsy: Novel susceptibility loci in idiopathic
generalized and focal epilepsies. PLoS Genetics, 6(5), e1000962.
Michel, M., Schmidt, M. J., & Mirnics, K. (2012). Immune system gene dysregulation in
autism and schizophrenia. Developmental Neurobiology, 72(10), 1277–1287.
Moehle, E. A., Rock, J. M., Lee, Y. L., Jouvenot, Y., DeKelver, R. C., Gregory, P. D., et al.
(2007). Targeted gene addition into a specified location in the human genome using
designed zinc finger nucleases. Proceedings of the National Academy of Sciences of the United
States of America, 104(9), 3055–3060.
Moles, A., Costantini, F., Garbugino, L., Zanettini, C., & D’Amato, F. R. (2007). Ultrasonic
vocalizations emitted during dyadic interactions in female mice: A possible index of
sociability? Behavioural Brain Research, 182(2), 223–230.
Monk, C. S., Peltier, S. J., Wiggins, J. L., Weng, S. J., Carrasco, M., Risi, S., et al. (2009).
Abnormalities of intrinsic functional connectivity in autism spectrum disorders.
NeuroImage, 47(2), 764–772.
Moorman, S., Gobes, S. M., Kuijpers, M., Kerkhofs, A., Zandbergen, M. A., & Bolhuis, J. J.
(2012). Human-like brain hemispheric dominance in birdsong learning. Proceedings of the
National Academy of Sciences of the United States of America, 109(31), 12782–12787.
Morice, R. D., & Igram, J. C. (1983). Language complexity and age of onset of schizophre-
nia. Psychiatry Research, 9(3), 233–242.
Mukamel, Z., Konopka, G., Wexler, E., Osborn, G. E., Dong, H., Bergman, M. Y., et al.
(2011). Regulation of MET by FOXP2, genes implicated in higher cognitive dysfunc-
tion and autism risk. Journal of Neuroscience, 31(32), 11437–11442.
Nair, A., Treiber, J. M., Shukla, D. K., Shih, P., & Muller, R. A. (2013). Impaired
thalamocortical connectivity in autism spectrum disorder: A study of functional and ana-
tomical connectivity. Brain, 136(Pt 6), 1942–1955.
Nelson, C. S., Fuller, C. K., Fordyce, P. M., Greninger, A. L., Li, H., & Derisi, J. L. (2013).
Microfluidic affinity and ChIP-seq analyses converge on a conserved FOXP2-binding
motif in chimp and human, which enables the detection of evolutionarily novel targets.
Nucleic Acids Research, 41(12), 5991–6004.
Newbury, D. F., Bonora, E., Lamb, J. A., Fisher, S. E., Lai, C. S., Baird, G., et al. (2002).
FOXP2 is not a major susceptibility gene for autism or specific language impairment.
American Journal of Human Genetics, 70(5), 1318–1327.
Ojemann, G. A. (1991). Cortical organization of language. Journal of Neuroscience, 11(8),
2281–2287.
O’Roak, B. J., Deriziotis, P., Lee, C., Vives, L., Schwartz, J. J., Girirajan, S., et al. (2011).
Exome sequencing in sporadic autism spectrum disorders identifies severe de novo muta-
tions. Nature Genetics, 43(6), 585–589.
O’Roak, B. J., Vives, L., Fu, W., Egertson, J. D., Stanaway, I. B., Phelps, I. G., et al. (2012).
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum
disorders. Science, 338(6114), 1619–1622.
130 Stephanie Lepp et al.
O’Roak, B. J., Vives, L., Girirajan, S., Karakoc, E., Krumm, N., Coe, B. P., et al. (2012).
Sporadic autism exomes reveal a highly interconnected protein network of de novo
mutations. Nature, 485(7397), 246–250.
Panaitof, S. C., Abrahams, B. S., Dong, H., Geschwind, D. H., & White, S. A. (2010).
Language-related Cntnap2 gene is differentially expressed in sexually dimorphic song
nuclei essential for vocal learning in songbirds. Journal of Comparative Neurology,
518(11), 1995–2018.
Pariani, M. J., Spencer, A., Graham, J. M., Jr., & Rimoin, D. L. (2009). A 785kb deletion of
3p14.1p13, including the FOXP1 gene, associated with speech delay, contractures,
hypertonia and blepharophimosis. European Journal of Medical Genetics, 52(2–3), 123–127.
Penagarikano, O., Abrahams, B. S., Herman, E. I., Winden, K. D., Gdalyahu, A., Dong, H.,
et al. (2011). Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities,
and core autism-related deficits. Cell, 147(1), 235–246.
Penn, D. C., Holyoak, K. J., & Povinelli, D. J. (2008). Darwin’s mistake: Explaining the dis-
continuity between human and nonhuman minds. Behavioral and Brain Sciences, 31(2),
109–130, discussion 130–178.
Penzes, P., Buonanno, A., Passafaro, M., Sala, C., & Sweet, R. A. (2013). Developmental
vulnerability of synapses and circuits associated with neuropsychiatric disorders. Journal
of Neurochemistry, 126(2), 165–182.
Petkov, C. I., & Jarvis, E. D. (2012). Birds, primates, and spoken language origins: Behavioral
phenotypes and neurobiological substrates. Frontiers in Evolutionary Neuroscience, 4, 12.
Pinker, S., & Jackendoff, R. (2005). The faculty of language: What’s special about it?
Cognition, 95(2), 201–236.
Poliak, S., Gollan, L., Martinez, R., Custer, A., Einheber, S., Salzer, J. L., et al. (1999).
Caspr2, a new member of the neurexin superfamily, is localized at the juxtaparanodes
of myelinated axons and associates with K þ channels. Neuron, 24(4), 1037–1047.
Poliak, S., Salomon, D., Elhanany, H., Sabanay, H., Kiernan, B., Pevny, L., et al. (2003).
Juxtaparanodal clustering of Shaker-like K þ channels in myelinated axons depends
on Caspr2 and TAG-1. Journal of Cell Biology, 162(6), 1149–1160.
Pomerantz, S. M., Nunez, A. A., & Bean, N. J. (1983). Female behavior is affected by male
ultrasonic vocalizations in house mice. Physiology and Behavior, 31(1), 91–96.
Poot, M., Beyer, V., Schwaab, I., Damatova, N., Van’t Slot, R., Prothero, J., et al. (2010).
Disruption of CNTNAP2 and additional structural genome changes in a boy with speech
delay and autism spectrum disorder. Neurogenetics, 11(1), 81–89.
Premack, D. (2007). Human and animal cognition: Continuity and discontinuity. Proceedings
of the National Academy of Sciences of the United States of America, 104(35), 13861–13867.
Rapoport, J. L., Addington, A. M., Frangou, S., & Psych, M. R. (2005). The neu-
rodevelopmental model of schizophrenia: Update 2005. Molecular Psychiatry, 10(5),
434–449.
Richler, E., Reichert, J. G., Buxbaum, J. D., & McInnes, L. A. (2006). Autism and ultra-
conserved non-coding sequence on chromosome 7q. Psychiatric Genetics, 16(1), 19–23.
Rogers, T. D., McKimm, E., Dickson, P. E., Goldowitz, D., Blaha, C. D., & Mittleman, G.
(2013). Is autism a disease of the cerebellum? An integration of clinical and pre-clinical
research. Frontiers in Systems Neuroscience, 7, 15.
Roll, P., Vernes, S. C., Bruneau, N., Cillario, J., Ponsole-Lenfant, M., Massacrier, A., et al.
(2010). Molecular networks implicated in speech-related disorders: FOXP2 regulates the
SRPX2/uPAR complex. Human Molecular Genetics, 19(24), 4848–4860.
Rousso, D. L., Gaber, Z. B., Wellik, D., Morrisey, E. E., & Novitch, B. G. (2008). Coor-
dinated actions of the forkhead protein Foxp1 and Hox proteins in the columnar orga-
nization of spinal motor neurons. Neuron, 59(2), 226–240.
Language and Autism 131
Sakai, Y., Shaw, C. A., Dawson, B. C., Dugas, D. V., Al-Mohtaseb, Z., Hill, D. E., et al.
(2011). Protein interactome reveals converging molecular pathways among autism dis-
orders. Science Translational Medicine, 3(86), 86ra49.
Sanjuan, J., Tolosa, A., Gonzalez, J. C., Aguilar, E. J., Molto, M. D., Najera, C., et al. (2005).
FOXP2 polymorphisms in patients with schizophrenia. Schizophrenia Research, 73(2–3),
253–256.
Sanjuan, J., Tolosa, A., Gonzalez, J. C., Aguilar, E. J., Perez-Tur, J., Najera, C., et al. (2006).
Association between FOXP2 polymorphisms and schizophrenia with auditory halluci-
nations. Psychiatric Genetics, 16(2), 67–72.
Schmeisser, M. J., Ey, E., Wegener, S., Bockmann, J., Stempel, A. V., Kuebler, A., et al.
(2012). Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/Shank2.
Nature, 486(7402), 256–260.
Scott-Van Zeeland, A. A., Abrahams, B. S., Alvarez-Retuerto, A. I., Sonnenblick, L. I.,
Rudie, J. D., Ghahremani, D., et al. (2010). Altered functional connectivity in frontal
lobe circuits is associated with variation in the autism risk gene CNTNAP2. Science Trans-
lational Medicine, 2(56), 56ra80.
Shepherd, G. M. (2013). Corticostriatal connectivity and its role in disease. Nature Reviews
Neuroscience, 14(4), 278–291.
Shi, F., Wang, L., Peng, Z., Wee, C. Y., & Shen, D. (2013). Altered modular organization of
structural cortical networks in children with autism. PLoS One, 8(5), e63131.
Shu, W., Cho, J. Y., Jiang, Y., Zhang, M., Weisz, D., Elder, G. A., et al. (2005). Altered
ultrasonic vocalization in mice with a disruption in the Foxp2 gene. Proceedings of the
National Academy of Sciences of the United States of America, 102(27), 9643–9648.
Shu, W., Yang, H., Zhang, L., Lu, M. M., & Morrisey, E. E. (2001). Characterization of a
new subfamily of winged-helix/forkhead (Fox) genes that are expressed in the lung and
act as transcriptional repressors. Journal of Biological Chemistry, 276(29), 27488–27497.
Spiteri, E., Konopka, G., Coppola, G., Bomar, J., Oldham, M., Ou, J., et al. (2007). Iden-
tification of the transcriptional targets of FOXP2, a gene linked to speech and language,
in developing human brain. American Journal of Human Genetics, 81(6), 1144–1157.
Spocter, M. A., Hopkins, W. D., Garrison, A. R., Bauernfeind, A. L., Stimpson, C. D.,
Hof, P. R., et al. (2010). Wernicke’s area homologue in chimpanzees (Pan troglodytes)
and its relation to the appearance of modern human language. Proceedings of the Biological
Sciences, 277(1691), 2165–2174.
State, M. W., & Levitt, P. (2011). The conundrums of understanding genetic risks for autism
spectrum disorders. Nature Neuroscience, 14(12), 1499–1506.
Strauss, K. A., Puffenberger, E. G., Huentelman, M. J., Gottlieb, S., Dobrin, S. E.,
Parod, J. M., et al. (2006). Recessive symptomatic focal epilepsy and mutant
contactin-associated protein-like 2. The New England Journal of Medicine, 354(13),
1370–1377.
Sugranyes, G., Kyriakopoulos, M., Corrigall, R., Taylor, E., & Frangou, S. (2011). Autism
spectrum disorders and schizophrenia: Meta-analysis of the neural correlates of social
cognition. PLoS One, 6(10), e25322.
Surmeli, G., Akay, T., Ippolito, G. C., Tucker, P. W., & Jessell, T. M. (2011). Patterns of
spinal sensory-motor connectivity prescribed by a dorsoventral positional template. Cell,
147(3), 653–665.
Takahashi, K., Liu, F. C., Hirokawa, K., & Takahashi, H. (2003). Expression of Foxp2, a
gene involved in speech and language, in the developing and adult striatum. Journal of
Neuroscience Research, 73(1), 61–72.
Takahashi, K., Liu, F. C., Hirokawa, K., & Takahashi, H. (2008). Expression of Foxp4 in the
developing and adult rat forebrain. Journal of Neuroscience Research, 86(14), 3106–3116.
132 Stephanie Lepp et al.
Takahashi, K., Liu, F. C., Oishi, T., Mori, T., Higo, N., Hayashi, M., et al. (2008). Expres-
sion of FOXP2 in the developing monkey forebrain: Comparison with the expression of
the genes FOXP1, PBX3, and MEIS2. Journal of Comparative Neurology, 509(2), 180–189.
Tang, B., Becanovic, K., Desplats, P. A., Spencer, B., Hill, A. M., Connolly, C., et al. (2012).
Forkhead box protein p1 is a transcriptional repressor of immune signaling in the CNS:
Implications for transcriptional dysregulation in Huntington disease. Human Molecular
Genetics, 21(14), 3097–3111.
Teramitsu, I., Kudo, L. C., London, S. E., Geschwind, D. H., & White, S. A. (2004). Parallel
FoxP1 and FoxP2 expression in songbird and human brain predicts functional interac-
tion. Journal of Neuroscience, 24(13), 3152–3163.
Tolosa, A., Sanjuan, J., Dagnall, A. M., Molto, M. D., Herrero, N., & de Frutos, R. (2010).
FOXP2 gene and language impairment in schizophrenia: Association and epigenetic
studies. BMC Medical Genetics, 11, 114.
Toma, C., Hervas, A., Torrico, B., Balmana, N., Salgado, M., Maristany, M., et al. (2013).
Analysis of two language-related genes in autism: A case-control association study of
FOXP2 and CNTNAP2. Psychiatric Genetics, 23(2), 82–85.
Tsui, D., Vessey, J. P., Tomita, H., Kaplan, D. R., & Miller, F. D. (2013). FoxP2 regulates
neurogenesis during embryonic cortical development. Journal of Neuroscience, 33(1),
244–258.
van Os, J., & Kapur, S. (2009). Schizophrenia. Lancet, 374(9690), 635–645.
Vargha-Khadem, F., Gadian, D. G., Copp, A., & Mishkin, M. (2005). FOXP2 and the neu-
roanatomy of speech and language. Nature Reviews Neuroscience, 6(2), 131–138.
Vargha-Khadem, F., Watkins, K., Alcock, K., Fletcher, P., & Passingham, R. (1995). Praxic
and nonverbal cognitive deficits in a large family with a genetically transmitted speech
and language disorder. Proceedings of the National Academy of Sciences of the United States
of America, 92(3), 930–933.
Vernes, S. C., Newbury, D. F., Abrahams, B. S., Winchester, L., Nicod, J., Groszer, M., et al.
(2008). A functional genetic link between distinct developmental language disorders. The
New England Journal of Medicine, 359(22), 2337–2345.
Vernes, S. C., Oliver, P. L., Spiteri, E., Lockstone, H. E., Puliyadi, R., Taylor, J. M., et al.
(2011). Foxp2 regulates gene networks implicated in neurite outgrowth in the develop-
ing brain. PLoS Genetics, 7(7), e1002145.
Vernes, S. C., Spiteri, E., Nicod, J., Groszer, M., Taylor, J. M., Davies, K. E., et al. (2007).
High-throughput analysis of promoter occupancy reveals direct neural targets of
FOXP2, a gene mutated in speech and language disorders. American Journal of Human
Genetics, 81(6), 1232–1250.
Villalobos, M. E., Mizuno, A., Dahl, B. C., Kemmotsu, N., & Muller, R. A. (2005).
Reduced functional connectivity between V1 and inferior frontal cortex associated with
visuomotor performance in autism. NeuroImage, 25(3), 916–925.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature,
474(7351), 380–384.
Walker, R. M., Hill, A. E., Newman, A. C., Hamilton, G., Torrance, H. S.,
Anderson, S. M., et al. (2012). The DISC1 promoter: Characterization and regulation
by FOXP2. Human Molecular Genetics, 21(13), 2862–2872.
Walsh, C. A., & Engle, E. C. (2010). Allelic diversity in human developmental neu-
rogenetics: Insights into biology and disease. Neuron, 68(2), 245–253.
Wang, G. Z., & Konopka, G. (2013). Decoding human gene expression signatures in the
brain. Transcription, 4(3), 102–108.
Wang, B., Weidenfeld, J., Lu, M. M., Maika, S., Kuziel, W. A., Morrisey, E. E., et al. (2004).
Foxp1 regulates cardiac outflow tract, endocardial cushion morphogenesis and myocyte
proliferation and maturation. Development, 131(18), 4477–4487.
Language and Autism 133
Wassink, T. H., Piven, J., Vieland, V. J., Pietila, J., Goedken, R. J., Folstein, S. E., et al.
(2002). Evaluation of FOXP2 as an autism susceptibility gene. American Journal of Medical
Genetics, 114(5), 566–569.
Watkins, K. E., Dronkers, N. F., & Vargha-Khadem, F. (2002). Behavioural analysis of an
inherited speech and language disorder: Comparison with acquired aphasia. Brain,
125(Pt. 3), 452–464.
Wei, C. L., Wu, Q., Vega, V. B., Chiu, K. P., Ng, P., Zhang, T., et al. (2006). A global map
of p53 transcription-factor binding sites in the human genome. Cell, 124(1), 207–219.
Werling, D. M., & Geschwind, D. H. (2013). Sex differences in autism spectrum disorders.
Current Opinion in Neurology, 26(2), 146–153.
Xu, L. M., Li, J. R., Huang, Y., Zhao, M., Tang, X., & Wei, L. (2011). AutismKB: An
evidence-based knowledgebase of autism genetics. Nucleic Acids Research, 40(Database
issue), D1016–D1022.
Yizhar, O., Fenno, L. E., Prigge, M., Schneider, F., Davidson, T. J., O’Shea, D. J., et al.
(2011). Neocortical excitation/inhibition balance in information processing and social
dysfunction. Nature, 477(7363), 171–178.
Zhang, B., & Horvath, S. (2005). A general framework for weighted gene co-expression net-
work analysis. Statistical Applications in Genetics and Molecular Biology, 4, Article17, E-pub.
http://dx.doi.org/10.2202/1544-6115.1128.
Zhang, J., Webb, D. M., & Podlaha, O. (2002). Accelerated protein evolution and origins of
human-specific features: Foxp2 as an example. Genetics, 162(4), 1825–1835.
Zweier, C., de Jong, E. K., Zweier, M., Orrico, A., Ousager, L. B., Collins, A. L., et al.
(2009). CNTNAP2 and NRXN1 are mutated in autosomal-recessive Pitt-Hopkins-like
mental retardation and determine the level of a common synaptic protein in Drosophila.
American Journal of Human Genetics, 85(5), 655–666.
CHAPTER FIVE
Contents
1. Introduction 135
2. MET Receptor Tyrosine Kinase-Mediated Signaling has a Pleiotropic Role in
Multiple Organ Ontogenesis 138
3. MET Signaling Plays a Role in a Large Number of Neurodevelopment Events 142
4. MET Receptor Tyrosine Kinase Expression in the Developing Brain 145
5. The Human MET Gene Emerges as a Prominent Autism Risk Factor 149
6. Implication of MET Signaling in Neural Development and Functional Connectivity 152
7. Concluding Remarks 156
Acknowledgments 158
References 158
Abstract
In this chapter, we will briefly discuss recent literature on the role of MET receptor tyro-
sine kinase (RTK) in brain development and how perturbation of MET signaling may alter
normal neurodevelopmental outcomes. Recent human genetic studies have
established MET as a risk factor for autism, and the molecular and cellular underpinnings
of this genetic risk are only beginning to emerge from obscurity. Unlike many autism risk
genes that encode synaptic proteins, the spatial and temporal expression pattern of
MET RTK indicates this signaling system is ideally situated to regulate neuronal growth,
functional maturation, and establishment of functional brain circuits, particularly in
those brain structures involved in higher levels of cognition, social skills, and executive
functions.
1. INTRODUCTION
Autism spectrum disorders (ASD), which include autistic disorder,
Asperger’s syndrome, and pervasive developmental disorder (PDD)-not
Durand et al., 2007; Penagarikano et al., 2011; Piggot et al., 2009; Tabuchi
et al., 2007; Yashiro et al., 2009). These molecules function by mediating
pre- and postsynaptic assembly, scaffolding the synaptic structure, control-
ling neurotransmitter release, and affecting the activity-dependent structural
changes, processes critical to sculpting our experience into neuronal circuits
to guide future behavior. Not surprisingly, pathogenic mutations of the pre-
viously mentioned ASD genes during development have been shown to lead
to synaptic dysfunction, impact the brain circuit, and disrupt the balanced
excitatory/inhibitory brain networks (Ebert & Greenberg, 2013;
Rubenstein & Merzenich, 2003; Tabuchi et al., 2007).
It is important to note, however, that synaptogenesis and neural circuit
dynamics are relatively late events during the neurodevelopmental timeline.
Prior to these events, the production and positioning of neurons in a correct
cellular and network context must take place in order for synaptogenesis and
circuit remodeling to occur. These early histogenic events are determined
by genetic programs encoding neurogenesis, migration, neurite outgrowth
and polarization, and axon guidance at critical developmental stages. At the
cellular level, once a neuron is born, it migrates a long distance before arriv-
ing at its destination and differentiating. Neurons extend two classes of
processes: a single axon to carry its output and several dendrites to collect
information input. Once this neuronal polarity is established, the axon
navigates through a complex environment to find its target, and dendrites
undergo extensive growth and branching. The last step in forming functional
circuitry is the establishment of synaptic connections between different neu-
rons (Bradke & Dotti, 2000; Craig & Banker, 1994; Mueller, 1999; Tessier-
Lavigne & Goodman, 1996). Two major types of synapses, excitatory and
inhibitory, coexist within any functional circuitry, and their balanced action
on the postsynaptic neurons shapes their functional output (Rubenstein &
Merzenich, 2003). Therefore, aberrant genetic programs during this early
extended timeline (as compared to impaired synaptic function at later stages)
may profoundly affect brain function as well. Consistently, autism risk genes
have been shown to control wide aspects of developmental events including
neurogenesis, synaptogenesis, glutamatergic transmission, endosomal traf-
ficking, and protein turnover (Ebert & Greenberg, 2013; Qiu, Aldinger, &
Levitt, 2012; Walsh et al., 2008). As diverse as these risk genes appear, they
may converge on a final common molecular pathway to disrupt developmen-
tal outcomes that perturb circuit formation and maturation.
The development of the central nervous system (CNS) is a complex pro-
cess driven by a myriad of factors including a large family of growth factors
138 Yun Peng et al.
and their receptors. Protein receptor tyrosine kinases (RTKs), which are
cell-surface receptors for many polypeptide growth factors, hormones,
and cytokines (Robinson, Wu, & Lin, 2000), regulate many aspects of neu-
ronal physiology, including neurogenesis and survival, differentiation and
migration, patterned connectivity, and plasticity. The human gene MET,
which encodes MET RTK (Cooper et al., 1984), has emerged as a prom-
inent risk factor for ASD (Campbell et al., 2006, 2009; Jackson et al., 2009;
Sousa et al., 2009; Thanseem et al., 2010). MET plays a pleiotropic role in
cell proliferation, motogenesis, differentiation, and survival in many tissue
types (Birchmeier, Birchmeier, Gherardi, & Vande Woude, 2003; Maina
et al., 1998). The ligand for MET receptor, hepatocyte growth factor
(HGF), is a polypeptide growth factor that activates MET (Naldini,
Weidner, et al., 1991). Both MET and HGF are expressed in the developing
brain, with distinct spatial and temporal profiles (Judson, Amaral, & Levitt,
2011; Judson, Bergman, Campbell, Eagleson, & Levitt, 2009; Jung et al.,
1994). Genetic studies from multiple laboratories have found that functional
MET promoter variants are associated with differential risks for ASD. Con-
sistently, clinical imaging and animal studies have provided evidence that
disrupted MET signaling levels produce both morphological and functional
alterations in neurons in those brain regions implicated in producing the
ASD endophenotypes. In this chapter, we will briefly discuss how MET sig-
naling might be ideally situated to regulate circuits and modify neuronal
function. We review recent literature and hypothesize that MET signaling
plays a critical role in balancing neuronal growth, functional maturation, and
establishing functional circuits.
Cooper et al., 1984; Naldini, Vigna, et al., 1991). Soon after, MET/HGF-
mediated signaling was found to be involved in a number of
normal physiological processes. The signaling system appears important in
mesenchymal–epithelial interactions during fetal development: genetic
inactivation of Met or Hgf in mice leads to embryonic lethality, resulting
from impaired liver development, loss of parenchymal cells, and failed devel-
opment of placenta trophoblast cells and muscles (Bladt, Riethmacher,
Isenmann, Aguzzi, & Birchmeier, 1995; Huh et al., 2004; Schmidt et al.,
1995; Uehara et al., 1995). The context in which MET function is best
understood is in cancer biology. HGF signaling through MET is said to
be morphogenic, motogenic, and mitogenic. The function of this signaling
extends to early steps of cell proliferation, survival, branching morphogen-
esis, neuronal induction, organ regeneration, angiogenesis, and tumor
metastasis (Furge, Zhang, & Vande Woude, 2000; Maina et al., 1998).
This pleiotropic role suggests that the molecular basis for MET signaling
is of broad significance. Human MET protein is produced as a 170 kD
single-chain precursor (Cooper et al., 1984; Faletto et al., 1992). The pre-
cursor is proteolytically processed, resulting in a highly glycosylated extra-
cellular a-subunit (50 kD) and a transmembrane b-subunit (145 kD) (Furge
et al., 2000; Tempest, Stratton, & Cooper, 1988) (Fig. 5.1). The two sub-
units are linked together by a disulfide bond. The b-subunit has extracellu-
lar, transmembrane, and intracellular domains. The extracellular domain of
both a- and b-subunits contains homology to semaphorins (Sema domain);
the b chain has cysteine-rich MET-related sequences, glycine-proline-rich
repeats, and four immunoglobulin-like domains (Ig domain). The intracel-
lular b-subunits contain motifs of tyrosine kinase domain and a multi-
substrate docking site. The function of both domains is dependent on
several critical tyrosine residues. Upon HGF activation, MET dimerizes
and transphosphorylation occurs on Tyr1234 and Tyr1235 within the activa-
tion loop of the tyrosine kinase domain, and this activates the intrinsic kinase
activity of the receptor (Naldini, Weidner, et al., 1991). Close to the
C-terminal region, two tyrosine residues (1349 and 1356), residing in the
multisubstrate docking site, are capable of recruiting downstream Src
homology-2 (SH2) domain-containing adaptor proteins (Ponzetto et al.,
1994). Some adaptor proteins, such as Grb2, Src, SHC, and PI3K, interact
with the multisubstrate docking site directly, whereas many other effects are
mediated through the large scaffolding protein Gab1, which is sequentially
tyrosine-phosphorylated and recruits a number of downstream effector pro-
teins such as PI3K, SHP2, and PLC-g (Faletto et al., 1992; Gual et al., 2000).
140 Yun Peng et al.
Figure 5.1 Potential molecular signaling pathways mediated by MET receptor tyrosine
kinase in neurons. The activation of the MET signaling pathway is initiated by hepato-
cyte growth factor (HGF) binding, which induces MET dimerization and trans-
phosphorylation of two critical tyrosine residues (Tyr1234 and Tyr1235) in the tyrosine
kinase domain to activate the intrinsic kinase activity of MET. The ensuing phosphory-
lation of two additional tyrosine residuals (Tyr1349 and Tyr1356) in the multisubstrate
docking sites recruits downstream adaptor proteins including Grb2, Gab1, and SHC
to activate cascades of downstream pathways that involve major signal transducers
such as PLCg, AKT, MAPK/Erk1/2, STAT3, focal adhesion kinase (FAK), and Rho family
of small GTPases (Rho, Rac1, and Cdc42). Note that PI3 kinase can be directly activated
by binding either to the multisubstrate docking site or downstream to Gab1 activation.
Although most of these signaling events are established in nonneuronal cells, it is pos-
sible that these molecular pathways cooperate in developing neurons to mediate the
outcome of neuronal survival, morphogenesis and proliferation, projection and motility,
and activity-dependent gene transcription. Indeed, there has been some experimental
evidence that MET signaling in neurons activates PI3K–AKT pathway and MAP kinase
pathway (indicated by shaded boxes). MET has been shown to directly interact with
other membrane-bound proteins in neurons, such as AXL, CD44, and plexins (which
bind to semaphorins). The extent to which these membrane protein interactions and
the intracellular signaling pathways in mediating the functional developmental out-
comes in neurons has yet to be ascertained.
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 141
Functional tests from the same study identified HGF and MET as mediators
of the conditioned medium-stimulated recovery. This protective effect is
due to HGF and MET promotion of neural cell development and
remyelination.
It was found that the expression of MET protein levels was tightly regulated
across the time domain in the forebrain. MET protein levels are relatively
low around the late embryonic stage in mouse (E16.5) but increase dramat-
ically during perinatal development (postnatal day 0, P0) to reach a peak at
P7. Thereafter, MET levels are relatively stable during the second postnatal
week but decline drastically after P21 to very low levels at adult stage. The
study has revealed that peak levels of MET expression coincide with prin-
cipal periods of neurite outgrowth and synaptogenesis. High-resolution
immunohistochemistry staining from the same study reveals that MET is
expressed by discrete subtypes of long-projecting neurons of the forebrain,
especially those of dorsal pallial origin. Interestingly, MET protein is found
to be enriched in the developing axons of these projection neurons. In P7–
P14 mouse, MET immunoreactivity is strongly distributed to axons and
neuropil throughout the anteroposterior axis in the cortex. The corpus cal-
losum has the highest level of MET expression. There is also a clear laminar
patterning of Met transcript and protein expression in the neocortex in
which the barrel cortex and layer IV of the cortex distinctly lack MET
expression. Because layer IV is the synaptic input layer from subcortical
structures, this is consistent with the observation that the majority of subcor-
tical region shows minimum MET immunoreactivity in mouse. Another
interesting finding from this study is the apparent discrepant patterns of
expression for Met transcripts and proteins in the striatum. For instance,
Western blot analysis showed that striatum tissue contains abundant MET
proteins, whereas in situ hybridization failed to reveal Met transcripts in
the striatum. Therefore, the presence of MET protein in the striatum is
exclusively attributed to cortical projecting axons. Since the medium spiny
neurons in the striatum do not express MET, changes to corticostriatal cir-
cuits following ablation of MET in dorsal pallium structures can be therefore
attributed to a presynaptic mechanism.
The brainstem circuitry is implicated in ASD pathophysiology and auto-
nomic function control, and a recent study (Wu & Levitt, 2013) has exam-
ined Met and Hgf mRNA expression in the developing rodent brainstem
using in situ hybridization and immunohistochemistry to probe protein
levels. This study revealed a highly selective expression pattern of MET
in the brainstem in a subpopulation of neurons in cranial motor nuclei,
the dorsal raphe, Barrington’s nucleus, and the nucleus of solitary tract.
All of these brainstem structures show strong Met transcripts and immuno-
reactivity, which indicates that MET signaling may influence the develop-
ment of brainstem circuits that control autonomic function, such as central
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 147
Walikonis (2006) had found that MET protein is localized at the postsynap-
tic dendritic site, suggesting MET could be part of the postsynaptic signaling
complex. In a recent study by Kawas, Benoist, Harding, Wayman, and Abu-
Lail (2013), it was found that MET protein levels are especially enriched in
brain regions that undergo extensive synaptic remodeling and plasticity,
such as the hippocampus CA1 region. Additionally, MET activation
increases the dendritic spine density and number of synapses. The authors
then used atomic force microscopy combined with a specific MET antibody
to address the question of subcellular localization of MET and found that the
activated multimeric form of MET is concentrated in the dendritic spine
compartment. In comparison, the inactivated monomeric form of MET
is prominent on the soma of neurons. This ultrastructural study provides
the first direct evidence of functional activation of MET in neurons.
A comprehensive morphology study by Eagleson, Milner, Xie, and Levitt
(2013) attempted to resolve the perisynaptic location of MET, that is,
whether MET is expressed in the presynaptic, postsynaptic, or glial compart-
ments. Combining immunoelectron microscopy and in situ proximity liga-
tion assay (PLA), the authors found that MET localization is rather dynamic,
depending on the postnatal age of mouse examined. In the striatum radiatum
layer of CA1 region of P7 mouse (peak stage of rapid neuronal dendritic
growth and morphogenesis), MET expression is equally located at both
pre- and postsynaptic compartments. At a later stage when extensive syn-
aptogenesis occurs, MET expression is predominantly presynaptic, with a
small proportion of immunoreactivity arising from glial cells at this time.
These morphological observations are consistent with their PLA analysis
in cultured neurons and Western blot analysis of MET levels in the sub-
synaptic compartments in brain tissues. This study provides conclusive evi-
dence that MET is enriched at synapses during development, and its
expression is dynamically regulated. The study also provides structural evi-
dence that signaling of MET can potentially recruit both pre- and postsyn-
aptic mechanisms.
A current important unanswered question is how MET receptor tyrosine
kinase is regulated to allow its spatiotemporal specificity in the developing
brain. Initial study by Campbell et al. (2006) has shown that the transcription
factors SP1 and PC4 (encoded by SUB1) bind to the 50 -transcriptional reg-
ulatory region of the MET gene, but the functional significance of this bind-
ing is not clear. A recent study (Mukamel et al., 2011) has identified
Forkhead box protein P2 (FOXP2) as a novel transcriptional repressor of
the MET gene. FOXP2 has been established as a regulatory repressor protein
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 149
the development of the brain (Beilmann et al., 2000; Gutierrez et al., 2004;
Ido et al., 2005; Ieraci et al., 2002; Maina et al., 1997; Okunishi et al., 2005;
Powell et al., 2001). Additionally, MET signaling plays a role in immune
function and gastrointestinal repair (Arthur et al., 2004; Tahara et al.,
2003), and both are impaired modalities seen in some ASD cases. Lastly,
there have been converging developmental biological studies indicating
hypomorphic MET signaling in the cortex results in abnormal interneuron
migration and decreased proliferation of granule cells in the cerebellum
(Eagleson et al., 2005; Ieraci et al., 2002; Powell, Campbell, et al., 2003).
The study by Campbell et al. (2006) analyzed the MET gene in a family-
based study of ASD including >1200 cases. The study revealed strong
genetic association of a common C allele (rs1858830 “C”) in the 50 -
transcriptional regulatory region of the MET gene in >200 autism families.
Additionally, in multiplex families with more than one autistic child, the
rs1858830 “C” allelic association is even stronger. Overall, the relative risk
for autism diagnosis was 2.27 (95% CI 1.41–3.65) for the CC genotype and
1.67 (95% CI 1.11–2.49) for the CG type compared with the GG type. The
autism risk susceptibility is correlated with MET promoter activity and the
promoter sequence’s binding for specific transcription factors SP1 and PC4
(encoded by SUB1) in a functional assay. A subsequent study following this
initial report by the same group examined MET expression levels in the
postmortem tissue from the temporal lobe of autism and control cases.
The study found decreased MET transcript and protein expression in indi-
viduals with ASD compared to matched controls (Campbell et al., 2007),
further supporting the notion that reduction or hypomorphic MET signaling
is a risk factor for autism.
Additional genetic and pathophysiological evidence that dysfunctional
MET signaling contributes to ASD risk has been complimentary to the orig-
inal findings. Campbell, Li, Sutcliffe, Persico, and Levitt (2008) tested
whether genes in the MET pathway (multiple genes encoding proteins that
regulate MET expression and activity), such as HGF, and PLAUR tran-
scripts are significantly altered in the ASD brain. The PLAUR gene encodes
the urokinase plasminogen activator receptor, which is required for the uro-
kinase plasminogen activator to process the HGF precursor into an active
form. In addition, the SERPINE1 gene, which encodes plasminogen acti-
vator inhibitor-1, was also examined. Both PLAUR and SERPINE1
exhibited significant association with autism (Campbell et al., 2008).
PLAUR promoter variant rs344781 T allele was associated with ASD by
both family-based association test and case–control analyses. There is also
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 151
from MET loss of function. Although this study did not reveal whether pre-
or postsynaptic mechanisms contribute to increased synaptic drive, the
enhanced synaptic connectivity seen in a major local synaptic circuit may
be reminiscent of hyperconnectivity of local brain regions seen in ASD
patients. It would be interesting to examine long-range circuit functionality
in future studies. Due to the limitations that LSPS mapping can only be done
in brain slices where intact long-range circuits cannot be preserved, adapta-
tion of new optogenetic tools into this technique provides a feasible
approach to map long-range circuits (Petreanu, Huber, Sobczyk, &
Svoboda, 2007). Lastly, although neuroanatomical and functional mapping
gained valuable insight into the static circuit property, neural circuits are
very dynamic in that they exhibit a remarkable ability to scale their activity
in response to changes of activity or experimental perturbations, a process
known as homeostatic plasticity (Pozo & Goda, 2010; Turrigiano &
Nelson, 2000). It would be interesting for future work to look at the prin-
cipal substrates of synaptic homeostasis, that is, the compensatory adaptations
in synaptic strength or intrinsic excitability, and how these components
respond to disrupted MET signaling.
7. CONCLUDING REMARKS
Translating the genetic contributions to neurodevelopmental disor-
ders, such as ASD, into pathophysiological mechanisms will bridge the cur-
rent knowledge gap and facilitate developing novel interventions and
treatments. Many of the most compelling candidate genes identified for
rare/syndromic and idiopathic forms of ASD so far are involved in brain
wiring and synaptic function by being an integral part of synaptic molecular
machinery, by regulating gene transcriptions, or by contributing to the
excitatory/inhibitory balances (Bourgeron, 2009). MET signaling may
be a unique mechanism capable of regulating a multitude of neuron
behavior, including differentiation, growth, neurite extension, and synapse
maturation, all of which are prerequisite steps in establishing brain circuits.
Therefore, efforts in deciphering the functional significance of MET at
molecular, cellular, and system levels are central to understanding how
MET contributes to ASD pathophysiology. A recent genetic study (Pinto
et al., 2010) focusing on the functional impact of global rare copy number
variations in ASD has reported “an enrichment of CNVs disrupting func-
tional gene sets involved in cellular proliferation, projection and motility,
and GTPase/Ras signaling.” Existing experimental evidence suggests that
MET signaling plays a role in each of these functional domains. MET
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 157
Figure 5.2 MET receptor tyrosine kinase as a synaptic player that balances neuronal
growth, synaptic plasticity, and functional maturation. The expression of MET protein
in the developing brain is tightly regulated in both spatial and temporal domains.
MET expression is turned on during the perinatal period in mouse and peaks during
the period of extensive neurite growth and synaptogenesis. This suggests that MET-
mediated signaling plays a role in these early processes of brain development (green
arrows). MET protein is dramatically reduced as the brain circuits undergo functional
maturation and synaptic plasticity (red ticks). Disturbances of MET signaling, such as car-
rying a hypofunctional MET allele, could have detrimental effects in the protracted neu-
ronal developmental timeline and contribute to impaired circuit function in the adult
brain.
158 Yun Peng et al.
ACKNOWLEDGMENTS
The authors thank Dr. Aaron McGee, Zhongming Lu, and Mariel Piechowicz for
proofreading and their critiques of this chapter.
REFERENCES
Abrahams, B. S., & Geschwind, D. H. (2008). Advances in autism genetics: On the threshold
of a new neurobiology. Nature Reviews Genetics, 9(5), 341–355.
Akimoto, M., Baba, A., Ikeda-Matsuo, Y., Yamada, M. K., Itamura, R., Nishiyama, N.,
et al. (2004). Hepatocyte growth factor as an enhancer of nmda currents and synaptic
plasticity in the hippocampus. Neuroscience, 128(1), 155–162.
Alarcon, M., Abrahams, B. S., Stone, J. L., Duvall, J. A., Perederiy, J. V., Bomar, J. M., et al.
(2008). Linkage, association, and gene-expression analyses identify CNTNAP2 as an
autism-susceptibility gene. American Journal of Human Genetics, 82(1), 150–159.
Aldinger, K. A., Plummer, J. T., Qiu, S., & Levitt, P. (2011). SnapShot: Genetics of autism.
Neuron, 72(2), 418, e411.
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 159
Anderson, J. S., Druzgal, T. J., Froehlich, A., DuBray, M. B., Lange, N., Alexander, A. L.,
et al. (2011). Decreased interhemispheric functional connectivity in autism. Cerebral Cor-
tex, 21(5), 1134–1146.
Arthur, L. G., Schwartz, M. Z., Kuenzler, K. A., & Birbe, R. (2004). Hepatocyte growth
factor treatment ameliorates diarrhea and bowel inflammation in a rat model of inflam-
matory bowel disease. Journal of Pediatric Surgery, 39(2), 139–143, discussion 139–143.
Bai, L., Lennon, D. P., Caplan, A. I., DeChant, A., Hecker, J., Kranso, J., et al. (2012). Hepa-
tocyte growth factor mediates mesenchymal stem cell-induced recovery in multiple scle-
rosis models. Nature Neuroscience, 15(6), 862–870.
Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E., et al. (1995).
Autism as a strongly genetic disorder: Evidence from a British twin study. Psychological
Medicine, 25(1), 63–77.
Beilmann, M., Vande Woude, G. F., Dienes, H. P., & Schirmacher, P. (2000). Hepatocyte
growth factor-stimulated invasiveness of monocytes. Blood, 95(12), 3964–3969.
Birchmeier, C., Birchmeier, W., Gherardi, E., & Vande Woude, G. F. (2003). Met, metas-
tasis, motility and more. Nature Reviews Molecular Cell Biology, 4(12), 915–925.
Bladt, F., Riethmacher, D., Isenmann, S., Aguzzi, A., & Birchmeier, C. (1995). Essential role
for the c-met receptor in the migration of myogenic precursor cells into the limb bud.
Nature, 376(6543), 768–771.
Boccaccio, C., Ando, M., Tamagnone, L., Bardelli, A., Michieli, P., Battistini, C., et al.
(1998). Induction of epithelial tubules by growth factor HGF depends on the STAT
pathway. Nature, 391(6664), 285–288.
Bottaro, D. P., Rubin, J. S., Faletto, D. L., Chan, A. M., Kmiecik, T. E., Vande
Woude, G. F., et al. (1991). Identification of the hepatocyte growth factor receptor
as the c-met proto-oncogene product. Science, 251(4995), 802–804.
Bourgeron, T. (2009). A synaptic trek to autism. Current Opinion in Neurobiology, 19(2), 231–234.
Bradke, F., & Dotti, C. G. (2000). Establishment of neuronal polarity: Lessons from cultured
hippocampal neurons. Current Opinion in Neurobiology, 10(5), 574–581.
Bronner-Fraser, M. (1995). Hepatocyte growth factor/scatter factor (HGF/SF) in early devel-
opment: Evidence for a role in neural induction. Trends in Genetics, 11(11), 423–425.
Campbell, D. B., Buie, T. M., Winter, H., Bauman, M., Sutcliffe, J. S., Perrin, J. M., et al.
(2009). Distinct genetic risk based on association of MET in families with co-occurring
autism and gastrointestinal conditions. Pediatrics, 123(3), 1018–1024.
Campbell, D. B., D’Oronzio, R., Garbett, K., Ebert, P. J., Mirnics, K., Levitt, P., et al.
(2007). Disruption of cerebral cortex MET signaling in autism spectrum disorder. Annals
of Neurology, 62(3), 243–250.
Campbell, D. B., Li, C., Sutcliffe, J. S., Persico, A. M., & Levitt, P. (2008). Genetic evidence
implicating multiple genes in the MET receptor tyrosine kinase pathway in autism spec-
trum disorder. Autism Research, 1(3), 159–168.
Campbell, D. B., Sutcliffe, J. S., Ebert, P. J., Militerni, R., Bravaccio, C., Trillo, S., et al.
(2006). A genetic variant that disrupts MET transcription is associated with autism. Pro-
ceedings of the National Academy of Sciences of the United States of America, 103(45),
16834–16839.
Caton, A., Hacker, A., Naeem, A., Livet, J., Maina, F., Bladt, F., et al. (2000). The branchial
arches and HGF are growth-promoting and chemoattractant for cranial motor axons.
Development, 127(8), 1751–1766.
Clement, J. P., Aceti, M., Creson, T. K., Ozkan, E. D., Shi, Y., Reish, N. J., et al. (2012).
Pathogenic SYNGAP1 mutations impair cognitive development by disrupting matura-
tion of dendritic spine synapses. Cell, 151(4), 709–723.
Constantino, J. N., Todorov, A., Hilton, C., Law, P., Zhang, Y., Molloy, E., et al. (2013).
Autism recurrence in half siblings: Strong support for genetic mechanisms of transmission
in ASD. Molecular Psychiatry, 18(2), 137–138.
160 Yun Peng et al.
Cooper, C. S., Park, M., Blair, D. G., Tainsky, M. A., Huebner, K., Croce, C. M., et al.
(1984). Molecular cloning of a new transforming gene from a chemically transformed
human cell line. Nature, 311(5981), 29–33.
Courchesne, E., & Pierce, K. (2005). Why the frontal cortex in autism might be talking only
to itself: Local over-connectivity but long-distance disconnection. Current Opinion in
Neurobiology, 15(2), 225–230.
Craig, A. M., & Banker, G. (1994). Neuronal polarity. Annual Review of Neuroscience, 17,
267–310.
Davey, F., Hilton, M., & Davies, A. M. (2000). Cooperation between HGF and CNTF in
promoting the survival and growth of sensory and parasympathetic neurons. Molecular
and Cellular Neurosciences, 15(1), 79–87.
Di Renzo, M. F., Bertolotto, A., Olivero, M., Putzolu, P., Crepaldi, T., Schiffer, D., et al.
(1993). Selective expression of the Met/HGF receptor in human central nervous system
microglia. Oncogene, 8(1), 219–222.
Durand, C. M., Betancur, C., Boeckers, T. M., Bockmann, J., Chaste, P., Fauchereau, F.,
et al. (2007). Mutations in the gene encoding the synaptic scaffolding protein SHANK3
are associated with autism spectrum disorders. Nature Genetics, 39(1), 25–27.
Eagleson, K. L., Bonnin, A., & Levitt, P. (2005). Region- and age-specific deficits in gamma-
aminobutyric acidergic neuron development in the telencephalon of the uPAR(-/-)
mouse. Journal of Comparative Neurology, 489(4), 449–466.
Eagleson, K. L., Milner, T. A., Xie, Z., & Levitt, P. (2013). Synaptic and extrasynaptic
location of the receptor tyrosine kinase Met during postnatal development in the mouse
neocortex and hippocampus. The Journal of Comparative Neurology, 521(14), 3241–3259.
Ebens, A., Brose, K., Leonardo, E. D., Hanson, M. G., Jr., Bladt, F., Birchmeier, C., et al.
(1996). Hepatocyte growth factor/scatter factor is an axonal chemoattractant and a neu-
rotrophic factor for spinal motor neurons. Neuron, 17(6), 1157–1172.
Ebert, D. H., & Greenberg, M. E. (2013). Activity-dependent neuronal signalling and autism
spectrum disorder. Nature, 493(7432), 327–337.
Faletto, D. L., Tsarfaty, I., Kmiecik, T. E., Gonzatti, M., Suzuki, T., & Vande Woude, G. F.
(1992). Evidence for non-covalent clusters of the c-met proto-oncogene product. Onco-
gene, 7(6), 1149–1157.
Fan, S., Ma, Y. X., Gao, M., Yuan, R. Q., Meng, Q., Goldberg, I. D., et al. (2001). The
multisubstrate adapter Gab1 regulates hepatocyte growth factor (scatter factor)-c-Met
signaling for cell survival and DNA repair. Molecular and Cellular Biology, 21(15),
4968–4984.
Furge, K. A., Zhang, Y. W., & Vande Woude, G. F. (2000). Met receptor tyrosine kinase:
Enhanced signaling through adapter proteins. Oncogene, 19(49), 5582–5589.
Garzotto, D., Giacobini, P., Crepaldi, T., Fasolo, A., & De Marchis, S. (2008). Hepatocyte
growth factor regulates migration of olfactory interneuron precursors in the rostral
migratory stream through Met-Grb2 coupling. The Journal of Neuroscience, 28(23),
5901–5909.
Geschwind, D. H., & Levitt, P. (2007). Autism spectrum disorders: Developmental discon-
nection syndromes. Current Opinion in Neurobiology, 17(1), 103–111.
Giacobini, P., Messina, A., Wray, S., Giampietro, C., Crepaldi, T., Carmeliet, P., et al.
(2007). Hepatocyte growth factor acts as a motogen and guidance signal for gonadotropin
hormone-releasing hormone-1 neuronal migration. The Journal of Neuroscience, 27(2),
431–445.
Gorski, J. A., Talley, T., Qiu, M., Puelles, L., Rubenstein, J. L., & Jones, K. R. (2002). Cor-
tical excitatory neurons and glia, but not GABAergic neurons, are produced in the
Emx1-expressing lineage. The Journal of Neuroscience, 22(15), 6309–6314.
Gual, P., Giordano, S., Williams, T. A., Rocchi, S., Van Obberghen, E., & Comoglio, P. M.
(2000). Sustained recruitment of phospholipase C-gamma to Gab1 is required for HGF-
induced branching tubulogenesis. Oncogene, 19(12), 1509–1518.
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 161
Gutierrez, H., Dolcet, X., Tolcos, M., & Davies, A. (2004). HGF regulates the development
of cortical pyramidal dendrites. Development, 131(15), 3717–3726.
Hong, S., Ke, X., Tang, T., Hang, Y., Chu, K., Huang, H., et al. (2011). Detecting abnor-
malities of corpus callosum connectivity in autism using magnetic resonance imaging and
diffusion tensor tractography. Psychiatry Research, 194(3), 333–339.
Huh, C. G., Factor, V. M., Sanchez, A., Uchida, K., Conner, E. A., & Thorgeirsson, S. S.
(2004). Hepatocyte growth factor/c-met signaling pathway is required for efficient liver
regeneration and repair. Proceedings of the National Academy of Sciences of the United States of
America, 101(13), 4477–4482.
Ido, A., Numata, M., Kodama, M., & Tsubouchi, H. (2005). Mucosal repair and growth
factors: Recombinant human hepatocyte growth factor as an innovative therapy for
inflammatory bowel disease. Journal of Gastroenterology, 40(10), 925–931.
Ieraci, A., Forni, P. E., & Ponzetto, C. (2002). Viable hypomorphic signaling mutant of the
Met receptor reveals a role for hepatocyte growth factor in postnatal cerebellar develop-
ment. Proceedings of the National Academy of Sciences of the United States of America, 99(23),
15200–15205.
IMGSAC (1998). A full genome screen for autism with evidence for linkage to a region on
chromosome 7q. International Molecular Genetic Study of Autism Consortium. Human
Molecular Genetics, 7(3), 571–578.
IMGSAC (2001). Further characterization of the autism susceptibility locus AUTS1 on chro-
mosome 7q. Human Molecular Genetics, 10(9), 973–982.
Ishigaki, A., Aoki, M., Nagai, M., Warita, H., Kato, S., Kato, M., et al. (2007). Intrathecal
delivery of hepatocyte growth factor from amyotrophic lateral sclerosis onset suppresses
disease progression in rat amyotrophic lateral sclerosis model. Journal of Neuropathology and
Experimental Neurology, 66(11), 1037–1044.
Jackson, P. B., Boccuto, L., Skinner, C., Collins, J. S., Neri, G., Gurrieri, F., et al. (2009).
Further evidence that the rs1858830 C variant in the promoter region of the MET gene
is associated with autistic disorder. Autism Research, 2(4), 232–236.
Judson, M. C., Amaral, D. G., & Levitt, P. (2011). Conserved subcortical and divergent cor-
tical expression of proteins encoded by orthologs of the autism risk gene MET. Cerebral
Cortex, 21(7), 1613–1626.
Judson, M. C., Bergman, M. Y., Campbell, D. B., Eagleson, K. L., & Levitt, P. (2009).
Dynamic gene and protein expression patterns of the autism-associated met receptor
tyrosine kinase in the developing mouse forebrain. Journal of Comparative Neurology,
513(5), 511–531.
Judson, M. C., Eagleson, K. L., & Levitt, P. (2011). A new synaptic player leading to autism
risk: Met receptor tyrosine kinase. Journal of Neurodevelopmental Disorders, 3(3), 282–292.
Judson, M. C., Eagleson, K. L., Wang, L., & Levitt, P. (2010). Evidence of cell-
nonautonomous changes in dendrite and dendritic spine morphology in the met-signaling-
deficient mouse forebrain. Journal of Comparative Neurology, 518(21), 4463–4478.
Jung, W., Castren, E., Odenthal, M., Vande Woude, G. F., Ishii, T., Dienes, H. P., et al.
(1994). Expression and functional interaction of hepatocyte growth factor-scatter factor
and its receptor c-met in mammalian brain. The Journal of Cell Biology, 126(2), 485–494.
Just, M. A., Cherkassky, V. L., Keller, T. A., Kana, R. K., & Minshew, N. J. (2007). Func-
tional and anatomical cortical underconnectivity in autism: Evidence from an FMRI
study of an executive function task and corpus callosum morphometry. Cerebral Cortex,
17(4), 951–961.
Just, M. A., Cherkassky, V. L., Keller, T. A., & Minshew, N. J. (2004). Cortical activation
and synchronization during sentence comprehension in high-functioning autism: Evi-
dence of underconnectivity. Brain, 127(Pt. 8), 1811–1821.
Kana, R. K., Keller, T. A., Cherkassky, V. L., Minshew, N. J., & Just, M. A. (2009). Atypical
frontal-posterior synchronization of Theory of Mind regions in autism during mental
state attribution. Social Neuroscience, 4(2), 135–152.
162 Yun Peng et al.
Kawas, L. H., Benoist, C. C., Harding, J. W., Wayman, G. A., & Abu-Lail, N. I. (2013).
Nanoscale mapping of the Met receptor on hippocampal neurons by AFM and confocal
microscopy. Nanomedicine, 9(3), 428–438.
Kitamura, K., Iwanami, A., Nakamura, M., Yamane, J., Watanabe, K., Suzuki, Y., et al.
(2007). Hepatocyte growth factor promotes endogenous repair and functional recovery
after spinal cord injury. Journal of Neuroscience Research, 85(11), 2332–2342.
Krasnoselsky, A., Massay, M. J., DeFrances, M. C., Michalopoulos, G., Zarnegar, R., &
Ratner, N. (1994). Hepatocyte growth factor is a mitogen for Schwann cells and is pre-
sent in neurofibromas. The Journal of Neuroscience, 14(12), 7284–7290.
Lai, C. S., Fisher, S. E., Hurst, J. A., Vargha-Khadem, F., & Monaco, A. P. (2001).
A forkhead-domain gene is mutated in a severe speech and language disorder. Nature,
413(6855), 519–523.
Levitt, P., & Campbell, D. B. (2009). The genetic and neurobiologic compass points toward
common signaling dysfunctions in autism spectrum disorders. The Journal of Clinical Inves-
tigation, 119(4), 747–754.
Lim, C. S., & Walikonis, R. S. (2008). Hepatocyte growth factor and c-Met promote den-
dritic maturation during hippocampal neuron differentiation via the Akt pathway. Cel-
lular Signalling, 20(5), 825–835.
Luo, L., Callaway, E. M., & Svoboda, K. (2008). Genetic dissection of neural circuits. Neu-
ron, 57(5), 634–660.
Maina, F., Casagranda, F., Audero, E., Simeone, A., Comoglio, P. M., Klein, R., et al.
(1996). Uncoupling of Grb2 from the Met receptor in vivo reveals complex roles in
muscle development. Cell, 87(3), 531–542.
Maina, F., Hilton, M. C., Andres, R., Wyatt, S., Klein, R., & Davies, A. M. (1998). Multiple
roles for hepatocyte growth factor in sympathetic neuron development. Neuron, 20(5),
835–846.
Maina, F., Hilton, M. C., Ponzetto, C., Davies, A. M., & Klein, R. (1997). Met receptor
signaling is required for sensory nerve development and HGF promotes axonal growth
and survival of sensory neurons. Genes and Development, 11(24), 3341–3350.
Maina, F., Pante, G., Helmbacher, F., Andres, R., Porthin, A., Davies, A. M., et al. (2001).
Coupling Met to specific pathways results in distinct developmental outcomes. Molecular
Cell, 7(6), 1293–1306.
Marshall, C. J. (1995). Specificity of receptor tyrosine kinase signaling: Transient versus
sustained extracellular signal-regulated kinase activation. Cell, 80(2), 179–185.
Marshall, C. R., Noor, A., Vincent, J. B., Lionel, A. C., Feuk, L., Skaug, J., et al. (2008).
Structural variation of chromosomes in autism spectrum disorder. American Journal of
Human Genetics, 82(2), 477–488.
Matsuzaki, M., Ellis-Davies, G. C., Nemoto, T., Miyashita, Y., Iino, M., & Kasai, H. (2001).
Dendritic spine geometry is critical for AMPA receptor expression in hippocampal CA1
pyramidal neurons. Nature Neuroscience, 4(11), 1086–1092.
Miyazawa, T., Matsumoto, K., Ohmichi, H., Katoh, H., Yamashima, T., & Nakamura, T.
(1998). Protection of hippocampal neurons from ischemia-induced delayed neuronal
death by hepatocyte growth factor: A novel neurotrophic factor. Journal of Cerebral Blood
Flow and Metabolism, 18(4), 345–348.
Monga, S. P., Mars, W. M., Pediaditakis, P., Bell, A., Mule, K., Bowen, W. C., et al. (2002).
Hepatocyte growth factor induces Wnt-independent nuclear translocation of beta-
catenin after Met-beta-catenin dissociation in hepatocytes. Cancer Research, 62(7),
2064–2071.
Moumen, A., Ieraci, A., Patane, S., Sole, C., Comella, J. X., Dono, R., et al. (2007). Met
signals hepatocyte survival by preventing Fas-triggered FLIP degradation in a PI3k-Akt-
dependent manner. Hepatology, 45(5), 1210–1217.
Mueller, B. K. (1999). Growth cone guidance: First steps towards a deeper understanding.
Annual Review of Neuroscience, 22, 351–388.
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 163
Mukamel, Z., Konopka, G., Wexler, E., Osborn, G. E., Dong, H., Bergman, M. Y., et al.
(2011). Regulation of MET by FOXP2, genes implicated in higher cognitive dysfunc-
tion and autism risk. The Journal of Neuroscience, 31(32), 11437–11442.
Nakatani, J., Tamada, K., Hatanaka, F., Ise, S., Ohta, H., Inoue, K., et al. (2009). Abnormal
behavior in a chromosome-engineered mouse model for human 15q11-13 duplication
seen in autism. Cell, 137(7), 1235–1246.
Naldini, L., Vigna, E., Ferracini, R., Longati, P., Gandino, L., Prat, M., et al. (1991). The
tyrosine kinase encoded by the MET proto-oncogene is activated by
autophosphorylation. Molecular and Cellular Biology, 11(4), 1793–1803.
Naldini, L., Weidner, K. M., Vigna, E., Gaudino, G., Bardelli, A., Ponzetto, C., et al. (1991).
Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET
receptor. The EMBO Journal, 10(10), 2867–2878.
O’Brien, L. E., Tang, K., Kats, E. S., Schutz-Geschwender, A., Lipschutz, J. H., &
Mostov, K. E. (2004). ERK and MMPs sequentially regulate distinct stages of epithelial
tubule development. Developmental Cell, 7(1), 21–32.
Okunishi, K., Dohi, M., Nakagome, K., Tanaka, R., Mizuno, S., Matsumoto, K., et al.
(2005). A novel role of hepatocyte growth factor as an immune regulator through
suppressing dendritic cell function. Journal of Immunology, 175(7), 4745–4753.
Penagarikano, O., Abrahams, B. S., Herman, E. I., Winden, K. D., Gdalyahu, A., Dong, H.,
et al. (2011). Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities,
and core autism-related deficits. Cell, 147(1), 235–246.
Petreanu, L., Huber, D., Sobczyk, A., & Svoboda, K. (2007). Channelrhodopsin-2-
assisted circuit mapping of long-range callosal projections. Nature Neuroscience,
10(5), 663–668.
Piggot, J., Shirinyan, D., Shemmassian, S., Vazirian, S., & Alarcon, M. (2009). Neural systems
approaches to the neurogenetics of autism spectrum disorders. Neuroscience, 164(1), 247–256.
Pinto, D., Pagnamenta, A. T., Klei, L., Anney, R., Merico, D., Regan, R., et al. (2010).
Functional impact of global rare copy number variation in autism spectrum disorders.
Nature, 466(7304), 368–372.
Ponzetto, C., Bardelli, A., Zhen, Z., Maina, F., dalla Zonca, P., Giordano, S., et al. (1994).
A multifunctional docking site mediates signaling and transformation by the hepatocyte
growth factor/scatter factor receptor family. Cell, 77(2), 261–271.
Ponzetto, C., Zhen, Z., Audero, E., Maina, F., Bardelli, A., Basile, M. L., et al. (1996). Spe-
cific uncoupling of GRB2 from the Met receptor. Differential effects on transformation
and motility. The Journal of Biological Chemistry, 271(24), 14119–14123.
Powell, E. M., Campbell, D. B., Stanwood, G. D., Davis, C., Noebels, J. L., & Levitt, P.
(2003). Genetic disruption of cortical interneuron development causes region- and
GABA cell type-specific deficits, epilepsy, and behavioral dysfunction. The Journal of
Neuroscience, 23(2), 622–631.
Powell, E. M., Mars, W. M., & Levitt, P. (2001). Hepatocyte growth factor/scatter factor is a
motogen for interneurons migrating from the ventral to dorsal telencephalon. Neuron,
30(1), 79–89.
Powell, E. M., Muhlfriedel, S., Bolz, J., & Levitt, P. (2003). Differential regulation of tha-
lamic and cortical axonal growth by hepatocyte growth factor/scatter factor. Develop-
mental Neuroscience, 25(2–4), 197–206.
Pozo, K., & Goda, Y. (2010). Unraveling mechanisms of homeostatic synaptic plasticity.
Neuron, 66(3), 337–351.
Qiu, S., Aldinger, K. A., & Levitt, P. (2012). Modeling of autism genetic variations in mice:
Focusing on synaptic and microcircuit dysfunctions. Developmental Neuroscience, 34(2–3),
88–100.
Qiu, S., Anderson, C. T., Levitt, P., & Shepherd, G. M. (2011). Circuit-specific intracortical
hyperconnectivity in mice with deletion of the autism-associated Met receptor tyrosine
kinase. The Journal of Neuroscience, 31(15), 5855–5864.
164 Yun Peng et al.
Robinson, D. R., Wu, Y. M., & Lin, S. F. (2000). The protein tyrosine kinase family of the
human genome. Oncogene, 19(49), 5548–5557.
Roccisana, J., Reddy, V., Vasavada, R. C., Gonzalez-Pertusa, J. A., Magnuson, M. A., &
Garcia-Ocana, A. (2005). Targeted inactivation of hepatocyte growth factor receptor
c-met in beta-cells leads to defective insulin secretion and GLUT-2 downregulation
without alteration of beta-cell mass. Diabetes, 54(7), 2090–2102.
Royal, I., Lamarche-Vane, N., Lamorte, L., Kaibuchi, K., & Park, M. (2000). Activation of
cdc42, rac, PAK, and rho-kinase in response to hepatocyte growth factor differentially
regulates epithelial cell colony spreading and dissociation. Molecular Biology of the Cell,
11(5), 1709–1725.
Rubenstein, J. L., & Merzenich, M. M. (2003). Model of autism: Increased ratio of
excitation/inhibition in key neural systems. Genes, Brain and Behavior, 2(5), 255–267.
Rudie, J. D., Hernandez, L. M., Brown, J. A., Beck-Pancer, D., Colich, N. L., Gorrindo, P.,
et al. (2012). Autism-associated promoter variant in MET impacts functional and struc-
tural brain networks. Neuron, 75(5), 904–915.
Sahyoun, C. P., Belliveau, J. W., Soulieres, I., Schwartz, S., & Mody, M. (2010). Neuroim-
aging of the functional and structural networks underlying visuospatial vs. linguistic rea-
soning in high-functioning autism. Neuropsychologia, 48(1), 86–95.
Salian-Mehta, S., Xu, M., & Wierman, M. E. (2013). AXL and MET crosstalk to promote
gonadotropin releasing hormone (GnRH) neuronal cell migration and survival. Molecular
and Cellular Endocrinology, 374(1–2), 92–100.
Schmidt, C., Bladt, F., Goedecke, S., Brinkmann, V., Zschiesche, W., Sharpe, M., et al.
(1995). Scatter factor/hepatocyte growth factor is essential for liver development. Nature,
373(6516), 699–702.
Sebat, J., Lakshmi, B., Malhotra, D., Troge, J., Lese-Martin, C., Walsh, T., et al. (2007).
Strong association of de novo copy number mutations with autism. Science,
316(5823), 445–449.
Segarra, J., Balenci, L., Drenth, T., Maina, F., & Lamballe, F. (2006). Combined signaling
through ERK, PI3K/AKT, and RAC1/p38 is required for met-triggered cortical neu-
ron migration. The Journal of Biological Chemistry, 281(8), 4771–4778.
Shukla, D. K., Keehn, B., & Muller, R. A. (2011). Tract-specific analyses of diffusion tensor
imaging show widespread white matter compromise in autism spectrum disorder. Journal
of Child Psychology and Psychiatry, 52(3), 286–295.
Shukla, D. K., Keehn, B., Smylie, D. M., & Muller, R. A. (2011). Microstructural abnor-
malities of short-distance white matter tracts in autism spectrum disorder.
Neuropsychologia, 49(5), 1378–1382.
Sousa, I., Clark, T. G., Toma, C., Kobayashi, K., Choma, M., Holt, R., et al. (2009). MET
and autism susceptibility: Family and case-control studies. European Journal of Human
Genetics, 17(6), 749–758.
Streit, A., Stern, C. D., Thery, C., Ireland, G. W., Aparicio, S., Sharpe, M. J., et al. (1995).
A role for HGF/SF in neural induction and its expression in Hensen’s node during gas-
trulation. Development, 121(3), 813–824.
Sun, W., Funakoshi, H., & Nakamura, T. (2002). Overexpression of HGF retards disease
progression and prolongs life span in a transgenic mouse model of ALS. The Journal of
Neuroscience, 22(15), 6537–6548.
Tabuchi, K., Blundell, J., Etherton, M. R., Hammer, R. E., Liu, X., Powell, C. M., et al.
(2007). A neuroligin-3 mutation implicated in autism increases inhibitory synaptic trans-
mission in mice. Science, 318(5847), 71–76.
Tahara, Y., Ido, A., Yamamoto, S., Miyata, Y., Uto, H., Hori, T., et al. (2003). Hepatocyte
growth factor facilitates colonic mucosal repair in experimental ulcerative colitis in rats.
The Journal of Pharmacology and Experimental Therapeutics, 307(1), 146–151.
MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor 165
Takaishi, K., Sasaki, T., Kato, M., Yamochi, W., Kuroda, S., Nakamura, T., et al. (1994).
Involvement of Rho p21 small GTP-binding protein and its regulator in the HGF-
induced cell motility. Oncogene, 9(1), 273–279.
Tempest, P. R., Stratton, M. R., & Cooper, C. S. (1988). Structure of the met protein and
variation of met protein kinase activity among human tumour cell lines. British Journal of
Cancer, 58(1), 3–7.
Tessier-Lavigne, M., & Goodman, C. S. (1996). The molecular biology of axon guidance.
Science, 274(5290), 1123–1133.
Thanseem, I., Nakamura, K., Miyachi, T., Toyota, T., Yamada, S., Tsujii, M., et al. (2010).
Further evidence for the role of MET in autism susceptibility. Neuroscience Research,
68(2), 137–141.
Thewke, D. P., & Seeds, N. W. (1999). The expression of mRNAs for hepatocyte growth
factor/scatter factor, its receptor c-met, and one of its activators tissue-type plasminogen
activator show a systematic relationship in the developing and adult cerebral cortex and
hippocampus. Brain Research, 821(2), 356–367.
Thomas, C., Humphreys, K., Jung, K. J., Minshew, N., & Behrmann, M. (2011). The anat-
omy of the callosal and visual-association pathways in high-functioning autism: A DTI
tractography study. Cortex, 47(7), 863–873.
Turrigiano, G. G., & Nelson, S. B. (2000). Hebb and homeostasis in neuronal plasticity.
Current Opinion in Neurobiology, 10(3), 358–364.
Tyndall, S. J., Patel, S. J., & Walikonis, R. S. (2007). Hepatocyte growth factor-induced
enhancement of dendritic branching is blocked by inhibitors of N-methyl-D-aspartate
receptors and calcium/calmodulin-dependent kinases. Journal of Neuroscience Research,
85(11), 2343–2351.
Tyndall, S. J., & Walikonis, R. S. (2006). The receptor tyrosine kinase Met and its ligand
hepatocyte growth factor are clustered at excitatory synapses and can enhance clustering
of synaptic proteins. Cell Cycle, 5(14), 1560–1568.
Uehara, Y., Minowa, O., Mori, C., Shiota, K., Kuno, J., Noda, T., et al. (1995). Placental
defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factor.
Nature, 373(6516), 702–705.
Walsh, C. A., Morrow, E. M., & Rubenstein, J. L. (2008). Autism and brain development.
Cell, 135(3), 396–400.
Wong, V., Glass, D. J., Arriaga, R., Yancopoulos, G. D., Lindsay, R. M., & Conn, G. (1997).
Hepatocyte growth factor promotes motor neuron survival and synergizes with ciliary
neurotrophic factor. The Journal of Biological Chemistry, 272(8), 5187–5191.
Wu, H. H., & Levitt, P. (2013). Prenatal expression of MET receptor tyrosine kinase in the
fetal mouse dorsal raphe nuclei and the visceral motor/sensory brainstem. Developmental
Neuroscience, 35(1), 1–16.
Yamamoto, Y., Livet, J., Pollock, R. A., Garces, A., Arce, V., deLapeyriere, O., et al. (1997).
Hepatocyte growth factor (HGF/SF) is a muscle-derived survival factor for a subpopu-
lation of embryonic motoneurons. Development, 124(15), 2903–2913.
Yang, X. M., Toma, J. G., Bamji, S. X., Belliveau, D. J., Kohn, J., Park, M., et al. (1998).
Autocrine hepatocyte growth factor provides a local mechanism for promoting axonal
growth. The Journal of Neuroscience, 18(20), 8369–8381.
Yashiro, K., Riday, T. T., Condon, K. H., Roberts, A. C., Bernardo, D. R., Prakash, R.,
et al. (2009). Ube3a is required for experience-dependent maturation of the neocortex.
Nature Neuroscience, 12(6), 777–783.
Yonan, A. L., Alarcon, M., Cheng, R., Magnusson, P. K., Spence, S. J., Palmer, A. A., et al.
(2003). A genomewide screen of 345 families for autism-susceptibility loci. American Jour-
nal of Human Genetics, 73(4), 886–897.
CHAPTER SIX
Transcriptional Dysregulation
of Neocortical Circuit Assembly
in ASD
Kenneth Y. Kwan1
Department of Human Genetics, Molecular & Behavioral Neuroscience Institute (MBNI), University of
Michigan, Ann Arbor, Michigan, USA
1
Corresponding author: e-mail address: kykwan@umich.edu
Contents
1. Introduction 168
1.1 The organization of the cerebral cortex 172
1.2 The generation and migration of neocortical projection neurons 174
1.3 Generation and migration of neocortical inhibitory interneurons 177
2. Transcriptional Regulation of ASD-related Layer-Dependent Identity
and Connectivity 177
2.1 T-box brain factor 1 (TBR1) 178
2.2 Sex-determining region Y-box 5 (SOX5) 183
2.3 FEZ family zinc finger 2 (FEZF2) 186
2.4 Special AT-rich sequence-binding protein 2 (SATB2) 189
3. Discussion 193
Acknowledgments 195
References 195
Abstract
Autism spectrum disorders (ASDs) impair social cognition and communication, key
higher-order functions centered in the human neocortex. The assembly of neocortical
circuitry is a precisely regulated developmental process susceptible to genetic alter-
ations that can ultimately affect cognitive abilities. Because ASD is an early onset neu-
rodevelopmental disorder that disrupts functions executed by the neocortex, miswiring
of neocortical circuits has been hypothesized to be an underlying mechanism of ASD.
This possibility is supported by emerging genetic findings and data from imaging stud-
ies. Recent research on neocortical development has identified transcription factors as
key determinants of neocortical circuit assembly, mediating diverse processes including
neuronal specification, migration, and wiring. Many of these TFs (TBR1, SOX5, FEZF2, and
SATB2) have been implicated in ASD. Here, I will discuss the functional roles of these
transcriptional programs in neocortical circuit development and their neurobiological
implications for the emerging etiology of ASD.
1. INTRODUCTION
Evolution of the neocortex is thought to underlie our species’ remark-
able cognitive, perceptive, and motor capabilities. It has been hypothesized
that evolutionary advances in neocortical organization and circuitry, while
enabling higher cognition, may have also increased our species’ susceptibility
to disorders that affect cognition. Autism spectrum disorders (ASDs) impair
higher cognitive functions executed by the human neocortex, including
social reciprocity and communication. Although the mechanisms underly-
ing ASD remain largely mysterious, emerging biological insights from
genetic and imaging studies have implicated abnormal neocortical circuit
assembly in ASD. The acquisition of neocortical organization and circuitry
requires the coordinated execution of a series of developmental processes,
including the specification of neuronal identity, neuronal migration,
and wiring of neural circuits. In recent studies, transcription factors
(TFs) have emerged as critical determinants of neocortical development
(Kwan, Sestan, & Anton, 2012; Leone, Srinivasan, Chen, Alcamo, &
McConnell, 2008; MacDonald et al., 2013; Molyneaux, Arlotta,
Menezes, & Macklis, 2007; Rash & Grove, 2006; Rubenstein, 2011). Inter-
estingly, many TFs that are required for the development of neocortical
circuitry have been implicated in ASD. In this chapter, I will review the
function of these ASD-implicated transcriptional mechanisms during
neocortical development and discuss the insights they provide into the neu-
robiology underpinning ASD.
Although ASD is a strongly heritable disorder, phenotypic and genetic
heterogeneity has impeded progress toward identifying loci that carry defin-
itive risk. Reliable genetic findings, however, have begun to emerge from
studies that utilized high-throughput methodologies to analyze
well-characterized populations of patients and families (Iossifov et al., 2012;
Jiang et al., 2013; Neale et al., 2012; O’Roak, Vives, Fu, et al., 2012;
O’Roak, Vives, Girirajan, et al., 2012; Sanders et al., 2011, 2012; Sebat
et al., 2007; Talkowski et al., 2012; Weiss et al., 2003). From these data, it
is now clear that no single locus accounts for more than 1% of ASD cases, with
contributing loci likely numbering in the hundreds. Perhaps somewhat iron-
ically, the genetic heterogeneity that has hindered progress in the previous
decades may now provide an opportunity to illuminate the biological under-
pinnings of ASD, since the increasing number of genes makes possible analyses
of convergent molecular pathways and cellular processes (State & Šestan,
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 169
2012). Indeed, this strategy has been used to intersect ASD-implicated genes
with those that interact with FMRP (Iossifov et al., 2012), the RNA-binding
protein that is lost in fragile X syndrome, which is the leading monogenic
cause of intellectual disability (ID) and syndromic autism (Chonchaiya,
Schneider, & Hagerman, 2009). This analysis found that a significant number
of ASD candidate genes are associated with FMRP, which is consistent with
the possibility that common molecular pathways underlie autism and fragile
X syndrome. In addition, weighted gene coexpression network analysis can
reveal previously unrecognized connections between ASD risk genes. One
such example of this unbiased approach was used to analyze gene expression
in differentiating normal human neuronal progenitors, which revealed a sig-
nificant overlap with ASD susceptibility genes annotated by the SFARI data-
base (http://gene.sfari.org) (Konopka, Wexler, et al., 2012). Future studies of
the relationship between loci that confer ASD risks are likely to lead to addi-
tional insights about the neurobiological underpinnings of ASD.
With accumulating genetic data, it may now be possible to better pin-
point the timing and location of the biological events most relevant to
the etiology of ASD. This possibility is facilitated by recent transcriptomic
studies from multiple groups that have focused on spatiotemporal analyses of
gene expression in the human brain (Colantuoni et al., 2011; Johnson et al.,
2009; Kang et al., 2011). Available data from one of these resources (http://
www.humanbraintranscriptome.org) revealed that many of the most reli-
able risk-carrying loci exhibit a sharp upregulation in the neocortex during
the mid-fetal period (red arrowhead in Fig. 6.1) (State & Šestan, 2012), a key
developmental window for the acquisition of neocortical organization and
neural circuits. This distinct mid-gestation developmental pattern, which is
not consistently observed in other brain regions (Fig. 6.1), suggests that
ASD-associated genes may converge on pathways that function during
the structural development and neural circuit wiring of the neocortex.
Additional evidence further implicates the mid-fetal period of neocorti-
cal development in ASD. Neuronal migration, the process by which new-
born neurons are positioned away from the germinal zones and toward their
correct mantle layer destinations (Angevine & Sidman, 1961; Caviness,
1982; Lambert de Rouvroit & Goffinet, 1998; Rakic, 1974), occurs during
early to mid-gestation and has been shown to be disrupted in some ASD
patients (Hutsler, Love, & Zhang, 2007; Peñagarikano et al., 2011;
Wegiel et al., 2010). Furthermore, there is evidence for abnormalities
in the minicolumnar organization of neocortical neurons in cases of
ASD and other neuropsychiatric disorders (Casanova, Buxhoeveden,
170 Kenneth Y. Kwan
11
10
9
8
7
6
CHD8 NRXN1 SCN2A
5 FOXP1 NRXN2 SHANK1
GRIN2B NRXN3 SHANK2
4 NLGN4X NTNG1 SHANK3
NR4A2 SCN1A TBR1
3
50 100 200 500 2000 10,000 30,000 50 100 200 500 2000 10,000 30,000
Birth Birth
Age (days post conception) Age (days post conception)
Figure 6.1 Developmental expression of 15 select ASD risk genes in the human neo-
cortex and other human brain regions. A collective upregulation in ASD risk gene
expression (red arrowhead) is present during mid-fetal development in the neocortex
but not other brain regions. Data from Kang et al. (2011).
CR Cajal–Retzius neuron
CP Cortical plate L1
En Embryonic day n (mouse)
IP Intermediate progenitor L2
IZ
WM
PP Corticospinal
Corticobulbar Subcortical
SVZ Corticotectal axon tracts
IP
MZ Corticothalamic
RGC E11
VZ
NP
SP L6 L5 L4 L2/3
<E10 E11 E12 E13 E14 E15/16 >E17
NP expansion Gliogenesis
Figure 6.2 Neocortical development in the mouse. Prior to neurogenesis (<E10), neur-
oepithelial progenitors (NPs) divide mostly symmetrically in the ventricular zone (VZ).
Starting at E11, NPs assume radial glial morphology to become radial glia cells
(RGCs), which divide asymmetrically to generate neurons and guide their migration
to the mantle layers. The first projection neurons settle in the preplate (PP) to form
the nascent cortical plate (CP), from which L2 to L6 would emerge. Incoming CP neurons
segregate the PP into the marginal zone (MZ) and subplate (SP). Projection neurons are
then generated sequentially through successive divisions of RGCs in the VZ, as well as
neurogenic divisions of intermediate progenitors (IPs) in the SVZ. The generation of pro-
jection neurons and their migration into the CP occurs in an inside-first, outside-last
manner; early-born neurons form the deep layers (SP, L6, and L5), whereas later-born
neurons migrate past older neurons to form more superficial layers (L4, L3, and L2).
At the end of neurogenesis, the radial scaffold is dismantled and gliogenesis occurs
(>E17).
from the deep layers (L5 and L6) and the subplate (SP), a neocortical struc-
ture that contains early-born neurons positioned between L6 and WM
(Allendoerfer & Shatz, 1994; Herrmann, Antonini, & Shatz, 1994;
Kostovic & Rakic, 1980, 1990; Molliver, Kostovic, & van der Loos,
1973; Rakic, 1976). Axons that innervate the thalamus, which form the cor-
ticothalamic tract, originate largely from SP and L6, whereas axons that pro-
ject subcerebrally, including the corticotectal, corticobulbar, and
corticospinal tracts, arise exclusively from L5. Projection neurons positioned
in the upper layers (L2–L4), in contrast, project only within the cortex,
either intrahemispherically or contralaterally. The corpus callosum, which
is formed by contralateral intracortical axons, enables communication
between the two cerebral hemispheres. Neural imaging studies have
174 Kenneth Y. Kwan
2008; MacDonald et al., 2013; Molyneaux et al., 2007; Polleux, Ince-Dunn, &
Ghosh, 2007; Rash & Grove, 2006). Although much remains unknown, genes
expressed in layer-selective or neuronal subtype-specific patterns are likely to be
important. TFs are known to play pivotal roles in processes including the early
patterning, sequential generation, arealization, dendritic morphology, and axo-
nal connectivity of distinct neuronal cell types (Guillemot, 2007; Hébert &
Fishell, 2008; Hevner, Hodge, Daza, & Englund, 2006; Jessell, 2000; Kwan,
Sestan, & Anton, 2012; MacDonald et al., 2013; Mallamaci & Stoykova,
2006; Monuki & Walsh, 2001; O’Leary & Sahara, 2008). In the neocortex, a
number of layer- and neuronal subtype-specific TFs have been identified and
characterized (Gray et al., 2004; Kwan, Sestan, & Anton, 2012; MacDonald
et al., 2013; Molyneaux et al., 2007).
Here, I will discuss four transcriptional mechanisms critical to neocortical
development that have been implicated in ASD. As revealed by recent
genetic findings, there exists considerable overlap of risk-carrying genes
in disorders that are phenotypically distinct (Marshall et al., 2008;
Mefford et al., 2010; Smoller et al., 2013; State & Šestan, 2012). Genes that
have been implicated in ASD have also been consistently implicated in epi-
lepsy, intellectual disability, schizophrenia, and bipolar disorder, suggesting
that shared genetic liabilities can lead to diverse clinical manifestations.
Therefore, in my discussion of genetic findings in the succeeding text, stud-
ies of neuropsychiatric disorders in addition to ASD will also be included.
L2-4 L2-4
L5 L5
L6 L6
SP SP
Mid
Th CC
St
St
Pons
Hyp
Misexpression Misexpression
Figure 6.3 Summary of neocortical gene expression and function for Tbr1, Sox5, Fezf2,
and Satb2.
TBR1
TANK TBR1 DPP4 FAP KCNH7 FIGN
PSMD14 SLC4A10 IFIH1
CNVs (loss)
Traylor et al. (2012)
Magri et al. (2011)
Krepischi et al. (2010)
Palumbo et al. (2012)
Takatsuki et al. (2010)
T-Box
SNVs
O’Roak, Vives, Ala136Profs X80
Fu, et al. (2012) Lys228Glu
O’Roak, Vives, Ser351 X
Girirajan, et al. (2012)
region (O’Roak, Vives, Fu, et al., 2012). The first (Lys228Glu) is a missense
mutation that affects a highly conserved residue within the T-box domain.
The second (Ser351X) is a nonsense mutation that truncates a significant por-
tion of the T-box domain. Both of these variants are predicted to be highly
deleterious to protein function.
Additional evidence of TBR1 involvement in molecular pathways rele-
vant to ASD includes its regulation of autism susceptibility candidate 2
(AUTS2) and Reelin (RELN) (Bedogni et al., 2010; Hevner et al., 2001).
Rare mutations in AUTS2 have been identified in ASD in studies from
numerous groups (Ben-David et al., 2011; Huang, Zou, Maher,
Newton, & Milunsky, 2010; Kalscheuer et al., 2007; Prasad et al., 2012;
Sultana et al., 2002; Talkowski et al., 2012). AUTS2 was further identified
as a hypermutable ASD-associated gene (Michaelson et al., 2012). The
expression of Auts2, which is highly enriched in frontal neocortex, is depen-
dent on Tbr1 (Bedogni et al., 2010). In the Tbr1-null mouse, Auts2 expres-
sion is severely decreased, suggesting that loss of AUTS2 may contribute
mechanistically to TBR1 dysfunction in ASD. TBR1 further controls the
expression of Reln (Hevner et al., 2001). Association studies have repeatedly
implicated RELN in ASD (Holt et al., 2010; Kelemenova et al., 2010; Li
et al., 2008; Persico et al., 2001; Serajee et al., 2006; Skaar et al., 2005).
Furthermore, rare variants in RELN have also been identified (Neale
et al., 2012). In the Tbr1-deficient neocortex, the early expression of Reln
in CR neurons of the MZ is greatly reduced (Hevner et al., 2001),
suggesting an additional candidate mechanism by which TBR1 may contrib-
ute to ASD.
The expression and function of Tbr1 may provide key insights into the
neurobiology of ASD. The expression of TBR1 is highly enriched in the
developing neocortex (Fig. 6.3) (Hevner et al., 2001), which is consistent
with a contribution of neocortical dysfunction to ASD. Within the neocor-
tex, TBR1 expression is restricted to the corticothalamic projection neurons
of L6 and the SP and to the CR neurons of the MZ from an early embryonic
age (E12.5) (Han et al., 2011; Hevner et al., 2001; McKenna et al., 2011).
TBR1, however, is absent from VZ and SVZ progenitor cells and therefore
functions postmitotically in neurons after their terminal division and during
their differentiation. In the neonatal mouse cortex, TBR1 is absent from the
upper-layer neurons that contribute to the majority of callosal axons. During
the first postnatal week, however, a number of upper-layer neurons begin to
upregulate TBR1 expression (Han et al., 2011; Hevner et al., 2001;
McKenna et al., 2011). This upper-layer expression of TBR1 likely serves
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 181
Hevner et al., 2001; McKenna et al., 2011). Therefore, TBR1 may function
cell autonomously in callosal neurons. Consistent with this possibility, the
expression of TBR1 in upper-layer neurons is sufficient to rescue the callosal
defects observed in the special AT-rich sequence-binding protein 2 (Satb2)
mutant neocortex (Srinivasan et al., 2012), which is further discussed later.
An additional explanation, however, may be found in TBR1 regulation of
early SP circuits, which are known to orchestrate the development of both
callosal and corticofugal connectivities (Allendoerfer & Shatz, 1994; Del
Rı́o, Martı́nez, Auladell, & Soriano, 2000; Herrmann et al., 1994;
Kostovic & Rakic, 1980, 1990; Molliver et al., 1973; Rakic, 1976).
The SP is a transient zone in the developing neocortex in which some of
the earliest synapses and pioneering circuits develop (Allendoerfer & Shatz,
1994; Del Rı́o et al., 2000; Herrmann et al., 1994; Kostovic & Rakic, 1980,
1990; Molliver et al., 1973; Rakic, 1976). The SP, the thickest zone in the
human mid-fetal cerebral wall, is thought to play critical roles in the migra-
tion and synaptogenesis of CP neurons, as well as in the assembly of proper
neocortical efferent and afferent axonal projections. Examination of Tbr1-
deficient mice revealed a severe disruption of SP formation, which is part
of a complex mismigration phenotype that is distinct compared to the
wholly inverted cortex of the Reeler mutant (Rice & Curran, 2001). In
the Tbr1-null neocortex, early-born SP neurons form an ectopic band in
the center of the CP. This “midplate” exhibits a rostral–caudal gradient,
being more deeply positioned in the rostral cortex (Han et al., 2011;
Hevner et al., 2001). These areal-dependent migration defects are consistent
with a role of Tbr1 in the control of regional identity (Bedogni et al., 2010).
In addition to mismigration of SP neurons, L5 neurons are also disrupted,
being more widely distributed throughout the CP, whereas upper-layer
neurons are distributed bimodally, with a majority positioned below the
ectopic band of SP neurons (Han et al., 2011; Hevner et al., 2001). These
complex migration defects suggest that, other than regulation of Reln
expression, Tbr1 is likely to control migration via additional mechanisms.
Interestingly, consistent with the central role of SP in orchestrating cor-
tical connectivites, recent data have implicated SP in ASD. Comprehensive
profiling of gene expression in the mouse SP through multiple developmen-
tal ages was combined with network analysis, which unbiasedly revealed a
significant enrichment of ASD- and schizophrenia-implicated genes being
expressed in the SP (Hoerder-Suabedissen et al., 2013). These data suggest
the possibility that disruption of early pioneering SP circuits can ultimately
lead to wider alterations of neocortical organization and circuitry in disease
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 183
SOX5
HMG CC
Rearrangement
Talkowski et al. (2012) t(11;12)(p13;p12.1)
necessary for homo- and heterodimerization (Lefebvre et al., 2007), and are
therefore predicted to be highly deleterious. Many of these patients suffer
from speech and language delays, moderate to severe ID, and behavioral def-
icits that include autistic-like features and stereotypies (Lamb et al., 2012;
Rosenfeld et al., 2010; Schanze et al., 2013; Talkowski et al., 2012). Fur-
thermore, several cases of larger deletions that span beyond the entire
SOX5 gene have also been reported (Gläser et al., 2003; Lu et al., 2009;
Nagai et al., 1995; Stumm et al., 2007). In addition to CNVs, SOX5 inter-
ruption by chromosomal rearrangement has been reported in ASD
(Talkowski et al., 2012). With a breakpoint located between the HMG
box and coiled-coil domain, this t(11;12)(p13;p12.1) translocation is also
predicted to lead to SOX5 loss of function.
The expression pattern of SOX5 suggests that it plays a role in esta-
blishing early neocortical circuits, perturbations of which may contribute
to ASD (Fig. 6.3). In the embryonic neocortex, SOX5 expression is highly
enriched in L6 and SP corticothalamic projection neurons and a subset of L5
subcerebral projection neurons, starting at around E14.5 (Kwan et al., 2008;
Lai et al., 2008). The absence of SOX5 from cortical VZ and SVZ progen-
itor cells throughout embryonic development indicates that it is likely to
function postmitotically (Kwan et al., 2008; Lai et al., 2008).
Consistent with this spatiotemporal expression, Sox5 cell autonomously
controls the migration, differentiation, and axonal projections of these early-
born neurons (Fig. 6.3) (Kwan et al., 2008; Lai et al., 2008). Analysis of
Sox5-deficient mice revealed marked disruption of layer-dependent
corticofugal connectivities. In the absence of Sox5, the vast majority of cor-
ticothalamic axons arising from SP and L6 neurons fail to reach the dorsal
thalamus and are instead misrouted to the hypothalamus (Kwan et al.,
2008). In addition, subcerebral axons originating from L5 neurons are
greatly reduced, with projections to the pons and spinal cord, including
the corticospinal tract (CST), being nearly completely abolished (Kwan
et al., 2008). Interestingly, although Sox5 is required for the normal forma-
tion of the CST, its misexpression in upper-layer neurons was not sufficient
to respecify their projectional fate (Kwan et al., 2008), suggesting that the
role of Sox5 is not instructive. The corpus callosum, which is known to
be altered in some cases of ASD, is grossly normal in the Sox5-null neocor-
tex. The remarkable defects in corticothalamic and corticospinal projections
in the Sox5 mutant mouse, however, are consistent with studies that show
changes to the internal capsule in ASD, which indicate alterations to
corticofugal connectivites (Ingalhalikar et al., 2011; Wolff et al., 2012).
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 185
FEZF2
ZFs
FEZF2
Sanders et al. (2012) Arg344Cys
Wang et al. (2009) rs3755827 (T/c)
Placental Mammal Basewise Conservation by PhyloP
Mammal
Cons
Multiz Alignments of Vertebrates
Rhesus
Mouse
Dog
Elephant
Opossum
Chicken
expression of Bcl11b, Etv1, Foxo1, Crym, Diap3, Clim1, Crim1, and other L5
markers are lost or severely reduced (Chen, Rasin, et al., 2005; Chen,
Schaevitz, & McConnell, 2005; Molyneaux et al., 2005). Accordingly,
Fezf2 is necessary for the formation of the corticospinal tract that originates
from these neurons. When Fezf2 is genetically removed (Chen, Schaevitz, &
McConnell, 2005; Molyneaux et al., 2005) or knocked down by RNAi
(Chen, Rasin, et al., 2005), corticospinal axons fail to enter the pons or spinal
cord and some of these axons aberrantly invade the anterior commissure
(Chen, Schaevitz, & McConnell, 2005; Molyneaux et al., 2005). Fezf2,
however, is not only necessary for normal corticospinal tract formation
but also sufficient to induce it ectopically. When Fezf2 is specifically mis-
expressed in upper-layer neurons that normally project only intracortically,
ectopic subcortical projections to the pons and spinal cord originate from
these neurons (Chen, Rasin, et al., 2005). Furthermore, Fezf2 is sufficient
to reprogram intracortical projection neurons to project subcortically in
the early postnatal neocortex (De la Rossa et al., 2013; Rouaux &
Arlotta, 2013). In addition to L5 identity and projections, Fezf2 also controls
the molecular development of L6 neurons and their corticothalamic projec-
tions (Chen, Rasin, et al., 2005; Chen, Schaevitz, & McConnell, 2005;
Molyneaux et al., 2005). Furthermore, Probst bundles containing callosal
axons that are unable to cross the midline have been reported in the
Fezf2-null cortex (Chen, Rasin, et al., 2005; Chen, Schaevitz, &
McConnell, 2005; Molyneaux et al., 2005). Although this Fezf2 phenotype
may not be cell autonomous, changes in callosal connectivity can contribute
to circuit alternations relevant to ASD. Furthermore, the loss of subcerebral
connectivity in the absence of Fezf2 is consistent with reduced internal cap-
sule innervation reported in some ASD patients (Ingalhalikar et al., 2011;
Wolff et al., 2012).
These diverse roles of Fezf2 on axon projections are mediated by its
involvement in complex transcription networks that mediate multiple
aspects of neocortical development (Chen et al., 2008; Han et al., 2011;
Kwan et al., 2008; Leone et al., 2008; McKenna et al., 2011; Shim et al.,
2012; Srinivasan et al., 2012). These networks include direct and indirect
interactions between FEZF2 and SOX5, TBR1, and SATB2, the other
ASD-implicated TFs discussed in this chapter. This central role of FEZF2
in neocortical transcriptional networks is consistent with its high
intramodular connectivity in a gene coexpression network, the cortical
development module, assembled unbiasedly from the developing human
brain transcriptome (Kang et al., 2011).
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 189
SATB2
CUT
SATB2 FTCDNL1 TYW5
SATB2-AS1 C2orf69
CNVs (loss)
Talkowski et al. (2012)
Rosenfeld et al. (2009)
Van Buggenhout et al. (2005)
Rearrangements
Marshall et al. (2008) t(2;6)(q32;p22)
Talkowski et al. (2012) t(2;6)(q33;q21)
SNVs
Leoyklang et al. (2007) Arg239X
Rauch et al. (2012) Val381Gly
Jiang et al. (2013) Pro655Ser
3. DISCUSSION
As the pace of genetic discoveries accelerates, functional studies are
required to understand how alterations in candidate risk genes can contrib-
ute to ASD. Mechanistic studies of ASD neurobiology, however, are chal-
lenging not only because of the significant degree of phenotypic and genetic
heterogeneity but also because the complex cognitive and social deficits in
ASD cannot be readily modeled in rodents.
Genetic studies in mice, however, do have the potential to illuminate
mechanisms of neural circuitry assembly relevant to ASD. Many fundamen-
tal aspects of neocortical development are well conserved between rodent
and primate species, including the establishment of primary neocortical areas
and major axon tracts and the genetic programs underlying their specifica-
tion. Indeed, the layer-dependent expression patterns of the four transcrip-
tional determinants discussed in this chapter (Tbr1, Sox5, Fezf2, and Satb2)
are conserved between the mouse and human neocortices (Ip, Bayatti,
Howard, Lindsay, & Clowry, 2011; Kwan, Lam, et al., 2012; Kwan
et al., 2008; Saito et al., 2011). This suggests not only that their highly spe-
cific gene expression patterns are controlled by the same upstream transcrip-
tional regulators in the two species but also that their functions during
neocortical development are shared. These common mechanisms should
be amenable to being modeled in the mouse. Furthermore, although the
mouse is an imperfect model, given the technical, logistical, and ethical con-
siderations, no better alternatives exist for experimental interrogation of
intact neural circuits.
Genetic removal of Tbr1, Sox5, Fezf2, or Satb2 from the mouse leads to
profound defects in the neocortex, which are not observed in human ASD
cases. It should be noted, however, that the mouse genetic studies discussed
in this chapter involved the complete, homozygous removal of the genes of
interest from the animal in order to reveal the most severe phenotypes. In
human cases, these genes are likely to be affected less severely. Often, the
reported ASD variants are SNVs or CNVs that do not remove the entire
gene. Furthermore, all of the ASD cases discussed in this chapter are hetero-
zygous at the locus in question, suggesting that one functional copy of the
gene is present. Subtle alterations in gene dosage or functional capacity may
more subtly affect neocortical circuit wiring in a manner consistent with
ASD. In future experiments, recapitulating patient mutations in the mouse
is likely to reveal mechanistic insights most relevant to human ASD.
194 Kenneth Y. Kwan
ACKNOWLEDGMENTS
I thank Yuka Imamura Kawasawa and Mingfeng Li for the help with the human brain
transcriptome data in Fig. 6.1. This work was supported by the National Institutes of
Health (MH096939).
REFERENCES
Alcamo, E., Chirivella, L., Dautzenberg, M., Dobreva, G., Fariñas, I., Grosschedl, R., et al.
(2008). Satb2 regulates callosal projection neuron identity in the developing cerebral cor-
tex. Neuron, 57, 364–377.
Alexander, A. L., Lee, J. E., Lazar, M., Boudos, R., DuBray, M. B., Oakes, T. R., et al.
(2007). Diffusion tensor imaging of the corpus callosum in Autism. NeuroImage, 34,
61–73.
Allendoerfer, K. L., & Shatz, C. J. (1994). The subplate, a transient neocortical structure: Its
role in the development of connections between thalamus and cortex. Annual Review of
Neuroscience, 17, 185–218.
Anderson, S. A., Eisenstat, D. D., Shi, L., & Rubenstein, J. L. (1997). Interneuron migration
from basal forebrain to neocortex: Dependence on Dlx genes. Science, 278, 474–476.
Ang, E. S., Haydar, T. F., Gluncic, V., & Rakic, P. (2003). Four-dimensional migratory
coordinates of GABAergic interneurons in the developing mouse cortex. The Journal
of Neuroscience, 23, 5805–5815.
Angevine, J. B., Jr., & Sidman, R. L. (1961). Autoradiographic study of cell migration during
histogenesis of cerebral cortex in the mouse. Nature, 192, 766–768.
Anthony, T. E., Klein, C., Fishell, G., & Heintz, N. (2004). Radial glia serve as neuronal
progenitors in all regions of the central nervous system. Neuron, 41, 881–890.
Anton, E. S., Kreidberg, J. A., & Rakic, P. (1999). Distinct functions of alpha3 and
alpha(v) integrin receptors in neuronal migration and laminar organization of the cerebral
cortex. Neuron, 22, 277–289.
Arlotta, P., Molyneaux, B. J., Chen, J., Inoue, J., Kominami, R., & Macklis, J. D. (2005).
Neuronal subtype-specific genes that control corticospinal motor neuron development
in vivo. Neuron, 45, 207–221.
Baranek, C., Dittrich, M., Parthasarathy, S., Bonnon, C. G., Britanova, O., Lanshakov, D.,
et al. (2012). Protooncogene Ski cooperates with the chromatin-remodeling factor Satb2
in specifying callosal neurons. Proceedings of the National Academy of Sciences of the United
States of America, 109, 3546–3551.
Bedogni, F., Hodge, R., Elsen, G., Nelson, B., Daza, R., Beyer, R., et al. (2010). Tbr1
regulates regional and laminar identity of postmitotic neurons in developing neocortex.
Proceedings of the National Academy of Sciences of the United States of America, 107,
13129–13134.
Ben-David, E., Granot-Hershkovitz, E., Monderer-Rothkoff, G., Lerer, E., Levi, S.,
Yaari, M., et al. (2011). Identification of a functional rare variant in autism using
genome-wide screen for monoallelic expression. Human Molecular Genetics, 20,
3632–3641.
Bentivoglio, M., & Mazzarello, P. (1999). The history of radial glia. Brain Research Bulletin,
49, 305–315.
Bowers, J. M., & Konopka, G. (2012). The role of the FOXP family of transcription factors in
ASD. Disease Markers, 33, 251–260.
Britanova, O., de Juan Romero, C., Cheung, A., Kwan, K. Y., Schwark, M., Gyorgy, A.,
et al. (2008). Satb2 is a postmitotic determinant for upper-layer neuron specification in
the neocortex. Neuron, 57, 378–392.
196 Kenneth Y. Kwan
Casanova, M. F., Buxhoeveden, D., & Gomez, J. (2003). Disruption in the inhibitory
architecture of the cell minicolumn: Implications for autism. The Neuroscientist, 9,
496–507.
Casanova, M. F., Buxhoeveden, D. P., Switala, A. E., & Roy, E. (2002). Minicolumnar
pathology in autism. Neurology, 58, 428–432.
Caviness, V. S., Jr. (1982). Neocortical histogenesis in normal and Reeler mice:
A developmental study based upon [3H]thymidine autoradiography. Brain Research,
256, 293–302.
Caviness, V. S., Jr., Takahashi, T., & Nowakowski, R. S. (1995). Numbers, time and neo-
cortical neuronogenesis: A general developmental and evolutionary model. Trends in
Neurosciences, 18, 379–383.
Chao, H. T., Chen, H., Samaco, R. C., Xue, M., Chahrour, M., Yoo, J., et al. (2010). Dys-
function in GABA signalling mediates autism-like stereotypies and Rett syndrome phe-
notypes. Nature, 468, 263–269.
Chen, J., Rasin, M., Kwan, K., & Sestan, N. (2005). Zfp312 is required for subcortical axonal
projections and dendritic morphology of deep-layer pyramidal neurons of the cerebral
cortex. Proceedings of the National Academy of Sciences of the United States of America,
102, 17792–17797.
Chen, B., Schaevitz, L., & McConnell, S. (2005). Fezl regulates the differentiation and axon
targeting of layer 5 subcortical projection neurons in cerebral cortex. Proceedings of the
National Academy of Sciences of the United States of America, 102, 17184–17189.
Chen, B., Wang, S., Hattox, A., Rayburn, H., Nelson, S., & McConnell, S. (2008). The
Fezf 2-Ctip2 genetic pathway regulates the fate choice of subcortical projection neurons
in the developing cerebral cortex. Proceedings of the National Academy of Sciences of the
United States of America, 105, 11382–11387.
Chonchaiya, W., Schneider, A., & Hagerman, R. J. (2009). Fragile X: A family of disorders.
Advances in Pediatrics, 56, 165–186.
Clancy, B., Kersh, B., Hyde, J., Darlington, R. B., Anand, K. J., & Finlay, B. L. (2007).
Web-based method for translating neurodevelopment from laboratory species to
humans. Neuroinformatics, 5, 79–94.
Colantuoni, C., Lipska, B. K., Ye, T., Hyde, T. M., Tao, R., Leek, J. T., et al. (2011). Tem-
poral dynamics and genetic control of transcription in the human prefrontal cortex.
Nature, 478, 519–523.
Comery, T. A., Harris, J. B., Willems, P. J., Oostra, B. A., Irwin, S. A., Weiler, I. J., et al.
(1997). Abnormal dendritic spines in fragile X knockout mice: Maturation and pruning
deficits. Proceedings of the National Academy of Sciences of the United States of America, 94,
5401–5404.
D’Arcangelo, G., & Curran, T. (1998). Reeler: New tales on an old mutant mouse. Bioessays,
20, 235–244.
De Carlos, J. A., & O’Leary, D. D. (1992). Growth and targeting of subplate axons and estab-
lishment of major cortical pathways. The Journal of Neuroscience, 12, 1194–1211.
DeFelipe, J., & Farinas, I. (1992). The pyramidal neuron of the cerebral cortex: Morpholog-
ical and chemical characteristics of the synaptic inputs. Progress in Neurobiology, 39,
563–607.
De la Rossa, A., Bellone, C., Golding, B., Vitali, I., Moss, J., Toni, N., et al. (2013). In vivo
reprogramming of circuit connectivity in postmitotic neocortical neurons. Nature
Neuroscience, 16, 193–200.
Del Rı́o, J., Martı́nez, A., Auladell, C., & Soriano, E. (2000). Developmental history of the
subplate and developing white matter in the murine neocortex. Neuronal organization
and relationship with the main afferent systems at embryonic and perinatal stages. Cerebral
Cortex, 10, 784–801.
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 197
Enard, W., Przeworski, M., Fisher, S. E., Lai, C. S., Wiebe, V., Kitano, T., et al. (2002).
Molecular evolution of FOXP2, a gene involved in speech and language. Nature,
418, 869–872.
Englund, C., Fink, A., Lau, C., Pham, D., Daza, R. A., Bulfone, A., et al. (2005). Pax6, Tbr2,
and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and
postmitotic neurons in developing neocortex. The Journal of Neuroscience, 25, 247–251.
Fischbach, G. D., & Lord, C. (2010). The Simons Simplex Collection: A resource for iden-
tification of autism genetic risk factors. Neuron, 68, 192–195.
Fishell, G., & Hanashima, C. (2008). Pyramidal neurons grow up and change their mind.
Neuron, 57, 333–338.
Geschwind, D., & Levitt, P. (2007). Autism spectrum disorders: Developmental disconnec-
tion syndromes. Current Opinion in Neurobiology, 17, 103–111.
Girirajan, S., Dennis, M. Y., Baker, C., Malig, M., Coe, B. P., Campbell, C. D., et al. (2013).
Refinement and discovery of new hotspots of copy-number variation associated with
autism spectrum disorder. American Journal of Human Genetics, 92, 221–237.
Gläser, B., Rossier, E., Barbi, G., Chiaie, L. D., Blank, C., Vogel, W., et al. (2003). Molec-
ular cytogenetic analysis of a constitutional de novo interstitial deletion of chromosome
12p in a boy with developmental delay and congenital anomalies. American Journal of
Medical Genetics Part A, 116A, 66–70.
Gray, P. A., Fu, H., Luo, P., Zhao, Q., Yu, J., Ferrari, A., et al. (2004). Mouse brain orga-
nization revealed through direct genome-scale TF expression analysis. Science, 306,
2255–2257.
Guillemot, F. (2007). Cell fate specification in the mammalian telencephalon. Progress in
Neurobiology, 83, 37–52.
Hamdan, F. F., Daoud, H., Rochefort, D., Piton, A., Gauthier, J., Langlois, M., et al. (2010).
De novo mutations in FOXP1 in cases with intellectual disability, autism, and language
impairment. American Journal of Human Genetics, 87, 671–678.
Han, W., Kwan, K. Y., Shim, S., Lam, M. M., Shin, Y., Xu, X., et al. (2011). TBR1 directly
represses Fezf 2 to control the laminar origin and development of the corticospinal tract.
Proceedings of the National Academy of Sciences of the United States of America, 108,
3041–3046.
Hashimoto, H., Yabe, T., Hirata, T., Shimizu, T., Bae, Y., Yamanaka, Y., et al. (2000).
Expression of the zinc finger gene fez-like in zebrafish forebrain. Mechanisms of Develop-
ment, 97, 191–195.
Haubensak, W., Attardo, A., Denk, W., & Huttner, W. B. (2004). Neurons arise in the basal
neuroepithelium of the early mammalian telencephalon: A major site of neurogenesis.
Proceedings of the National Academy of Sciences of the United States of America, 101,
3196–3201.
Hébert, J. M., & Fishell, G. (2008). The genetics of early telencephalon patterning: Some
assembly required. Nature Reviews Neuroscience, 9, 678–685.
Herrmann, K., Antonini, A., & Shatz, C. J. (1994). Ultrastructural evidence for synaptic
interactions between thalamocortical axons and subplate neurons. The European Journal
of Neuroscience, 6, 1729–1742.
Hevner, R. F., Hodge, R. D., Daza, R. A., & Englund, C. (2006). Transcription factors in
glutamatergic neurogenesis: Conserved programs in neocortex, cerebellum, and adult
hippocampus. Neuroscience Research, 55, 223–233.
Hevner, R., Shi, L., Justice, N., Hsueh, Y., Sheng, M., Smiga, S., et al. (2001). Tbr1 regulates
differentiation of the preplate and layer 6. Neuron, 29, 353–366.
Hirata, T., Nakazawa, M., Muraoka, O., Nakayama, R., Suda, Y., & Hibi, M. (2006). Zinc-
finger genes Fez and Fez-like function in the establishment of diencephalon subdivisions.
Development, 133, 3993–4004.
198 Kenneth Y. Kwan
Hirata, T., Suda, Y., Nakao, K., Narimatsu, M., Hirano, T., & Hibi, M. (2004). Zinc finger
gene fez-like functions in the formation of subplate neurons and thalamocortical axons.
Developmental Dynamics, 230, 546–556.
Hoerder-Suabedissen, A., Oeschger, F. M., Krishnan, M. L., Belgard, T. G., Wang, W. Z.,
Lee, S., et al. (2013). Expression profiling of mouse subplate reveals a dynamic gene net-
work and disease association with autism and schizophrenia. Proceedings of the National
Academy of Sciences of the United States of America, 110, 3555–3560.
Holt, R., Barnby, G., Maestrini, E., Bacchelli, E., Brocklebank, D., Sousa, I., et al. (2010).
Linkage and candidate gene studies of autism spectrum disorders in European
populations. European Journal of Human Genetics, 18, 1013–1019.
Huang, X. L., Zou, Y. S., Maher, T. A., Newton, S., & Milunsky, J. M. (2010). A de novo
balanced translocation breakpoint truncating the autism susceptibility candidate 2
(AUTS2) gene in a patient with autism. American Journal of Medical Genetics Part A,
152A, 2112–2114.
Hutsler, J. J., Love, T., & Zhang, H. (2007). Histological and magnetic resonance imaging
assessment of cortical layering and thickness in autism spectrum disorders. Biological
Psychiatry, 61, 449–457.
Hutsler, J. J., & Zhang, H. (2010). Increased dendritic spine densities on cortical projection
neurons in autism spectrum disorders. Brain Research, 1309, 83–94.
Ingalhalikar, M., Parker, D., Bloy, L., Roberts, T. P., & Verma, R. (2011). Diffusion based
abnormality markers of pathology: Toward learned diagnostic prediction of ASD.
NeuroImage, 57, 918–927.
Iossifov, I., Ronemus, M., Levy, D., Wang, Z., Hakker, I., Rosenbaum, J., et al. (2012). De
novo gene disruptions in children on the autistic spectrum. Neuron, 74, 285–299.
Ip, B. K., Bayatti, N., Howard, N. J., Lindsay, S., & Clowry, G. J. (2011). The corticofugal
neuron-associated genes ROBO1, SRGAP1, and CTIP2 exhibit an anterior to posterior
gradient of expression in early fetal human neocortex development. Cerebral Cortex, 21,
1395–1407.
Irwin, S. A., Galvez, R., & Greenough, W. T. (2000). Dendritic spine structural anomalies in
fragile-X mental retardation syndrome. Cerebral Cortex, 10, 1038–1044.
Jessell, T. (2000). Neuronal specification in the spinal cord: Inductive signals and transcrip-
tional codes. Nature Reviews Genetics, 1, 20–29.
Jiang, Y. H., Yuen, R. K., Jin, X., Wang, M., Chen, N., Wu, X., et al. (2013). Detection of
clinically relevant genetic variants in autism spectrum disorder by whole-genome
sequencing. American Journal of Human Genetics, 93, 249–263.
Johnson, M. B., Kawasawa, Y. I., Mason, C. E., Krsnik, Z., Coppola, G., Bogdanovic, D.,
et al. (2009). Functional and evolutionary insights into human brain development
through global transcriptome analysis. Neuron, 62, 494–509.
Jones, E. G. (1986). Neurotransmitters in the cerebral cortex. Journal of Neurosurgery, 65,
135–153.
Joshi, P. S., Molyneaux, B. J., Feng, L., Xie, X., Macklis, J. D., & Gan, L. (2008). Bhlhb5
regulates the postmitotic acquisition of area identities in layers II-V of the developing
neocortex. Neuron, 60, 258–272.
Just, M. A., Keller, T. A., Malave, V. L., Kana, R. K., & Varma, S. (2012). Autism as a neural
systems disorder: A theory of frontal-posterior underconnectivity. Neuroscience and
Biobehavioral Reviews, 36, 1292–1313.
Kalscheuer, V. M., FitzPatrick, D., Tommerup, N., Bugge, M., Niebuhr, E.,
Neumann, L. M., et al. (2007). Mutations in autism susceptibility candidate 2
(AUTS2) in patients with mental retardation. Human Genetics, 121, 501–509.
Kang, H. J., Kawasawa, Y. I., Cheng, F., Zhu, Y., Xu, X., Li, M., et al. (2011). Spatio-
temporal transcriptome of the human brain. Nature, 478, 483–489.
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 199
Kelemenova, S., Schmidtova, E., Ficek, A., Celec, P., Kubranska, A., & Ostatnikova, D.
(2010). Polymorphisms of candidate genes in Slovak autistic patients. Psychiatric Genetics,
20, 137–139.
Keller, T. A., Kana, R. K., & Just, M. A. (2007). A developmental study of the structural
integrity of white matter in autism. Neuroreport, 18, 23–27.
Kerjan, G., & Gleeson, J. G. (2007). Genetic mechanisms underlying abnormal neuronal
migration in classical lissencephaly. Trends in Genetics, 23, 623–630.
Konopka, G., Bomar, J. M., Winden, K., Coppola, G., Jonsson, Z. O., Gao, F., et al. (2009).
Human-specific transcriptional regulation of CNS development genes by FOXP2.
Nature, 462, 213–217.
Konopka, G., Friedrich, T., Davis-Turak, J., Winden, K., Oldham, M. C., Gao, F., et al.
(2012). Human-specific transcriptional networks in the brain. Neuron, 75, 601–617.
Konopka, G., Wexler, E., Rosen, E., Mukamel, Z., Osborn, G. E., Chen, L., et al. (2012).
Modeling the functional genomics of autism using human neurons. Molecular Psychiatry,
17, 202–214.
Kostovic, I., & Rakic, P. (1980). Cytology and time of origin of interstitial neurons in the
white matter in infant and adult human and monkey telencephalon. Journal of
Neurocytology, 9, 219–242.
Kostovic, I., & Rakic, P. (1990). Developmental history of the transient subplate zone in the
visual and somatosensory cortex of the macaque monkey and human brain. The Journal of
Comparative Neurology, 297, 441–470.
Kowalczyk, T., Pontious, A., Englund, C., Daza, R. A., Bedogni, F., Hodge, R., et al.
(2009). Intermediate neuronal progenitors (basal progenitors) produce pyramidal-
projection neurons for all layers of cerebral cortex. Cerebral Cortex, 19, 2439–2450.
Krepischi, A. C., Knijnenburg, J., Bertola, D. R., Kim, C. A., Pearson, P. L., Bijlsma, E.,
et al. (2010). Two distinct regions in 2q24.2-q24.3 associated with idiopathic epilepsy.
Epilepsia, 51, 2457–2460.
Kumar, A., Sundaram, S. K., Sivaswamy, L., Behen, M. E., Makki, M. I., Ager, J., et al.
(2010). Alterations in frontal lobe tracts and corpus callosum in young children with
autism spectrum disorder. Cerebral Cortex, 20, 2103–2113.
Kwan, K. Y., Lam, M. M., Johnson, M. B., Dube, U., Shim, S., Rašin, M. R., et al. (2012).
Species-dependent posttranscriptional regulation of NOS1 by FMRP in the developing
cerebral cortex. Cell, 149, 899–911.
Kwan, K. Y., Lam, M. M., Krsnik, Z., Kawasawa, Y. I., Lefebvre, V., & Sestan, N. (2008).
SOX5 postmitotically regulates migration, postmigratory differentiation, and projections
of subplate and deep-layer neocortical neurons. Proceedings of the National Academy of
Sciences of the United States of America, 105, 16021–16026.
Kwan, K. Y., Sestan, N., & Anton, E. S. (2012). Transcriptional co-regulation of neuronal
migration and laminar identity in the neocortex. Development, 139, 1535–1546.
Lai, C. S., Fisher, S. E., Hurst, J. A., Vargha-Khadem, F., & Monaco, A. P. (2001).
A forkhead-domain gene is mutated in a severe speech and language disorder. Nature,
413, 519–523.
Lai, T., Jabaudon, D., Molyneaux, B., Azim, E., Arlotta, P., Menezes, J., et al. (2008). SOX5
controls the sequential generation of distinct corticofugal neuron subtypes. Neuron, 57,
232–247.
Lamb, A. N., Rosenfeld, J. A., Neill, N. J., Talkowski, M. E., Blumenthal, I., Girirajan, S.,
et al. (2012). Haploinsufficiency of SOX5 at 12p12.1 is associated with developmental
delays with prominent language delay, behavior problems, and mild dysmorphic features.
Human Mutation, 33, 728–740.
Lambert de Rouvroit, C., & Goffinet, A. M. (1998). A new view of early cortical develop-
ment. Biochemical Pharmacology, 56, 1403–1409.
200 Kenneth Y. Kwan
Lange, N., Dubray, M. B., Lee, J. E., Froimowitz, M. P., Froehlich, A., Adluru, N., et al.
(2010). Atypical diffusion tensor hemispheric asymmetry in autism. Autism Research, 3,
350–358.
Lee, J. E., Bigler, E. D., Alexander, A. L., Lazar, M., DuBray, M. B., Chung, M. K., et al.
(2007). Diffusion tensor imaging of white matter in the superior temporal gyrus and tem-
poral stem in autism. Neuroscience Letters, 424, 127–132.
Lefebvre, V., Dumitriu, B., Penzo-Méndez, A., Han, Y., & Pallavi, B. (2007). Control of cell
fate and differentiation by Sry-related high-mobility-group box (Sox) transcription fac-
tors. The International Journal of Biochemistry & Cell Biology, 39, 2195–2214.
Leone, D. P., Srinivasan, K., Chen, B., Alcamo, E., & McConnell, S. K. (2008). The deter-
mination of projection neuron identity in the developing cerebral cortex. Current Opin-
ion in Neurobiology, 18, 28–35.
Leoyklang, P., Suphapeetiporn, K., Siriwan, P., Desudchit, T., Chaowanapanja, P.,
Gahl, W. A., et al. (2007). Heterozygous nonsense mutation SATB2 associated with cleft
palate, osteoporosis, and cognitive defects. Human Mutation, 28, 732–738.
Li, H., Li, Y., Shao, J., Li, R., Qin, Y., Xie, C., et al. (2008). The association analysis of
RELN and GRM8 genes with autistic spectrum disorder in Chinese Han population.
American Journal of Medical Genetics Part B, Neuropsychiatric Genetics, 147B, 194–200.
Liu, J. S. (2011). Molecular genetics of neuronal migration disorders. Current Neurology and
Neuroscience Reports, 11, 171–178.
Liu, X., Novosedlik, N., Wang, A., Hudson, M. L., Cohen, I. L., Chudley, A. E., et al.
(2009). The DLX1and DLX2 genes and susceptibility to autism spectrum disorders.
European Journal of Human Genetics, 17, 228–235.
Lodato, S., Rouaux, C., Quast, K. B., Jantrachotechatchawan, C., Studer, M.,
Hensch, T. K., et al. (2011). Excitatory projection neuron subtypes control the distribu-
tion of local inhibitory interneurons in the cerebral cortex. Neuron, 69, 763–779.
Lu, H. Y., Cui, Y. X., Shi, Y. C., Xia, X. Y., Liang, Q., Yao, B., et al. (2009). A girl with
distinctive features of borderline high blood pressure, short stature, characteristic
brachydactyly, and 11.47 Mb deletion in 12p11.21-12p12.2 by oligonucleotide array
CGH. American Journal of Medical Genetics Part A, 149A, 2321–2323.
Lui, J. H., Hansen, D. V., & Kriegstein, A. R. (2011). Development and evolution of the
human neocortex. Cell, 146, 18–36.
MacDonald, J. L., Fame, R. M., Azim, E., Shnider, S. J., Molyneaux, B. J., Arlotta, P., et al.
(2013). Specification of cortical projection neurons. In J. Rubenstein & P. Rakic (Eds.),
Comprehensive developmental neuroscience: patterning and cell type specification in the developing
CNS and PNS (pp. 475–502). Amsterdam: Elsevier.
Magri, C., Piovani, G., Pilotta, A., Michele, T., Buzi, F., & Barlati, S. (2011). De novo dele-
tion of chromosome 2q24.2 region in a mentally retarded boy with muscular hypotonia.
European Journal of Medical Genetics, 54, 361–364.
Malatesta, P., Hack, M. A., Hartfuss, E., Kettenmann, H., Klinkert, W., Kirchhoff, F., et al.
(2003). Neuronal or glial progeny: Regional differences in radial glia fate. Neuron, 37,
751–764.
Mallamaci, A., & Stoykova, A. (2006). Gene networks controlling early cerebral cortex
arealization. The European Journal of Neuroscience, 23, 847–856.
Manzini, M. C., & Walsh, C. A. (2011). What disorders of cortical development tell us about
the cortex: One plus one does not always make two. Current Opinion in Genetics & Devel-
opment, 21, 333–339.
Marı́n, O., & Rubenstein, J. L. (2003). Cell migration in the forebrain. Annual Review of
Neuroscience, 26, 441–483.
Marin-Padilla, M. (1971). Early prenatal ontogenesis of the cerebral cortex (neocortex) of the
cat (Felis domestica). A Golgi study. I. The primordial neocortical organization.
Zeitschrift für Anatomie und Entwicklungsgeschichte, 134, 117–145.
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 201
Marin-Padilla, M. (1978). Dual origin of the mammalian neocortex and evolution of the cor-
tical plate. Anatomy and Embryology (Berlin), 152, 109–126.
Marı́n-Padilla, M. (1992). Ontogenesis of the pyramidal cell of the mammalian neocortex
and developmental cytoarchitectonics: A unifying theory. The Journal of Comparative Neu-
rology, 321, 223–240.
Markram, H., Toledo-Rodriguez, M., Wang, Y., Gupta, A., Silberberg, G., & Wu, C.
(2004). Interneurons of the neocortical inhibitory system. Nature Reviews Neuroscience,
5, 793–807.
Marshall, C. R., Noor, A., Vincent, J. B., Lionel, A. C., Feuk, L., Skaug, J., et al. (2008).
Structural variation of chromosomes in autism spectrum disorder. American Journal of
Human Genetics, 82, 477–488.
McKenna, W. L., Betancourt, J., Larkin, K. A., Abrams, B., Guo, C., Rubenstein, J. L., et al.
(2011). Tbr1 and Fezf2 regulate alternate corticofugal neuronal identities during neocor-
tical development. The Journal of Neuroscience, 31, 549–564.
Mefford, H. C., Muhle, H., Ostertag, P., von Spiczak, S., Buysse, K., Baker, C., et al. (2010).
Genome-wide copy number variation in epilepsy: Novel susceptibility loci in idiopathic
generalized and focal epilepsies. PLoS Genetics, 6, e1000962.
Michaelson, J. J., Shi, Y., Gujral, M., Zheng, H., Malhotra, D., Jin, X., et al. (2012). Whole-
genome sequencing in autism identifies hot spots for de novo germline mutation. Cell,
151, 1431–1442.
Miyata, T., Kawaguchi, A., Saito, K., Kawano, M., Muto, T., & Ogawa, M. (2004). Asym-
metric production of surface-dividing and non-surface-dividing cortical progenitor cells.
Development, 131, 3133–3145.
Miyoshi, G., Butt, S. J., Takebayashi, H., & Fishell, G. (2007). Physiologically distinct tem-
poral cohorts of cortical interneurons arise from telencephalic Olig2-expressing precur-
sors. The Journal of Neuroscience, 27, 7786–7798.
Miyoshi, G., & Fishell, G. (2011). GABAergic interneuron lineages selectively sort into spe-
cific cortical layers during early postnatal development. Cerebral Cortex, 21, 845–852.
Miyoshi, G., Hjerling-Leffler, J., Karayannis, T., Sousa, V. H., Butt, S. J., Battiste, J., et al.
(2010). Genetic fate mapping reveals that the caudal ganglionic eminence produces a
large and diverse population of superficial cortical interneurons. The Journal of Neurosci-
ence, 30, 1582–1594.
Molliver, M. E., Kostovic, I., & van der Loos, H. (1973). The development of synapses in
cerebral cortex of the human fetus. Brain Research, 50, 403–407.
Molyneaux, B., Arlotta, P., Hirata, T., Hibi, M., & Macklis, J. (2005). Fezl is required for the
birth and specification of corticospinal motor neurons. Neuron, 47, 817–831.
Molyneaux, B., Arlotta, P., Menezes, J., & Macklis, J. (2007). Neuronal subtype specification
in the cerebral cortex. Nature Reviews Neuroscience, 8, 427–437.
Monuki, E. S., & Walsh, C. A. (2001). Mechanisms of cerebral cortical patterning in mice
and humans. Nature Neuroscience, 4(Suppl.), 1199–1206.
Mountcastle, V. B. (1997). The columnar organization of the neocortex. Brain, 120(Pt. 4),
701–722.
Nadarajah, B., & Parnavelas, J. G. (2002). Modes of neuronal migration in the developing
cerebral cortex. Nature Reviews Neuroscience, 3, 423–432.
Nagae, L. M., Zarnow, D. M., Blaskey, L., Dell, J., Khan, S. Y., Qasmieh, S., et al. (2012).
Elevated mean diffusivity in the left hemisphere superior longitudinal fasciculus in autism
spectrum disorders increases with more profound language impairment. AJNR—
American Journal of Neuroradiology, 33, 1720–1725.
Nagai, T., Nishimura, G., Kato, R., Hasegawa, T., Ohashi, H., & Fukushima, Y. (1995).
Del(12)(p11.21p12.2) associated with an asphyxiating thoracic dystrophy or cho-
ndroectodermal dysplasia-like syndrome. American Journal of Medical Genetics, 55,
16–18.
202 Kenneth Y. Kwan
Nakashima, N., Yamagata, T., Mori, M., Kuwajima, M., Suwa, K., & Momoi, M. Y. (2010).
Expression analysis and mutation detection of DLX5 and DLX6 in autism. Brain & Develop-
ment, 32, 98–104.
Neale, B. M., Kou, Y., Liu, L., Ma’ayan, A., Samocha, K. E., Sabo, A., et al. (2012). Patterns
and rates of exonic de novo mutations in autism spectrum disorders. Nature, 485,
242–245.
Nieuwenhuys, R. (1994). The neocortex. An overview of its evolutionary development,
structural organization and synaptology. Anatomy and Embryology (Berlin), 190, 307–337.
Noctor, S. C., Martinez-Cerdeno, V., Ivic, L., & Kriegstein, A. R. (2004). Cortical neurons
arise in symmetric and asymmetric division zones and migrate through specific phases.
Nature Neuroscience, 7, 136–144.
Noctor, S. C., Martı́nez-Cerdeño, V., & Kriegstein, A. R. (2008). Distinct behaviors of neu-
ral stem and progenitor cells underlie cortical neurogenesis. The Journal of Comparative
Neurology, 508, 28–44.
Northcutt, R. G., & Kaas, J. H. (1995). The emergence and evolution of mammalian neo-
cortex. Trends in Neurosciences, 18, 373–379.
Oldham, M. C., Konopka, G., Iwamoto, K., Langfelder, P., Kato, T., Horvath, S., et al.
(2008). Functional organization of the transcriptome in human brain. Nature Neuroscience,
11, 1271–1282.
O’Leary, D. D., & Koester, S. E. (1993). Development of projection neuron types, axon
pathways, and patterned connections of the mammalian cortex. Neuron, 10, 991–1006.
O’Leary, D. D., & Sahara, S. (2008). Genetic regulation of arealization of the neocortex. Cur-
rent Opinion in Neurobiology, 18, 90–100.
O’Roak, B. J., Deriziotis, P., Lee, C., Vives, L., Schwartz, J. J., Girirajan, S., et al. (2011).
Exome sequencing in sporadic autism spectrum disorders identifies severe de novo muta-
tions. Nature Genetics, 43, 585–589.
O’Roak, B. J., Vives, L., Fu, W., Egertson, J. D., Stanaway, I. B., Phelps, I. G., et al. (2012).
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum
disorders. Science, 338, 1619–1622.
O’Roak, B. J., Vives, L., Girirajan, S., Karakoc, E., Krumm, N., Coe, B. P., et al. (2012).
Sporadic autism exomes reveal a highly interconnected protein network of de novo
mutations. Nature, 485, 246–250.
Palumbo, O., D’Agruma, L., Minenna, A. F., Palumbo, P., Stallone, R., Palladino, T., et al.
(2013). 3p14.1 de novo microdeletion involving the FOXP1 gene in an adult patient
with autism, severe speech delay and deficit of motor coordination. Gene, 516, 107–113.
Palumbo, O., Palumbo, P., Palladino, T., Stallone, R., Zelante, L., & Carella, M. (2012).
A novel deletion in 2q24.1q24.2 in a girl with mental retardation and generalized hypo-
tonia: A case report. Molecular Cytogenetics, 5, 1.
Peñagarikano, O., Abrahams, B. S., Herman, E. I., Winden, K. D., Gdalyahu, A., Dong, H.,
et al. (2011). Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities,
and core autism-related deficits. Cell, 147, 235–246.
Persico, A. M., D’Agruma, L., Maiorano, N., Totaro, A., Militerni, R., Bravaccio, C., et al.
(2001). Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder.
Molecular Psychiatry, 6, 150–159.
Peters, A. (2010). The morphology of minicolumns. In G. Blatt (Ed.), The neurochemical basis
of autism (pp. 45–68). Berlin: Springer.
Polleux, F., Ince-Dunn, G., & Ghosh, A. (2007). Transcriptional regulation of vertebrate
axon guidance and synapse formation. Nature Reviews Neuroscience, 8, 331–340.
Prasad, A., Merico, D., Thiruvahindrapuram, B., Wei, J., Lionel, A. C., Sato, D., et al.
(2012). A discovery resource of rare copy number variations in individuals with autism
spectrum disorder. G3 (Bethesda), 2, 1665–1685.
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 203
Rakic, P. (1971). Guidance of neurons migrating to the fetal monkey neocortex. Brain
Research, 33, 471–476.
Rakic, P. (1974). Neurons in rhesus monkey visual cortex: Systematic relation between time
of origin and eventual disposition. Science, 183, 425–427.
Rakic, P. (1976). Prenatal genesis of connections subserving ocular dominance in the rhesus
monkey. Nature, 261, 467–471.
Rakic, P. (1988). Specification of cerebral cortical areas. Science, 241, 170–176.
Rakic, P. (1995). A small step for the cell, a giant leap for mankind: A hypothesis of neocor-
tical expansion during evolution. Trends in Neurosciences, 18, 383–388.
Rash, B., & Grove, E. (2006). Area and layer patterning in the developing cerebral cortex.
Current Opinion in Neurobiology, 16, 25–34.
Rauch, A., Wieczorek, D., Graf, E., Wieland, T., Endele, S., Schwarzmayr, T., et al. (2012).
Range of genetic mutations associated with severe non-syndromic sporadic intellectual
disability: An exome sequencing study. Lancet, 380, 1674–1682.
Rice, D., & Curran, T. (2001). Role of the reelin signaling pathway in central nervous system
development. Annual Review of Neuroscience, 24, 1005–1039.
Rosenfeld, J. A., Ballif, B. C., Lucas, A., Spence, E. J., Powell, C., Aylsworth, A. S., et al.
(2009). Small deletions of SATB2 cause some of the clinical features of the 2q33.1 micro-
deletion syndrome. PLoS One, 4, e6568.
Rosenfeld, J. A., Ballif, B. C., Torchia, B. S., Sahoo, T., Ravnan, J. B., Schultz, R., et al.
(2010). Copy number variations associated with autism spectrum disorders contribute to
a spectrum of neurodevelopmental disorders. Genetics in Medicine, 12, 694–702.
Rouaux, C., & Arlotta, P. (2013). Direct lineage reprogramming of post-mitotic callosal
neurons into corticofugal neurons in vivo. Nature Cell Biology, 15, 214–221.
Rubenstein, J. L. (2011). Annual research review: Development of the cerebral cortex:
Implications for neurodevelopmental disorders. Journal of Child Psychology and Psychiatry,
52, 339–355.
Rubenstein, J. L., & Merzenich, M. M. (2003). Model of autism: Increased ratio of
excitation/inhibition in key neural systems. Genes, Brain, and Behavior, 2, 255–267.
Saito, T., Hanai, S., Takashima, S., Nakagawa, E., Okazaki, S., Inoue, T., et al. (2011). Neo-
cortical layer formation of human developing brains and lissencephalies: Consideration
of layer-specific marker expression. Cerebral Cortex, 21, 588–596.
Sanders, S. J., Ercan-Sencicek, A. G., Hus, V., Luo, R., Murtha, M. T., Moreno-De-
Luca, D., et al. (2011). Multiple recurrent de novo CNVs, including duplications of
the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron,
70, 863–885.
Sanders, S. J., Murtha, M. T., Gupta, A. R., Murdoch, J. D., Raubeson, M. J., Willsey, A. J.,
et al. (2012). De novo mutations revealed by whole-exome sequencing are strongly asso-
ciated with autism. Nature, 485, 237–241.
Schaaf, C. P., Sabo, A., Sakai, Y., Crosby, J., Muzny, D., Hawes, A., et al. (2011). Oligogenic
heterozygosity in individuals with high-functioning autism spectrum disorders. Human
Molecular Genetics, 20, 3366–3375.
Schanze, I., Schanze, D., Bacino, C. A., Douzgou, S., Kerr, B., & Zenker, M. (2013).
Haploinsufficiency of SOX5, a member of the SOX (SRY-related HMG-box) family
of transcription factors is a cause of intellectual disability. European Journal of Medical
Genetics, 56, 108–113.
Sebat, J., Lakshmi, B., Malhotra, D., Troge, J., Lese-Martin, C., Walsh, T., et al. (2007).
Strong association of de novo copy number mutations with autism. Science, 316,
445–449.
Serajee, F. J., Zhong, H., & Mahbubul Huq, A. H. (2006). Association of Reelin gene poly-
morphisms with autism. Genomics, 87, 75–83.
204 Kenneth Y. Kwan
Sessa, A., Mao, C. A., Colasante, G., Nini, A., Klein, W. H., & Broccoli, V. (2010). Tbr2-
positive intermediate (basal) neuronal progenitors safeguard cerebral cortex expansion by
controlling amplification of pallial glutamatergic neurons and attraction of subpallial
GABAergic interneurons. Genes & Development, 24, 1816–1826.
Shim, S., Kwan, K. Y., Li, M., Lefebvre, V., & Sestan, N. (2012). Cis-regulatory control of
corticospinal system development and evolution. Nature, 486, 74–79.
Shukla, D. K., Keehn, B., Lincoln, A. J., & Müller, R. A. (2010). White matter compromise
of callosal and subcortical fiber tracts in children with autism spectrum disorder:
A diffusion tensor imaging study. Journal of the American Academy of Child and Adolescent
Psychiatry, 49, 1269–1278, 1278.e1261–1278.e1262.
Skaar, D. A., Shao, Y., Haines, J. L., Stenger, J. E., Jaworski, J., Martin, E. R., et al. (2005).
Analysis of the RELN gene as a genetic risk factor for autism. Molecular Psychiatry, 10,
563–571.
Smoller, J. W., Craddock, N., Kendler, K., Lee, P. H., Neale, B. M., Nurnberger, J. I., et al.
(2013). Identification of risk loci with shared effects on five major psychiatric disorders:
A genome-wide analysis. Lancet, 381, 1371–1379.
Srinivasan, K., Leone, D. P., Bateson, R. K., Dobreva, G., Kohwi, Y., Kohwi-
Shigematsu, T., et al. (2012). A network of genetic repression and derepression specifies
projection fates in the developing neocortex. Proceedings of the National Academy of Sciences
of the United States of America, 109, 19071–19078.
State, M. W., & Šestan, N. (2012). Neuroscience. The emerging biology of autism spectrum
disorders. Science, 337, 1301–1303.
Strauss, K. A., Puffenberger, E. G., Huentelman, M. J., Gottlieb, S., Dobrin, S. E.,
Parod, J. M., et al. (2006). Recessive symptomatic focal epilepsy and mutant
contactin-associated protein-like 2. The New England Journal of Medicine, 354,
1370–1377.
Stumm, M., Klopocki, E., Gasiorek-Wiens, A., Knoll, U., Wirjadi, D., Sarioglu, N., et al.
(2007). Molecular cytogenetic characterisation of an interstitial deletion 12p detected by
prenatal diagnosis. Prenatal Diagnosis, 27, 475–478.
Sultana, R., Yu, C. E., Yu, J., Munson, J., Chen, D., Hua, W., et al. (2002). Identification of
a novel gene on chromosome 7q11.2 interrupted by a translocation breakpoint in a pair
of autistic twins. Genomics, 80, 129–134.
Sundaram, S. K., Kumar, A., Makki, M. I., Behen, M. E., Chugani, H. T., & Chugani, D. C.
(2008). Diffusion tensor imaging of frontal lobe in autism spectrum disorder. Cerebral
Cortex, 18, 2659–2665.
Tabata, H., Kanatani, S., & Nakajima, K. (2009). Differences of migratory behavior between
direct progeny of apical progenitors and basal progenitors in the developing cerebral cor-
tex. Cerebral Cortex, 19, 2092–2105.
Takatsuki, S., Nakamura, R., Haga, Y., Mitsui, K., Hashimoto, T., Shimojima, K., et al.
(2010). Severe pulmonary emphysema in a girl with interstitial deletion of
2q24.2q24.3 including ITGB6. American Journal of Medical Genetics Part A, 152A,
1020–1025.
Talkowski, M. E., Rosenfeld, J. A., Blumenthal, I., Pillalamarri, V., Chiang, C., Heilbut, A.,
et al. (2012). Sequencing chromosomal abnormalities reveals neurodevelopmental loci
that confer risk across diagnostic boundaries. Cell, 149, 525–537.
Tarabykin, V., Stoykova, A., Usman, N., & Gruss, P. (2001). Cortical upper layer neurons
derive from the subventricular zone as indicated by Svet1 gene expression. Development,
128, 1983–1993.
The Boulder Committee (1970). Embryonic vertebrate central nervous system: Revised ter-
minology. The Boulder Committee. The Anatomical Record, 166, 257–261.
Thornton, G. K., & Woods, C. G. (2009). Primary microcephaly: Do all roads lead to Rome?
Trends in Genetics, 25, 501–510.
Transcriptional Dysregulation of Neocortical Circuit Assembly in ASD 205
Tissir, F., & Goffinet, A. M. (2003). Reelin and brain development. Nature Reviews Neuro-
science, 4, 496–505.
Traylor, R. N., Dobyns, W. B., Rosenfeld, J. A., Wheeler, P., Spence, J. E.,
Bandholz, A. M., et al. (2012). Investigation of TBR1 hemizygosity: Four individuals
with 2q24 microdeletions. Molecular Syndromology, 3, 102–112.
Uppal, N., & Hof, P. R. (2013). Discrete cortical neuropathology in autism spectrum dis-
orders. In J. D. Buxbaum (Ed.), The neuroscience of autism spectrum disorders (pp. 313–325).
San Diego: Academic Press.
Van Buggenhout, G., Van Ravenswaaij-Arts, C., Mc Maas, N., Thoelen, R., Vogels, A.,
Smeets, D., et al. (2005). The del(2)(q32.2q33) deletion syndrome defined by clinical and
molecular characterization of four patients. European Journal of Medical Genetics, 48,
276–289.
Valiente, M., & Marı́n, O. (2010). Neuronal migration mechanisms in development and
disease. Current Opinion in Neurobiology, 20, 68–78.
Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner, J. T., Abrahams, B. S., et al. (2009).
Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature,
459, 528–533.
Wegiel, J., Kuchna, I., Nowicki, K., Imaki, H., Marchi, E., Ma, S. Y., et al. (2010). The
neuropathology of autism: Defects of neurogenesis and neuronal migration, and dysplas-
tic changes. Acta Neuropathologica, 119, 755–770.
Weinstein, M., Ben-Sira, L., Levy, Y., Zachor, D. A., Ben Itzhak, E., Artzi, M., et al. (2011).
Abnormal white matter integrity in young children with autism. Human Brain Mapping,
32, 534–543.
Weiss, L. A., Escayg, A., Kearney, J. A., Trudeau, M., MacDonald, B. T., Mori, M., et al.
(2003). Sodium channels SCN1A, SCN2A and SCN3A in familial autism. Molecular
Psychiatry, 8, 186–194.
Wolff, J. J., Gu, H., Gerig, G., Elison, J. T., Styner, M., Gouttard, S., et al. (2012). Differ-
ences in white matter fiber tract development present from 6 to 24 months in infants with
autism. The American Journal of Psychiatry, 169, 589–600.
Wonders, C. P., & Anderson, S. A. (2006). The origin and specification of cortical interneu-
rons. Nature Reviews Neuroscience, 7, 687–696.
Xu, Q., Cobos, I., De La Cruz, E., Rubenstein, J. L., & Anderson, S. A. (2004). Origins of
cortical interneuron subtypes. The Journal of Neuroscience, 24, 2612–2622.
Yokota, Y., Gashghaei, H. T., Han, C., Watson, H., Campbell, K. J., & Anton, E. S. (2007).
Radial glial dependent and independent dynamics of interneuronal migration in the
developing cerebral cortex. PLoS One, 2, e794.
Zhang, L., Song, N. N., Chen, J. Y., Huang, Y., Li, H., & Ding, Y. Q. (2011). Satb2 is
required for dendritic arborization and soma spacing in mouse cerebral cortex. Cerebral
Cortex, 22(7), 1510–1519.
CHAPTER SEVEN
Contents
1. Why Study Motor Skill in Autism? 208
1.1 Motor skill as foundational in development 209
1.2 Early signs are motor 210
1.3 Correlation with social and communication skill 211
2. Overview of Motor Skill Deficits 212
2.1 Gross motor skills 212
2.2 Fine motor skills 215
2.3 Dyspraxia 216
2.4 Eye movements 217
3. Mapping Autism Motor Skill Findings into a Useful Framework 218
3.1 Cortico–subcortical loops for motor control 218
3.2 How do autism motor skill deficits fit in this framework? 225
4. Can We Intervene? 228
4.1 Motor skill training in rodents 230
4.2 Motor training for children with ASD? 232
4.3 Motor training for older adults with ASD? 233
5. Summary 234
References 235
Abstract
The earliest observable symptoms of autism spectrum disorders (ASDs) involve motor
behavior. There is a growing awareness of the developmental importance of impaired
motor function in ASD and its association with social skill. Compromised motor function
requires increased attention, leaving fewer resources available for processing environ-
mental stimuli and learning. This knowledge suggests that the motor system—which
we know to be trainable—may be a gateway to improving outcomes of individuals liv-
ing with ASD. In this review, we suggest a framework borrowed from machine learning
to examine where, why, and how motor skills are different in individuals with ASD.
frequently observed include motor skill delays, deficits, and atypical move-
ment patterns (Fournier, Hass, et al., 2010; Jansiewicz et al., 2006; Maski,
Jeste, & Spence, 2011). Children with ASD showed greater impairments
in motor skill on a standardized motor testing battery when compared to
children diagnosed with other neurodevelopmental disorders (Dewey,
Cantell, & Crawford, 2007). The children with autism were also the only
group to show impairments in gestural skills. An insightful recent study of
Hilton and Constantino (Hilton et al., 2012) examined the performance
of 83 sibling pairs, some concordant and some discordant for autism, using
a comprehensive and standardized motor skill battery and found that motor
performance was strongly correlated with the diagnostic status but not with
sibship. The authors argue that given the highly heritable nature of ASD,
this finding suggests that motor measures should factor into the diagnosis
of autism.
Fahey, Rawicki, & Boyd, 2010). Taken together, these studies suggest that
one might consider atypical motor development as a critical factor in the
further development of symptoms that characterize autism in children aged
two and older.
recently, Landa and colleagues (Flanagan, Landa, Bhat, & Bauman, 2012)
reported that 6-month-olds at high risk for ASD had more head lag when
pulled up to a seated position. Both Ozonoff et al. (2010) and Landa and
Garrett-Mayer (2006) observed an increasing trend toward atypicality
between the first and second year, with gross and fine motor differences
in the high-risk ASD siblings emerging in one study (Landa & Garrett-
Mayer, 2006) that measured them. High- and low-risk infants were not sta-
tistically distinguishable by motor skill differences at 6 months, but over
the following 8 months, low- and high-risk groups became more distinct.
At 24 months, the ASD-diagnosed group also differed in motor skill from
children who were diagnosed with language delay.
It also appears that early motor skill development is a strong predictor of
ASD outcome in later childhood. Sutera et al. (2007) reported that motor
skills at 2 years old were the best predictors of outcome in ASD at 8 years
old. This is an important research that needs further study, as there are very
few solid predictors of outcome in ASD.
2.1.1 Balance
Balance deficits have been measured using scientific grade force plates across
a wide age range of individuals with ASD and normal intelligence (Chang,
Wade, Stoffregen, Hsu, & Pan, 2010; Fournier, Kimberg, et al., 2010;
Kohen-Raz, Volkmar, & Cohen, 1992; Minshew, Sung, Jones, &
Furman, 2004; Molloy, Dietrich, & Bhattacharya, 2003). In an early study
from Minshew et al. (2004), the impairments were most pronounced in con-
ditions that perturbed either visual (eyes closed or sway-referenced sur-
roundings) or somatosensory stimulation (sway-referenced platform).
Recently, Travers et al. (2013) demonstrated impaired balance in adoles-
cents and adults with ASD using the low-cost Wii Balance Board
(WBB), which has been favorably evaluated with respect to the scientific
grade force plate (Clark et al., 2010; Huurnink, Fransz, Kingma, & van
Dieen, 2013), had excellent test–retest reliability, and performed better than
the widely used Balance Error Scoring System (Chang, Levy, Seay, & Goble,
2013). Here too, individuals were found to have impaired balance under
“challenged” conditions, in this case while standing on one leg.
2.1.2 Gait
Static and dynamic balance skills are essential to functional gait performance.
Balance skill reflects sensorimotor status through the complex integration of
sensory feedback and coordinated motor responses to keep one’s center of mass
over the body’s base of support. Several studies of gait have reported atypical
gait in individuals with ASD. Qualitative evaluations have revealed a lack of
smoothness or overall coordination, atypical trunk and arm postures, or asym-
metrical gait (Esposito, Venuti, Apicella, & Muratori, 2011; Hallett et al., 1993;
Rinehart et al., 2006; Shetreat-Klein, Shinnar, & Rapin, 2012). Quantitative
evaluations have confirmed the lack of smoothness and irregular trunk move-
ments and in addition have shown significant differences in other spatial and
temporal gait parameters (Nobile et al., 2011; Vernazza-Martin et al., 2005;
Vilensky, Damasio, & Maurer, 1981; Weiss et al., 2013). While the specific gait
parameters identified as abnormal vary from study to study, shorter step/stride
length in ASD is a common finding. Atypical gait might result directly from
difficulties in balance (Lajoie, Teasdale, Bard, & Fleury, 1993), although hypo-
tonia may also play a role (Calhoun, Longworth, & Chester, 2011).
2.1.3 Reaching
There are relatively few studies of true reaching behavior in ASD; however,
several findings bear on reaching. Children with ASD appear to be slower in
214 Leanne Chukoskie et al.
2.3. Dyspraxia
The term dyspraxia (or apraxia for the more severe form) refers to difficulty
in organizing, planning, or executing skilled movement, which impairs
movement fluidity and speed, and importantly is out of proportion to
any underlying motor deficits. Not surprisingly, this has been difficult to
assess clearly in ASD because the contribution of “underlying motor def-
icits” remains to be concretely characterized. What is that “something
extra” that is dyspraxia? Several batteries have been used to measure
impairment with the “conceptualization” of the movement (Weimer,
Schatz, Lincoln, Ballantyne, & Trauner, 2001). These batteries typically
include subtests to assess spatial orientation, movement selection and
sequencing, imitation, spontaneous tool use, limb kinetics, and oral–facial
skills. These batteries emphasize learned, purposeful movements that are
often performed as a sequence, and it is perhaps the motor sequence, which
is “something extra.” Using one of these test batteries, Mostofsky and col-
leagues reported lower levels of simple motor skill in addition to dyspraxia
(Dowell, Mahone, & Mostofsky, 2009). This suggests that there may be
deficits in both simple motor skill and the “something extra” that is
dyspraxia. Oral–motor dyspraxia such as difficulty in imitating mouth
shapes and noises or spontaneously producing them by name is relatively
common, and perhaps, it includes the failure of the incredibly complex
machinery of the mouth and tongue to engage both sequentially and in
concert in order to produce comprehensible speech (Belmonte et al.,
2013; Larson & Mostofsky, 2008). It is intriguing to consider how a deeper
understanding of oral–motor praxis could be translated to interventions for
those that are minimally verbal.
Motor Skill in Autism Spectrum Disorders 217
There are many ways to mark eye movement latency and doing so can be
difficult in atypical eye movements. Since pursuit and saccade latency deficits
were expected given the other pattern of deficits and saccade latency delays
were observed elsewhere, it would be useful to see a replication of this result.
Figure 7.1 Regions of the brain with substantial contributions to motor skill and the
computations believed to be central to each region. Reproduced with permission from
Doya (2000).
220 Leanne Chukoskie et al.
1994; Hashimoto et al., 1995; Kates et al., 1998; Kaufmann et al., 2003;
Levitt et al., 1999; Saitoh, Courchesne, Egaas, Lincoln, & Schreibman,
1995; Zilbovicius et al., 1995) or hemispheres (Courchesne et al., 2001)
or in overall cerebellar gray matter (McAlonan et al., 2002). In some few
cases, cerebellar size reduction is so substantial as to be detected by visual
inspection (Miles & Hillman, 2000). Some studies have associated anatom-
ical abnormality with severity of symptoms (Ecker et al., 2012; O’Halloran,
Kinsella, & Storey, 2012). Functional imaging studies of the cerebellum
reveal an unfortunate mix of results—increases, decreases, and unchanged
activation in children with ASD. These discrepancies could emerge from
methodological differences, age-related differences between typical and
neurodevelopmentally delayed populations, and artifact-creating movement
during scanning (Brown & Jernigan, 2012). Looking at motor production
during a finger-tapping task that was designed to be cognitively simple,
thereby isolating movement, has revealed an increase in activity in the ipsi-
lateral anterior cerebellum along with cerebellar regions not typically rec-
ruited for finger tapping (Allen, Muller, & Courchesne, 2004). However,
during another finger-tapping task, Mostofsky and colleagues showed
decreased ipsilateral anterior cerebellar activity and increased supplementary
motor cortical activity in children with high-functioning autism (Mostofsky
et al., 2009).
Which system should be engaged for a given scenario? Daw, Niv, and Dayan
(2005) proposed a method for choosing which action system is the best by
minimizing the uncertainty of predictions produced by the goal or habit
systems.
typically developing children and also typical adults (Di Martino et al.,
2011) and consistently found atypically increased resting-state connectivity
in children with ASD. Interestingly, a study of connectivity in children,
who had ASD, ADHD, or both, found that the children diagnosed with
both had dysfunctional striatal circuitry, but not the children diagnosed
with ASD alone (Di Martino et al., 2013). This result suggests that comor-
bid symptoms are likely contributing to the heterogeneity of responses
observed in ASD. Fractional anisotropy (FA) of white matter tracts con-
necting the putamen with the frontal cortex was lower in adults with
autism (Langen et al., 2012). Performance on a go/no-go task was corre-
lated with FA in this white matter tract, suggesting that the inhibitory con-
trol needed for a go/no-go task is at least partially mediated by this tract.
A recent report showed dramatically decreased task-based connectivity
from reward-related regions of the basal ganglia to the posterior superior
temporal sulcus, a region believed to be involved in speech sound percep-
tion (Abrams et al., 2013).
Both basal ganglia volume and connectivity measures have been com-
pared with motor performance, again with mixed findings. Hardan,
Kilpatrick, Keshavan, and Minshew (2003) examined motor performance
with the grooved pegboard task, grip strength, and finger tapping. The
authors aimed to compare motor deficits observed in ASD to basal ganglia
volume estimates, but found no volume differences between the children
with ASD and typically developing children. Takarae and colleagues
showed increased bilateral activation of frontostriatal circuitry during a visu-
ally guided saccade task (Takarae, Minshew, Luna, & Sweeney, 2007).
Mostofsky and colleagues (Qiu et al., 2010) reported that the shape of the
basal ganglia is predictive of motor dysfunction using the PANESS assess-
ment battery and also social and communication skills. A correlation
between repetitive behaviors and the volume of the right caudate and total
putamen was reported (Dichter, 2012; Estes et al., 2011; Hollander
et al., 2005).
which they both looped. However, we now know that the basal ganglia and
the cerebellum are reciprocally connected through disynaptic subcortical
connections (Bostan et al., 2013). These newly identified pathways recipro-
cally connect motor and associative regions of the cerebellum and basal
ganglia. Importantly, connections between the basal ganglia and cerebellum
bring “reinforcement learning” machinery together with “supervised learn-
ing” machinery.
What benefits would such a connection bring? There is not a precise
answer to that question, but optimal control theory offers a useful perspec-
tive. Todorov and Jordan have argued that we make movements to reach a
more rewarding state (Todorov & Jordan, 2002). Optimal control theory
describes a formal way to link motor costs, expected rewards, noise from
sensory feedback, and internal models of a movement. In this framework,
the basal ganglia and cerebellum are both engaged in feedback loops to opti-
mize motor control. The basal ganglia calculate expected costs of motor
commands and expected rewards of sensory feedback, whereas the cerebel-
lum, through internal models, predicts the visual and proprioceptive feed-
back expected as a consequence of a particular motor command. Through
feedback, both of these areas contribute to the refinement of future motor
commands, which would be essential in learning a motor skill and especially
a sequence of motor skills.
with prism goggles and a novel-reaching task with forces imposed via a
robotic arm (Gidley Larson, Bastian, Donchin, Shadmehr, & Mostofsky,
2008), children with ASD learned quickly and exhibited typical aftereffects,
suggesting that a failure to update the internal model is not the source of
motor skill deficits in autism. However, recent data suggest that this is indeed
an area of concern in individuals with ASD, at least where eye movements
are concerned. In a recent saccade adaptation experiments in individuals
with ASD (Mosconi et al., 2013), approximately 30% of the subjects with
ASD did not adapt at all (compared with 6% of control subjects). Those indi-
viduals with ASD who did adapt did so more slowly and also showed
increased trial-to-trial variability in saccade amplitudes. Another recent sac-
cade adaptation experiment revealed similar results but notably also exam-
ined adaptation of children with Asperger’s syndrome ( Johnson, Rinehart,
White, Millist, & Fielding, 2013). The authors found weaker adaptation in
children with autism and Asperger’s syndrome compared to typically devel-
oped children. These results indicate deficits in the learning mechanisms of
the oculomotor vermis; however, the increased amplitude variability could
result in “noisy” information feeding into the calculated error signal
(Havermann & Lappe, 2010).
discount rate for children nicely matches their faster saccades for any given
amplitude. Similar data exist for individuals with schizophrenia (who also
have a higher temporal discount rate (Shadmehr et al., 2010). However,
we still lack data that span a range of directions and amplitudes in a matched
sample of individuals with ASD and typically developed individuals.
Some regions of the basal ganglia are more specialized for “goals” versus
“habits” (Redgrave et al., 2010). By examining the activity of particular
regions of the basal ganglia with respect to each other, one might learn
whether individuals with ASD have a biased use of basal ganglia circuitry,
for example, they may be more goal-directed in their actions versus habitual.
Referring back to Daw and colleagues’ perspective on using uncertainty to
choose which action system to use (Daw et al., 2005), it is possible that goal-
directed or habitual actions might be noisier in individuals with ASD, bias-
ing the choice toward the more reliable system.
4. CAN WE INTERVENE?
What happens in the brain during motor skill learning? This question
can be asked at many levels, including synaptic and subsynaptic, functional
motor maps and activity, and at a range of temporal scales. Figure 7.2 rep-
resents a summary of motor skill training effects modifying the efficacy of
synapses in the cerebellum and basal ganglia based on animal research
findings.
Motor Skill in Autism Spectrum Disorders 229
A
Climbing
Parallel fibers
fiber
Purkinje cells
B
Pyramidal cells
Cortex
Striatum
MSNs
Interneuron
Figure 7.2 (A) Summary of acrobatic motor skill training-induced plasticity in the cerebel-
lum. Yellow stars indicate putative sites for training-induced synaptic strengthening. Par-
allel fiber (red) to Purkinje cell (blue) synapses were significantly increased, while climbing
fiber (yellow) synapses showed a trend toward increasing. (B) Stars indicate putative sites of
training-induced plasticity in the basal ganglia. Medium spiny neurons of the direct (green)
and indirect (blue) pathways are depicted along with descending input from cortical pyra-
midal cells (black, purple) and striatal interneurons (yellow). Adapted with permission from
Kreitzer and Malenka (2008).
230 Leanne Chukoskie et al.
and between direct and indirect pathway MSNs (see Fig. 7.2B). These phys-
iological measures of synaptic plasticity echo the structural changes reported
in the cerebellum and motor cortex during acrobatic training. This is still a
rather active area of research in which the recent development of
optogenetic techniques applied to the basal ganglia (Kravitz et al., 2010)
is especially promising for unifying our understanding of the changes motor
learning creates across different brain regions.
Training motor skills may have additional benefits beyond those specific
to motor function. Recent findings in typically developing infants show that
early infant milestones such as learning to reach are foundational for later
social skill components such as social gaze preference (Libertus &
Needham, 2011). Coordination and speed of movement correlate positively
with cognitive measures in typical preschool aged boys and girls (Planinsec,
2002). Studies of DCD, in which there is substantial overlap with ASD
(Kopp, Beckung, & Gillberg, 2010; Piek & Dyck, 2004), show poorer social
skills in individuals with greater motor deficits. A recent paper also reveals
that poorer balance skills are associated with increased repetitive behaviors
(Radonovich, Fournier, & Hass, 2013). These data suggest that training-
induced improvements in a foundational motor skill, such as balance, might
have positive effects on other autism symptoms.
5. SUMMARY
This review of data on motor skills with anatomical and connectivity
differences in individuals with ASD places us in a position to consider what
questions, if answered, would take us to the next step in understanding the
motor skill in ASD. The questions in Box 7.1 are the proverbial tip of the
iceberg, but attempting to answer them will take us to the next stage of our
understanding.
Despite the increased interest in motor skills in autism, most of the
reports described earlier contain primarily descriptive data. If we are to
use motor skill differences to understand more about the nervous system
in autism, we need more quantitative and computational characterizations
of the motor control differences in individuals with ASD. In this review,
we have examined the data from the perspective of subcortical motor
centers—the cerebellum and basal ganglia and their cortical loops—as a
starting point for understanding the acquisition and performance of skilled
movements in ASD. We have also pointed to areas of movement research
in typically developing individuals that might be beneficial to consider for
understanding motor skill in ASD. By thinking about the types of compu-
tational roles performed by the different motor regions, we can design
future motor skill experiments that probe these roles explicitly and,
through greater understanding of motor skill deficits, design effective
interventions.
BOX 7.1 Open questions about motor skill in individuals with ASD
What does the developmental trajectory of motor skill look like in ASD?
Where are the delays?
Where are the deficits?
How reliable is the sensory and corollary discharge feedback in
individuals ASD?
Is there a bias for goal-directed or habitual movements in individuals
with ASD?
What differences exist in terms of motor costs and sensory rewards in ASD
compared with typical development?
Motor Skill in Autism Spectrum Disorders 235
REFERENCES
Abrahams, B. S., & Geschwind, D. H. (2010). Connecting genes to brain in the autism spec-
trum disorders. Archives of Neurology, 67(4), 395–399. http://dx.doi.org/10.1001/
archneurol.2010.47.
Abrams, D. A., Lynch, C. J., Cheng, K. M., Phillips, J., Supekar, K., Ryali, S., et al. (2013).
Underconnectivity between voice-selective cortex and reward circuitry in children with
autism. Proceedings of the National Academy of Sciences of the United States of America, 110(29),
12060–12065. http://dx.doi.org/10.1073/pnas.1302982110.
Allen, G., Buxton, R. B., Wong, E. C., & Courchesne, E. (1997). Attentional activation of
the cerebellum independent of motor involvement. Science, 275(5308), 1940–1943.
Allen, G., Muller, R. A., & Courchesne, E. (2004). Cerebellar function in autism: Functional
magnetic resonance image activation during a simple motor task. Biological Psychiatry,
56(4), 269–278. http://dx.doi.org/10.1016/j.biopsych.2004.06.005.
Ameis, S. H., Fan, J., Rockel, C., Voineskos, A. N., Lobaugh, N. J., Soorya, L., et al. (2011).
Impaired structural connectivity of socio-emotional circuits in autism spectrum disor-
ders: A diffusion tensor imaging study. PLoS One, 6(11), e28044. http://dx.doi.org/
10.1371/journal.pone.0028044.
Asperger, H., & Frith, U. T. (1991). ‘Autistic psychopathology’ in childhood. In U. Frith
(Ed.), Autism and Asperger syndrome (pp. 37–92). New York, NY: Cambridge
University Press.
Association, A. P. (2013). Diagnostic and statistical manual of mental health disorders: DSM-5 (5th
ed.). Washington, DC: American Psychiatric Publishing.
Bailey, A., Luthert, P., Dean, A., Harding, B., Janota, I., Montgomery, M., et al. (1998).
A clinicopathological study of autism. Brain, 121(5), 889–905.
Balleine, B. W., Delgado, M. R., & Hikosaka, O. (2007). The role of the dorsal striatum in
reward and decision-making. The Journal of Neuroscience, 27(31), 8161–8165. http://dx.
doi.org/10.1523/JNEUROSCI.1554-07.2007.
Baranek, G. T. (1999). Autism during infancy: A retrospective video analysis of sensory-
motor and social behaviors at 9-12 months of age. Journal of Autism and Developmental
Disorders, 29(3), 213–224.
Baranek, G. T. (2002). Efficacy of sensory and motor interventions for children with autism.
Journal of Autism and Developmental Disorders, 32(5), 397–422.
Bauman, M. L., & Kemper, T. L. (1994). Neuroanatomic observations of the brain in autism.
In M. L. Bauman & T. L. Kemper (Eds.), The neurobiology of autism (pp. 119–145).
Baltimore: John Hopkins University Press.
Bauman, M. L., & Kemper, T. L. (2005). Neuroanatomic observations of the brain in autism:
A review and future directions. International Journal of Developmental Neuroscience, 23(2–3),
183–187. http://dx.doi.org/10.1016/j.ijdevneu.2004.09.006.
Belmonte, M. K., Allen, G., Beckel-Mitchener, A., Boulanger, L. M., Carper, R. A., &
Webb, S. J. (2004). Autism and abnormal development of brain connectivity. The Journal
of Neuroscience, 24(42), 9228–9231. http://dx.doi.org/10.1523/JNEUROSCI.3340-
04.2004.
Belmonte, M. K., Saxena-Chandhok, T., Cherian, R., Muneer, R., George, L., &
Karanth, P. (2013). Oral motor deficits in speech-impaired children with autism. Frontiers
in Integrative Neuroscience, 7, 47. http://dx.doi.org/10.3389/fnint.2013.00047.
Berg, P., Becker, T., Martian, A., Primrose, K. D., & Wingen, J. (2012). Motor control out-
comes following Nintendo Wii use by a child with Down syndrome. Pediatric Physical
Therapy, 24(1), 78–84. http://dx.doi.org/10.1097/PEP.0b013e31823e05e6.
Bhat, A. N., Galloway, J. C., & Landa, R. J. (2012). Relation between early motor delay and
later communication delay in infants at risk for autism. Infant Behavior & Development,
35(4), 838–846. http://dx.doi.org/10.1016/j.infbeh.2012.07.019.
236 Leanne Chukoskie et al.
Bilder, D., Botts, E. L., Smith, K. R., Pimentel, R., Farley, M., Viskochil, J., et al. (2013).
Excess mortality and causes of death in autism spectrum disorders: A follow up of the
1980s Utah/UCLA autism epidemiologic study. Journal of Autism and Developmental Dis-
orders, 43(5), 1196–1204. http://dx.doi.org/10.1007/s10803-012-1664-z.
Black, J. E., Isaacs, K. R., Anderson, B. J., Alcantara, A. A., & Greenough, W. T. (1990).
Learning causes synaptogenesis, whereas motor activity causes angiogenesis, in cerebellar
cortex of adult rats. Proceedings of the National Academy of Sciences of the United States of
America, 87(14), 5568–5572.
Bostan, A. C., Dum, R. P., & Strick, P. L. (2013). Cerebellar networks with the cerebral
cortex and basal ganglia. Trends in Cognitive Sciences, 17(5), 241–254. http://dx.doi.
org/10.1016/j.tics.2013.03.003.
Brown, T. T., & Jernigan, T. L. (2012). Brain development during the preschool years. Neu-
ropsychology Review, 22(4), 313–333. http://dx.doi.org/10.1007/s11065-012-9214-1.
Bushnell, E. W., & Boudreau, J. P. (1993). Motor development and the mind: The potential
role of motor abilities as a determinant of aspects of perceptual development. Child Devel-
opment, 64(4), 1005–1021.
Calhoun, M., Longworth, M., & Chester, V. L. (2011). Gait patterns in children with autism
[Research Support, Non-U.S. Gov’t]. Clinical Biomechanics, 26(2), 200–206. http://dx.
doi.org/10.1016/j.clinbiomech.2010.09.013.
Carper, R. A., & Courchesne, E. (2000). Inverse correlation between frontal lobe and cer-
ebellum sizes in children with autism. Brain, 123, 836–844.
Cattaneo, L., Fabbri-Destro, M., Boria, S., Pieraccini, C., Monti, A., Cossu, G., et al. (2007).
Impairment of actions chains in autism and its possible role in intention understanding.
Proceedings of the National Academy of Sciences of the United States of America, 104(45),
17825–17830. http://dx.doi.org/10.1073/pnas.0706273104.
Chang, J. O., Levy, S. S., Seay, S. W., & Goble, D. J. (2013). An alternative to the balance
error scoring system: Using a low-cost balance board to improve the validity/reliability
of sports-related concussion balance testing. Clinical Journal of Sports Medicine (in press).
Chang, C. H., Wade, M. G., Stoffregen, T. A., Hsu, C. Y., & Pan, C. Y. (2010). Visual tasks and
postural sway in children with and without autism spectrum disorders. Research in Develop-
mental Disabilities, 31(6), 1536–1542. http://dx.doi.org/10.1016/j.ridd.2010.06.003.
Cherng, R. J., Liang, L. Y., Chen, Y. J., & Chen, J. Y. (2009). The effects of a motor and a
cognitive concurrent task on walking in children with developmental coordination disor-
der. Gait & Posture, 29(2), 204–207. http://dx.doi.org/10.1016/j.gaitpost.2008.08.003.
Chukoskie, L., & Movshon, J. A. (2009). Modulation of visual signals in macaque MT and
MST neurons during pursuit eye movement. Journal of Neurophysiology, 102(6),
3225–3233. http://dx.doi.org/10.1152/jn.90692.2008.
Chukoskie, L., Snider, J., Mozer, M. C., Krauzlis, R. J., & Sejnowski, T. J. (2013). Learning
where to look for a hidden target. Proceedings of the National Academy of Sciences of the
United States of America, 110(Suppl. 2), 10438–10445. http://dx.doi.org/10.1073/
pnas.1301216110.
Ciesielski, K. T., & Akshoomoff, E. (1990). Hypoplasia of cerebellar vermis in autism and
childhood leukemia. In: Paper presented at the 5th International Child Neurology Congress,
Tokyo, Japan.
Ciesielski, K. T., Harris, R. J., Hart, B. L., & Pabst, H. F. (1997). Cerebellar hypoplasia and
frontal lobe cognitive deficits in disorders of early childhood. Neuropsychologia, 35(5),
643–655, S0028-3932(96)00119-4.
Ciesielski, K. T., & Knight, J. E. (1994). Cerebellar abnormality in autism: A nonspecific
effect of early brain damage? Acta Neurobiologiae Experimentalis, 54(2), 151–154.
Clark, R. A., Bryant, A. L., Pua, Y., McCrory, P., Bennell, K., & Hunt, M. (2010). Validity
and reliability of the Nintendo Wii Balance Board for assessment of standing balance.
Gait & Posture, 31(3), 307–310. http://dx.doi.org/10.1016/j.gaitpost.2009.11.012.
Motor Skill in Autism Spectrum Disorders 237
Clemson, L., Fiatarone Singh, M. A., Bundy, A., Cumming, R. G., Manollaras, K.,
O’Loughlin, P., et al. (2012). Integration of balance and strength training into daily life
activity to reduce rate of falls in older people (the LiFE study): Randomised parallel trial.
British Medical Journal, 345, e4547. http://dx.doi.org/10.1136/bmj.e4547.
Conner, J. M., Culberson, A., Packowski, C., Chiba, A. A., & Tuszynski, M. H. (2003).
Lesions of the basal forebrain cholinergic system impair task acquisition and abolish
cortical plasticity associated with motor skill learning. Neuron, 38(5), 819–829,
S0896627303002885.
Courchesne, E., Hesselink, J. R., Jernigan, T. L., & Yeung-Courchesne, R. (1987). Abnor-
mal neuroanatomy in a nonretarded person with autism. Unusual findings with magnetic
resonance imaging. Archives of Neurology, 44(3), 335–341.
Courchesne, E., Karns, C. M., Davis, H. R., Ziccardi, R., Carper, R. A., Tigue, Z. D., et al.
(2001). Unusual brain growth patterns in early life in patients with autistic disorder: An
MRI study. Neurology, 57(2), 245–254.
Courchesne, E., Saitoh, O., Yeung-Courchesne, R., Press, G. A., Lincoln, A. J.,
Haas, R. H., et al. (1994). Abnormality of cerebellar vermian lobules VI and VII in
patients with infantile autism: Identification of hypoplastic and hyperplastic subgroups
with MR imaging. AJR: American Journal of Roentgenology, 162(1), 123–130.
Courchesne, E., Yeung-Courchesne, R., Press, G. A., Hesselink, J. R., & Jernigan, T. L.
(1988). Hypoplasia of cerebellar vermal lobules VI and VII in autism. New England Journal
of Medicine, 318(21), 1349–1354.
David, F. J., Baranek, G. T., Wiesen, C., Miao, A. F., & Thorpe, D. E. (2012). Coordination
of precision grip in 2-6 years-old children with autism spectrum disorders compared to
children developing typically and children with developmental disabilities. Frontiers in
Integrative Neuroscience, 6, 122. http://dx.doi.org/10.3389/fnint.2012.00122.
Daw, N. D., Niv, Y., & Dayan, P. (2005). Uncertainty-based competition between prefron-
tal and dorsolateral striatal systems for behavioral control. Nature Neuroscience, 8(12),
1704–1711. http://dx.doi.org/10.1038/nn1560.
Dayan, E., & Cohen, L. G. (2011). Neuroplasticity subserving motor skill learning. Neuron,
72(3), 443–454. http://dx.doi.org/10.1016/j.neuron.2011.10.008.
DeLorey, T. M., Sahbaie, P., Hashemi, E., Homanics, G. E., & Clark, J. D. (2008). Gabrb3
gene deficient mice exhibit impaired social and exploratory behaviors, deficits in non-
selective attention and hypoplasia of cerebellar vermal lobules: A potential model of
autism spectrum disorder. Behavioural Brain Research, 187(2), 207–220. http://dx.doi.
org/10.1016/j.bbr.2007.09.009.
Desmond, J. E., & Fiez, J. A. (1998). Neuroimaging studies of the cerebellum: Language, learn-
ing and memory. Trends in Cognitive Sciences, 2(9), 355–362, S1364-6613(98)01211-X.
Desrochers, T. M., Jin, D. Z., Goodman, N. D., & Graybiel, A. M. (2010). Optimal habits
can develop spontaneously through sensitivity to local cost. Proceedings of the National
Academy of Sciences of the United States of America, 107(47), 20512–20517. http://dx.
doi.org/10.1073/pnas.1013470107.
Deutsch, J. E., Borbely, M., Filler, J., Huhn, K., & Guarrera-Bowlby, P. (2008). Use of a
low-cost, commercially available gaming console (Wii) for rehabilitation of an adolescent
with cerebral palsy. Physical Therapy, 88(10), 1196–1207. http://dx.doi.org/10.2522/
ptj.20080062.
Dewey, D., Cantell, M., & Crawford, S. G. (2007). Motor and gestural performance in chil-
dren with autism spectrum disorders, developmental coordination disorder, and/or atten-
tion deficit hyperactivity disorder. Journal of the International Neuropsychological Society, 13(2),
246–256. http://dx.doi.org/10.1017/S1355617707070270.
Di Martino, A., Kelly, C., Grzadzinski, R., Zuo, X. N., Mennes, M., Mairena, M. A., et al.
(2011). Aberrant striatal functional connectivity in children with autism. Biological Psy-
chiatry, 69(9), 847–856. http://dx.doi.org/10.1016/j.biopsych.2010.10.029.
238 Leanne Chukoskie et al.
Di Martino, A., Zuo, X. N., Kelly, C., Grzadzinski, R., Mennes, M., Schvarcz, A., et al.
(2013). Shared and distinct intrinsic functional network centrality in autism and
attention-deficit/hyperactivity disorder. Biological Psychiatry, 74(8), 623–632. http://
dx.doi.org/10.1016/j.biopsych.2013.02.011.
Dichter, G. S. (2012). Functional magnetic resonance imaging of autism spectrum disorders.
Dialogues in Clinical Neuroscience, 14(3), 319–351.
Dowell, L. R., Mahone, E. M., & Mostofsky, S. H. (2009). Associations of postural knowl-
edge and basic motor skill with dyspraxia in autism: Implication for abnormalities in dis-
tributed connectivity and motor learning. Neuropsychology, 23(5), 563–570. http://dx.
doi.org/10.1037/a0015640.
Doya, K. (1999). What are the computations of the cerebellum, the basal ganglia and the
cerebral cortex? Neural Networks, 12(7–8), 961–974, S0893-6080(99)00046-5.
Doya, K. (2000). Complementary roles of basal ganglia and cerebellum in learning and motor
control. Current Opinion in Neurobiology, 10(6), 732–739, S0959-4388(00)00153-7.
Doyon, J., Penhune, V., & Ungerleider, L. G. (2003). Distinct contribution of the cortico-
striatal and cortico-cerebellar systems to motor skill learning. Neuropsychologia, 41(3),
252–262, S0028393202001586.
Durisko, C., & Fiez, J. A. (2010). Functional activation in the cerebellum during working
memory and simple speech tasks. Cortex, 46(7), 896–906. http://dx.doi.org/10.1016/
j.cortex.2009.09.009.
Dziuk, M. A., Gidley Larson, J. C., Apostu, A., Mahone, E. M., Denckla, M. B., &
Mostofsky, S. H. (2007). Dyspraxia in autism: Association with motor, social, and com-
municative deficits. Developmental Medicine and Child Neurology, 49(10), 734–739. http://
dx.doi.org/10.1111/j.1469-8749.2007.00734.x.
Ecker, C., Suckling, J., Deoni, S. C., Lombardo, M. V., Bullmore, E. T., Baron-Cohen, S.,
et al. (2012). Brain anatomy and its relationship to behavior in adults with autism spec-
trum disorder: A multicenter magnetic resonance imaging study. Archives of General Psy-
chiatry, 69(2), 195–209. http://dx.doi.org/10.1001/archgenpsychiatry.2011.1251.
Elsabbagh, M., Volein, A., Holmboe, K., Tucker, L., Csibra, G., Baron-Cohen, S., et al.
(2009). Visual orienting in the early broader autism phenotype: Disengagement and facil-
itation. Journal of Child Psychology and Psychiatry, 50(5), 637–642. http://dx.doi.org/
10.1111/j.1469-7610.2008.02051.x.
Esposito, G., Venuti, P., Apicella, F., & Muratori, F. (2011). Analysis of unsupported gait in
toddlers with autism. Brain & development, 33(5), 367–373. http://dx.doi.org/10.1016/j.
braindev.2010.07.006.
Estes, A., Shaw, D. W., Sparks, B. F., Friedman, S., Giedd, J. N., Dawson, G., et al. (2011).
Basal ganglia morphometry and repetitive behavior in young children with
autism spectrum disorder. Autism Research, 4(3), 212–220. http://dx.doi.org/
10.1002/aur.193.
Fabbri-Destro, M., Cattaneo, L., Boria, S., & Rizzolatti, G. (2009). Planning actions in
autism. Experimental Brain Research, 192(3), 521–525. http://dx.doi.org/10.1007/
s00221-008-1578-3.
Fatemi, S. H., Aldinger, K. A., Ashwood, P., Bauman, M. L., Blaha, C. D., Blatt, G. J., et al.
(2012). Consensus paper: Pathological role of the cerebellum in autism. Cerebellum,
11(3), 777–807. http://dx.doi.org/10.1007/s12311-012-0355-9.
Fehlow, P., Bernstein, K., Tennstedt, A., & Walther, F. (1993). Early infantile autism and
excessive aerophagy with symptomatic megacolon and ileus in a case of Ehlers-Danlos
syndrome. Pädiatrie und Grenzgebiete, 31(4), 259–267.
Fink, G. R., Marshall, J. C., Shah, N. J., Weiss, P. H., Halligan, P. W., Grosse-Ruyken, M.,
et al. (2000). Line bisection judgments implicate right parietal cortex and cerebellum as
assessed by fMRI. Neurology, 54(6), 1324–1331.
Motor Skill in Autism Spectrum Disorders 239
Flanagan, J. E., Landa, R., Bhat, A., & Bauman, M. (2012). Head lag in infants at risk for
autism: A preliminary study. The American Journal of Occupational Therapy, 66(5),
577–585. http://dx.doi.org/10.5014/ajot.2012.004192.
Fournier, K. A., Hass, C. J., Naik, S. K., Lodha, N., & Cauraugh, J. H. (2010). Motor coor-
dination in autism spectrum disorders: A synthesis and meta-analysis. Journal of Autism and
Developmental Disorders, 40(10), 1227–1240. http://dx.doi.org/10.1007/s10803-010-
0981-3.
Fournier, K. A., Kimberg, C. I., Radonovich, K. J., Tillman, M. D., Chow, J. W.,
Lewis, M. H., et al. (2010). Decreased static and dynamic postural control in children
with autism spectrum disorders. Gait & Posture, 32(1), 6–9. http://dx.doi.org/
10.1016/j.gaitpost.2010.02.007.
Fu, M., Yu, X., Lu, J., & Zuo, Y. (2012). Repetitive motor learning induces coordinated
formation of clustered dendritic spines in vivo. Nature, 483(7387), 92–95. http://dx.
doi.org/10.1038/nature10844.
Fuentes, C. T., Mostofsky, S. H., & Bastian, A. J. (2009). Children with autism show specific
handwriting impairments. Neurology, 73(19), 1532–1537. http://dx.doi.org/10.1212/
WNL.0b013e3181c0d48c.
Fulkerson, S. C., & Freeman, W. M. (1980). Perceptual-motor deficiency in autistic children.
Perceptual and Motor Skills, 50(1), 331–336.
Gaffney, G. R., Tsai, L. Y., Kuperman, S., & Minchin, S. (1987). Cerebellar structure in
autism. American Journal of Diseases of Children, 141(12), 1330–1332.
Geschwind, D. H., & Levitt, P. (2007). Autism spectrum disorders: Developmental discon-
nection syndromes. Current Opinion in Neurobiology, 17(1), 103–111. http://dx.doi.org/
10.1016/j.conb.2007.01.009.
Ghaziuddin, M., & Butler, E. (1998). Clumsiness in autism and Asperger syndrome: A further
report. Journal of Intellectual Disability Research, 42(Pt. 1), 43–48.
Ghaziuddin, M., Butler, E., Tsai, L., & Ghaziuddin, N. (1994). Is clumsiness a marker for
Asperger syndrome? Journal of Intellectual Disability Research, 38(Pt. 5), 519–527.
Gidley Larson, J. C., Bastian, A. J., Donchin, O., Shadmehr, R., & Mostofsky, S. H. (2008).
Acquisition of internal models of motor tasks in children with autism. Brain, 131(Pt. 11),
2894–2903. http://dx.doi.org/10.1093/brain/awn226.
Gillberg, C., Billstedt, E., Sundh, V., & Gillberg, I. C. (2010). Mortality in autism:
A prospective longitudinal community-based study. Journal of Autism and Developmental
Disorders, 40(3), 352–357. http://dx.doi.org/10.1007/s10803-009-0883-4.
Glazebrook, C. M., Elliott, D., & Lyons, J. (2006). A kinematic analysis of how young adults
with and without autism plan and control goal-directed movements. Motor Control,
10(3), 244–264.
Glazebrook, C. M., Elliott, D., & Szatmari, P. (2008). How do individuals with autism plan
their movements? Journal of Autism and Developmental Disorders, 38(1), 114–126. http://
dx.doi.org/10.1007/s10803-007-0369-1.
Glazebrook, C., Gonzalez, D., Hansen, S., & Elliott, D. (2009). The role of vision for online
control of manual aiming movements in persons with autism spectrum disorders. Autism,
13(4), 411–433. http://dx.doi.org/10.1177/1362361309105659.
Granacher, U., Gollhofer, A., Hortobagyi, T., Kressig, R. W., & Muehlbauer, T. (2013).
The importance of trunk muscle strength for balance, functional performance, and fall
prevention in seniors: A systematic review. Sports Medicine, 43(7), 627–641. http://dx.
doi.org/10.1007/s40279-013-0041-1.
Graybiel, A. M. (1995a). The basal ganglia. Trends in Neurosciences, 18(2), 60–62, 0166-2236
(95)80019-X.
Graybiel, A. M. (1995b). Building action repertoires: Memory and learning functions of the
basal ganglia. Current Opinion in Neurobiology, 5(6), 733–741, 0959-4388(95)80100-6.
240 Leanne Chukoskie et al.
Graybiel, A. M., Aosaki, T., Flaherty, A. W., & Kimura, M. (1994). The basal ganglia and
adaptive motor control. Science, 265(5180), 1826–1831.
Green, D., Charman, T., Pickles, A., Chandler, S., Loucas, T., Simonoff, E., et al. (2009).
Impairment in movement skills of children with autistic spectrum disorders. Developmen-
tal Medicine and Child Neurology, 51(4), 311–316. http://dx.doi.org/10.1111/j.1469-
8749.2008.03242.x.
Greenough, W. T., Larson, J. R., & Withers, G. S. (1985). Effects of unilateral and bilateral
training in a reaching task on dendritic branching of neurons in the rat motor-sensory
forelimb cortex. Behavioral and Neural Biology, 44(2), 301–314.
Greenspan, S. I., Brazelton, T. B., Cordero, J., Solomon, R., Bauman, M. L., Robinson, R.,
et al. (2008). Guidelines for early identification, screening, and clinical management of
children with autism spectrum disorders. Pediatrics, 121(4), 828–830. http://dx.doi.org/
10.1542/peds.2007-3833.
Greffou, S., Bertone, A., Hahler, E. M., Hanssens, J. M., Mottron, L., & Faubert, J. (2012).
Postural hypo-reactivity in autism is contingent on development and visual environ-
ment: A fully immersive virtual reality study. Journal of Autism and Developmental Disorders,
42(6), 961–970. http://dx.doi.org/10.1007/s10803-011-1326-6.
Grissmer, D., Grimm, K. J., Aiyer, S. M., Murrah, W. M., & Steele, J. S. (2010). Fine motor
skills and early comprehension of the world: Two new school readiness indicators. Devel-
opmental Psychology, 46(5), 1008–1017. http://dx.doi.org/10.1037/a0020104.
Haas, R. H., Townsend, J., Courchesne, E., Lincoln, A. J., Schreibman, L., & Yeung-
Courchesne, R. (1996). Neurologic abnormalities in infantile autism. Journal of Child
Neurology, 11(2), 84–92.
Hallett, M., Lebiedowska, M. K., Thomas, S. L., Stanhope, S. J., Denckla, M. B., &
Rumsey, J. (1993). Locomotion of autistic adults [Comparative Study]. Archives of Neu-
rology, 50(12), 1304–1308.
Hallgato, E., Gyori-Dani, D., Pekar, J., Janacsek, K., & Nemeth, D. (2013). The differential
consolidation of perceptual and motor learning in skill acquisition. Cortex, 49(4),
1073–1081. http://dx.doi.org/10.1016/j.cortex.2012.01.002.
Happe, F., & Charlton, R. A. (2012). Aging in autism spectrum disorders: A mini-review.
Gerontology, 58(1), 70–78. http://dx.doi.org/10.1159/000329720.
Hardan, A. Y., Kilpatrick, M., Keshavan, M. S., & Minshew, N. J. (2003). Motor perfor-
mance and anatomic magnetic resonance imaging (MRI) of the basal ganglia in autism.
Journal of Child Neurology, 18(5), 317–324.
Hashimoto, T., Tayama, M., Murakawa, K., Yoshimoto, T., Miyazaki, M., Harada, M.,
et al. (1995). Development of the brainstem and cerebellum in autistic patients. Journal
of Autism and Developmental Disorders, 25, 1–18.
Haswell, C. C., Izawa, J., Dowell, L. R., Mostofsky, S. H., & Shadmehr, R. (2009). Rep-
resentation of internal models of action in the autistic brain. Nature Neuroscience, 12(8),
970–972. http://dx.doi.org/10.1038/nn.2356.
Hautzel, H., Mottaghy, F. M., Specht, K., Muller, H. W., & Krause, B. J. (2009). Evidence of
a modality-dependent role of the cerebellum in working memory? An fMRI study com-
paring verbal and abstract n-back tasks. NeuroImage, 47(4), 2073–2082. http://dx.doi.
org/10.1016/j.neuroimage.2009.06.005.
Havermann, K., & Lappe, M. (2010). The influence of the consistency of postsaccadic visual
errors on saccadic adaptation. Journal of Neurophysiology, 103(6), 3302–3310. http://dx.
doi.org/10.1152/jn.00970.2009.
Hikosaka, O., Nakamura, K., Sakai, K., & Nakahara, H. (2002). Central mechanisms of motor
skill learning. Current Opinion in Neurobiology, 12(2), 217–222, S0959438802003070.
Hilton, C. L., Zhang, Y., Whilte, M. R., Klohr, C. L., & Constantino, J. (2012). Motor
impairment in sibling pairs concordant and discordant for autism spectrum disorders.
Autism, 16(4), 430–441. http://dx.doi.org/10.1177/1362361311423018.
Motor Skill in Autism Spectrum Disorders 241
Hollander, E., Anagnostou, E., Chaplin, W., Esposito, K., Haznedar, M. M., Licalzi, E., et al.
(2005). Striatal volume on magnetic resonance imaging and repetitive behaviors in
autism. Biological Psychiatry, 58(3), 226–232. http://dx.doi.org/10.1016/j.
biopsych.2005.03.040.
Houk, J. C., & Wise, S. P. (1995). Distributed modular architectures linking basal ganglia,
cerebellum, and cerebral cortex: Their role in planning and controlling action. Cerebral
Cortex, 5(2), 95–110.
Hsu, H. C., Chen, C. L., Cheng, P. T., Chen, C. H., Chong, C. Y., & Lin, Y. Y. (2004). The
relationship of social function with motor and speech functions in children with autism.
Chang Gung Medical Journal, 27(10), 750–757, 2710/271006.
Huang, H. J., & Mercer, V. S. (2001). Dual-task methodology: Applications in studies of
cognitive and motor performance in adults and children. Pediatric Physical Therapy,
13(3), 133–140, 00001577-200110000-00005.
Huurnink, A., Fransz, D. P., Kingma, I., & van Dieen, J. H. (2013). Comparison of a lab-
oratory grade force platform with a Nintendo Wii Balance Board on measurement of
postural control in single-leg stance balance tasks. Journal of Biomechanics, 46(7),
1392–1395. http://dx.doi.org/10.1016/j.jbiomech.2013.02.018.
Hwang, E. J. (2013). The basal ganglia, the ideal machinery for the cost-benefit analysis of
action plans. Front Neural Circuits, 7, 121. http://dx.doi.org/10.3389/fncir.2013.00121.
Ilg, W., Schatton, C., Schicks, J., Giese, M. A., Schols, L., & Synofzik, M. (2012). Video
game-based coordinative training improves ataxia in children with degenerative ataxia.
Neurology, 79(20), 2056–2060. http://dx.doi.org/10.1212/WNL.0b013e3182749e67.
Iverson, J. M., & Goldin-Meadow, S. (2005). Gesture paves the way for language develop-
ment. Psychological Science, 16(5), 367–371. http://dx.doi.org/10.1111/j.0956-
7976.2005.01542.x.
Izawa, J., Pekny, S. E., Marko, M. K., Haswell, C. C., Shadmehr, R., & Mostofsky, S. H.
(2012). Motor learning relies on integrated sensory inputs in ADHD, but over-
selectively on proprioception in autism spectrum conditions. Autism Research, 5(2),
124–136. http://dx.doi.org/10.1002/aur.1222.
Janicki, M. P., Henderson, C. M., & Rubin, I. L. (2008). Neurodevelopmental conditions
and aging: Report on the Atlanta Study Group Charrette on Neurodevelopmental Con-
ditions and Aging. Disability and Health Journal, 1(2), 116–124. http://dx.doi.org/
10.1016/j.dhjo.2008.02.004.
Jansiewicz, E. M., Goldberg, M. C., Newschaffer, C. J., Denckla, M. B., Landa, R., &
Mostofsky, S. H. (2006). Motor signs distinguish children with high functioning autism
and Asperger’s syndrome from controls. Journal of Autism and Developmental Disorders,
36(5), 613–621. http://dx.doi.org/10.1007/s10803-006-0109-y.
Jelsma, J., Pronk, M., Ferguson, G., & Jelsma-Smit, D. (2013). The effect of the Nintendo
Wii Fit on balance control and gross motor function of children with spastic hemiplegic
cerebral palsy. Developmental Neurorehabilitation, 16(1), 27–37. http://dx.doi.org/
10.3109/17518423.2012.711781.
Johnson, B. P., Rinehart, N. J., Papadopoulos, N., Tonge, B., Millist, L., White, O., et al.
(2012). A closer look at visually guided saccades in autism and Asperger’s disorder. Fron-
tiers in Integrative Neuroscience, 6, 99. http://dx.doi.org/10.3389/fnint.2012.00099.
Johnson, B. P., Rinehart, N. J., White, O., Millist, L., & Fielding, J. (2013). Saccade adap-
tation in autism and Asperger’s disorder. Neuroscience, 243, 76–87. http://dx.doi.org/
10.1016/j.neuroscience.2013.03.051.
Kanner, L. (1943). Autistic disturbances of affective contact. Nervous Child, 2(3), 217–250.
Kates, W. R., Mostofsky, S. H., Zimmerman, A. W., Mazzocco, M. M., Landa, R.,
Warsofsky, I. S., et al. (1998). Neuroanatomical and neurocognitive differences in a pair
of monozygous twins discordant for strictly defined autism. Annals of Neurology, 43(6),
782–791.
242 Leanne Chukoskie et al.
Kaufmann, W. E., Cooper, K. L., Mostofsky, S. H., Capone, G. T., Kates, W. R.,
Newschaffer, C. J., et al. (2003). Specificity of cerebellar vermian abnormalities in
autism: A quantitative magnetic resonance imaging study. Journal of Child Neurology,
18(7), 463–470.
Kawakubo, Y., Kasai, K., Okazaki, S., Hosokawa-Kakurai, M., Watanabe, K.,
Kuwabara, H., et al. (2007). Electrophysiological abnormalities of spatial attention in
adults with autism during the gap overlap task. Clinical Neurophysiology, 118(7),
1464–1471. http://dx.doi.org/10.1016/j.clinph.2007.04.015.
Kellermann, T., Regenbogen, C., De Vos, M., Mossnang, C., Finkelmeyer, A., & Habel, U.
(2012). Effective connectivity of the human cerebellum during visual attention. The Journal
of Neuroscience, 32(33), 11453–11460. http://dx.doi.org/10.1523/JNEUROSCI.0678-
12.2012.
Kemper, T. L., & Bauman, M. (1998). Neuropathology of infantile autism. Journal of Neu-
ropathology and Experimental Neurology, 57(7), 645–652.
Kleim, J. A., Swain, R. A., Armstrong, K. A., Napper, R. M., Jones, T. A., &
Greenough, W. T. (1998). Selective synaptic plasticity within the cerebellar cortex fol-
lowing complex motor skill learning. Neurobiology of Learning and Memory, 69(3),
274–289. http://dx.doi.org/10.1006/nlme.1998.3827.
Klin, A., Jones, W., Schultz, R., Volkmar, F., & Cohen, D. (2002). Visual fixation patterns
during viewing of naturalistic social situations as predictors of social competence in indi-
viduals with autism. Archives of General Psychiatry, 59(9), 809–816, yoa10221.
Klintsova, A. Y., Cowell, R. M., Swain, R. A., Napper, R. M., Goodlett, C. R., &
Greenough, W. T. (1998). Therapeutic effects of complex motor training on motor per-
formance deficits induced by neonatal binge-like alcohol exposure in rats. I. Behavioral
results. Brain Research, 800(1), 48–61, S0006-8993(98)00495-8.
Klintsova, A. Y., Scamra, C., Hoffman, M., Napper, R. M., Goodlett, C. R., &
Greenough, W. T. (2002). Therapeutic effects of complex motor training on motor per-
formance deficits induced by neonatal binge-like alcohol exposure in rats: II.
A quantitative stereological study of synaptic plasticity in female rat cerebellum. Brain
Research, 937(1–2), 83–93, S0006899302024927.
Kohen-Raz, R., Volkmar, F. R., & Cohen, D. J. (1992). Postural control in children with
autism. Journal of Autism and Developmental Disorders, 22(3), 419–432.
Kopp, S., Beckung, E., & Gillberg, C. (2010). Developmental coordination disorder and
other motor control problems in girls with autism spectrum disorder and/or
attention-deficit/hyperactivity disorder. Research in Developmental Disabilities, 31(2),
350–361. http://dx.doi.org/10.1016/j.ridd.2009.09.017.
Kording, K. P., & Wolpert, D. M. (2006). Probabilistic mechanisms in sensorimotor control.
Novartis Foundation Symposium, 270, 191–198, discussion 198-202, 232-197.
Kravitz, A. V., Freeze, B. S., Parker, P. R., Kay, K., Thwin, M. T., Deisseroth, K., et al.
(2010). Regulation of Parkinsonian motor behaviours by optogenetic control of basal
ganglia circuitry. Nature, 466(7306), 622–626. http://dx.doi.org/10.1038/nature09159.
Kravitz, A. V., & Kreitzer, A. C. (2012). Striatal mechanisms underlying movement, rein-
forcement, and punishment. Physiology (Bethesda), 27(3), 167–177. http://dx.doi.org/
10.1152/physiol.00004.2012.
Kreitzer, A. C., & Malenka, R. C. (2007). Endocannabinoid-mediated rescue of striatal LTD
and motor deficits in Parkinson’s disease models. Nature, 445(7128), 643–647. http://dx.
doi.org/10.1038/nature05506.
Kreitzer, A. C., & Malenka, R. C. (2008). Striatal plasticity and basal ganglia circuit function.
Neuron, 60(4), 543–554. http://dx.doi.org/10.1016/j.neuron.2008.11.005.
Kuhn, G., Kourkoulou, A., & Leekam, S. R. (2010). How magic changes our expectations
about autism. Psychological Science, 21(10), 1487–1493. http://dx.doi.org/10.1177/
0956797610383435.
Motor Skill in Autism Spectrum Disorders 243
Kushki, A., Chau, T., & Anagnostou, E. (2011). Handwriting difficulties in children with
autism spectrum disorders: A scoping review. Journal of Autism and Developmental Disor-
ders, 41(12), 1706–1716. http://dx.doi.org/10.1007/s10803-011-1206-0.
Lajoie, Y., Teasdale, N., Bard, C., & Fleury, M. (1993). Attentional demands for static and
dynamic equilibrium. Experimental Brain Research, 97(1), 139–144.
Landa, R., & Garrett-Mayer, E. (2006). Development in infants with autism spectrum dis-
orders: A prospective study. Journal of Child Psychology and Psychiatry, 47(6), 629–638.
http://dx.doi.org/10.1111/j.1469-7610.2006.01531.x.
Landry, R., & Bryson, S. E. (2004). Impaired disengagement of attention in young children
with autism. Journal of Child Psychology and Psychiatry, 45(6), 1115–1122. http://dx.doi.
org/10.1111/j.1469-7610.2004.00304.x.
Langen, M., Durston, S., Staal, W. G., Palmen, S. J., & van Engeland, H. (2007). Caudate
nucleus is enlarged in high-functioning medication-naive subjects with autism. Biological
Psychiatry, 62(3), 262–266. http://dx.doi.org/10.1016/j.biopsych.2006.09.040.
Langen, M., Leemans, A., Johnston, P., Ecker, C., Daly, E., Murphy, C. M., et al. (2012).
Fronto-striatal circuitry and inhibitory control in autism: Findings from diffusion tensor
imaging tractography. Cortex, 48(2), 183–193. http://dx.doi.org/10.1016/j.
cortex.2011.05.018.
Larson, J. C. G., & Mostofsky, S. H. (2008). Evidence that the pattern of visuomotor
sequence learning is altered in children with autism. Autism Research, 1(6), 341–353.
http://dx.doi.org/10.1002/Aur.54.
Laufer, Y., Ashkenazi, T., & Josman, N. (2008). The effects of a concurrent cognitive task on
the postural control of young children with and without developmental coordination
disorder. Gait & Posture, 27(2), 347–351. http://dx.doi.org/10.1016/j.gaitpost.
2007.04.013.
Le, T. H., Pardo, J. V., & Hu, X. (1998). 4T-fMRI study of nonspatial shifting of selective
attention: Cerebellar and parietal contributions. Journal of Neurophysiology, 79(3),
1535–1548.
Lee, K. J., Park, I. S., Kim, H., Greenough, W. T., Pak, D. T., & Rhyu, I. J. (2013). Motor
skill training induces coordinated strengthening and weakening between neighboring
synapses. The Journal of Neuroscience, 33(23), 9794–9799. http://dx.doi.org/10.1523/
JNEUROSCI.0848-12.2013.
Levitt, J. G., Blanton, R., Capetillo-Cunliffe, L., Guthrie, D., Toga, A., & McCracken, J. T.
(1999). Cerebellar vermis lobules VIII-X in autism. Progress in Neuro-Psychopharmacology
and Biological Psychiatry, 23(4), 625–633.
Libertus, K., & Needham, A. (2010). Teach to reach: The effects of active vs. passive reaching
experiences on action and perception. Vision Research, 50(24), 2750–2757. http://dx.doi.
org/10.1016/j.visres.2010.09.001.
Libertus, K., & Needham, A. (2011). Reaching experience increases face preference in
3-month-old infants. Developmental Science, 14(6), 1355–1364. http://dx.doi.org/
10.1111/j.1467-7687.2011.01084.x.
Lord, C., Petkova, E., Hus, V., Gan, W., Lu, F., Martin, D. M., et al. (2012). A multisite
study of the clinical diagnosis of different autism spectrum disorders. Archives of General
Psychiatry, 69(3), 306–313. http://dx.doi.org/10.1001/archgenpsychiatry.2011.148.
Luna, B., Doll, S. K., Hegedus, S. J., Minshew, N. J., & Sweeney, J. A. (2007). Maturation of
executive function in autism. Biological Psychiatry, 61(4), 474–481. http://dx.doi.org/
10.1016/j.biopsych.2006.02.030.
Manjiviona, J., & Prior, M. (1995). Comparison of Asperger syndrome and high-functioning
autistic children on a test of motor impairment. Journal of Autism and Developmental Dis-
orders, 25(1), 23–39.
Mari, M., Castiello, U., Marks, D., Marraffa, C., & Prior, M. (2003). The reach-to-grasp
movement in children with autism spectrum disorder. Philosophical Transactions of the
244 Leanne Chukoskie et al.
Murphy, D. G., Beecham, J., Craig, M., & Ecker, C. (2011). Autism in adults. New biolog-
ical findings and their translational implications to the cost of clinical services. Brain
Research, 1380, 22–33. http://dx.doi.org/10.1016/j.brainres.2010.10.042.
Nakamura, K., & Hikosaka, O. (2006). Role of dopamine in the primate caudate nucleus in
reward modulation of saccades. The Journal of Neuroscience, 26(20), 5360–5369. http://dx.
doi.org/10.1523/JNEUROSCI.4853-05.2006.
Naviaux, R. K., Zolkipli, Z., Wang, L., Nakayama, T., Naviaux, J. C., Le, T. P., et al.
(2013). Antipurinergic therapy corrects the autism-like features in the poly(IC) mouse
model. PLoS One, 8(3), e57380. http://dx.doi.org/10.1371/journal.pone.0057380.
Nayate, A., Tonge, B. J., Bradshaw, J. L., McGinley, J. L., Iansek, R., & Rinehart, N. J.
(2012). Differentiation of high-functioning autism and Asperger’s disorder based on
neuromotor behaviour. Journal of Autism and Developmental Disorders, 42(5), 707–717.
http://dx.doi.org/10.1007/s10803-011-1299-5.
Nazarali, N., Glazebrook, C. M., & Elliott, D. (2009). Movement planning and repro-
gramming in individuals with autism. Journal of Autism and Developmental Disorders,
39(10), 1401–1411. http://dx.doi.org/10.1007/s10803-009-0756-x.
Niv, Y., Daw, N. D., Joel, D., & Dayan, P. (2007). Tonic dopamine: Opportunity costs and
the control of response vigor. Psychopharmacology, 191(3), 507–520. http://dx.doi.org/
10.1007/s00213-006-0502-4.
Nobile, M., Perego, P., Piccinini, L., Mani, E., Rossi, A., Bellina, M., et al. (2011). Further
evidence of complex motor dysfunction in drug naive children with autism using auto-
matic motion analysis of gait [Research Support, Non-U.S. Gov’t]. Autism: The Interna-
tional Journal of Research and Practice, 15(3), 263–283. http://dx.doi.org/10.1177/
1362361309356929.
Nowinski, C. V., Minshew, N. J., Luna, B., Takarae, Y., & Sweeney, J. A. (2005). Oculo-
motor studies of cerebellar function in autism. Psychiatry Research, 137(1–2), 11–19.
http://dx.doi.org/10.1016/j.psychres.2005.07.005.
O’Halloran, C. J., Kinsella, G. J., & Storey, E. (2012). The cerebellum and neuropsycholog-
ical functioning: A critical review. Journal of Clinical and Experimental Neuropsychology,
34(1), 35–56. http://dx.doi.org/10.1080/13803395.2011.614599.
Ozonoff, S., Iosif, A. M., Baguio, F., Cook, I. C., Hill, M. M., Hutman, T., et al. (2010).
A prospective study of the emergence of early behavioral signs of autism. Journal of
the American Academy of Child and Adolescent Psychiatry, 49(3), 256–266, e251-252,
00004583-201003000-00009.
Ozonoff, S., Young, G. S., Carter, A., Messinger, D., Yirmiya, N., Zwaigenbaum, L., et al.
(2011). Recurrence risk for autism spectrum disorders: A Baby Siblings Research Consor-
tium Study. Pediatrics, 128(3), e488–e495. http://dx.doi.org/10.1542/peds.2010-2825.
Ozonoff, S., Young, G. S., Goldring, S., Greiss-Hess, L., Herrera, A. M., Steele, J., et al.
(2008). Gross motor development, movement abnormalities, and early identification
of autism. Journal of Autism and Developmental Disorders, 38(4), 644–656. http://dx.doi.
org/10.1007/s10803-007-0430-0.
Perry, W., Minassian, A., Lopez, B., Maron, L., & Lincoln, A. (2007). Sensorimotor gating
deficits in adults with autism. Biological Psychiatry, 61(4), 482–486. http://dx.doi.org/
10.1016/j.biopsych.2005.09.025.
Piek, J. P., & Dyck, M. J. (2004). Sensory-motor deficits in children with developmental coor-
dination disorder, attention deficit hyperactivity disorder and autistic disorder. Human
Movement Science, 23(3–4), 475–488. http://dx.doi.org/10.1016/j.humov.2004.08.019.
Piven, J., & Rabins, P. (2011). Autism spectrum disorders in older adults: Toward defining a
research agenda. Journal of the American Geriatrics Society, 59(11), 2151–2155. http://dx.
doi.org/10.1111/j.1532-5415.2011.03632.x.
Planinsec, J. (2002). Relations between the motor and cognitive dimensions of preschool
girls and boys. Perceptual and Motor Skills, 94(2), 415–423.
246 Leanne Chukoskie et al.
Provost, B., Heimerl, S., & Lopez, B. R. (2007). Levels of gross and fine motor development
in young children with autism spectrum disorder. Physical & Occupational Therapy in Pedi-
atrics, 27(3), 21–36.
Qiu, A., Adler, M., Crocetti, D., Miller, M. I., & Mostofsky, S. H. (2010). Basal ganglia
shapes predict social, communication, and motor dysfunctions in boys with autism spec-
trum disorder. Journal of the American Academy of Child and Adolescent Psychiatry, 49(6),
539–551. http://dx.doi.org/10.1016/j.jaac.2010.02.012, 551.e1-4.
Radonovich, K. J., Fournier, K. A., & Hass, C. J. (2013). Relationship between postural con-
trol and restricted, repetitive behaviors in autism spectrum disorders. Frontiers in Integra-
tive Neuroscience, 7, 28. http://dx.doi.org/10.3389/fnint.2013.00028.
Ramstrand, N., & Lygnegard, F. (2012). Can balance in children with cerebral palsy improve
through use of an activity promoting computer game? Technology and Health Care, 20(6),
501–510. http://dx.doi.org/10.3233/THC-2012-0696.
Redgrave, P., Rodriguez, M., Smith, Y., Rodriguez-Oroz, M. C., Lehericy, S.,
Bergman, H., et al. (2010). Goal-directed and habitual control in the basal ganglia: Impli-
cations for Parkinson’s disease. Nature Reviews Neuroscience, 11(11), 760–772. http://dx.
doi.org/10.1038/nrn2915.
Reilly, D. S., van Donkelaar, P., Saavedra, S., & Woollacott, M. H. (2008). Interaction between
the development of postural control and the executive function of attention. Journal of Motor
Behavior, 40(2), 90–102. http://dx.doi.org/10.3200/Jmbr.40.2.90-102.
Rinehart, N. J., Bellgrove, M. A., Tonge, B. J., Brereton, A. V., Howells-Rankin, D., &
Bradshaw, J. L. (2006). An examination of movement kinematics in young people with
high-functioning autism and Asperger’s disorder: Further evidence for a motor planning
deficit. Journal of Autism and Developmental Disorders, 36(6), 757–767. http://dx.doi.org/
10.1007/s10803-006-0118-x.
Rinehart, N. J., Tonge, B. J., Bradshaw, J. L., Iansek, R., Enticott, P. G., & McGinley, J.
(2006). Gait function in high-functioning autism and Asperger’s disorder: Evidence for
basal-ganglia and cerebellar involvement? [Research Support, Non-U.S. Gov’t]. Euro-
pean Child & Adolescent Psychiatry, 15(5), 256–264. http://dx.doi.org/10.1007/s00787-
006-0530-y.
Ritvo, E. R., Freeman, B. J., Scheibel, A. B., Duong, T., Robinson, H., Guthrie, D., et al.
(1986). Lower Purkinje cell counts in the cerebella of four autistic subjects: Initial find-
ings of the UCLA-NSAC Autopsy Research Report. The American Journal of Psychiatry,
143(7), 862–866.
Rodier, P. M., Ingram, J. L., Tisdale, B., Nelson, S., & Romano, J. (1996). Embryological
origin for autism: Developmental anomalies of the cranial nerve motor nuclei. The Journal
of Comparative Neurology, 370(2), 247–261. http://dx.doi.org/10.1002/(SICI)1096-9861
(19960624)370:2<247::AID-CNE8>3.0.CO;2-2.
Saitoh, O., Courchesne, E., Egaas, B., Lincoln, A. J., & Schreibman, L. (1995). Cross-
sectional area of the posterior hippocampus in autistic patients with cerebellar and corpus
callosum abnormalities. Neurology, 45(2), 317–324.
Schlosser, R., Hutchinson, M., Joseffer, S., Rusinek, H., Saarimaki, A., Stevenson, J., et al.
(1998). Functional magnetic resonance imaging of human brain activity in a verbal flu-
ency task. Journal of Neurology, Neurosurgery, and Psychiatry, 64(4), 492–498. http://dx.doi.
org/10.1136/jnnp.64.4.492.
Schmitz, C., Martineau, J., Barthelemy, C., & Assaiante, C. (2003). Motor control and chil-
dren with autism: Deficit of anticipatory function? Neuroscience Letters, 348(1), 17–20,
S030439400300644X.
Scott-Van Zeeland, A. A., Dapretto, M., Ghahremani, D. G., Poldrack, R. A., &
Bookheimer, S. Y. (2010). Reward processing in autism. Autism Research, 3(2),
53–67. http://dx.doi.org/10.1002/aur.122.
Motor Skill in Autism Spectrum Disorders 247
Sears, L. L., Vest, C., Mohamed, S., Bailey, J., Ranson, B. J., & Piven, J. (1999). An MRI
study of the basal ganglia in autism. Progress in Neuro-Psychopharmacology & Biological Psy-
chiatry, 23(4), 613–624, S0278584699000202.
Sejnowski, T. J. (2010). Learning optimal strategies in complex environments. Proceedings of
the National Academy of Sciences of the United States of America, 107(47), 20151–20152.
http://dx.doi.org/10.1073/pnas.1014954107.
Shadmehr, R. (2010). Control of movements and temporal discounting of reward. Current
Opinion in Neurobiology, 20(6), 726–730. http://dx.doi.org/10.1016/j.conb.2010.08.017.
Shadmehr, R., & Krakauer, J. W. (2008). A computational neuroanatomy for motor control.
Experimental Brain Research, 185(3), 359–381. http://dx.doi.org/10.1007/s00221-008-
1280-5.
Shadmehr, R., Orban de Xivry, J. J., Xu-Wilson, M., & Shih, T. Y. (2010). Temporal dis-
counting of reward and the cost of time in motor control. The Journal of Neuroscience,
30(31), 10507–10516. http://dx.doi.org/10.1523/JNEUROSCI.1343-10.2010.
Shavelle, R. M., Strauss, D. J., & Pickett, J. (2001). Causes of death in autism. Journal of Autism
and Developmental Disorders, 31(6), 569–576.
Sherrington, C., Tiedemann, A., Fairhall, N., Close, J. C., & Lord, S. R. (2011). Exercise to
prevent falls in older adults: An updated meta-analysis and best practice recommenda-
tions. New South Wales Public Health Bulletin, 22(3–4), 78–83. http://dx.doi.org/
10.1071/NB10056.
Shetreat-Klein, M., Shinnar, S., & Rapin, I. (2012). Abnormalities of joint mobility and gait
in children with autism spectrum disorders. Brain & Development, http://dx.doi.org/
10.1016/j.braindev.2012.02.005.
Shi, L., Smith, S. E., Malkova, N., Tse, D., Su, Y., & Patterson, P. H. (2009). Activation
of the maternal immune system alters cerebellar development in the offspring. Brain,
Behavior, and Immunity, 23(1), 116–123. http://dx.doi.org/10.1016/j.bbi.2008.07.012.
Shih, C. H., Shih, C. T., & Chu, C. L. (2010). Assisting people with multiple disabilities
actively correct abnormal standing posture with a Nintendo Wii balance board through
controlling environmental stimulation. Research in Developmental Disabilities, 31(4),
936–942. http://dx.doi.org/10.1016/j.ridd.2010.03.004.
Shmuelof, L., & Krakauer, J. W. (2011). Are we ready for a natural history of motor learning?
Neuron, 72(3), 469–476. http://dx.doi.org/10.1016/j.neuron.2011.10.017.
Shukla, D. K., Keehn, B., & Muller, R. A. (2010). Regional homogeneity of fMRI time
series in autism spectrum disorders. Neuroscience Letters, 476(1), 46–51. http://dx.doi.
org/10.1016/j.neulet.2010.03.080.
Stoodley, C. J., Valera, E. M., & Schmahmann, J. D. (2012). Functional topography of the
cerebellum for motor and cognitive tasks: An fMRI study. NeuroImage, 59(2),
1560–1570. http://dx.doi.org/10.1016/j.neuroimage.2011.08.065.
Sutera, S., Pandey, J., Esser, E. L., Rosenthal, M. A., Wilson, L. B., Barton, M., et al. (2007).
Predictors of optimal outcome in toddlers diagnosed with autism spectrum disorders.
Journal of Autism and Developmental Disorders, 37(1), 98–107. http://dx.doi.org/
10.1007/s10803-006-0340-6.
Takarae, Y., Minshew, N. J., Luna, B., Krisky, C. M., & Sweeney, J. A. (2004). Pursuit eye
movement deficits in autism. Brain, 127(Pt. 12), 2584–2594. http://dx.doi.org/10.1093/
brain/awh307.
Takarae, Y., Minshew, N. J., Luna, B., & Sweeney, J. A. (2004). Oculomotor abnormalities
parallel cerebellar histopathology in autism. Journal of Neurology, Neurosurgery, and Psychi-
atry, 75(9), 1359–1361. http://dx.doi.org/10.1136/jnnp.2003.022491.
Takarae, Y., Minshew, N. J., Luna, B., & Sweeney, J. A. (2007). Atypical involvement of
frontostriatal systems during sensorimotor control in autism. Psychiatry Research,
156(2), 117–127. http://dx.doi.org/10.1016/j.pscychresns.2007.03.008.
248 Leanne Chukoskie et al.
Teitelbaum, O., Benton, T., Shah, P. K., Prince, A., Kelly, J. L., & Teitelbaum, P. (2004).
Eshkol-Wachman movement notation in diagnosis: The early detection of Asperger’s
syndrome. Proceedings of the National Academy of Sciences of the United States of America,
101(32), 11909–11914. http://dx.doi.org/10.1073/pnas.0403919101.
Teitelbaum, P., Teitelbaum, O., Nye, J., Fryman, J., & Maurer, R. G. (1998). Movement
analysis in infancy may be useful for early diagnosis of autism. Proceedings of the National
Academy of Sciences of the United States of America, 95(23), 13982–13987.
Todorov, E., & Jordan, M. I. (2002). Optimal feedback control as a theory of motor coor-
dination. Nature Neuroscience, 5(11), 1226–1235. http://dx.doi.org/10.1038/nn963.
Travers, B. G., Powell, P. S., Klinger, L. G., & Klinger, M. R. (2013). Motor difficulties in
autism spectrum disorder: Linking symptom severity and postural stability. Journal of
Autism and Developmental Disorders, 43(7), 1568–1583. http://dx.doi.org/10.1007/
s10803-012-1702-x.
Tsai, P. T., Hull, C., Chu, Y., Greene-Colozzi, E., Sadowski, A. R., Leech, J. M., et al.
(2012). Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant
mice. Nature, 488(7413), 647–651. http://dx.doi.org/10.1038/nature11310.
Tsai, C. L., Pan, C. Y., Cherng, R. J., & Wu, S. K. (2009). Dual-task study of cognitive and
postural interference: A preliminary investigation of the automatization deficit hypoth-
esis of developmental co-ordination disorder. Child: Care, Health and Development, 35(4),
551–560. http://dx.doi.org/10.1111/j.1365-2214.2009.00974.x.
van Batenburg-Eddes, T., Henrichs, J., Schenk, J. J., Sincer, I., de Groot, L., Hofman, A.,
et al. (2013). Early infant neuromotor assessment is associated with language and non-
verbal cognitive function in toddlers: the generation R study. Jurnal of Developmental
and Behavioral Pediatrics, 34(5), 326–334.
van Swieten, L. M., van Bergen, E., Williams, J. H., Wilson, A. D., Plumb, M. S.,
Kent, S. W., et al. (2010). A test of motor (not executive) planning in developmental
coordination disorder and autism. Journal of Experimental Psychology Human Perception
and Performance, 36(2), 493–499. http://dx.doi.org/10.1037/a0017177.
Vernazza-Martin, S., Martin, N., Vernazza, A., Lepellec-Muller, A., Rufo, M., Massion, J.,
et al. (2005). Goal directed locomotion and balance control in autistic children. Journal of
Autism and Developmental Disorders, 35(1), 91–102.
Vilensky, J. A., Damasio, A. R., & Maurer, R. G. (1981). Gait disturbances in patients with
autistic behavior: A preliminary study. Archives of Neurology, 38(10), 646–649.
Weimer, A. K., Schatz, A. M., Lincoln, A., Ballantyne, A. O., & Trauner, D. A. (2001).
“Motor” impairment in Asperger syndrome: Evidence for a deficit in proprioception.
Journal of Developmental and Behavioral Pediatrics, 22(2), 92–101.
Weiss, M. J., Moran, M. F., Parker, M. E., & Foley, J. T. (2013). Gait analysis of teenagers
and young adults diagnosed with autism and severe verbal communication disorders.
Frontiers in Integrative Neuroscience, 7, 33. http://dx.doi.org/10.3389/fnint.2013.00033.
Whittingham, K., Fahey, M., Rawicki, B., & Boyd, R. (2010). The relationship between
motor abilities and early social development in a preschool cohort of children with cere-
bral palsy. Research in Developmental Disabilities, 31(6), 1346–1351. http://dx.doi.org/
10.1016/j.ridd.2010.07.006.
Williams, R. S., Hauser, S. L., Purpura, D. P., DeLong, G. R., & Swisher, C. N. (1980).
Autism and mental retardation: Neuropathologic studies performed in four retarded per-
sons with autistic behavior. Archives of Neurology, 37(12), 749–753.
Wolff, J. J., Gu, H., Gerig, G., Elison, J. T., Styner, M., Gouttard, S., et al. (2012). Differ-
ences in white matter fiber tract development present from 6 to 24 months in infants with
autism. The American Journal of Psychiatry, 169(6), 589–600. http://dx.doi.org/10.1176/
appi.ajp.2011.11091447.
Motor Skill in Autism Spectrum Disorders 249
Wuang, Y. P., Chiang, C. S., Su, C. Y., & Wang, C. C. (2011). Effectiveness of virtual reality
using Wii gaming technology in children with Down syndrome. Research in Developmen-
tal Disabilities, 32(1), 312–321. http://dx.doi.org/10.1016/j.ridd.2010.10.002.
Xiang, H., Lin, C., Ma, X., Zhang, Z., Bower, J. M., Weng, X., et al. (2003). Involvement of
the cerebellum in semantic discrimination: An fMRI study. Human Brain Mapping, 18(3),
208–214. http://dx.doi.org/10.1002/hbm.10095.
Yin, H. H., Knowlton, B. J., & Balleine, B. W. (2005). Blockade of NMDA receptors in the
dorsomedial striatum prevents action-outcome learning in instrumental conditioning.
The European Journal of Neuroscience, 22(2), 505–512. http://dx.doi.org/10.1111/
j.1460-9568.2005.04219.x.
Zikopoulos, B., & Barbas, H. (2010). Changes in prefrontal axons may disrupt the network in
autism. The Journal of Neuroscience, 30(44), 14595–14609. http://dx.doi.org/10.1523/
JNEUROSCI.2257-10.2010.
Zilbovicius, M., Murayama, N., Garreau, B., Leroy-Willig, A., Barthelemy, C.,
Salamon, G., et al. (1995). Hypoplasia of vermal lobules I-V, but not of lobules
VI-VII, in childhood autism. Neurology, 45(Suppl. 4), A162.
Zwaigenbaum, L., Bryson, S., Rogers, T., Roberts, W., Brian, J., & Szatmari, P. (2005).
Behavioral manifestations of autism in the first year of life. International Journal of Develop-
mental Neuroscience, 23(2–3), 143–152. http://dx.doi.org/10.1016/j.ijdevneu.2004.05.001.
CHAPTER EIGHT
Orchestration of
Neurodevelopmental Programs
by RBFOX1: Implications for
Autism Spectrum Disorder
Brent R. Bill*, Jennifer K. Lowe*,†, Christina T. DyBuncio*,†,
Brent L. Fogel†,1
*Department of Psychiatry, David Geffen School of Medicine, Center for Autism Research and Treatment
and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University
of California, Los Angeles, California, USA
†
Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of
California, Los Angeles, California, USA
1
Corresponding author: e-mail address: bfogel@ucla.edu
Contents
1. Neurodevelopment and the RBFOX1 RNA Splicing Factor 252
2. RBFOX1 Genetic Variation and Autism Spectrum Disorder 253
3. The Contributions of RBFOX1 Model Systems to Molecular Pathogenesis 258
4. A Model for the Dysregulation of RBFOX1 in Human Neurodevelopmental Disease 259
Acknowledgments 263
References 263
Abstract
Neurodevelopmental and neuropsychiatric disorders result from complex interactions
between critical genetic factors and as-yet-unknown environmental components. To
gain clinical insight, it is critical to develop a comprehensive understanding of these
genetic components. RBFOX1, an RNA splicing factor, regulates expression of large
genetic networks during early neuronal development, and haploinsufficiency causes
severe neurodevelopmental phenotypes including autism spectrum disorder (ASD),
intellectual disability, and epilepsy. Genomic testing in individuals and large patient
cohorts has identified phenotypically similar cases possessing copy number variations
in RBFOX1, implicating the gene as an important cause of neurodevelopmental dis-
ease. However, a significant proportion of the observed structural variation is inherited
from phenotypically normal individuals, raising questions regarding overall pathoge-
nicity of variation at the RBFOX1 locus. In this chapter, we discuss the molecular, cel-
lular, and clinical evidence supporting the role of RBFOX1 in neurodevelopment and
present a comprehensive model for the contribution of structural variation in RBFOX1
to ASD.
Figure 8.1 RBFOX1 genomic architecture and copy number variation associated with
autism spectrum disorder. (A) RBFOX1 gene structure is complex with multiple transcrip-
tional start sites, translational start sites, multiple isoforms, and alternate endings. The
distribution of CNVs associated with autism spectrum disorder in the RBFOX1 locus
(HG19-chr16: 5289804–7763340) is shown and compared to the database of genomic
variants. (B) Exons 1A–1E. (C) Exons 8–21. Observed structural variation is shown clus-
tered at the 5' untranslated region between exons 1 and 7. Single nucleotide polymor-
phisms related to the locus and showing genome-wide associated in ASD also follow a
similar distribution pattern (green). Red bars, deletions; blue bars, duplications; brown
bars, insertions and deletions; black bars, unknown feature.
RBFOX1 and ASD 255
with developmental delay but without epilepsy was given a clinical diagnosis
of autism (Zhao, 2013). The extent of the clinical evaluation for ASD each
patient received in this cohort was unfortunately not reported, so it cannot
be assumed the other patients lacked such phenotypes.
In general, it has been presumed that the aforementioned CNVs lead to
reduced RBFOX1 expression. If true, then it would be consistent with the
observations that RBFOX1 haploinsufficiency results in a syndrome charac-
terized primarily by neurodevelopmental and neurological phenotypes
including ASD, intellectual disability, and epilepsy (Bhalla et al., 2004;
Martin et al., 2007). Data from knockdown studies in human neural progen-
itor cell lines modeling haploinsufficiency during neuronal differentiation
demonstrate widespread changes in RNA splicing and gene expression
(Fogel et al., 2012), and studies of the Rbfox1 neural-specific knockout
mouse show alterations in synaptic transmission, increased membrane excit-
ability, and a predisposition to seizures (Gehman et al., 2011). Interestingly,
whole transcriptome analysis in the brains of autistic patients reveals
decreased levels of RBFOX1 and dysregulation of RBFOX1-dependent
alternative splicing (Voineagu et al., 2011), similar to the effects seen in
haploinsufficient neuronal cell lines (Fogel et al., 2012). However, in the
majority of cases, the impact of the identified CNV to RBFOX1 expression
or function is unclear, as evidenced by the presence of identical CNVs in
controls and unaffected family members (Fig. 8.1B and C).
To better understand the contribution of structural variation in RBFOX1
to the development of autism and related disorders, we compiled all publi-
shed CNVs including those from two of the largest ASD cohorts, the Autism
Genetic Resource Exchange (AGRE), a primarily multiplex cohort of fam-
ilies with multiple affected siblings, and the Simons Simplex Cohort (SSC),
which contains families with only a single affected child (Fig. 8.1B and C). In
the AGRE cohort, we found that 2.2% of patients carried a CNV in the
RBFOX1 locus compared to 0.7% of unaffected siblings (OR 3.19,
p ¼ 0.006, 95% CI (1.27, 10.28)). In contrast, the SSC did not show a signif-
icantly increased odds ratio, with 2.6% of probands and 2.4% of their normal
siblings having a CNV (OR 1.11, p ¼ 0.77, 95% CI (0.59, 2.12)). While we
cannot rule out ascertainment bias or differences in population structure,
these data demonstrate a significant enrichment of RBFOX1 CNVs in a mul-
tiplex but not in a simplex cohort. This analysis reveals two additional striking
features. The first is that, irrespective of cohort, CNVs in RBFOX1 tend to be
inherited from an unaffected parent. Second, we observe a locational bias of
CNVs toward the 50 untranslated exons 1–7 (Fig. 8.1B) compared to the
RBFOX1 and ASD 257
Cellular stress
Adaptive Adaptive
transcriptional alternative
regulation splicing
Altered
RBFOX1 RBFOX1
function
adapts cell
to stressor
RNA splicing
Autism
spectrum
disorder
Transcription
Routine 1. Impaired routine function
function (e.g., Haploinsufficiency)
2. Impaired adaptive response
(e.g., Potential role for CNVs?)
Normal brain
development
Figure 8.2 Proposed model for RBFOX1 dysregulation in autism spectrum disorder. Dur-
ing neurodevelopment, RBFOX1 regulates large genetic networks via direct effects on
RNA splicing, mRNA stability, and transcriptional regulation as well as indirect effects on
gene transcription leading to normal neuronal development. If this function is disrupted
(e.g., by haploinsufficiency of RBFOX1), development is sufficiently perturbed resulting
in autism spectrum disorder (ASD). As discussed in the text, RBFOX1 may play a further
role in the adaptive response to cellular stress by regulating RNA splicing or transcrip-
tional effects in response to environmental stimuli to maintain normal function. If this
process is perturbed, this may also result in ASD. Inherited structural variation (CNVs)
may damage RBFOX1 RNA processing or transcriptional signals and thus impair this
adaptive response, thereby increasing the risk of ASD occurring upon exposure to cer-
tain environmental stressors during critical stages in neurodevelopment.
via effects on RNA stability (Pistoni et al., 2013) and alternative splicing
(Damianov & Black, 2010; Lee et al., 2009). Rbfox1 can autoregulate the
splicing of its own RNA-binding domain creating a functional dominant
negative, and thus regulate its own activity (Damianov & Black, 2010). Fur-
thermore, Lee et al. have demonstrated that cellular depolarization, known
to widely affect RNA splicing, causes alternative splicing of murine Rbfox1
in neurons, resulting in a change in subcellular localization of the protein and
subsequent reversal of the effects of depolarization on the splicing of other
Rbfox1 targets, which they interpret as a novel adaptive mechanism to
chronic stimuli (Lee et al., 2009). Regulation of RBFOX1 alternative splic-
ing in response to cellular stressors could therefore play a meaningful role in
neurodevelopment and provide another possible regulatory mechanism
CNVs may affect (Fig. 8.2).
Given the extensive networks of genes regulated both directly and indi-
rectly by RBFOX1 (Fogel et al., 2012; Ray et al., 2013), it is likely that var-
ious stimuli could occur throughout the course of neuronal development
that require an adaptive response from RBFOX1, alone or in conjunction
with other factors, to maintain normal expression profiles. Therefore, it
may be supposed that mutations in RBFOX1 could exist which impair
these adaptive responses while having no, or minimal, effect on routine
function. Applying this scenario to human neurodevelopment, noncoding
variation that directly disrupts RBFOX1 expression would act to cause
haploinsufficiency and therefore disease, whereas variation altering accessory
regulatory responses (e.g., various target binding sequences) would only lead
to disease under cellular conditions requiring function of those elements. If
such conditions are rare, or only transient, then the ability to mount a limited
adaptive response may be tolerated, but if the condition is such that the adap-
tive response is essential for normal brain development, this inability to
respond could result in disease. This synergizes well with the suspected con-
tribution of environmental factors to ASD if exposure occurs during critical
points in brain development (Herbert, 2010) and suggests a mechanism
whereby variation in the RBFOX1 gene could modify ASD risk under cer-
tain conditions, but not others. Families tend to stay in the same environ-
ment for long periods of time, and, therefore, children are often exposed
to the same environmental stressors as their siblings. If RBFOX1 CNVs con-
fer ASD risk in a particular environmental context, the pathogenicity of such
variants would be most apparent in families with multiple affected children.
This model could explain why enrichment of RBFOX1 CNVs is seen in a
multiplex, but not a simplex, cohort, as shown earlier. Further work will be
RBFOX1 and ASD 263
ACKNOWLEDGMENTS
The authors wish to thank Stephan Sanders for his invaluable assistance with the assessment of
copy number variation in the autism cohorts as well as Daniel H. Geschwind (D.H.G.) for his
support and helpful suggestions. Data in this chapter were obtained from the ISCA
Consortium database (www.iscaconsortium.org), which generates this information using
NCBI’s database of genomic structural variation (dbVar, www.ncbi.nlm.nih.gov/dbvar/),
study nstd37. Samples and associated phenotype data were provided by ISCA. We
gratefully acknowledge the resources provided by the Autism Genetic Resource
Exchange (AGRE) Consortium and the participating AGRE families. The Autism
Genetic Resource Exchange is a program of Autism Speaks and is supported, in part, by
Grant 1U24MH081810 from the National Institute of Mental Health to Clara M.
Lajonchere (PI). This work was supported by the National Institutes of Health
(9R01MH100027 to D.H.G. and K08MH086297 to B.L.F.) and Simons SFARI award
206744 to D.H.G.
REFERENCES
Amir-Zilberstein, L., Blechman, J., Sztainberg, Y., Norton, W. H., Reuveny, A.,
Borodovsky, N., et al. (2012). Homeodomain protein otp and activity-dependent splic-
ing modulate neuronal adaptation to stress. Neuron, 73(2), 279–291.
Andersen, C. L., Lamy, P., Thorsen, K., Kjeldsen, E., Wikman, F., Villesen, P., et al. (2011).
Frequent genomic loss at chr16p13.2 is associated with poor prognosis in colorectal can-
cer. International Journal of Cancer, 129(8), 1848–1858.
Bajpai, R., Sambrani, N., Stadelmayer, B., & Shashidhara, L. S. (2004). Identification of a
novel target of D/V signaling in Drosophila wing disc: Wg-independent function of
the organizer. Gene Expression Patterns, 5(1), 113–121.
Bass, A. J., Lawrence, M. S., Brace, L. E., Ramos, A. H., Drier, Y., Cibulskis, K., et al.
(2011). Genomic sequencing of colorectal adenocarcinomas identifies a recurrent
VTI1A-TCF7L2 fusion. Nature Genetics, 43(10), 964–968.
Bhalla, K., Phillips, H. A., Crawford, J., McKenzie, O. L., Mulley, J. C., Eyre, H., et al.
(2004). The de novo chromosome 16 translocations of two patients with abnormal phe-
notypes (mental retardation and epilepsy) disrupt the A2BP1 gene. Journal of Human
Genetics, 49(6), 308–311.
Bill, B. R., & Geschwind, D. H. (2009). Genetic advances in autism: Heterogeneity and con-
vergence on shared pathways. Current Opinion in Genetics & Development, 19(3), 271–278.
Chen-Plotkin, A. S., Geser, F., Plotkin, J. B., Clark, C. M., Kwong, L. K., Yuan, W., et al.
(2008). Variations in the progranulin gene affect global gene expression in
frontotemporal lobar degeneration. Human Molecular Genetics, 17(10), 1349–1362.
264 Brent R. Bill et al.
Damianov, A., & Black, D. L. (2010). Autoregulation of Fox protein expression to produce
dominant negative splicing factors. RNA, 16(2), 405–416.
Davis, L. K., Maltman, N., Mosconi, M. W., Macmillan, C., Schmitt, L., Moore, K., et al.
(2012). Rare inherited A2BP1 deletion in a proband with autism and developmental
hemiparesis. American Journal of Medical Genetics Part A, 158A(7), 1654–1661.
Day, A., Carlson, M. R., Dong, J., O’Connor, B. D., & Nelson, S. F. (2007). Celsius:
A community resource for Affymetrix microarray data. Genome Biology, 8(6), R112.
Day, A., Dong, J., Funari, V. A., Harry, B., Strom, S. P., Cohn, D. H., et al. (2009). Disease
gene characterization through large-scale co-expression analysis. PLoS ONE, 4(12),
e8491.
Elia, J., Gai, X., Xie, H. M., Perin, J. C., Geiger, E., Glessner, J. T., et al. (2010). Rare struc-
tural variants found in attention-deficit hyperactivity disorder are preferentially associ-
ated with neurodevelopmental genes. Molecular Psychiatry, 15(6), 637–646.
Fatemi, S. H., Aldinger, K. A., Ashwood, P., Bauman, M. L., Blaha, C. D., Blatt, G. J., et al.
(2012). Consensus paper: Pathological role of the cerebellum in autism. Cerebellum,
11(3), 777–807.
Fogel, B. L., & Geschwind, D. H. (2012). Clinical neurogenetics. In R. Daroff, G. Fenichel,
J. Jankovic, & J. Mazziotta (Eds.), Neurology in clinical practice (6th ed., pp. 704–734).
Philadelphia, PA: Elsevier.
Fogel, B. L., & Perlman, S. (2011). Cerebellar disorders: Balancing the approach to cerebellar
ataxia. In N. Gálvez-Jiménez & P. J. Tuite (Eds.), Uncommon causes of movement disorders
(1st ed., pp. 198–216). Cambridge, NY: Cambridge University Press.
Fogel, B. L., Wexler, E., Wahnich, A., Friedrich, T., Vijayendran, C., Gao, F., et al. (2012).
RBFOX1 regulates both splicing and transcriptional networks in human neuronal devel-
opment. Human Molecular Genetics, 21(19), 4171–4186.
Gallagher, T. L., Arribere, J. A., Geurts, P. A., Exner, C. R., McDonald, K. L., Dill, K. K.,
et al. (2011). Rbfox-regulated alternative splicing is critical for zebrafish cardiac and skel-
etal muscle functions. Developmental Biology, 359(2), 251–261.
Gehman, L. T., Stoilov, P., Maguire, J., Damianov, A., Lin, C. H., Shiue, L., et al. (2011).
The splicing regulator Rbfox1 (A2BP1) controls neuronal excitation in the mammalian
brain. Nature Genetics, 43(7), 706–711.
Hamshere, M. L., Green, E. K., Jones, I. R., Jones, L., Moskvina, V., Kirov, G., et al. (2009).
Genetic utility of broadly defined bipolar schizoaffective disorder as a diagnostic concept.
The British Journal of Psychiatry, 195(1), 23–29.
Herbert, M. R. (2010). Contributions of the environment and environmentally vulnerable
physiology to autism spectrum disorders. Current Opinion in Neurology, 23(2), 103–110.
Hodgkin, J., Zellan, J. D., & Albertson, D. G. (1994). Identification of a candidate primary
sex determination locus, fox-1, on the X chromosome of Caenorhabditis elegans.
Development, 120(12), 3681–3689.
Holsboer, F., & Ising, M. (2008). Central CRH system in depression and anxiety—Evidence
from clinical studies with CRH1 receptor antagonists. European Journal of Pharmacology,
583(2–3), 350–357.
Hu, J., Ho, A. L., Yuan, L., Hu, B., Hua, S., Hwang, S. S., et al. (2013). Neutralization of
terminal differentiation in gliomagenesis. Proceedings of the National Academy of Sciences of
the United States of America, 110(36), 14520–14527.
Iafrate, A. J., Feuk, L., Rivera, M. N., Listewnik, M. L., Donahoe, P. K., Qi, Y., et al. (2004).
Detection of large-scale variation in the human genome. Nature Genetics, 36(9), 949–951.
International Schizophrenia Consortium. (2008). Rare chromosomal deletions and duplica-
tions increase risk of schizophrenia. Nature, 455(7210), 237–241.
Jin, Y., Suzuki, H., Maegawa, S., Endo, H., Sugano, S., Hashimoto, K., et al. (2003).
A vertebrate RNA-binding protein Fox-1 regulates tissue-specific splicing via the
pentanucleotide GCAUG. The EMBO Journal, 22(4), 905–912.
RBFOX1 and ASD 265
Kohannim, O., Hibar, D. P., Jahanshad, N., Stein, J. L., Hua, X., Toga, A. W., et al. (2012).
Predicting temporal lobe volume on Mri from genotypes using L(1)–L(2) regularized
regression. Proceedings of IEEE International Symposium on Biomedical Imaging, 1160–1163.
Kuroyanagi, H. (2009). Fox-1 family of RNA-binding proteins. Cellular and Molecular Life
Sciences, 66(24), 3895–3907.
Lal, D., Trucks, H., Moller, R. S., Hjalgrim, H., Koeleman, B. P., de Kovel, C. G., et al.
(2013). Rare exonic deletions of the RBFOX1 gene increase risk of idiopathic gener-
alized epilepsy. Epilepsia, 54(2), 265–271.
Lee, Y., Mattai, A., Long, R., Rapoport, J. L., Gogtay, N., & Addington, A. M. (2012).
Microduplications disrupting the MYT1L gene (2p25.3) are associated with schizophre-
nia. Psychiatric Genetics, 22(4), 206–209.
Lee, J. A., Tang, Z. Z., & Black, D. L. (2009). An inducible change in Fox-1/A2BP1 splicing
modulates the alternative splicing of downstream neuronal target exons. Genes & Devel-
opment, 23(19), 2284–2293.
Le-Niculescu, H., Patel, S. D., Bhat, M., Kuczenski, R., Faraone, S. V., Tsuang, M. T., et al.
(2009). Convergent functional genomics of genome-wide association data for bipolar
disorder: Comprehensive identification of candidate genes, pathways and mechanisms.
American Journal of Medical Genetics Part B, Neuropsychiatric Genetics, 150B(2), 155–181.
Lim, C., & Allada, R. (2013). ATAXIN-2 activates PERIOD translation to sustain circadian
rhythms in Drosophila. Science, 340(6134), 875–879.
Lim, J., Hao, T., Shaw, C., Patel, A. J., Szabo, G., Rual, J. F., et al. (2006). A protein-protein
interaction network for human inherited ataxias and disorders of Purkinje cell degener-
ation. Cell, 125(4), 801–814.
Linnebacher, M., Ostwald, C., Koczan, D., Salem, T., Schneider, B., Krohn, M., et al.
(2013). Single nucleotide polymorphism array analysis of microsatellite-stable,
diploid/near-diploid colorectal carcinomas without the CpG island methylator pheno-
type. Oncology Letters, 5(1), 173–178.
Ma, L., Hanson, R. L., Traurig, M. T., Muller, Y. L., Kaur, B. P., Perez, J. M., et al. (2010).
Evaluation of A2BP1 as an obesity gene. Diabetes, 59(11), 2837–2845.
Martin, C. L., Duvall, J. A., Ilkin, Y., Simon, J. S., Arreaza, M. G., Wilkes, K., et al. (2007).
Cytogenetic and molecular characterization of A2BP1/FOX1 as a candidate gene for
autism. American Journal of Medical Genetics Part B, Neuropsychiatric Genetics, 144(7), 869–876.
Mazzone, L., Ruta, L., & Reale, L. (2012). Psychiatric comorbidities in asperger syndrome and
high functioning autism: Diagnostic challenges. Annals of General Psychiatry, 11(1), 16.
Melhem, N., Middleton, F., McFadden, K., Klei, L., Faraone, S. V., Vinogradov, S., et al.
(2011). Copy number variants for schizophrenia and related psychotic disorders in Oce-
anic Palau: Risk and transmission in extended pedigrees. Biological Psychiatry, 70(12),
1115–1121.
Meyer, K. J., Axelsen, M. S., Sheffield, V. C., Patil, S. R., & Wassink, T. H. (2012).
Germline mosaic transmission of a novel duplication of PXDN and MYT1L to two male
half-siblings with autism. Psychiatric Genetics, 22(3), 137–140.
Mikhail, F. M., Lose, E. J., Robin, N. H., Descartes, M. D., Rutledge, K. D.,
Rutledge, S. L., et al. (2011). Clinically relevant single gene or intragenic deletions
encompassing critical neurodevelopmental genes in patients with developmental delay,
mental retardation, and/or autism spectrum disorders. American Journal of Medical Genetics
Part A, 155A(10), 2386–2396.
Obayashi, T., & Kinoshita, K. (2011). COXPRESdb: A database to compare gene
coexpression in seven model animals. Nucleic Acids Research, 39(Database issue),
D1016–D1022.
Pescosolido, M. F., Yang, U., Sabbagh, M., & Morrow, E. M. (2012). Lighting a path:
Genetic studies pinpoint neurodevelopmental mechanisms in autism and related disor-
ders. Dialogues in Clinical Neuroscience, 14(3), 239–252.
266 Brent R. Bill et al.
Pistoni, M., Shiue, L., Cline, M. S., Bortolanza, S., Neguembor, M. V., Xynos, A., et al.
(2013). Rbfox1 downregulation and altered calpain 3 splicing by FRG1 in a mouse
model of Facioscapulohumeral muscular dystrophy (FSHD). PLoS Genetics, 9(1),
e1003186.
Priebe, L., Degenhardt, F., Strohmaier, J., Breuer, R., Herms, S., Witt, S. H., et al. (2013).
Copy number variants in German patients with schizophrenia. PLoS ONE, 8(7),
e64035.
Ray, D., Kazan, H., Cook, K. B., Weirauch, M. T., Najafabadi, H. S., Li, X., et al. (2013).
A compendium of RNA-binding motifs for decoding gene regulation. Nature,
499(7457), 172–177.
Richdale, A. L., & Schreck, K. A. (2009). Sleep problems in autism spectrum disorders: Prev-
alence, nature, & possible biopsychosocial aetiologies. Sleep Medicine Reviews, 13(6),
403–411.
Sebat, J., Lakshmi, B., Malhotra, D., Troge, J., Lese-Martin, C., Walsh, T., et al. (2007).
Strong association of de novo copy number mutations with autism. Science,
316(5823), 445–449.
Shibata, H., Huynh, D. P., & Pulst, S. M. (2000). A novel protein with RNA-binding motifs
interacts with ataxin-2. Human Molecular Genetics, 9(9), 1303–1313.
Stambolian, D., Wojciechowski, R., Oexle, K., Pirastu, M., Li, X., Raffel, L. J., et al. (2013).
Meta-analysis of genome-wide association studies in five cohorts reveals common var-
iants in RBFOX1, a regulator of tissue-specific splicing, associated with refractive error.
Human Molecular Genetics, 22(13), 2754–2764.
Stevens, S. J., van Ravenswaaij-Arts, C. M., Janssen, J. W., Klein Wassink-Ruiter, J. S., van
Essen, A. J., Dijkhuizen, T., et al. (2011). MYT1L is a candidate gene for intellectual
disability in patients with 2p25.3 (2pter) deletions. American Journal of Medical Genetics
Part A, 155A(11), 2739–2745. http://dx.doi.org/10.1002/ajmg.a.34274.
Sun, S., Zhang, Z., Fregoso, O., & Krainer, A. R. (2012). Mechanisms of activation and
repression by the alternative splicing factors RBFOX1/2. RNA, 18(2), 274–283.
Takahashi, K., & Yamanaka, S. (2006). Induction of pluripotent stem cells from mouse
embryonic and adult fibroblast cultures by defined factors. Cell, 126(4), 663–676.
Underwood, J. G., Boutz, P. L., Dougherty, J. D., Stoilov, P., & Black, D. L. (2005). Homo-
logues of the Caenorhabditis elegans Fox-1 protein are neuronal splicing regulators in
mammals. Molecular and Cellular Biology, 25(22), 10005–10016.
Usha, N., & Shashidhara, L. S. (2010). Interaction between Ataxin-2 binding protein 1 and
Cubitus-interruptus during wing development in Drosophila. Developmental Biology,
341(2), 389–399.
Van Den Bossche, M. J., Strazisar, M., Cammaerts, S., Liekens, A. M., Vandeweyer, G.,
Depreeuw, V., et al. (2013). Identification of rare copy number variants in high burden
schizophrenia families. American Journal of Medical Genetics Part B, Neuropsychiatric Genetics,
162B(3), 273–282. http://dx.doi.org/10.1002/ajmg.b.32146.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature,
474(7351), 380–384.
Vrijenhoek, T., Buizer-Voskamp, J. E., van der Stelt, I., Strengman, E., Sabatti, C., Geurts
van Kessel, A., et al. (2008). Recurrent CNVs disrupt three candidate genes in schizo-
phrenia patients. American Journal of Human Genetics, 83(4), 504–510.
Wang, E. T., Sandberg, R., Luo, S., Khrebtukova, I., Zhang, L., Mayr, C., et al. (2008).
Alternative isoform regulation in human tissue transcriptomes. Nature, 456(7221),
470–476.
Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner, J. T., Abrahams, B. S., et al. (2009).
Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature,
459(7246), 528–533.
RBFOX1 and ASD 267
Wintle, R. F., Lionel, A. C., Hu, P., Ginsberg, S. D., Pinto, D., Thiruvahindrapduram, B.,
et al. (2011). A genotype resource for postmortem brain samples from the Autism Tissue
Program. Autism Research, 4(2), 89–97.
Xu, B., Roos, J. L., Levy, S., van Rensburg, E. J., Gogos, J. A., & Karayiorgou, M. (2008).
Strong association of de novo copy number mutations with sporadic schizophrenia.
Nature Genetics, 40(7), 880–885.
Yeo, G. W., Nostrand, E. L., & Liang, T. Y. (2007). Discovery and analysis of evolutionarily
conserved intronic splicing regulatory elements. PLoS Genetics, 3(5), e85.
Yeo, G. W., Xu, X., Liang, T. Y., Muotri, A. R., Carson, C. T., Coufal, N. G., et al. (2007).
Alternative splicing events identified in human embryonic stem cells and neural progen-
itors. PLoS Computational Biology, 3(10), 1951–1967.
Yi, S., & Li, W. H. (2005). Molecular evolution of recombination hotspots and highly
recombining pseudoautosomal regions in hominoids. Molecular Biology and Evolution,
22(5), 1223–1230.
Zhang, C., Zhang, Z., Castle, J., Sun, S., Johnson, J., Krainer, A. R., et al. (2008). Defining
the regulatory network of the tissue-specific splicing factors Fox-1 and Fox-2. Genes &
Development, 22(18), 2550–2563.
Zhao, W. W. (2013). Intragenic deletion of RBFOX1 associated with neurodevelopmental/
neuropsychiatric disorders and possibly other clinical presentations. Molecular Cytogenetics,
6(1), 26.
Zhou, H. L., & Lou, H. (2008). Repression of prespliceosome complex formation at two
distinct steps by Fox-1/Fox-2 proteins. Molecular and Cellular Biology, 28(17),
5507–5516.
CHAPTER NINE
Contents
1. Introduction: The Autism Spectrum 270
2. Genetic and Environmental Contributions to ASD 272
3. Immune Activation as a Primary Risk Factor for ASD 273
3.1 Maternal immune activation 273
3.2 Maternal autoantibody production 276
4. Immune-Related Genetic Risk Factors for ASD 278
4.1 Major histocompatibility complex molecules 279
5. Postnatal Immune Dysregulation in ASD 280
5.1 Neuroimmune abnormalities 280
5.2 Peripheral immune abnormalities in ASD 285
6. Immune Contributions to ASD Pathogenesis 287
7. Immune-Related Therapies for ASD 289
8. Conclusion 290
References 292
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder diagnosed based
on the presence and severity of core abnormalities in social communication and repet-
itive behavior, yet several studies converge on immune dysregulation as a feature of
ASD. Widespread alterations in immune molecules and responses are seen in the brains
and periphery of ASD individuals, and early life immune disruptions are associated with
ASD. This chapter discusses immune-related environmental and genetic risk factors for
ASD, emphasizing population-wide studies and animal research that reveal potential
mechanistic pathways involved in the development of ASD-related symptoms. It further
reviews immunologic pathologies seen in ASD individuals and how such abnormalities
can impact neurodevelopment and behavior. Finally, it evaluates emerging evidence for
an immune contribution to the pathogenesis of ASD and a potential role for immuno-
modulatory effects in current treatments for ASD.
Intellectual Immune
disability dysfunction
Seizures/ Gastrointestinal
epilepsy symptoms
Repetitive/
Impaired social Metabolic
stereotyped
Aggression communication abnormalities
behavior
Hyperactivity Macrocephaly
Sleep
Anxiety
deficiency
Figure 9.1 Comorbidities of ASD. Although ASD is diagnosed based on the presence
of stereotypical behavior and impairments in social communication, a wide variety of
medical comorbidities are observed in ASD individuals. The striking heterogeneity
of ASD points to the need to tailor research to well-defined subsets of ASD individuals.
and genetic risk factors for ASD are examined, drawing from both human
and animal studies that reveal a role for immune activation in the etiology of
ASD-related behavioral and neuropathologic abnormalities. In addition,
several postnatal immune disturbances that have been reported in ASD indi-
viduals are reviewed and evaluated in the context of emerging evidence
supporting a primary role for immune molecules in the regulation of neu-
rodevelopmental processes and an increasing appreciation for the influence
of peripheral immunity on the brain and behavior.
of active infection; injection of microbial antigens, such as the cell wall com-
ponent lipopolysaccharide (LPS), or of viral mimics, such as the double-
stranded RNA poly(I:C), sufficiently yields offspring with neuropathologic,
behavioral, and peripheral abnormalities analogous to those observed in
human ASD (Harvey & Boksa, 2012; Hsiao & Patterson, 2012a). Moreover,
maternal exposure to particular recombinant cytokines alone can recapitu-
late many features of MIA in fetal and adult offspring, including brain gene
expression profiles and ASD-related behavioral deficits (Garay, Hsiao,
Patterson, & McAllister, 2012; Patterson, 2011). Altogether, these several
large epidemiological studies and corroborative animal models point to a
primary role for MIA in the etiology of ASD.
Animal models of MIA exhibit strong face and construct validity for
human ASD. Importantly, offspring of immune-activated mothers exhibit
the cardinal diagnostic symptoms of ASD; in mice, MIA offspring exhibit
decreased number and quality of ultrasonic vocalizations, as a primary mode
of communication, in addition to altered olfactory communication,
impaired social interactions, and elevated repetitive marble burying and
self-grooming, among other ASD-related behavioral abnormalities
(Malkova, Yu, Hsiao, Moore, & Patterson, 2012). Similar impairments
are replicated in monkey models for MIA, where rhesus macaque offspring
of immune-activated mothers display abnormal eye tracking, decreased
vocalizations, abnormal social behavior, and increased stereotypical behavior
(Bauman et al., 2011). In addition, numerous ASD-related neurochemical
alterations, synaptic abnormalities, and neuropathologies are observed in
rodent models of MIA (Baharnoori, Bhardwaj, and Srivastava, 2013;
Forrest et al., 2012; Harvey & Boksa, 2012).
An increasing number of findings from animal models are uncovering
potential mechanisms by which MIA can lead to core symptoms of ASD
in the offspring. It is now well understood that maternal responses to
immune insults are rapidly transferred to the developing embryo; shortly
after maternal injection with LPS or poly(I:C), proinflammatory cytokines
are elevated in the placenta, amniotic fluid, and fetal brain itself (Arrode-
Bruses & Bruses, 2012; Mandal, Marzouk, Donnelly, & Ponzio, 2011;
Meyer, Feldon, & Fatemi, 2009; Meyer et al., 2006). In the placenta, the
cytokine interleukin (IL)-6 is highly upregulated, and its signaling through
placental trophoblast cells alters levels of endocrine factors critical toward
normal embryonic development (Hsiao & Patterson, 2011). In addition,
elevated levels of placental IL-1b and tumor necrosis factor (TNF)-a after
MIA cause severe placental damage, resulting in dysregulated vascular
Immune Dysregulation in Autism Spectrum Disorder 275
et al., 2013). Similar mechanisms have been identified across various species,
including humans, leading to the notion that MHC is central to social sig-
naling and the formation of social memories (Ruff, Nelson, Kubinak, &
Potts, 2012). It is interesting to speculate that ASD-associated HLA poly-
morphisms might therefore influence abnormal social interactions, as a core
diagnostic domain of autism. Directly testing this hypothesis is difficult,
however, in light of challenges inherent to translating HLA genotypes to
animal models. Advances in the development of “humanized” mouse
models containing HLA alleles have been useful for studying T-cell
responses restricted by human MHC molecules, but no such systems have
been developed for the study of neural phenotypes (Kotturi et al., 2009).
Future studies aimed toward assessing the influence of particular MHC dis-
ruptions on autism-related symptoms will be important for better under-
standing the neurological bases for subtypes of ASD.
Altogether, both environmental and genetic risk factors support a role for
immune dysregulation in the development of ASD symptoms. MIA and
maternal autoantibody production represent immune-related environmen-
tal risk factors whose influences during prenatal or early postnatal life can
contribute to the development of ASD in the offspring. Polymorphisms
in the HLA system, and in other immune-related genes, can have severe
consequences on neurodevelopment, as well as the developing immune sys-
tem. Understanding how early life immune insults impact fetal brain devel-
opment and ultimately result in cardinal symptoms of ASD will shed light on
potential targets for better diagnosis and treatment of subsets of ASD
individuals.
(Napolioni et al., 2013). Specific cytokine alterations are seen, however, upon
subclassification of ASD individuals into those exhibiting nonverbal commu-
nication, early regression, and gastrointestinal complications.
Aside from cytokine and chemokines, altered levels of specific Ig sub-
types, and decreased levels of total Ig, are reported in ASD, despite no dif-
ferences in general functionality of Ig-producing B-cells (Heuer, Rose,
Ashwood, & Van de Water, 2012). Moreover, increased levels of comple-
ment proteins, including the lytic component C1q (Corbett et al., 2007) and
complement factor I (Momeni et al., 2012), and decreased levels of circu-
lating adhesion molecules platelet endothelial adhesion molecule-1
(PECAM-1) and P-selectin (Onore, Nordahl, et al., 2012), are reported
in sera of ASD individuals, altogether pointing to several blood
immunophenotypes relevant to ASD.
social interactions in animal models for ASD (Deutsch et al., 2012) and also
reduces social withdrawal in a small cohort of ASD children (Posey et al.,
2004). Whether these effects are mediated by its anti-inflammatory proper-
ties and/or by its role as a partial NMDA-receptor agonist is not clear.
In addition, several of the antidepressant and antipsychotic drugs com-
monly used to treat ASD are known to exhibit immunomodulatory prop-
erties. Risperidone and aripiprazole, the only two drugs FDA-approved for
the treatment of irritability in ASD, both have immunologic effects, altering
T-cell differentiation, microglial activation, and serum cytokine levels,
among other immunologic responses (Cecchelli, Grassi, & Pallanti, 2010;
Chen et al., 2011, 2012; Ching & Pringsheim, 2012; Kim et al., 2001;
Richtand et al., 2012; Zhang et al., 2004). The selective serotonin reuptake
inhibitor, fluoxetine, reduces stereotypical behavior in a double-blind
placebo-controlled trial of adults with ASD (Hollander et al., 2012) and also
confers several effects on innate and adaptive cellular immune responses of
T cells and NK cells (Basterzi et al., 2010; Frick, Rapanelli, Cremaschi, &
Genaro, 2009; Nunez et al., 2006; Rogoz, Kubera, Rogoz, Basta-Kaim, &
Budziszewska, 2009). In addition, the acetylcholinesterase inhibitors
galantamine and donepezil ameliorate social impairment, irritability, and
inattention in ASD children (Hohnadel, Bouchard, & Terry, 2007;
Nicolson, Craven-Thuss, & Smith, 2006) and exhibit primary effects on
activation of the cholinergic anti-inflammatory pathway (Hardan &
Handen, 2002; Pavlov et al., 2009). Further studies are needed to determine
whether the immunomodulatory properties of these drugs are necessary to
confer beneficial effects on ASD symptoms.
8. CONCLUSION
Increasing evidence highlights widespread immune dysregulation
as an important component of ASD (Fig. 9.2). Several prenatal immune
insults and postnatal immune abnormalities may be involved in the devel-
opment and/or persistence of ASD symptoms. The study of MIA and mater-
nal autoantibody production as primary immune-related environmental
risk factors for ASD can uncover critical pathways underlying the develop-
ment of autism symptoms and key targets for improving the diagnosis
and treatment of significant subsets of ASD. The mechanisms discovered
might also apply broadly to other prenatal risk factors for ASD, such as
premature birth, advanced paternal age, and maternal thalidomide, VPA,
Immune Dysregulation in Autism Spectrum Disorder 291
REFERENCES
Abazyan, B., Nomura, J., Kannan, G., Ishizuka, K., Tamashiro, K. L., Nucifora, F., et al.
(2010). Prenatal interaction of mutant DISC1 and immune activation produces adult
psychopathology. Biological Psychiatry, 68, 1172–1181.
Abdallah, M. W., Hougaard, D. M., Norgaard-Pedersen, B., Grove, J., Bonefeld-
Jorgensen, E. C., & Mortensen, E. L. (2012). Infections during pregnancy and after birth,
and the risk of autism spectrum disorders: A register-based study utilizing a Danish his-
toric birth cohort. Turk psikiyatri dergisi—Turkish Journal of Psychiatry, 23, 229–236.
Abrahams, B. S., & Geschwind, D. H. (2010). Connecting genes to brain in the autism spec-
trum disorders. Archives of Neurology, 67, 395–399.
Al-Ayadhi, L. Y., & Mostafa, G. A. (2012). Elevated serum levels of interleukin-17A in chil-
dren with autism. Journal of Neuroinflammation, 9, 158.
Arrode-Bruses, G., & Bruses, J. L. (2012). Maternal immune activation by poly I:C induces
expression of cytokines IL-1beta and IL-13, chemokine MCP-1 and colony stimulating
factor VEGF in fetal mouse brain. Journal of Neuroinflammation, 9, 83.
Ashwood, P., Corbett, B. A., Kantor, A., Schulman, H., Van de Water, J., & Amaral, D. G.
(2011). In search of cellular immunophenotypes in the blood of children with autism.
PLoS One, 6, e19299.
Ashwood, P., Enstrom, A., Krakowiak, P., Hertz-Picciotto, I., Hansen, R. L., Croen, L. A.,
et al. (2008). Decreased transforming growth factor beta1 in autism: A potential link
between immune dysregulation and impairment in clinical behavioral outcomes. Journal
of Neuroimmunology, 204, 149–153.
Ashwood, P., Krakowiak, P., Hertz-Picciotto, I., Hansen, R., Pessah, I., & Van de Water, J.
(2011a). Elevated plasma cytokines in autism spectrum disorders provide evidence
of immune dysfunction and are associated with impaired behavioral outcome. Brain,
Behavior, and Immunity, 25, 40–45.
Ashwood, P., Krakowiak, P., Hertz-Picciotto, I., Hansen, R., Pessah, I. N., & Van de
Water, J. (2011b). Associations of impaired behaviors with elevated plasma chemokines
in autism spectrum disorders. Journal of Neuroimmunology, 232, 196–199.
Atladottir, H. O., Henriksen, T. B., Schendel, D. E., & Parner, E. T. (2012). Autism after
infection, febrile episodes, and antibiotic use during pregnancy: An exploratory study.
Pediatrics, 130, e1447–e1454.
Atladottir, H. O., Pedersen, M. G., Thorsen, P., Mortensen, P. B., Deleuran, B.,
Eaton, W. W., et al. (2009). Association of family history of autoimmune diseases and
autism spectrum disorders. Pediatrics, 124, 687–694.
Atladottir, H. O., Thorsen, P., Ostergaard, L., Schendel, D. E., Lemcke, S., Abdallah, M.,
et al. (2010). Maternal infection requiring hospitalization during pregnancy and autism
spectrum disorders. Journal of Autism and Developmental Disorders, 40, 1423–1430.
Autism and Developmental Disabilities Monitoring Network Surveillance Year 2008 Prin-
ciple Investigators; Centers for Disease Control and Prevention. (2012). Prevalence of
autism spectrum disorders—Autism and Developmental Disabilities Monitoring Net-
work, 14 sites, United States, 2008. MMWR Surveillance Summaries, 61, 1–19.
Baharnoori, M., Bhardwaj, S. K., & Srivastava, L. K. (2013). Effect of maternal lipopolysac-
charide administration on the development of dopaminergic receptors and transporter in
the rat offspring. PLoS One, 8, e54439.
Immune Dysregulation in Autism Spectrum Disorder 293
Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E., et al. (1995).
Autism as a strongly genetic disorder: Evidence from a British twin study. Psychological
Medicine, 25, 63–77.
Basterzi, A. D., Yazici, K., Buturak, V., Cimen, B., Yazici, A., Eskandari, G., et al. (2010).
Effects of venlafaxine and fluoxetine on lymphocyte subsets in patients with major
depressive disorder: A flow cytometric analysis. Progress in Neuro-Psychopharmacology &
Biological Psychiatry, 34, 70–75.
Bauman, M. L. (2010). Medical comorbidities in autism: Challenges to diagnosis and treat-
ment. Neurotherapeutics, 7, 320–327.
Bauman, M. D., Iosif, A. M., Ashwood, P., Braunschweig, D., Lee, A., Schumann, C. M.,
et al. (2013). Maternal antibodies from mothers of children with autism alter brain
growth and social behavior development in the rhesus monkey. Translational Psychiatry,
3, e278.
Bauman, M. D., Iosif, A. M., Smith, S. E., Bregere, C., Zadran, S., Amaral, D. G., et al.
(2011). A nonhuman primate model of maternal immune activation. In: Program No.
778.06/Y29. 2011 Neuroscience Meeting Planner. Paper presented at Society for Neuroscience,
Washington, DC.
Bilbo, S. D. (2013). Frank A. Beach Award: Programming of neuroendocrine function by
early-life experience: A critical role for the immune system. Hormones and Behavior,
63, 684–691.
Bitzer-Quintero, O. K., & Gonzalez-Burgos, I. (2012). Immune system in the brain:
A modulatory role on dendritic spine morphophysiology? Neural Plasticity, 2012,
348642.
Braunschweig, D., Duncanson, P., Boyce, R., Hansen, R., Ashwood, P., Pessah, I. N., et al.
(2012). Behavioral correlates of maternal antibody status among children with autism.
Journal of Autism and Developmental Disorders, 42, 1435–1445.
Braunschweig, D., Golub, M. S., Koenig, C. M., Qi, L., Pessah, I. N., Van de Water, J., et al.
(2012). Maternal autism-associated IgG antibodies delay development and produce anx-
iety in a mouse gestational transfer model. Journal of Neuroimmunology, 252, 56–65.
Braunschweig, D., Krakowiak, P., Duncanson, P., Boyce, R., Hansen, R. L., Ashwood, P.,
et al. (2013). Autism-specific maternal autoantibodies recognize critical proteins in
developing brain. Translational Psychiatry, 3, e277.
Braunschweig, D., & Van de Water, J. (2012). Maternal autoantibodies in autism. Archives of
Neurology, 69, 693–699.
Breece, E., Paciotti, B., Nordahl, C. W., Ozonoff, S., Van de Water, J. A., Rogers, S. J., et al.
(2013). Myeloid dendritic cells frequencies are increased in children with autism spec-
trum disorder and associated with amygdala volume and repetitive behaviors. Brain,
Behavior, and Immunity, 31, 69–75.
Brown, R. S. (2009). Autoimmune thyroid disease: Unlocking a complex puzzle. Current
Opinion in Pediatrics, 21, 523–528.
Brown, A. S. (2012). Epidemiologic studies of exposure to prenatal infection and risk of
schizophrenia and autism. Developmental Neurobiology, 72, 1272–1276.
Brown, A. S., Sourander, A., Hinkka-Yli-Salomaki, S., McKeague, I. W., Sundvall, J., &
Surcel, H. M. (2013). Elevated maternal C-reactive protein and autism in a national birth
cohort. Molecular Psychiatry, http://dx.doi.org/10.1038/mp.2012.197.
Brynskikh, A., Warren, T., Zhu, J., & Kipnis, J. (2008). Adaptive immunity affects learning
behavior in mice. Brain, Behavior, and Immunity, 22, 861–869.
Campbell, D. B., Buie, T. M., Winter, H., Bauman, M., Sutcliffe, J. S., Perrin, J. M., et al.
(2009). Distinct genetic risk based on association of MET in families with co-occurring
autism and gastrointestinal conditions. Pediatrics, 123, 1018–1024.
Careaga, M., & Ashwood, P. (2012). Autism spectrum disorders: From immunity to
behavior. Methods in Molecular Biology, 934, 219–240.
294 Elaine Y. Hsiao
Carpentier, P. A., Dingman, A. L., & Palmer, T. D. (2011). Placental TNF-alpha signaling in
illness-induced complications of pregnancy. The American Journal of Pathology, 178,
2802–2810.
Cecchelli, C., Grassi, G., & Pallanti, S. (2010). Aripiprazole improves depressive symptoms
and immunological response to antiretroviral therapy in an HIV-infected subject with
resistant depression. Case Report Medical, 2010, 836214.
Chaste, P., & Leboyer, M. (2012). Autism risk factors: Genes, environment, and gene-
environment interactions. Dialogues in Clinical Neuroscience, 14, 281–292.
Chen, M. L., Tsai, T. C., Lin, Y. Y., Tsai, Y. M., Wang, L. K., Lee, M. C., et al. (2011).
Antipsychotic drugs suppress the AKT/NF-kappaB pathway and regulate the differen-
tiation of T-cell subsets. Immunology Letters, 140, 81–91.
Chen, M. L., Tsai, T. C., Wang, L. K., Lin, Y. Y., Tsai, Y. M., Lee, M. C., et al. (2012).
Risperidone modulates the cytokine and chemokine release of dendritic cells and induces
TNF-alpha-directed cell apoptosis in neutrophils. International Immunopharmacology, 12,
197–204.
Chen, S. K., Tvrdik, P., Peden, E., Cho, S., Wu, S., Spangrude, G., et al. (2010). Hema-
topoietic origin of pathological grooming in Hoxb8 mutant mice. Cell, 141, 775–785.
Chess, S. (1971). Autism in children with congenital rubella. Journal of Autism and Childhood
Schizophrenia, 1, 33–47.
Ching, H., & Pringsheim, T. (2012). Aripiprazole for autism spectrum disorders (ASD).
Cochrane Database of Systematic Reviews, 5, CD009043.
Chow, M. L., Pramparo, T., Winn, M. E., Barnes, C. C., Li, H. R., Weiss, L., et al. (2012).
Age-dependent brain gene expression and copy number anomalies in autism suggest dis-
tinct pathological processes at young versus mature ages. PLoS Genetics, 8, e1002592.
Comi, A. M., Zimmerman, A. W., Frye, V. H., Law, P. A., & Peeden, J. N. (1999). Familial
clustering of autoimmune disorders and evaluation of medical risk factors in autism.
Journal of Child Neurology, 14, 388–394.
Constantino, J. N., Zhang, Y., Frazier, T., Abbacchi, A. M., & Law, P. (2010). Sibling recur-
rence and the genetic epidemiology of autism. The American Journal of Psychiatry, 167,
1349–1356.
Corbett, B. A., Kantor, A. B., Schulman, H., Walker, W. L., Lit, L., Ashwood, P., et al.
(2007). A proteomic study of serum from children with autism showing differential
expression of apolipoproteins and complement proteins. Molecular Psychiatry, 12,
292–306.
Coury, D. L., Ashwood, P., Fasano, A., Fuchs, G., Geraghty, M., Kaul, A., et al. (2012).
Gastrointestinal conditions in children with autism spectrum disorder: Developing a
research agenda. Pediatrics, 130(Suppl. 2), S160–S168.
Crespi, B. J., & Thiselton, D. L. (2011). Comparative immunogenetics of autism and schizo-
phrenia. Genes, Brain, and Behavior, 10, 689–701.
Cushman, J., Lo, J., Huang, Z., Wasserfall, C., & Petitto, J. M. (2003). Neurobehavioral
changes resulting from recombinase activation gene 1 deletion. Clinical and Diagnostic
Laboratory Immunology, 10, 13–18.
Dalton, P., Deacon, R., Blamire, A., Pike, M., McKinlay, I., Stein, J., et al. (2003). Maternal
neuronal antibodies associated with autism and a language disorder. Annals of Neurology,
53, 533–537.
Dantzer, R., O’Connor, J. C., Freund, G. G., Johnson, R. W., & Kelley, K. W. (2008).
From inflammation to sickness and depression: When the immune system subjugates
the brain. Nature Reviews Neuroscience, 9, 46–56.
de Lacy, N., & King, B. H. (2013). Revisiting the relationship between autism and schizo-
phrenia: Toward an integrated neurobiology. Annual Review of Clinical Psychology, 9,
555–587.
Immune Dysregulation in Autism Spectrum Disorder 295
Delorme, R., Ey, E., Toro, R., Leboyer, M., Gillberg, C., & Bourgeron, T. (2013). Progress
toward treatments for synaptic defects in autism. Nature Medicine, 19, 685–694.
Derecki, N. C., Cronk, J. C., Lu, Z., Xu, E., Abbott, S. B., Guyenet, P. G., et al. (2012).
Wild-type microglia arrest pathology in a mouse model of Rett syndrome. Nature, 484,
105–109.
Deutsch, S. I., Pepe, G. J., Burket, J. A., Winebarger, E. E., Herndon, A. L., & Benson, A. D.
(2012). D-cycloserine improves sociability and spontaneous stereotypic behaviors in
4-week old mice. Brain Research, 1439, 96–107.
Deverman, B. E., & Patterson, P. H. (2009). Cytokines and CNS development. Neuron, 64,
61–78.
Ehninger, D., Sano, Y., de Vries, P. J., Dies, K., Franz, D., Geschwind, D. H., et al. (2012).
Gestational immune activation and Tsc2 haploinsufficiency cooperate to disrupt fetal
survival and may perturb social behavior in adult mice. Molecular Psychiatry, 17, 62–70.
Elmer, B. M., & McAllister, A. K. (2012). Major histocompatibility complex class I proteins
in brain development and plasticity. Trends in Neurosciences, 35, 660–670.
Enstrom, A. M., Lit, L., Onore, C. E., Gregg, J. P., Hansen, R. L., Pessah, I. N., et al. (2009).
Altered gene expression and function of peripheral blood natural killer cells in children
with autism. Brain, Behavior, and Immunity, 23, 124–133.
Fidler, D. J., Bailey, J. N., & Smalley, S. L. (2000). Macrocephaly in autism and other pervasive
developmental disorders. Developmental Medicine and Child Neurology, 42, 737–740.
Folstein, S., & Rutter, M. (1977). Infantile autism: A genetic study of 21 twin pairs. Journal of
Child Psychology and Psychiatry, and Allied Disciplines, 18, 297–321.
Forrest, C. M., Khalil, O. S., Pisar, M., Smith, R. A., Darlington, L. G., & Stone, T. W.
(2012). Prenatal activation of Toll-like receptors-3 by administration of the viral mimetic
poly(I:C) changes synaptic proteins, N-methyl-D-aspartate receptors and neurogenesis
markers in offspring. Molecular Brain, 5, 22.
Fox, E., Amaral, D., & Van de Water, J. (2012). Maternal and fetal antibrain antibodies in
development and disease. Developmental Neurobiology, 72, 1327–1334.
Frick, L. R., Rapanelli, M., Cremaschi, G. A., & Genaro, A. M. (2009). Fluoxetine directly
counteracts the adverse effects of chronic stress on T cell immunity by compensatory and
specific mechanisms. Brain, Behavior, and Immunity, 23, 36–40.
Garay, P. A., Hsiao, E. Y., Patterson, P. H., & McAllister, A. K. (2012). Maternal immune
activation causes age- and region-specific changes in brain cytokines in offspring
throughout development. Brain, Behavior, and Immunity, 31, 54–68.
Garay, P. A., & McAllister, A. K. (2010). Novel roles for immune molecules in neural devel-
opment: Implications for neurodevelopmental disorders. Frontiers in Synaptic Neuroscience,
2, 136.
Garbett, K., Ebert, P. J., Mitchell, A., Lintas, C., Manzi, B., Mirnics, K., et al. (2008).
Immune transcriptome alterations in the temporal cortex of subjects with autism.
Neurobiology of Disease, 30, 303–311.
Garbett, K. A., Hsiao, E. Y., Kalman, S., Patterson, P. H., & Mirnics, K. (2012). Effects of
maternal immune activation on gene expression patterns in the fetal brain. Translation
Psychiatry, 2, e98.
Ghahramani Seno, M. M., Hu, P., Gwadry, F. G., Pinto, D., Marshall, C. R., Casallo, G.,
et al. (2011). Gene and miRNA expression profiles in autism spectrum disorders. Brain
Research, 1380, 85–97.
Ginsberg, M. R., Rubin, R. A., Falcone, T., Ting, A. H., & Natowicz, M. R. (2012). Brain
transcriptional and epigenetic associations with autism. PLoS One, 7, e44736.
Giovanoli, S., Engler, H., Engler, A., Richetto, J., Voget, M., Willi, R., et al. (2013). Stress
in puberty unmasks latent neuropathological consequences of prenatal immune activa-
tion in mice. Science, 339, 1095–1099.
296 Elaine Y. Hsiao
Girard, S., Tremblay, L., Lepage, M., & Sebire, G. (2010). IL-1 receptor antagonist protects
against placental and neurodevelopmental defects induced by maternal inflammation.
Journal of Immunology, 184, 3997–4005.
Glatt, S. J., Tsuang, M. T., Winn, M., Chandler, S. D., Collins, M., Lopez, L., et al. (2012).
Blood-based gene expression signatures of infants and toddlers with autism. Journal of the
American Academy of Child and Adolescent Psychiatry, 51(934–944), e932.
Goines, P. E., & Ashwood, P. (2013). Cytokine dysregulation in autism spectrum disorders
(ASD): Possible role of the environment. Neurotoxicology and Teratology, 36, 67–81.
Goines, P., Haapanen, L., Boyce, R., Duncanson, P., Braunschweig, D., Delwiche, L., et al.
(2011). Autoantibodies to cerebellum in children with autism associate with behavior.
Brain, Behavior, and Immunity, 25, 514–523.
Gregg, J. P., Lit, L., Baron, C. A., Hertz-Picciotto, I., Walker, W., Davis, R. A., et al. (2008).
Gene expression changes in children with autism. Genomics, 91, 22–29.
Grigorenko, E. L., Han, S. S., Yrigollen, C. M., Leng, L., Mizue, Y., Anderson, G. M., et al.
(2008). Macrophage migration inhibitory factor and autism spectrum disorders. Pediatrics,
122, e438–e445.
Gupta, S., Samra, D., & Agrawal, S. (2010). Adaptive and innate immune responses in autism:
Rationale for therapeutic use of intravenous immunoglobulin. Journal of Clinical Immu-
nology, 30, 90–96.
Hagerman, R., Lauterborn, J., Au, J., & Berry-Kravis, E. (2012). Fragile syndrome and
targeted treatment trials. Results and Problems in Cell Differentiation, 54, 297–335.
Hallmayer, J., Cleveland, S., Torres, A., Phillips, J., Cohen, B., Torigoe, T., et al. (2011).
Genetic heritability and shared environmental factors among twin pairs with autism.
Archives of General Psychiatry, 68, 1095–1102.
Hamlyn, J., Duhig, M., McGrath, J., & Scott, J. (2013). Modifiable risk factors for schizo-
phrenia and autism—Shared risk factors impacting on brain development. Neurobiology of
Disease, 53, 3–9.
Hardan, A. Y., & Handen, B. L. (2002). A retrospective open trial of adjunctive donepezil in
children and adolescents with autistic disorder. Journal of Child and Adolescent Psychophar-
macology, 12, 237–241.
Harvey, L., & Boksa, P. (2012). Prenatal and postnatal animal models of immune activation:
Relevance to a range of neurodevelopmental disorders. Developmental Neurobiology Special
Issue: Neuroimmunology in Development and Disease, 72, 1335–1348.
Heo, Y., Zhang, Y., Gao, D., Miller, V. M., & Lawrence, D. A. (2011). Aberrant immune
responses in a mouse with behavioral disorders. PLoS One, 6, e20912.
Heuer, L., Ashwood, P., Schauer, J., Goines, P., Krakowiak, P., Hertz-Picciotto, I., et al.
(2008). Reduced levels of immunoglobulin in children with autism correlates with
behavioral symptoms. Autism Research, 1, 275–283.
Heuer, L., Braunschweig, D., Ashwood, P., Van de Water, J., & Campbell, D. B. (2011).
Association of a MET genetic variant with autism-associated maternal autoantibodies
to fetal brain proteins and cytokine expression. Translation Psychiatry, 1, e48.
Heuer, L. S., Rose, M., Ashwood, P., & Van de Water, J. (2012). Decreased levels of total
immunoglobulin in children with autism are not a result of B cell dysfunction. Journal of
Neuroimmunology, 251, 94–102.
Hohnadel, E., Bouchard, K., & Terry, A. V., Jr. (2007). Galantamine and donepezil attenuate
pharmacologically induced deficits in prepulse inhibition in rats. Neuropharmacology, 52,
542–551.
Hollander, E., Soorya, L., Chaplin, W., Anagnostou, E., Taylor, B. P., Ferretti, C. J., et al.
(2012). A double-blind placebo-controlled trial of fluoxetine for repetitive behaviors and
global severity in adult autism spectrum disorders. The American Journal of Psychiatry, 169,
292–299.
Immune Dysregulation in Autism Spectrum Disorder 297
Hsiao, E. Y., McBride, S. W., Chow, J., Mazmanian, S. K., & Patterson, P. H. (2012).
Modeling an autism risk factor in mice leads to permanent immune dysregulation.
Proceedings of the National Academy of Sciences of the United States of America, 109,
12776–12781.
Hsiao, E. Y., & Patterson, P. H. (2011). Activation of the maternal immune system induces
endocrine changes in the placenta via IL-6. Brain, Behavior, and Immunity, 25, 604–615.
Hsiao, E. Y., & Patterson, P. H. (2012a). Immune involvement in autism spectrum disorder
as a basis for animal models. Autism, S1.
Hsiao, E. Y., & Patterson, P. H. (2012b). Placental regulation of maternal-fetal interactions
and brain development. Developmental Neurobiology, 72, 1317–1326.
Hu, V. W., Nguyen, A., Kim, K. S., Steinberg, M. E., Sarachana, T., Scully, M. A., et al.
(2009). Gene expression profiling of lymphoblasts from autistic and nonaffected sib pairs:
Altered pathways in neuronal development and steroid biosynthesis. PLoS One, 4, e5775.
Hu, V. W., Sarachana, T., Kim, K. S., Nguyen, A., Kulkarni, S., Steinberg, M. E., et al.
(2009). Gene expression profiling differentiates autism case-controls and phenotypic var-
iants of autism spectrum disorders: Evidence for circadian rhythm dysfunction in severe
autism. Autism Research, 2, 78–97.
Irish Schizophrenia Genomics Consortium and the Wellcome Trust Case Control Consor-
tium 2. (2012). Genome-wide association study implicates HLA-C*01:02 as a risk factor
at the major histocompatibility complex locus in schizophrenia. Biological Psychiatry, 72,
620–628.
Ivarsson, S. A., Bjerre, I., Vegfors, P., & Ahlfors, K. (1990). Autism as one of several disabil-
ities in two children with congenital cytomegalovirus infection. Neuropediatrics, 21,
102–103.
Jia, P., Wang, L., Fanous, A. H., Pato, C. N., Edwards, T. L., & Zhao, Z. (2012). Network-
assisted investigation of combined causal signals from genome-wide association studies in
schizophrenia. PLoS Computational Biology, 8, e1002587.
Jonakait, G. M., Pratt, L., Acevedo, G., & Ni, L. (2012). Microglial regulation of cholinergic
differentiation in the basal forebrain. Developmental Neurobiology, 72, 857–864.
Kanne, S. M., & Mazurek, M. O. (2011). Aggression in children and adolescents with ASD:
Prevalence and risk factors. Journal of Autism and Developmental Disorders, 41, 926–937.
Kanner, L. (1968). Autistic disturbances of affective contact. Acta Paedopsychiatrica, 35,
100–136.
Keil, A., Daniels, J. L., Forssen, U., Hultman, C., Cnattingius, S., Soderberg, K. C., et al.
(2010). Parental autoimmune diseases associated with autism spectrum disorders in off-
spring. Epidemiology, 21, 805–808.
Kim, D. J., Kim, W., Yoon, S. J., Go, H. J., Choi, B. M., Jun, T. Y., et al. (2001). Effect of
risperidone on serum cytokines. The International Journal of Neuroscience, 111, 11–19.
Kipnis, J., Cohen, H., Cardon, M., Ziv, Y., & Schwartz, M. (2004). T cell deficiency leads to
cognitive dysfunction: Implications for therapeutic vaccination for schizophrenia and
other psychiatric conditions. Proceedings of the National Academy of Sciences of the United
States of America, 101, 8180–8185.
Kohane, I. S., McMurry, A., Weber, G., MacFadden, D., Rappaport, L., Kunkel, L., et al.
(2012). The co-morbidity burden of children and young adults with autism spectrum
disorders. PLoS One, 7, e33224.
Kong, S. W., Collins, C. D., Shimizu-Motohashi, Y., Holm, I. A., Campbell, M. G.,
Lee, I. H., et al. (2012). Characteristics and predictive value of blood transcriptome sig-
nature in males with autism spectrum disorders. PLoS One, 7, e49475.
Kotturi, M. F., Assarsson, E., Peters, B., Grey, H., Oseroff, C., Pasquetto, V., et al. (2009). Of
mice and humans: How good are HLA transgenic mice as a model of human immune
responses? Immunome Research, 5, 3.
298 Elaine Y. Hsiao
Lee, B. K., Dalman, C., Newschaffer, C. J., Burstyn, I., Blomstrom, A., Idring, S., et al.
(2012). Maternal hospitalization for infection during pregnancy and risk of autism spec-
trum disorders. In: Paper presented at IMFAR, Toronto, Canada.
Li, X., Chauhan, A., Sheikh, A. M., Patil, S., Chauhan, V., Li, X. M., et al. (2009). Elevated
immune response in the brain of autistic patients. Journal of Neuroimmunology, 207,
111–116.
Malkova, N. V., Yu, C. Z., Hsiao, E. Y., Moore, M. J., & Patterson, P. H. (2012). Maternal
immune activation yields offspring displaying mouse versions of the three core symptoms
of autism. Brain, Behavior, and Immunity, 26, 607–616.
Mandal, M., Marzouk, A. C., Donnelly, R., & Ponzio, N. M. (2011). Maternal immune
stimulation during pregnancy affects adaptive immunity in offspring to promote devel-
opment of TH17 cells. Brain, Behavior, and Immunity, 25, 863–871.
Martin, L. A., Ashwood, P., Braunschweig, D., Cabanlit, M., Van de Water, J., &
Amaral, D. G. (2008). Stereotypies and hyperactivity in rhesus monkeys exposed to
IgG from mothers of children with autism. Brain, Behavior, and Immunity, 22, 806–816.
McCusker, R. H., & Kelley, K. W. (2013). Immune-neural connections: How the immune
system’s response to infectious agents influences behavior. The Journal of Experimental
Biology, 216, 84–98.
Meyer, U., Feldon, J., & Fatemi, S. H. (2009). In-vivo rodent models for the experimental
investigation of prenatal immune activation effects in neurodevelopmental brain disor-
ders. Neuroscience and Biobehavioral Reviews, 33, 1061–1079.
Meyer, U., Nyffeler, M., Engler, A., Urwyler, A., Schedlowski, M., Knuesel, I., et al. (2006).
The time of prenatal immune challenge determines the specificity of inflammation-
mediated brain and behavioral pathology. The Journal of Neuroscience, 26, 4752–4762.
Michel, M., Schmidt, M. J., & Mirnics, K. (2012). Immune system gene dysregulation in
autism and schizophrenia. Developmental Neurobiology, 72, 1277–1287.
Mombaerts, P., Mizoguchi, E., Ljunggren, H. G., Iacomini, J., Ishikawa, H., Wang, L., et al.
(1994). Peripheral lymphoid development and function in TCR mutant mice. Interna-
tional Immunology, 6, 1061–1070.
Momeni, N., Brudin, L., Behnia, F., Nordstrom, B., Yosefi-Oudarji, A., Sivberg, B., et al.
(2012). High complement factor I activity in the plasma of children with autism spectrum
disorders. Autism Research and Treatment, 2012, 868576.
Morgan, J. T., Chana, G., Abramson, I., Semendeferi, K., Courchesne, E., & Everall, I. P.
(2012). Abnormal microglial-neuronal spatial organization in the dorsolateral prefrontal
cortex in autism. Brain Research, 1456, 72–81.
Morgan, J. T., Chana, G., Pardo, C. A., Achim, C., Semendeferi, K., Buckwalter, J., et al.
(2010). Microglial activation and increased microglial density observed in the dorsolateral
prefrontal cortex in autism. Biological Psychiatry, 68, 368–376.
Morris, C. M., Zimmerman, A. W., & Singer, H. S. (2009). Childhood serum anti-fetal brain
antibodies do not predict autism. Pediatric Neurology, 41, 288–290.
Mostafa, G. A., & Al-Ayadhi, L. Y. (2011). A lack of association between hyperserotonemia
and the increased frequency of serum anti-myelin basic protein auto-antibodies in autistic
children. Journal of Neuroinflammation, 8, 71.
Mostafa, G. A., Shehab, A. A., & Al-Ayadhi, L. Y. (2013). The link between some alleles on
human leukocyte antigen system and autism in children. Journal of Neuroimmunology, 255,
70–74.
Napolioni, V., Ober-Reynolds, B., Szelinger, S., Corneveaux, J. J., Pawlowski, T., Ober-
Reynolds, S., et al. (2013). Plasma cytokine profiling in sibling pairs discordant for autism
spectrum disorder. Journal of Neuroinflammation, 10, 38.
Needleman, L. A., & McAllister, A. K. (2012). The major histocompatibility complex and
autism spectrum disorder. Developmental Neurobiology, 72, 1288–1301.
Immune Dysregulation in Autism Spectrum Disorder 299
Nicolson, R., Craven-Thuss, B., & Smith, J. (2006). A prospective, open-label trial of
galantamine in autistic disorder. Journal of Child and Adolescent Psychopharmacology, 16,
621–629.
Nishimura, Y., Martin, C. L., Vazquez-Lopez, A., Spence, S. J., Alvarez-Retuerto, A. I.,
Sigman, M., et al. (2007). Genome-wide expression profiling of lymphoblastoid cell lines
distinguishes different forms of autism and reveals shared pathways. Human Molecular
Genetics, 16, 1682–1698.
Noh, H. J., Ponting, C. P., Boulding, H. C., Meader, S., Betancur, C., Buxbaum, J. D., et al.
(2013). Network topologies and convergent aetiologies arising from deletions and dupli-
cations observed in individuals with autism. PLoS Genetics, 9, e1003523.
Nordahl, C. W., Braunschweig, D., Iosif, A. M., Lee, A., Rogers, S., Ashwood, P., et al.
(2013). Maternal autoantibodies are associated with abnormal brain enlargement in a
subgroup of children with autism spectrum disorder. Brain, Behavior, and Immunity,
30, 61–65.
Nunez, M. J., Balboa, J., Rodrigo, E., Brenlla, J., Gonzalez-Peteiro, M., & Freire-Garabal, M.
(2006). Effects of fluoxetine on cellular immune response in stressed mice. Neuroscience
Letters, 396, 247–251.
Onore, C., Careaga, M., & Ashwood, P. (2012). The role of immune dysfunction in the
pathophysiology of autism. Brain, Behavior, and Immunity, 26, 383–392.
Onore, C. E., Nordahl, C. W., Young, G. S., Van de Water, J. A., Rogers, S. J., &
Ashwood, P. (2012). Levels of soluble platelet endothelial cell adhesion molecule-1
and P-selectin are decreased in children with autism spectrum disorder. Biological
Psychiatry, 72, 1020–1025.
Patterson, P. H. (2011). Maternal infection and immune involvement in autism. Trends in
Molecular Medicine, 17, 389–394.
Patterson, P. H. (2012). Maternal infection and autism. Brain, Behavior, and Immunity, 26, 393.
Pavlov, V. A., Parrish, W. R., Rosas-Ballina, M., Ochani, M., Puerta, M., Ochani, K., et al.
(2009). Brain acetylcholinesterase activity controls systemic cytokine levels through the
cholinergic anti-inflammatory pathway. Brain, Behavior, and Immunity, 23, 41–45.
Posey, D. J., Kem, D. L., Swiezy, N. B., Sweeten, T. L., Wiegand, R. E., & McDougle, C. J.
(2004). A pilot study of D-cycloserine in subjects with autistic disorder. The American
Journal of Psychiatry, 161, 2115–2117.
Ramos, P. S., Sajuthi, S., Langefeld, C. D., & Walker, S. J. (2012). Immune function genes
CD99L2, JARID2 and TPO show association with autism spectrum disorder. Molecular
Autism, 3, 4.
Ratnayake, U., Quinn, T. A., Castillo-Melendez, M., Dickinson, H., & Walker, D. W.
(2012). Behaviour and hippocampus-specific changes in spiny mouse neonates after
treatment of the mother with the viral-mimetic Poly I:C at mid-pregnancy. Brain, Behav-
ior, and Immunity, 26, 1288–1299.
Richtand, N. M., Ahlbrand, R., Horn, P., Tambyraja, R., Grainger, M., Bronson, S. L.,
et al. (2012). Fluoxetine and aripiprazole treatment following prenatal immune activa-
tion exert longstanding effects on rat locomotor response. Physiology & Behavior, 106,
171–177.
Rogoz, Z., Kubera, M., Rogoz, K., Basta-Kaim, A., & Budziszewska, B. (2009). Effect of
co-administration of fluoxetine and amantadine on immunoendocrine parameters in rats
subjected to a forced swimming test. Pharmacological Reports, 61, 1050–1060.
Rosen, N. J., Yoshida, C. K., & Croen, L. A. (2007). Infection in the first 2 years of life and
autism spectrum disorders. Pediatrics, 119, e61–e69.
Rosenberg, R. E., Law, J. K., Yenokyan, G., McGready, J., Kaufmann, W. E., & Law, P. A.
(2009). Characteristics and concordance of autism spectrum disorders among 277 twin
pairs. Archives of Pediatrics & Adolescent Medicine, 163, 907–914.
300 Elaine Y. Hsiao
Ross, H. E., Guo, Y., Coleman, K., Ousley, O., & Miller, A. H. (2013). Association of
IL-12p70 and IL-6:IL-10 ratio with autism-related behaviors in 22q11.2 deletion syn-
drome: A preliminary report. Brain, Behavior, and Immunity, 31, 76–81.
Rotschafer, S. E., Trujillo, M. S., Dansie, L. E., Ethell, I. M., & Razak, K. A. (2012). Min-
ocycline treatment reverses ultrasonic vocalization production deficit in a mouse model
of Fragile Syndrome. Brain Research, 1439, 7–14.
Rout, U. K., Mungan, N. K., & Dhossche, D. M. (2012). Presence of GAD65 autoanti-
bodies in the serum of children with autism or ADHD. European Child & Adolescent
Psychiatry, 21, 141–147.
Ruff, J. S., Nelson, A. C., Kubinak, J. L., & Potts, W. K. (2012). MHC signaling during social
communication. Advances in Experimental Medicine and Biology, 738, 290–313.
Sacco, R., Lenti, C., Saccani, M., Curatolo, P., Manzi, B., Bravaccio, C., et al. (2012). Clus-
ter analysis of autistic patients based on principal pathogenetic components. Autism
Research, 5, 137–147.
Sarachana, T., Zhou, R., Chen, G., Manji, H. K., & Hu, V. W. (2010). Investigation of post-
transcriptional gene regulatory networks associated with autism spectrum disorders by
microRNA expression profiling of lymphoblastoid cell lines. Genome Medicine, 2, 23.
Saxena, V., Ramdas, S., Ochoa, C. R., Wallace, D., Bhide, P., & Kohane, I. (2012). Struc-
tural, genetic, and functional signatures of disordered neuro-immunological develop-
ment in autism spectrum disorder. PLoS One, 7, e48835.
Schneider, T., Roman, A., Basta-Kaim, A., Kubera, M., Budziszewska, B., Schneider, K.,
et al. (2008). Gender-specific behavioral and immunological alterations in an animal
model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology,
33, 728–740.
Shatz, C. J. (2009). MHC class I: An unexpected role in neuronal plasticity. Neuron, 64,
40–45.
Singer, H. S., Morris, C., Gause, C., Pollard, M., Zimmerman, A. W., & Pletnikov, M.
(2009). Prenatal exposure to antibodies from mothers of children with autism produces
neurobehavioral alterations: A pregnant dam mouse model. Journal of Neuroimmunology,
211, 39–48.
Spence, S. J., & Schneider, M. T. (2009). The role of epilepsy and epileptiform EEGs in
autism spectrum disorders. Pediatric Research, 65, 599–606.
Steffenburg, S., Gillberg, C., Hellgren, L., Andersson, L., Gillberg, I. C., Jakobsson, G., et al.
(1989). A twin study of autism in Denmark, Finland, Iceland, Norway and Sweden.
Journal of Child Psychology and Psychiatry, and Allied Disciplines, 30, 405–416.
Stephan, A. H., Barres, B. A., & Stevens, B. (2012). The complement system: An unexpected
role in synaptic pruning during development and disease. Annual Review of Neuroscience,
35, 369–389.
Sturm, T., Leinders-Zufall, T., Macek, B., Walzer, M., Jung, S., Pommerl, B., et al. (2013).
Mouse urinary peptides provide a molecular basis for genotype discrimination by nasal
sensory neurons. Nature Communications, 4, 1616.
Suzuki, K., Sugihara, G., Ouchi, Y., Nakamura, K., Futatsubashi, M., Takebayashi, K., et al.
(2013). Microglial activation in young adults with autism spectrum disorder. JAMA
Psychiatry, 70, 49–58.
Swedo, S. E., Buckley, A. W., Thurm, A., Lee, L. C., Azhagira, A., & Pardo, C. (2010).
Pilot study of minocycline treatment for autism. In: Paper presented at IMFAR, Philadel-
phia, PA.
Sweeten, T. L., Posey, D. J., & McDougle, C. J. (2004). Brief report: Autistic disorder in
three children with cytomegalovirus infection. Journal of Autism and Developmental
Disorders, 34, 583–586.
Tang, B., Jia, H., Kast, R. J., & Thomas, E. A. (2013). Epigenetic changes at gene promoters
in response to immune activation in utero. Brain, Behavior, and Immunity, 30, 168–175.
Immune Dysregulation in Autism Spectrum Disorder 301
Tetreault, N. A., Hakeem, A. Y., Jiang, S., Williams, B. A., Allman, E., Wold, B. J., et al.
(2012). Microglia in the cerebral cortex in autism. Journal of Autism and Developmental
Disorders, 42, 2569–2584.
Toro, R., Konyukh, M., Delorme, R., Leblond, C., Chaste, P., Fauchereau, F., et al. (2010).
Key role for gene dosage and synaptic homeostasis in autism spectrum disorders. Trends in
Genetics: TIG, 26, 363–372.
Torres, A. R., Westover, J. B., & Rosenspire, A. J. (2012). HLA immune function genes in
autism. Autism Research and Treatment, 2012, 959073.
Tremblay, M. E., Stevens, B., Sierra, A., Wake, H., Bessis, A., & Nimmerjahn, A. (2011).
The role of microglia in the healthy brain. The Journal of Neuroscience, 31, 16064–16069.
Vargas, D. L., Nascimbene, C., Krishnan, C., Zimmerman, A. W., & Pardo, C. A. (2005).
Neuroglial activation and neuroinflammation in the brain of patients with autism. Annals
of Neurology, 57, 67–81.
Vijai, J., Kirchhoff, T., Schrader, K. A., Brown, J., Dutra-Clarke, A. V., Manschreck, C.,
et al. (2013). Susceptibility loci associated with specific and shared subtypes of lymphoid
malignancies. PLoS Genetics, 9, e1003220.
Vincent, A., Bien, C. G., Irani, S. R., & Waters, P. (2011). Autoantibodies associated with
diseases of the CNS: New developments and future challenges. Lancet Neurology, 10,
759–772.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature, 474,
380–384.
Vojdani, A., Mumper, E., Granpeesheh, D., Mielke, L., Traver, D., Bock, K., et al. (2008).
Low natural killer cell cytotoxic activity in autism: The role of glutathione, IL-2 and
IL-15. Journal of Neuroimmunology, 205, 148–154.
Vuillermot, S., Joodmardi, E., Perlmann, T., Ogren, S. O., Feldon, J., & Meyer, U. (2012).
Prenatal immune activation interacts with genetic Nurr1 deficiency in the development
of attentional impairments. The Journal of Neuroscience, 32, 436–451.
Warren, R. P., Foster, A., & Margaretten, N. C. (1987). Reduced natural killer cell
activity in autism. Journal of the American Academy of Child and Adolescent Psychiatry,
26, 333–335.
Wei, H., Zou, H., Sheikh, A. M., Malik, M., Dobkin, C., Brown, W. T., et al. (2011). IL-6 is
increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and
synaptic formation. Journal of Neuroinflammation, 8, 52.
Wong, C. C., Meaburn, E. L., Ronald, A., Price, T. S., Jeffries, A. R., Schalkwyk, L. C.,
et al. (2013). Methylomic analysis of monozygotic twins discordant for autism spectrum
disorder and related behavioural traits. Molecular Psychiatry, http://dx.doi.org/10.1038/
mp.2013.41.
Yamashita, Y., Fujimoto, C., Nakajima, E., Isagai, T., & Matsuishi, T. (2003). Possible asso-
ciation between congenital cytomegalovirus infection and autistic disorder. Journal of
Autism and Developmental Disorders, 33, 455–459.
Yeargin-Allsopp, M. (2002). Past and future perspectives in autism epidemiology. Molecular
Psychiatry, 7(Suppl. 2), S9–S11.
Young, A. M., Campbell, E., Lynch, S., Suckling, J., & Powis, S. J. (2011). Aberrant
NF-kappaB expression in autism spectrum condition: A mechanism for neuro-
inflammation. Frontiers in Psychiatry, 2, 27.
Zerbo, O., Iosif, A. M., Walker, C., Ozonoff, S., Hansen, R. L., & Hertz-Picciotto, I.
(2012). Is maternal influenza or fever during pregnancy associated with autism or devel-
opmental delays? Results from the CHARGE (CHildhood Autism Risks from Genetics
and Environment) Study. Journal of Autism and Developmental Disorders, 43, 25–33.
Zerrate, M. C., Pletnikov, M., Connors, S. L., Vargas, D. L., Seidler, F. J.,
Zimmerman, A. W., et al. (2007). Neuroinflammation and behavioral abnormalities after
302 Elaine Y. Hsiao
neonatal terbutaline treatment in rats: Implications for autism. The Journal of Pharmacology
and Experimental Therapeutics, 322, 16–22.
Zhang, X. Y., Zhou, D. F., Cao, L. Y., Zhang, P. Y., Wu, G. Y., & Shen, Y. C. (2004).
Changes in serum interleukin-2, -6, and -8 levels before and during treatment with
risperidone and haloperidol: Relationship to outcome in schizophrenia. The Journal of
Clinical Psychiatry, 65, 940–947.
Ziats, M. N., & Rennert, O. M. (2013). Sex-biased gene expression in the developing brain:
Implications for autism spectrum disorders. Molecular Autism, 4, 10.
CHAPTER TEN
Contents
1. Introduction 303
2. ASD Susceptibility Genes 305
3. ASD Brain Transcriptome Studies 307
4. Transcriptional Properties of ASD Genes in the Normal Human Brain 308
5. Conclusions and Further Directions 315
Acknowledgments 315
References 315
Abstract
Autism is the most severe end of a spectrum of neurodevelopmental conditions, autism
spectrum disorders (ASD). ASD are genetically heterogeneous, and hundreds of genes
have been implicated in the etiology of the disease. Here, we discuss the contribution of
brain transcriptome studies in advancing our understanding of the genetic mechanisms
of ASD and review recent work characterizing the spatial and temporal variation of the
human brain transcriptome, with a focus on the relevance of these data to autism
susceptibility genes.
1. INTRODUCTION
Autism is a neurodevelopmental disorder characterized by language
deficits, difficulties with social interactions, and repetitive and restrictive
behaviors (Kanner, 1968). It has a prevalence of 1 in 166 individuals
(Fombonne, 2009) and occurs more frequently in males than in females.
The clinical picture is highly heterogeneous, with large differences in symp-
tom severity between patients. In addition, autism patients often suffer from
comorbid conditions such as epilepsy, intellectual disability, anxiety, and
depression (Kim & Lord, 2013). Thus, autism is currently conceptualized
and temporal gene expression variation in the normal human brain and dis-
cuss the relevance of these data for understanding the transcriptional varia-
tion of ASD susceptibility genes.
of individuals included in this study was small (two ASD cases and two con-
trols), it identified several hundreds of long noncoding RNAs differentially
expressed between ASD brain and controls. The findings highlighted the
informative value of noncoding RNA expression in the brain (discussed
in more detail in this volume, Chapter 2). Chow et al. focused on prefrontal
cortex (PFC) and investigated gene expression changes in ASD brain in dif-
ferent age groups (Chow et al., 2012). This study found evidence for the
dysregulation of functional pathways regulating cell number, cortical pat-
terning, and differentiation in young autistic PFC and dysregulation of sig-
naling and repair pathways in adult autistic PFC.
Transcriptome studies of ASD brain are just beginning to emerge, and a
complete picture of the transcriptional changes occurring in ASD brain
would require larger sample sizes than currently available and extensive
characterization of the brain transcriptome, including noncoding RNAs,
alternative promoters, and alternative splicing isoform regulation. In addi-
tion, understanding the significance of ASD-associated transcriptional
changes is still limited due to the yet incomplete understanding of the normal
human brain transcriptome variation. In the following section, we discuss
recent progress in characterizing the spatial and temporal variation in the
normal human brain and its genetic control, as well as insights from these
studies into the transcriptional regulation of ASD genes.
Table 10.1 Temporal variation of ASD gene expression in human prefrontal cortex
Gene Fetal Postnatal
symbol Probe ID development development Teen/adult
ADA HEEBO-086- 0.27 0.21 0.35
HCA86H10
ADSL HEEBO-098- 0.23 0.18 0.36
HCA98B15
AHI1 HEEBO-028- 0.15 0.43 0.34
HCC28J10
ALDH5A1 HEEBO-020- 0.35 0.21 0.40
HCC20P12
ARX HEEBO-053- 0.04 0.51 0.04
HCC53O18
ASTN2 HEEBO-099- 0.39 0.02 0.14
HCA99O1
ATP10A HEEBO-036- 0.08 0.05 0.13
HCC36A3
AUTS2 HEEBO-009- 0.12 0.74 0.32
HCC9K20
CACNA1C HEEBO-008- 0.70 0.84 0.13
HCC8D3
CACNA1H HEEBO-015- 0.00 0.72 0.65
HCC15B22
CDKL5 HEEBO-049- 0.05 0.40 0.19
HCC49I11
CNTN4 HEEBO-106- 0.45 0.61 0.01
HCA106E4
CNTNAP2 HEEBO-023- 0.51 0.27 0.19
HCC23G17
DMD HEEBO-016- 0.00 0.02 0.17
HCC16E16
DMPK HEEBO-043- 0.15 0.23 0.27
HCC43J4
FMR1 HEEBO-017- 0.22 0.58 0.31
HCC17G21
FOXP2 HEEBO-108- 0.58 0.59 0.19
HCE108M2
Continued
312 Shingo Miyauchi and Irina Voineagu
Table 10.1 Temporal variation of ASD gene expression in human prefrontal cortex—
cont'd
Gene Fetal Postnatal
symbol Probe ID development development Teen/adult
GRIK2 HEEBO-050- 0.27 0.79 0.06
HCC50I12
HOXA1 HEEBO-104- 0.32 0.61 0.09
HCA104I3
LAMB1 HEEBO-012- 0.18 0.20 0.37
HCC12B20
MECP2 HEEBO-020- 0.10 0.02 0.28
HCC20K22
MET HEEBO-009- 0.30 0.50 0.00
HCC9C1
NF1 HEEBO-050- 0.14 0.51 0.05
HCC50L6
NLGN3 HEEBO-028- 0.20 0.57 0.31
HCC28J4
NLGN4X HEEBO-009- 0.67 0.81 0.45
HCC9P3
NRXN1 HEEBO-092- 0.35 0.82 0.08
HCA92O3
OXTR HEEBO-047- 0.39 0.15 0.11
HCC47J2
PTCHD1 HEEBO-013- 0.80 0.84 0.22
HCC13H16
RAI1 HEEBO-031- 0.14 0.33 0.03
HCC31M8
RBFOX1 HEEBO-060- 0.01 0.37 0.08
HCC60A14
RELN HEEBO-013- 0.39 0.48 0.29
HCC13P23
SCN1A HEEBO-048- 0.02 0.19 0.38
HCC48F10
SEMA5A HEEBO-068- 0.60 0.51 0.39
HCC68P24
SHANK3 HEEBO-068- 0.56 0.40 0.06
HCC68M15
Autism Susceptibility Genes and the Transcriptional Landscape of the Human Brain 313
Table 10.1 Temporal variation of ASD gene expression in human prefrontal cortex—
cont'd
Gene Fetal Postnatal
symbol Probe ID development development Teen/adult
SLC9A6 HEEBO-039- 0.60 0.23 0.23
HCC39I18
TSC2 HEEBO-085- 0.09 0.49 0.20
HCA85M3
UBE3A HEEBO-069- 0.12 0.43 0.13
HCC69P17
The table lists Spearman correlation coefficients of normalized expression values versus age. Age groups:
fetal development, postnatal development (age <10 years), and teen/adult (age 10 years). For ASD
genes covered by multiple probes, the probe showing the highest expression correlation with age
is shown.
Data from Colantuoni et al. (2011).
Figure 10.1 ASD gene expression variation in the normal human brain. Y-axis: Spearman
correlation coefficients from Table 10.1. Data from Colantuoni et al. (2011).
1.5 1.0 0
0.5 1
–1
0.5 0.0
–2 0
–1.0 –3
–0.5 –1
–4
–1.5 –2.0 –2
–5
0 20 40 60 80 0 20 40 60 80 0 20 40 60 80 0 20 40 60 80
Age (years) Age (years) Age (years) Age (years)
0 20 40 60 80 0 20 40 60 80 0 20 40 60 80 0 20 40 60 80
Age (years) Age (years) Age (years) Age (years)
Figure 10.2 Scatter plots of ASD gene expression values versus age for selected genes. Y-axis: normalized expression values from Colantuoni
et al. (2011). Distinct colors reflect distinct 10-year age intervals.
Autism Susceptibility Genes and the Transcriptional Landscape of the Human Brain 315
ACKNOWLEDGMENTS
We would like to thank Monica Nguyen for proof-reading the manuscript. This work has
been supported by a NARSAD Young Investigator Grant (IV).
REFERENCES
Abrahams, B. S., & Geschwind, D. H. (2008). Advances in autism genetics: On the threshold
of a new neurobiology. Nature Reviews Genetics, 9, 341–355.
Anitha, A., Nakamura, K., Thanseem, I., Yamada, K., Iwayama, Y., Toyota, T., et al. (2012).
Brain region-specific altered expression and association of mitochondria-related genes in
autism. Molecular Autism, 3, 12.
Anney, R., Klei, L., Pinto, D., Regan, R., Conroy, J., Magalhaes, T. R., et al. (2010).
A genome-wide scan for common alleles affecting risk for autism. Human Molecular
Genetics, 19, 4072–4082.
316 Shingo Miyauchi and Irina Voineagu
Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E., et al. (1995).
Autism as a strongly genetic disorder: Evidence from a British twin study. Psychological
Medicine, 25, 63–77.
Banerjee-Basu, S., & Packer, A. (2010). SFARI Gene: An evolving database for the autism
research community. Disease Models & Mechanisms, 3, 133–135.
Cantor, R. M. (2009). Molecular genetics of autism. Current Psychiatry Reports, 11, 137–142.
Carayol, J., Sacco, R., Tores, F., Rousseau, F., Lewin, P., Hager, J., et al. (2011). Converging
evidence for an association of ATP2B2 allelic variants with autism in male subjects. Bio-
logical Psychiatry, 70, 880–887.
Chow, M. L., Pramparo, T., Winn, M. E., Barnes, C. C., Li, H. R., Weiss, L., et al. (2012).
Age-dependent brain gene expression and copy number anomalies in autism suggest dis-
tinct pathological processes at young versus mature ages. PLoS Genetics, 8, e1002592.
Colantuoni, C., Lipska, B. K., Ye, T., Hyde, T. M., Tao, R., Leek, J. T., et al. (2011).
Temporal dynamics and genetic control of transcription in the human prefrontal cortex.
Nature, 478, 519–523.
Cook, E. H., Jr., & Scherer, S. W. (2008). Copy-number variations associated with neuro-
psychiatric conditions. Nature, 455, 919–923.
Dolen, G., Osterweil, E., Shankaranarayana Rao, B. S., Smith, G. B., Auerbach, B. D.,
Chattarji, S., et al. (2007). Correction of fragile X syndrome in mice. Neuron, 56, 955–962.
Enstrom, A. M., Lit, L., Onore, C. E., Gregg, J. P., Hansen, R. L., Pessah, I. N., et al. (2009).
Altered gene expression and function of peripheral blood natural killer cells in children
with autism. Brain, Behavior, and Immunity, 23, 124–133.
Fombonne, E. (2009). Epidemiology of pervasive developmental disorders. Pediatric Research,
65, 591–598.
Freitag, C. M. (2007). The genetics of autistic disorders and its clinical relevance: A review of
the literature. Molecular Psychiatry, 12, 2–22.
Garbett, K., Ebert, P. J., Mitchell, A., Lintas, C., Manzi, B., Mirnics, K., et al. (2008).
Immune transcriptome alterations in the temporal cortex of subjects with autism. Neu-
robiology of Disease, 30, 303–311.
Ginsberg, M. R., Rubin, R. A., & Natowicz, M. R. (2013). Patterning of regional gene
expression in autism: New complexity. Scientific Reports, 3, 1831.
Gregg, J. P., Lit, L., Baron, C. A., Hertz-Picciotto, I., Walker, W., Davis, R. A., et al. (2008).
Gene expression changes in children with autism. Genomics, 91, 22–29.
Guryev, V., Saar, K., Adamovic, T., Verheul, M., van Heesch, S. A., Cook, S., et al. (2008).
Distribution and functional impact of DNA copy number variation in the rat. Nature
Genetics, 40, 538–545.
Hawrylycz, M. J., Lein, E. S., Guillozet-Bongaarts, A. L., Shen, E. H., Ng, L., Miller, J. A.,
et al. (2012). An anatomically comprehensive atlas of the adult human brain trans-
criptome. Nature, 489, 391–399.
Henrichsen, C. N., Vinckenbosch, N., Zöllner, S., Chaignat, E., Pradervand, S., Schütz, F.,
et al. (2009). Segmental copy number variation shapes tissue transcriptomes. Nature
Genetics, 41, 424–429.
Hu, V. W., Frank, B. C., Heine, S., Lee, N. H., & Quackenbush, J. (2006). Gene expression
profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity
of autism reveals differential regulation of neurologically relevant genes. BMC Genomics,
7, 118.
Hu, V. W., Nguyen, A., Kim, K. S., Steinberg, M. E., Sarachana, T., Scully, M. A., et al.
(2009). Gene expression profiling of lymphoblasts from autistic and nonaffected sib pairs:
Altered pathways in neuronal development and steroid biosynthesis. PLoS One, 4, e5775.
Hu, V. W., Sarachana, T., Kim, K. S., Nguyen, A., Kulkarni, S., Steinberg, M. E., et al.
(2009). Gene expression profiling differentiates autism case-controls and phenotypic var-
iants of autism spectrum disorders: Evidence for circadian rhythm dysfunction in severe
autism. Autism Research, 2, 78–97.
Autism Susceptibility Genes and the Transcriptional Landscape of the Human Brain 317
Kang, H. J., Kawasawa, Y. I., Cheng, F., Zhu, Y., Xu, X., Li, M., et al. (2011). Spatio-
temporal transcriptome of the human brain. Nature, 478, 483–489.
Kanner, L. (1968). Autistic disturbances of affective contact. Acta Paedopsychiatrica, 35, 100–136.
Kim, S. H., & Lord, C. (2013). The behavioral manifestations of autism spectrum disorders.
In The neuroscience of autism spectrum disorders (pp. 25–37). Oxford, UK: Elsevier.
Li, X., Zou, H., & Brown, W. T. (2012). Genes associated with autism spectrum disorder.
Brain Research Bulletin, 88, 543–552.
Lintas, C., Sacco, R., & Persico, A. M. (2010). Genome-wide expression studies in Autism
spectrum disorder, Rett syndrome, and Down syndrome. Neurobiology of Disease, 45(1),
57–68.
Lord, C., Petkova, E., Hus, V., Gan, W., Lu, F., Martin, D. M., et al. (2012). A multisite study
of the clinical diagnosis of different autism spectrum disorders. Archives of General Psychiatry,
69, 306–313.
Morrow, E. M. (2010). Genomic copy number variation in disorders of cognitive develop-
ment. Journal of the American Academy of Child and Adolescent Psychiatry, 49, 1091–1104.
Murdoch, J. D., & State, M. W. (2013). Recent developments in the genetics of autism spec-
trum disorders. Current Opinion in Genetics & Development, 23(3), 310–315.
Neale, B. M., Kou, Y., Liu, L., Ma’ayan, A., Samocha, K. E., Sabo, A., et al. (2012). Patterns and
rates of exonic de novo mutations in autism spectrum disorders. Nature, 485, 242–245.
Nishimura, Y., Martin, C. L., Vazquez-Lopez, A., Spence, S. J., Alvarez-Retuerto, A. I.,
Sigman, M., et al. (2007). Genome-wide expression profiling of lymphoblastoid cell lines
distinguishes different forms of autism and reveals shared pathways. Human Molecular
Genetics, 16, 1682–1698.
Nord, A. S., Roeb, W., Dickel, D. E., Walsh, T., Kusenda, M., O’Connor, K. L., et al.
(2011). Reduced transcript expression of genes affected by inherited and de novo CNVs
in autism. European Journal of Human Genetics, 19(6), 727–731.
O’Roak, B. J., Deriziotis, P., Lee, C., Vives, L., Schwartz, J. J., Girirajan, S., et al. (2011).
Exome sequencing in sporadic autism spectrum disorders identifies severe de novo muta-
tions. Nature Genetics, 43, 585–589.
O’Roak, B. J., Vives, L., Girirajan, S., Karakoc, E., Krumm, N., Coe, B. P., et al. (2012).
Sporadic autism exomes reveal a highly interconnected protein network of de novo
mutations. Nature, 485, 246–250.
Pinto, D., Pagnamenta, A. T., Klei, L., Anney, R., Merico, D., Regan, R., et al. (2010).
Functional impact of global rare copy number variation in autism spectrum disorders.
Nature, 466, 368–372.
Provenzano, G., Zunino, G., Genovesi, S., Sgado, P., & Bozzi, Y. (2012). Mutant mouse
models of autism spectrum disorders. Disease Markers, 33, 225–239.
Purcell, A. E., Jeon, O. H., Zimmerman, A. W., Blue, M. E., & Pevsner, J. (2001).
Postmortem brain abnormalities of the glutamate neurotransmitter system in autism.
Neurology, 57, 1618–1628.
Sanders, S. J., Murtha, M. T., Gupta, A. R., Murdoch, J. D., Raubeson, M. J., Jeremy
Willsey, A., et al. (2012). De novo mutations revealed by whole-exome sequencing
are strongly associated with autism. Nature, 485, 237–241.
Sebat, J., Lakshmi, B., Malhotra, D., Troge, J., Lese-Martin, C., Walsh, T., et al. (2007).
Strong association of de novo copy number mutations with autism. Science, 316, 445–449.
Skuse, D. H. (2012). DSM-5’s conceptualization of autistic disorders. Journal of the American
Academy of Child and Adolescent Psychiatry, 51, 344–346.
Song, R. R., Zou, L., Zhong, R., Zheng, X. W., Zhu, B. B., Chen, W., et al. (2011). An
integrated meta-analysis of two variants in HOXA1/HOXB1 and their effect on the risk
of autism spectrum disorders. PLoS One, 6, e25603.
Splawski, I., Timothy, K. W., Sharpe, L. M., Decher, N., Kumar, P., Bloise, R., et al. (2004).
Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhyth-
mia and autism. Cell, 119, 19–31.
318 Shingo Miyauchi and Irina Voineagu
State, M. W. (2010). The genetics of child psychiatric disorders: Focus on autism and
Tourette syndrome. Neuron, 68, 254–269.
State, M. W., & Levitt, P. (2011). The conundrums of understanding genetic risks for autism
spectrum disorders. Nature Neuroscience, 14, 1499–1506.
Steffenburg, S., Gillberg, C., Hellgren, L., Andersson, L., Gillberg, I. C., Jakobsson, G., et al.
(1989). A twin study of autism in Denmark, Finland, Iceland, Norway and Sweden. Jour-
nal of Child Psychology and Psychiatry, 30, 405–416.
Voineagu, I. (2012). Gene expression studies in autism: Moving from the genome to the
transcriptome and beyond. Neurobiology of Disease, 45, 69–75.
Voineagu, I., Wang, X., Johnston, P., Lowe, J. K., Tian, Y., Horvath, S., et al. (2011). Trans-
criptomic analysis of autistic brain reveals convergent molecular pathology. Nature, 474,
380–384.
Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner, J. T., Abrahams, B. S., et al. (2009).
Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature,
459, 528–533.
Weiss, L. A., Arking, D. E., Daly, M. J., & Chakravarti, A. (2009). A genome-wide linkage
and association scan reveals novel loci for autism. Nature, 461, 802–808.
Wiznitzer, M. (2004). Autism and tuberous sclerosis. Journal of Child Neurology, 19, 675–679.
Xu, L. M., Li, J. R., Huang, Y., Zhao, M., Tang, X., & Wei, L. (2012). AutismKB: An
evidence-based knowledgebase of autism genetics. Nucleic Acids Research, 40,
D1016–D1022.
Yu, T. W., Chahrour, M. H., Coulter, M. E., Jiralerspong, S., Okamura-Ikeda, K.,
Ataman, B., et al. (2013). Using whole-exome sequencing to identify inherited causes
of autism. Neuron, 77, 259–273.
Zhou, X., Xu, Y., Wang, J., Zhou, H., Liu, X., Ayub, Q., et al. (2011). Replication of the
association of a MET variant with autism in a Chinese Han population. PLoS One, 6,
e27428.
Ziats, M. N., & Rennert, O. M. (2013). Aberrant expression of long noncoding RNAs in
autistic brain. Journal of Molecular Neuroscience, 49, 589–593.
INDEX
319
320 Index
N Neurogenesis
ncRNAs. See Noncoding RNAs defects, 176
(ncRNAs) early cortical, mouse, 175–176
Neocortical circuit assembly, transcriptional gliogenesis, 173f
dysregulation. See Transcriptional neocortical projection neurons, 175
dysregulation Neuroimaging
Neocortical inhibitory interneurons, 177 cerebellar hypoplasia, 10
Neocortical projection neurons vermal lobules, 10
CNTNAP2 absence, 176–177 voxel-based morphometry studies,
neurogenesis, 175–176 10–11, 11f
postmigratory neurons, 175 Neuroimmune abnormalities
preplate (PP), 174–175 brain cytokine dysregulation, 282–283
Reeler mutation, 176 immune molecules, brain development,
RGCs, 174 283–285, 284t
subtypes, 174 microglia, 280–281
Neural circuit wiring, 169, 193 transcriptome changes, 281–282
Neural development, MET Neuronal migration
disconnection syndrome and defects, 176–177, 192
pathophysiology, 152 definition, 169–170
functional and structural imaging studies, neocortical minicolumns, 169–170
154–155 Reeler mutant, 176
GABAergic interneurons, 152 Satb2 absence, 192
intracellular mechanisms, 152 Noncoding RNAs (ncRNAs)
laser scanning photostimulation (LSPS), categories, 36
155–156 long (see Long noncoding RNAs
mature synapses, 152–153 (LncRNAs))
Metfx/fx and Emx1cre neurons, small, 36–38
153–154
neuroanatomical and functional mapping, P
155–156 Peripheral immune abnormalities, ASD
neurogenetic and neuroanatomical blood protein markers, 285–286
approaches, 155–156 blood transcriptome, 286–287
neurological dysfunction, 153–154 leukocyte abundance and function, 286
physiological role, 152–153 Postnatal immune dysregulation, ASD
synaptic proteins, 152–153 neuroimmune abnormalities
Neurodegenerative diseases (see Neuroimmune abnormalities)
BC200 RNA, 44 peripheral immune abnormalities,
lncRNAs, 44 285–287
Neurodevelopmental disorders
FMR1 genes, 45 R
HAR1, 46–47 Radial glial cells (RGCs), 173f, 174
RMST, 46 RAR-related orphan receptor alpha
schizophrenia, 45 (ROR-alpha), 15
UBE3A-ATS, 46 RBFOX1 gene
Neurodevelopmental programs, RBFOX1. dysregulation, 259–263
See RBFOX1 gene genetic variation and ASD, 253–258
Neurodevelopment, immune molecules, model systems, 258–259
283–285, 284t RNA splicing factor, 252
326 Index
gene expression and age, correlation, 311t manipulation, pup, 116, 118f
gene expression variation in human brain, mutations, 116–118
313f NLGN4 knockout mice, 116–118
genes involved in synapse development, SHANK2 knockout females, 118
309–310 speech and language disorders, 118–119
rate of change in gene expression, 309 Urokinase-type plasminogen activator
temporal and spatial dynamics, 309–310 receptor (uPAR), 142–143, 152
temporally regulated gene expression, 310
temporal variation, 310–314 V
Translating ribosome affinity purification Voxel-based morphometry studies, 10–11,
(TRAP), 74 11f
TRAP. See Translating ribosome affinity
purification (TRAP)
W
U Weighted gene coexpression network
Ultrasonic vocalizations (USVs) analysis (WGCNA), 120–122
autistic-like communication, 116–118
communication, 116 X
experimental set up, 116, 117f X-chromosome inactivation (XCI), 38–40
Foxp2 mutant mice, 116, 118–119 X-inactive specific transcript (Xist), 38–40
CONTENTS OF RECENT VOLUMES
329
330 Contents of Recent Volumes
Free Radicals, Calcium, and the Synaptic Vesicle Recycling at the Drosophila Neuromuscu-
Plasticity-Cell Death Continuum: Emerging lar Junction
Roles of the Trascription Factor NFkB Daniel T. Stimson and Mani Ramaswami
Mark P. Mattson
Ionic Currents in Larval Muscles of Drosophila
AP-I Transcription Factors: Short- and Long- Satpal Singh and Chun-Fang Wu
Term Modulators of Gene Expression in the Brain
Development of the Adult Neuromuscular
Keith Pennypacker
System
Ion Channels in Epilepsy Joyce J. Fernandes and Haig Keshishian
Istvan Mody
Controlling the Motor Neuron
Posttranslational Regulation of Ionotropic Gluta- James R. Trimarchi, Ping Jin, and Rodney
mate Receptors and Synaptic Plasticity K. Murphey
Xiaoning Bi, Steve Standley, and Michel Baudry
Heritable Mutations in the Glycine, GABAA, and
Nicotinic Acetylcholine Receptors Provide New
Insights into the Ligand-Gated Ion Channel
Volume 44
Receptor Superfamily Human Ego-Motion Perception
Behnaz Vafa and Peter R. Schofield A. V. van den Berg
INDEX Optic Flow and Eye Movements
M. Lappe and K.-P. Hoffman
The Role of MST Neurons during Ocular Track-
Volume 43 ing in 3D Space
K. Kawano, U. Inoue, A. Takemura, Y. Kodaka,
Early Development of the Drosophila Neuromus-
and F. A. Miles
cular Junction: A Model for Studying Neuronal
Networks in Development Visual Navigation in Flying Insects
Akira Chiba M. V. Srinivasan and S.-W. Zhang
Development of Larval Body Wall Muscles Neuronal Matched Filters for Optic Flow
Michael Bate, Matthias Landgraf, and Mar Ruiz Processing in Flying Insects
Gómez Bate H. G. Krapp
Development of Electrical Properties and Synaptic A Common Frame of Reference for the Analysis
Transmission at the Embryonic Neuromuscular of Optic Flow and Vestibular Information
Junction B. J. Frost and D. R. W. Wylie
Kendal S. Broadie
Optic Flow and the Visual Guidance of
Ultrastructural Correlates of Neuromuscular Locomotion in the Cat
Junction Development H. Sherk and G. A. Fowler
Mary B. Rheuben, Motojiro Yoshihara, and
Stages of Self-Motion Processing in Primate
Yoshiaki Kidokoro
Posterior Parietal Cortex
Assembly and Maturation of the Drosophila Larval F. Bremmer, J.-R. Duhamel, S. B. Hamed, and
Neuromuscular Junction W. Graf
L. Sian Gramates and Vivian Budnik
Optic Flow Analysis for Self-Movement
Second Messenger Systems Underlying Plasticity Perception
at the Neuromuscular Junction C. J. Duffy
Frances Hannan and Yi Zhong
Neural Mechanisms for Self-Motion Perception
Mechanisms of Neurotransmitter Release in Area MST
J. Troy Littleton, Leo Pallanck, and Barry R. A. Andersen, K. V. Shenoy, J. A. Crowell,
Ganetzky and D. C. Bradley
Contents of Recent Volumes 333
Volume 51 INDEX
Volume 56
Volume 55
Behavioral Mechanisms and the Neurobiology of
Section I: Virsu Vectors For Use in the Nervous Conditioned Sexual Responding
System Mark Krause
Non-Neurotropic Adenovirus: a Vector for Gene NMDA Receptors in Alcoholism
Transfer to the Brain and Gene Therapy of Neu- Paula L. Hoffman
rological Disorders
P. R. Lowenstein, D. Suwelack, J. Hu, X. Yuan, Processing and Representation of Species-Specific
M. Jimenez-Dalmaroni, S. Goverdhama, and Communication Calls in the Auditory System of
M.G. Castro Bats
George D. Pollak, Achim Klug, and Eric E. Bauer
Adeno-Associated Virus Vectors
E. Lehtonen and L. Tenenbaum Central Nervous System Control of Micturition
Gert Holstege and Leonora J. Mouton
Problems in the Use of Herpes Simplex Virus as a
Vector The Structure and Physiology of the Rat Auditory
L. T. Feldman System: An Overview
Manuel Malmierca
Lentiviral Vectors
J. Jakobsson, C. Ericson, N. Rosenquist, and Neurobiology of Cat and Human Sexual Behavior
C. Lundberg Gert Holstege and J. R. Georgiadis
Dopamine Transporter Network and Pathways Neuroimaging Studies in Bipolar Children and
Rajani Maiya and R. Dayne Mayfield Adolescents
Rene L. Olvera, David C. Glahn, Sheila
Proteomic Approaches in Drug Discovery
C. Caetano, Steven R. Pliszka, and Jair C. Soares
and Development
Holly D. Soares, Stephen A. Williams, Peter Chemosensory G-Protein-Coupled Receptor
J. Snyder, Feng Gao, Tom Stiger, Christian Rohlff, Signaling in the Brain
Athula Herath, Trey Sunderland, Karen Putnam, Geoffrey E. Woodard
and W. Frost White
Disturbances of Emotion Regulation after Focal
Section III: Informatics Brain Lesions
Antoine Bechara
Proteomic Informatics
Steven Russell, William Old, Katheryn Resing, The Use of Caenorhabditis elegans in Molecular
and Lawrence Hunter Neuropharmacology
Jill C. Bettinger, Lucinda Carnell, Andrew
Section IV: Changes in the Proteome by Disease
G. Davies, and Steven L. McIntire
Proteomics Analysis in Alzheimer’s Disease: New
INDEX
Insights into Mechanisms of Neurodegeneration
D. Allan Butterfield and Debra Boyd-Kimball
Proteomics and Alcoholism
Volume 63
Frank A. Witzmann and Wendy N. Strother Mapping Neuroreceptors at work: On the Defini-
tion and Interpretation of Binding Potentials after
Proteomics Studies of Traumatic Brain Injury
20 years of Progress
Kevin K. W. Wang, Andrew Ottens,
Albert Gjedde, Dean F. Wong, Pedro Rosa-Neto,
William Haskins, Ming Cheng Liu, Firas
and Paul Cumming
Kobeissy, Nancy Denslow, SuShing Chen, and
Ronald L. Hayes Mitochondrial Dysfunction in Bipolar Disorder:
From 31P-Magnetic Resonance Spectroscopic
Influence of Huntington’s Disease on the Human
Findings to Their Molecular Mechanisms
and Mouse Proteome
Tadafumi Kato
Claus Zabel and Joachim Klose
Large-Scale Microarray Studies of Gene Expres-
Section V: Overview of the Neuroproteome
sion in Multiple Regions of the Brain in Schizo-
Proteomics—Application to the Brain phrenia and Alzeimer’s Disease
Katrin Marcus, Oliver Schmidt, Heike Schaefer, Pavel L. Katsel, Kenneth L. Davis, and Vahram
Michael Hamacher, AndrÅ van Hall, and Helmut Haroutunian
E. Meyer
Regulation of Serotonin 2C Receptor PRE-
INDEX mRNA Editing By Serotonin
Claudia Schmauss
The Dopamine Hypothesis of Drug Addiction:
Volume 62 Hypodopaminergic State
Miriam Melis, Saturnino Spiga, and Marco Diana
GABAA Receptor Structure–Function Studies:
A Reexamination in Light of New Acetylcholine Human and Animal Spongiform Encephalopa-
Receptor Structures thies are Autoimmune Diseases: A Novel Theory
Myles H. Akabas and Its supporting Evidence
Bao Ting Zhu
Dopamine Mechanisms and Cocaine Reward
Aiko Ikegami and Christine L. Duvauchelle Adenosine and Brain Function
Bertil B. Fredholm, Jiang-Fan Chen, Rodrigo
Proteolytic Dysfunction in Neurodegenerative
A. Cunha, Per Svenningsson, and Jean-Marie Vaugeois
Disorders
Kevin St. P. McNaught INDEX
342 Contents of Recent Volumes
Effects of Genes and Stress on the Neurobiology of Artistic Changes in Alzheimer’s Disease
Depression Sebastian J. Crutch and Martin N. Rossor
J. John Mann and Dianne Currier
Section IV: Cerebrovascular Disease
Quantitative Imaging with the Micropet Small-
Stroke in Painters
Animal Pet Tomograph
H. Bäzner and M. Hennerici
Paul Vaska, Daniel J. Rubins, David L. Alexoff,
and Wynne K. Schiffer Visuospatial Neglect in Lovis Corinth’s Self-
Portraits
Understanding Myelination through Studying its
Olaf Blanke
Evolution
Rüdiger Schweigreiter, Betty I. Roots, Art, Constructional Apraxia, and the Brain
Christine Bandtlow, and Robert M. Gould Louis Caplan
INDEX Section V: Genetic Diseases
Neurogenetics in Art
Alan E. H. Emery
Volume 74 A Naı̈ve Artist of St Ives
Evolutionary Neurobiology and Art F. Clifford Rose
C. U. M. Smith
Van Gogh’s Madness
Section I: Visual Aspects F. Clifford Rose
Perceptual Portraits Absinthe, The Nervous System and Painting
Nicholas Wade Tiina Rekand
The Neuropsychology of Visual Art: Conferring Section VI: Neurologists as Artists
Capacity
Anjan Chatterjee Sir Charles Bell, KGH, FRS, FRSE
(1774–1842)
Vision, Illusions, and Reality Christopher Gardner-Thorpe
Christopher Kennard
Section VII: Miscellaneous
Localization in the Visual Brain
Peg Leg Frieda
George K. York
Espen Dietrichs
Section II: Episodic Disorders
The Deafness of Goya (1746–1828)
Neurology, Synaesthesia, and Painting F. Clifford Rose
Amy Ione
INDEX
Fainting in Classical Art
Philip Smith
Migraine Art in the Internet: A Study of 450
Contemporary Artists
Klaus Podoll
Volume 75
Introduction on the Use of the Drosophila Embry-
Sarah Raphael’s Migraine with Aura as Inspiration
onic/Larval Neuromuscular Junction as a Model
for the Foray of Her Work into Abstraction
System to Study Synapse Development and
Klaus Podoll and Debbie Ayles
Function, and a Brief Summary of Pathfinding
The Visual Art of Contemporary Artists with and Target Recognition
Epilepsy Catalina Ruiz-Cañada and Vivian Budnik
Steven C. Schachter
Development and Structure of Motoneurons
Section III: Brain Damage Matthias Landgraf and Stefan Thor
Creativity in Painting and Style in Brain- The Development of the Drosophila Larval Body
Damaged Artists Wall Muscles
Julien Bogousslavsky Karen Beckett and Mary K. Baylies
Contents of Recent Volumes 347
Organization of the Efferent System and Structure ID, Ego, and Temporal Lobe Revisited
of Neuromuscular Junctions in Drosophila Shirley M. Ferguson and Mark Rayport
Andreas Prokop
Section II: Stereotaxic Studies
Development of Motoneuron Electrical Proper-
Olfactory Gustatory Responses Evoked by
ties and Motor Output
Electrical Stimulation of Amygdalar Region in
Richard A. Baines
Man Are Qualitatively Modifiable by Interview
Transmitter Release at the Neuromuscular Content: Case Report and Review
Junction Mark Rayport, Sepehr Sani, and Shirley M. Ferguson
Thomas L. Schwarz
Section III: Controversy in Definition of Behav-
Vesicle Trafficking and Recycling at the Neuro- ioral Disturbance
muscular Junction: Two Pathways for Endocytosis
Pathogenesis of Psychosis in Epilepsy. The
Yoshiaki Kidokoro
“Seesaw” Theory: Myth or Reality?
Glutamate Receptors at the Drosophila Neuromus- Shirley M. Ferguson and Mark Rayport
cular Junction
Section IV: Outcome of Temporal Lobectomy
Aaron DiAntonio
Memory Function After Temporal Lobectomy for
Scaffolding Proteins at the Drosophila Neuromus-
Seizure Control: A Comparative Neuropsy chi-
cular Junction
atric and Neuropsychological Study
Bulent Ataman, Vivian Budnik, and Ulrich Thomas
Shirley M. Ferguson, A. John McSweeny, and Mark
Synaptic Cytoskeleton at the Neuromuscular Rayport
Junction
Life After Surgery for Temporolimbic Seizures
Catalina Ruiz-Cañada and Vivian Budnik
Shirley M. Ferguson, Mark Rayport, and Carolyn
Plasticity and Second Messengers During Synapse A. Schell
Development
Appendix I
Leslie C. Griffith and Vivian Budnik
Mark Rayport
Retrograde Signaling that Regulates Synaptic
Appendix II: Conceptual Foundations of Studies
Development and Function at the Drosophila Neu-
of Patients Undergoing Temporal Lobe Surgery
romuscular Junction
for Seizure Control
Guillermo Marqués and Bing Zhang
Mark Rayport
Activity-Dependent Regulation of Transcription
INDEX
During Development of Synapses
Subhabrata Sanyal and Mani Ramaswami
Experience-Dependent Potentiation of Larval
Neuromuscular Synapses
Volume 77
Christoph M. Schuster Regenerating the Brain
David A. Greenberg and Kunlin Jin
Selected Methods for the Anatomical Study of
Drosophila Embryonic and Larval Neuromuscular Serotonin and Brain: Evolution, Neuroplasticity,
Junctions and Homeostasis
Vivian Budnik, Michael Gorczyca, and Andreas Efrain C. Azmitia
Prokop
INDEX
Therapeutic Approaches to Promoting Axonal
Regeneration in the Adult Mammalian Spinal Cord
Volume 76 Sari S. Hannila, Mustafa M. Siddiq, and Marie
T. Filbin
Section I: Physiological Correlates of Freud’s
Evidence for Neuroprotective Effects of Antipsy-
Theories
chotic Drugs: Implications for the Pathophysio-
The ID, the Ego, and the Temporal Lobe logy and Treatment of Schizophrenia
Shirley M. Ferguson and Mark Rayport Xin-Min Li and Haiyun Xu
348 Contents of Recent Volumes
Neurogenesis and Neuroenhancement in the Patho- Schizophrenia and the a7 Nicotinic Acetylcholine
physiology and Treatment of Bipolar Disorder Receptor
Robert J. Schloesser, Guang Chen, and Husseini Laura F. Martin and Robert Freedman
K. Manji
Histamine and Schizophrenia
Neuroreplacement, Growth Factor, and Small Jean-Michel Arrang
Molecule Neurotrophic Approaches for Treating
Cannabinoids and Psychosis
Parkinson’s Disease
Deepak Cyril D’Souza
Michael J. O’Neill, Marcus J. Messenger, Viktor
Lakics, Tracey K. Murray, Eric H. Karran, Philip Involvement of Neuropeptide Systems in Schizo-
G. Szekeres, Eric S. Nisenbaum, and Kalpana phrenia: Human Studies
M. Merchant Ricardo Cáceda, Becky Kinkead, and Charles
B. Nemeroff
Using Caenorhabditis elegans Models of Neuro-
degenerative Disease to Identify Neuroprotective Brain-Derived Neurotrophic Factor in Schizo-
Strategies phrenia and Its Relation with Dopamine
Brian Kraemer and Gerard D. Schellenberg Olivier Guillin, Caroline Demily, and Florence
Thibaut
Neuroprotection and Enhancement of Neurite
Outgrowth With Small Molecular Weight Com- Schizophrenia Susceptibility Genes: In Search of a
pounds From Screens of Chemical Libraries Molecular Logic and Novel Drug Targets for a
Donard S. Dwyer and Addie Dickson Devastating Disorder
Joseph A. Gogos
INDEX
INDEX
Volume 78
Neurobiology of Dopamine in Schizophrenia
Olivier Guillin, Anissa Abi-Dargham, and Marc Volume 79
Laruelle
The Destructive Alliance: Interactions of
The Dopamine System and the Pathophysiology Leukocytes, Cerebral Endothelial Cells, and the
of Schizophrenia: A Basic Science Perspective Immune Cascade in Pathogenesis of Multiple
Yukiori Goto and Anthony A. Grace Sclerosis
Alireza Minagar, April Carpenter, and J. Steven
Glutamate and Schizophrenia: Phencyclidine,
Alexander
N-methyl-D-aspartate Receptors, and Dopamine–
Glutamate Interactions Role of B Cells in Pathogenesis of Multiple
Daniel C. Javitt Sclerosis
Behrouz Nikbin, Mandana Mohyeddin Bonab,
Deciphering the Disease Process of Schizophrenia:
Farideh Khosravi, and Fatemeh Talebian
The Contribution of Cortical GABA Neurons
David A. Lewis and Takanori Hashimoto The Role of CD4 T Cells in the Pathogenesis of
Multiple Sclerosis
Alterations of Serotonin Transmission in
Tanuja Chitnis
Schizophrenia
Anissa Abi-Dargham The CD8 T Cell in Multiple Sclerosis: Suppressor
Cell or Mediator of Neuropathology?
Serotonin and Dopamine Interactions in Rodents
Aaron J. Johnson, Georgette L. Suidan, Jeremiah
and Primates: Implications for Psychosis and Anti-
McDole, and Istvan Pirko
psychotic Drug Development
Gerard J. Marek Immunopathogenesis of Multiple Sclerosis
Smriti M. Agrawal and V. Wee Yong
Cholinergic Circuits and Signaling in the Patho-
physiology of Schizophrenia Molecular Mimicry in Multiple Sclerosis
Joshua A. Berman, David A. Talmage, and Lorna Jane E. Libbey, Lori L. McCoy, and Robert
W. Role S. Fujinami
Contents of Recent Volumes 349
Life and Death of Neurons in the Aging Recruitment and Retention in Clinical Trials of
Cerebral Cortex the Elderly
John H. Morrison and Patrick R. Hof Flavia M. Macias, R. Eugene Ramsay, and
A. James Rowan
An In Vitro Model of Stroke-Induced Epilepsy:
Elucidation of the Roles of Glutamate and Treatment of Convulsive Status Epilepticus
Calcium in the Induction and Maintenance of David M. Treiman
Stroke-Induced Epileptogenesis Treatment of Nonconvulsive Status Epilepticus
Robert J. DeLorenzo, David A. Sun, Robert Matthew C. Walker
E. Blair, and Sompong Sambati
Antiepileptic Drug Formulation and Treatment
Mechanisms of Action of Antiepileptic Drugs in the Elderly: Biopharmaceutical Considerations
H. Steve White, Misty D. Smith, and Barry E. Gidal
Karen S. Wilcox
INDEX
Epidemiology and Outcomes of Status Epilepticus
in the Elderly
Alan R. Towne
New Insights into the Roles of Metalloproteinases Differential Modulation of Type 1 and Type 2
in Neurodegeneration and Neuroprotection Cannabinoid Receptors Along the Neuroimmune
A. J. Turner and N. N. Nalivaeva Axis
Sergio Oddi, Paola Spagnuolo, Monica Bari,
Relevance of High-Mobility Group Protein Antonella D’Agostino, and Mauro Maccarrone
Box 1 to Neurodegeneration
Silvia Fossati and Alberto Chiarugi Effects of the HIV-1 Viral Protein Tat on Central
Neurotransmission: Role of Group I Meta-
Early Upregulation of Matrix Metalloproteinases botropic Glutamate Receptors
Following Reperfusion Triggers Neuro-
Elisa Neri, Veronica Musante, and Anna Pittaluga
inflammatory Mediators in Brain Ischemia in Rat
Diana Amantea, Rossella Russo, Micaela Gliozzi, Evidence to Implicate Early Modulation of Inter-
Vincenza Fratto, Laura Berliocchi, G. Bagetta, leukin-1b Expression in the Neuroprotection
G. Bernardi, and M. Tiziana Corasaniti Afforded by 17b-Estradiol in Male Rats Under-
gone Transient Middle Cerebral Artery Occlusion
The (Endo)Cannabinoid System in Multiple Olga Chiappetta, Micaela Gliozzi, Elisa Siviglia,
Sclerosis and Amyotrophic Lateral Sclerosis
Diana Amantea, Luigi A. Morrone, Laura
Diego Centonze, Silvia Rossi, Alessandro Berliocchi, G. Bagetta, and M. Tiziana Corasaniti
Finazzi-Agrò, Giorgio Bernardi, and Mauro
Maccarrone A Role for Brain Cyclooxygenase-2 and Prosta-
glandin-E2 in Migraine: Effects of Nitroglycerin
Chemokines and Chemokine Receptors: Multi- Cristina Tassorelli, Rosaria Greco, Marie Therèse
purpose Players in Neuroinflammation
Armentero, Fabio Blandini, Giorgio Sandrini, and
Richard M. Ransohoff, LiPing Liu, and Astrid Giuseppe Nappi
E. Cardona
The Blockade of K+-ATP Channels has Neuro-
Systemic and Acquired Immune Responses in protective Effects in an In Vitro Model of Brain
Alzheimer’s Disease Ischemia
Markus Britschgi and Tony Wyss-Coray
Robert Nisticò, Silvia Piccirilli, L. Sebastianelli,
Neuroinflammation in Alzheimer’s Disease and Giuseppe Nisticò, G. Bernardi, and N. B. Mercuri
Parkinson’s Disease: Are Microglia Pathogenic
Retinal Damage Caused by High Intraocular
in Either Disorder?
Pressure-Induced Transient Ischemia is Prevented
Joseph Rogers, Diego Mastroeni, Brian Leonard,
by Coenzyme Q10 in Rat
Jeffrey Joyce, and Andrew Grover
Carlo Nucci, Rosanna Tartaglione, Angelica
Cytokines and Neuronal Ion Channels in Health Cerulli, R. Mancino, A. Spanò, Federica Cavaliere,
and Disease Laura Rombolà, G. Bagetta, M. Tiziana
Barbara Viviani, Fabrizio Gardoni, and Marina Corasaniti, and Luigi A. Morrone
Marinovich
Evidence Implicating Matrix Metalloproteinases
Cyclooxygenase-2, Prostaglandin E2, and Micro- in the Mechanism Underlying Accumulation of
glial Activation in Prion Diseases IL-1b and Neuronal Apoptosis in the Neocortex
Luisa Minghetti and Maurizio Pocchiari of HIV/gp120-Exposed Rats
Rossella Russo, Elisa Siviglia, Micaela Gliozzi,
Glia Proinflammatory Cytokine Upregulation as a
Diana Amantea, Annamaria Paoletti,
Therapeutic Target for Neurodegenerative
Laura Berliocchi, G. Bagetta, and
Diseases: Function-Based and Target-Based
M. Tiziana Corasaniti
Discovery Approaches
Linda J. Van Eldik, Wendy L. Thompson, Neuroprotective Effect of Nitroglycerin in a
Hantamalala Ralay Ranaivo, Heather A. Behanna, Rodent Model of Ischemic Stroke: Evaluation
and D. Martin Watterson of Bcl-2 Expression
Rosaria Greco, Diana Amantea, Fabio Blandini,
Oxidative Stress and the Pathogenesis of Neuro-
Giuseppe Nappi, Giacinto Bagetta, M. Tiziana
degenerative Disorders
Corasaniti, and Cristina Tassorelli
Ashley Reynolds, Chad Laurie, R. Lee Mosley, and
Howard E. Gendelman INDEX
Contents of Recent Volumes 353
Involvement of the Prefrontal Cortex in Problem Bv8/Prokineticins and their Receptors: A New
Solving Pronociceptive System
Hajime Mushiake, Kazuhiro Sakamoto, Naohiro Lucia Negri, Roberta Lattanzi, Elisa Giannini,
Saito, Toshiro Inui, Kazuyuki Aihara, and Jun Michela Canestrelli, Annalisa Nicotra,
Tanji and Pietro Melchiorri
Contents of Recent Volumes 355
Bidirectional Interfaces with the Peripheral Section Four: Brain-Machine Interfaces and
Nervous System Space
Silvestro Micera and Xavier Navarro Adaptive Changes of Rhythmic EEG Oscillations
in Space: Implications for Brain–Machine
Interfacing Insect Brain for Space Applications
Interface Applications
Giovanni Di Pino, Tobias Seidl,
G. Cheron, A. M. Cebolla, M. Petieau,
Antonella Benvenuto, Fabrizio Sergi, Domenico
A. Bengoetxea, E. Palmero-Soler, A. Leroy, and
Campolo, Dino Accoto, Paolo Maria Rossini,
B. Dan
and Eugenio Guglielmelli
Validation of Brain–Machine Interfaces During
Section Two: Meet the Brain
Parabolic Flight
Meet the Brain: Neurophysiology
José del R. Millán, Pierre W. Ferrez, and Tobias
John Rothwell
Seidl
Fundamentals of Electroencefalography, Magne-
Matching Brain–Machine Interface Performance
toencefalography, and Functional Magnetic
to Space Applications
Resonance Imaging
Luca Citi, Oliver Tonet, and Martina Marinelli
Claudio Babiloni, Vittorio Pizzella, Cosimo Del
Gratta, Antonio Ferretti, and Gian Luca Romani Brain–Machine Interfaces for Space
Applications—Research, Technological Devel-
Implications of Brain Plasticity to Brain–Machine
opment, and Opportunities
Interfaces Operation: A Potential Paradox?
Leopold Summerer, Dario Izzo, and Luca Rossini
Paolo Maria Rossini
INDEX
Section Three: Brain Machine Interfaces, A New
Brain-to-Environment Communication Channel
An Overview of BMIs
Francisco Sepulveda Volume 87
Neurofeedback and Brain–Computer Interface: Peripheral Nerve Repair and Regeneration
Clinical Applications Research: A Historical Note
Niels Birbaumer, Ander Ramos Murguialday, Bruno Battiston, Igor Papalia, Pierluigi Tos, and
Cornelia Weber, and Pedro Montoya Stefano Geuna
Flexibility and Practicality: Graz Brain–Computer Development of the Peripheral Nerve
Interface Approach Suleyman Kaplan, Ersan Odaci, Bunyami Unal,
Reinhold Scherer, Gernot R. Müller-Putz, and Bunyamin Sahin, and Michele Fornaro
Gert Pfurtscheller
Histology of the Peripheral Nerve and Changes
On the Use of Brain–Computer Interfaces Out- Occurring During Nerve Regeneration
side Scientific Laboratories: Toward an Applica- Stefano Geuna, Stefania Raimondo, Giulia Ronchi,
tion in Domotic Environments Federica Di Scipio, Pierluigi Tos, Krzysztof Czaja,
F. Babiloni, F. Cincotti, M. Marciani, S. Salinari, and Michele Fornaro
L. Astolfi, F. Aloise, F. De Vico Fallani, and
Methods and Protocols in Peripheral Nerve
D. Mattia
Regeneration Experimental Research:
Brain–Computer Interface Research at the Part I—Experimental Models
Wadsworth Center: Developments in Noninva- Pierluigi Tos, Giulia Ronchi, Igor Papalia,
sive Communication and Control Vera Sallen, Josette Legagneux, Stefano Geuna, and
Dean J. Krusienski and Jonathan R. Wolpaw Maria G. Giacobini-Robecchi
Watching Brain TV and Playing Brain Ball: Methods and Protocols in Peripheral Nerve
Exploring Novel BCL Strategies Using Real– Regeneration Experimental Research: Part
Time Analysis of Human Intercranial Data II—Morphological Techniques
Karim Jerbi, Samson Freyermuth, Lorella Minotti, Stefania Raimondo, Michele Fornaro, Federica Di
Philippe Kahane, Alain Berthoz, and Jean-Philippe Scipio, Giulia Ronchi, Maria G. Giacobini-
Lachaux Robecchi, and Stefano Geuna
Contents of Recent Volumes 357
Deciphering Rett Syndrome With Mouse Genet- Part III—Transcranial Sonography in other
ics, Epigenomics, and Human Neurons Movement Disorders and Depression
Jifang Tao, Hao Wu, and Yi Eve Sun
Transcranial Sonography in Brain Disorders with
INDEX Trace Metal Accumulation
Uwe Walter
Transcranial Sonography in Dystonia
Volume 90 Alexandra Gaenslen
Part I: Introduction Transcranial Sonography in Essential Tremor
Heike Stockner and Isabel Wurster
Introductory Remarks on the History and Current
Applications of TCS VII—Transcranial Sonography in Restless Legs
Matthew B. Stern Syndrome
Jana Godau and Martin Sojer
Method and Validity of Transcranial Sonography
in Movement Disorders Transcranial Sonography in Ataxia
David Školoudı´k and Uwe Walter Christos Krogias, Thomas Postert and Jens Eyding
Transcranial Sonography—Anatomy Transcranial Sonography in Huntington’s Disease
Heiko Huber Christos Krogias, Jens Eyding and Thomas Postert
Transcranial Sonography in Depression
Part II: Transcranial Sonography in Parkinsons Milija D. Mijajlovic
Disease
Transcranial Sonography in Relation to SPECT Part IV: Future Applications and Conclusion
and MIBG Transcranial Sonography-Assisted Stereotaxy and
Yoshinori Kajimoto, Hideto Miwa and Tomoyoshi Follow-Up of Deep Brain Implants in Patients
Kondo with Movement Disorders
Diagnosis of Parkinson’s Disease—Transcranial Uwe Walter
Sonography in Relation to MRI Conclusions
Ludwig Niehaus and Kai Boelmans Daniela Berg
Early Diagnosis of Parkinson’s Disease INDEX
Alexandra Gaenslen and Daniela Berg
Transcranial Sonography in the Premotor Diag-
nosis of Parkinson’s Disease
Stefanie Behnke, Ute Schroder and Daniela Berg Volume 91
Pathophysiology of Transcranial Sonography Sig- The Role of microRNAs in Drug Addiction:
nal Changes in the Human Substantia Nigra A Big Lesson from Tiny Molecules
K. L. Double, G. Todd and S. R. Duma Andrzej Zbigniew Pietrzykowski
Transcranial Sonography for the Discrimination of The Genetics of Behavioral Alcohol Responses in
Idiopathic Parkinson’s Disease from the Atypical Drosophila
Parkinsonian Syndromes Aylin R. Rodan and Adrian Rothenfluh
A. E. P. Bouwmans, A. M. M. Vlaar, K. Srulijes,
Neural Plasticity, Human Genetics, and Risk for
W. H. Mess AND W. E. J. Weber
Alcohol Dependence
Transcranial Sonography in the Discrimination of Shirley Y. Hill
Parkinson’s Disease Versus Vascular Parkinsonism
Using Expression Genetics to Study the Neurobi-
Pablo Venegas-Francke
ology of Ethanol and Alcoholism
TCS in Monogenic Forms of Parkinson’s Disease Sean P. Farris, Aaron R. Wolen and Michael
Kathrin Brockmann and Johann Hagenah F. Miles
360 Contents of Recent Volumes
Genetic Variation and Brain Gene Expression in Neuroimaging of Dreaming: State of the Art and
Rodent Models of Alcoholism: Implications for Limitations
Medication Development Caroline Kussé, Vincenzo Muto, Laura Mascetti,
Karl Björk, Anita C. Hansson and Luca Matarazzo, Ariane Foret, Anahita Shaffii-Le
W. olfgang H. Sommer Bourdiec and Pierre Maquet
Identifying Quantitative Trait Loci (QTLs) and Memory Consolidation, The Diurnal Rhythm of
Genes (QTGs) for Alcohol-Related Phenotypes Cortisol, and The Nature of Dreams: A New
in Mice Hypothesis
Lauren C. Milner and Kari J. Buck Jessica D. Payne
Glutamate Plasticity in the Drunken Amygdala: Characteristics and Contents of Dreams
The Making of an Anxious Synapse Michael Schredl
Brian A. Mccool, Daniel T. Christian, Marvin
Trait and Neurobiological Correlates of Individ-
R. Diaz and Anna K. Läck
ual Differences in Dream Recall and Dream
Ethanol Action on Dopaminergic Neurons in Content
the Ventral Tegmental Area: Interaction with Mark Blagrove and Edward F. Pace-Schott
Intrinsic Ion Channels and Neurotransmitter
Consciousness in Dreams
Inputs
David Kahn and Tzivia Gover
Hitoshi Morikawa and Richard
A. Morrisett The Underlying Emotion and the Dream: Relat-
ing Dream Imagery to the Dreamer’s Underlying
Alcohol and the Prefrontal Cortex
Emotion can Help Elucidate the Nature of
Kenneth Abernathy, L. Judson Chandler and John
Dreaming
J. Woodward
Ernest Hartmann
BK Channel and Alcohol, A Complicated Affair
Dreaming, Handedness, and Sleep Architecture:
Gilles Erwan Martin
Interhemispheric Mechanisms
A Review of Synaptic Plasticity at Purkinje Neu- Stephen D. Christman and Ruth E. Propper
rons with a Focus on Ethanol-Induced Cerebellar
To What Extent Do Neurobiological Sleep-
Dysfunction
Waking Processes Support Psychoanalysis?
C. Fernando Valenzuela, Britta Lindquist and
Claude Gottesmann
Paula A. Zflmudio-Bulcock
The Use of Dreams in Modern Psychotherapy
INDEX
Clara E. Hill and Sarah Knox
INDEX
Volume 92
The Development of the Science of Dreaming Volume 93
Claude Gottesmann
Underlying Brain Mechanisms that Regulate
Dreaming as Inspiration: Evidence from Religion, Sleep-Wakefulness Cycles
Philosophy, Literature, and Film Irma Gvilia
Kelly Bulkeley
What Keeps Us Awake?—the Role of Clocks and
Developmental Perspective: Dreaming Across the Hourglasses, Light, and Melatonin
Lifespan and What This Tells Us Christian Cajochen, Sarah Chellappa and Christina
Melissa M. Burnham and Christian Conte Schmidt
REM and NREM Sleep Mentation Suprachiasmatic Nucleus and Autonomic Nervous
Patrick Mcnamara, Patricia Johnson, Deirdre System Influences on Awakening From Sleep
McLaren, Erica Harris,Catherine Beauharnais and Andries Kalsbeek, Chun-xia Yi, Susanne E. la
Sanford Auerbach Fleur, Ruud m. Buijs, and Eric Fliers
Contents of Recent Volumes 361
Volume 97 Volume 98
Behavioral Pharmacology of Orofacial Movement
An Introduction to Dyskinesia—the Clinical
Disorders
Spectrum
Noriaki Koshikawa, Satoshi Fujita and Kazunori
Ainhi Ha and Joseph Jankovic
Adachi
L-dopa-induced Dyskinesia—Clinical Presenta-
Regulation of Orofacial Movement: Dopamine
tion, Genetics, And Treatment
Receptor Mechanisms and Mutant Models
L.K. Prashanth, Susan Fox and Wassilios
John L. Waddington, Gerard J. O’Sullivan and
G. Meissner
Katsunori Tomiyama
Experimental Models of L-DOPA-induced
Regulation of Orofacial Movement: Amino Acid
Dyskinesia
Mechanisms and Mutant Models
Tom H. Johnston and Emma L. Lane
Katsunori Tomiyama, Colm M.P. O’Tuathaigh,
and John L. Waddington Molecular Mechanisms of L-DOPA-induced
Dyskinesia
The Trigeminal Circuits Responsible for
Gilberto Fisone and Erwan Bezard
Chewing
Karl-Gunnar Westberg and Arlette Kolta New Approaches to Therapy
Jonathan Brotchie and Peter Jenner
Ultrastructural Basis for Craniofacial Sensory
Processing in the Brainstem Surgical Approach to L-DOPA-induced
Yong Chul Bae and Atsushi Yoshida Dyskinesias
Tejas Sankar and Andres M. Lozano
Mechanisms of Nociceptive Transduction and
Transmission: A Machinery for Pain Sensation Clinical and Experimental Experiences of
and Tools for Selective Analgesia Graft-induced Dyskinesia
Alexander M. Binshtok Emma L. Lane
Contents of Recent Volumes 363
Multimodal Drugs and their Future for Abnormalities in Metabolism and Hypothalamic–
Alzheimer’s and Parkinson’s Disease Pituitary–Adrenal Axis Function in Schizophrenia
Cornelis J. Van der Schyf and Werner J. Geldenhuys Paul C. Guest, Daniel Martins-de-Souza,
Natacha Vanattou-Saifoudine, Laura W. Harris
Neuroprotective Profile of the Multitarget Drug
and Sabine Bahn
Rasagiline in Parkinson’s Disease
Orly Weinreb, Tamar Amit, Peter Riederer, Immune and Neuroimmune Alterations in Mood
Moussa B.H. Youdim and Silvia A. Mandel Disorders and Schizophrenia
Roosmarijn C. Drexhage, Karin Weigelt, Nico van
Rasagiline in Parkinson’s Disease
Beveren, Dan Cohen, Marjan A. Versnel, Willem
L.M. Chahine and M.B. Stern
A. Nolen and Hemmo A. Drexhage
Selective Inhibitors of Monoamine Oxidase Type
Behavioral and Molecular Biomarkers in Transla-
B and the “Cheese Effect”
tional Animal Models for Neuropsychiatric
John P.M. Finberg and Ken Gillman
Disorders
A Novel Anti-Alzheimer’s Disease Drug, Ladostigil: Zoltán Sarnyai, Murtada Alsaif, Sabine Bahn,
Neuroprotective, Multimodal Brain-Selective Agnes Ernst, Paul C. Guest, Eva Hradetzky,
Monoamine Oxidase and Cholinesterase Inhibitor Wolfgang Kluge, Viktoria Stelzhammer and
Orly Weinreb, Tamar Amit, Orit Bar-Am and Hendrik Wesseling
Moussa B.H. Youdim
Stem Cell Models for Biomarker Discovery in
Novel MAO-B Inhibitors: Potential Therapeutic Brain Disease
Use of the Selective MAO-B Inhibitor PF9601N Alan Mackay-Sim, George Mellick and Stephen
in Parkinson’s Disease Wood
Mercedes Unzeta and Elisenda Sanz
The Application of Multiplexed Assay Systems for
INDEX Molecular Diagnostics
Emanuel Schwarz, Nico J.M. VanBeveren,
Paul C. Guest, Rauf Izmailov and
Volume 101 Sabine Bahn
General Overview: Biomarkers in Neuroscience Algorithm Development for Diagnostic Bio-
Research marker Assays
Michaela D. Filiou and Christoph W. Turck Rauf Izmailov, Paul C. Guest, Sabine Bahn and
Emanuel Schwarz
Imaging Brain Microglial Activation Using
Positron Emission Tomography and Translocator Challenges of Introducing New Biomarker Prod-
Protein-Specific Radioligands ucts for Neuropsychiatric Disorders into the
David R.J. Owen and Paul M. Matthews Market
The Utility of Gene Expression in Blood Cells for Sabine Bahn, Richard Noll, Anthony Barnes,
Diagnosing Neuropsychiatric Disorders Emanuel Schwarz and Paul C. Guest
Christopher H. Woelk, Akul Singhania, Josué Toward Personalized Medicine in the Neuropsy-
Pérez-Santiago, Stephen J. Glatt and Ming chiatric Field
T. Tsuang Erik H.F. Wong, Jayne C. Fox, Mandy
Proteomic Technologies for Biomarker Studies in Y.M. Ng and Chi-Ming Lee
Psychiatry: Advances and Needs Clinical Utility of Serum Biomarkers for Major
Daniel Martins-de-Souza, Paul C. Guest, Psychiatric Disorders
Natacha Vanattou-Saifoudine, Laura W. Harris Nico J.M. van Beveren and Witte
and Sabine Bahn J.G. Hoogendijk
Converging Evidence of Blood-Based Biomarkers
The Future: Biomarkers, Biosensors, Neu-
for Schizophrenia: An update
roinformatics, and E-Neuropsychiatry
Man K. Chan, Paul C. Guest, Yishai Levin,
Christopher R. Lowe
Yagnesh Umrania, Emanuel Schwarz, Sabine Bahn
and Hassan Rahmoune SUBJECT INDEX
Contents of Recent Volumes 365
Neurophysiology of Deep Brain Stimulation Bone Marrow Mesenchymal Stem Cell Trans-
Manuela Rosa, Gaia Giannicola, Sara Marceglia, plantation for Improving Nerve Regeneration
Manuela Fumagalli, Sergio Barbieri, and Alberto Priori Júlia Teixeira Oliveira, Klauss Mostacada, Silmara
de Lima, and Ana Maria Blanco Martinez
Neurophysiology of Cortical Stimulation
Jean-Pascal Lefaucheur Perspectives of Employing Mesenchymal Stem
Cells from the Wharton’s Jelly of the Umbilical
Neural Mechanisms of Spinal Cord Stimulation
Cord for Peripheral Nerve Repair
Robert D. Foreman and Bengt Linderoth
Jorge Ribeiro, Andrea Gartner, Tiago Pereira,
Magnetoencephalography and Neuromodulation Raquel Gomes, Maria Ascensão Lopes,
Alfons Schnitzler and Jan Hirschmann Carolina Gonçalves, Artur Varejão, Ana Lúcia
Luı´s, and Ana Colette Maurı´cio
Current Challenges to the Clinical Translation of
Brain Machine Interface Technology Adipose-Derived Stem Cells and Nerve Regener-
Charles W. Lu, Parag G. Patil, and Cynthia A. ation: Promises and Pitfalls
Chestek Alessandro Faroni, Giorgio Terenghi, and
Adam J. Reid
Nanotechnology in Neuromodulation
Russell J. Andrews The Pros and Cons of Growth Factors and Cyto-
kines in Peripheral Axon Regeneration
Optogenetic Neuromodulation
Lars Klimaschewski, Barbara Hausott, and Doychin
Paul S. A. Kalanithi and Jaimie M. Henderson
N. Angelov
Diffusion Tensor Imaging and Neuromodulation:
Role of Inflammation and Cytokines in Peripheral
DTI as Key Technology for Deep Brain
Nerve Regeneration
Stimulation
P. Dubový, R. Jancˇálek, and T. Kubek
Volker Arnd Coenen, Thomas E. Schlaepfer, Niels
Allert, and Burkhard Mädler Ghrelin: A Novel Neuromuscular Recovery Pro-
moting Factor?
DBS and Electrical Neuro-Network Modulation
Raimondo Stefania, Ronchi Giulia, Geuna Stefano,
to Treat Neurological Disorders
Pascal Davide, Reano Simone, Filigheddu Nicoletta,
Amanda Thompson, Takashi Morishita, and
and Graziani Andrea
Michael S. Okun
Neuregulin 1 Role in Schwann Cell Regulation
Neuromodulation in Psychiatric Disorders
and Potential Applications to Promote Peripheral
Yasin Temel, Sarah A. Hescham, Ali Jahanshahi,
Nerve Regeneration
Marcus L. F. Janssen, Sonny K. H. Tan, Jacobus
Giovanna Gambarotta, Federica Fregnan, Sara
J. van Overbeeke, Linda Ackermans, Mayke
Gnavi, and Isabelle Perroteau
Oosterloo, Annelien Duits, Albert F. G. Leentjens,
and LeeWei Lim Extracellular Matrix Components in Peripheral
Nerve Regeneration
Ethical Aspects of Neuromodulation
Francisco Gonzalez-Perez, Esther Udina, and
Christiane Woopen
Xavier Navarro
SUBJECT INDEX
SUBJECT INDEX
Volume 108
Tissue Engineering and Regenerative Medicine:
Volume 109
Past, Present, and Future The Use of Chitosan-Based Scaffold to Enhance
António J. Salgado, Joaquim M. Oliveira, Albino Regeneration in the Nervous System
Martins, Fábio G. Teixeira, Nuno A. Silva, Sara Gnavi, Christina Barwig, Thomas Freier,
Nuno M. Neves, Nuno Sousa, and Rui L. Reis Kirsten Haarstert-Talini, Claudia Grothe, and
Stefano Geuna
Tissue Engineering and Peripheral Nerve Recon-
struction: An Overview Interfaces with the Peripheral Nerve for the Con-
Stefano Geuna, S. Gnavi, I. Perroteau, trol of Neuroprostheses
Pierluigi Tos, and B. Battiston Jaume del Valle and Xavier Navarro
368 Contents of Recent Volumes
The Use of Shock Waves in Peripheral Nerve The Neuropathology of Neurodegeneration with
Regeneration: New Perspectives? Brain Iron Accumulation
Thomas Hausner and Antal Nógrádi Michael C. Kruer
Phototherapy and Nerve Injury: Focus on Muscle Imaging of Iron
Response Petr Dusek, Monika Dezortova, and Jens Wuerfel
Shimon Rochkind, Stefano Geuna, and Asher
The Role of Iron Imaging in Huntington’s Disease
Shainberg
S.J.A. van den Bogaard, E.M. Dumas, and
Electrical Stimulation for Promoting Peripheral R.A.C. Roos
Nerve Regeneration
Lysosomal Storage Disorders and Iron
Kirsten Haastert-Talini and Claudia Grothe
Jose Miguel Bras
Role of Physical Exercise for Improving Post-
Manganese and the Brain
traumatic Nerve Regeneration
Karin Tuschl, Philippa B. Mills, and Peter T. Clayton
Paulo A.S. Armada-da-Silva, Cátia Pereira,
SandraAmado, and António P. Veloso Update on Wilson Disease
Aggarwal Annu and Bhatt Mohit
The Role of Timing in Nerve Reconstruction
Lars B. Dahlin An Update on Primary Familial Brain Calcification
R.R. Lemos, J.B.M.M. Ferreira, M.P. Keasey,
Future Perspectives in Nerve Repair and
and J.R.M. Oliveira
Regeneration
Pierluigi Tos, Giulia Ronchi, Stefano Geuna, and INDEX
Bruno Battiston
INDEX
Volume 111
Volume 110 History of Acupuncture Research
Yi Zhuang, Jing-jing Xing, Juan Li, Bai-Yun Zeng,
The Relevance of Metals in the Pathophysiology of and Fan-rong Liang
Neurodegeneration, Pathological Considerations
Effects of Acupuncture Needling with Specific
Kurt A. Jellinger
Sensation on Cerebral Hemodynamics and
Pantothenate Kinase-Associated Neurodegener- Autonomic Nervous Activity in Humans
ation (PKAN) and PLA2G6-Associated Neuro- Kouich Takamoto, Susumu Urakawa, Kazushige
degeneration (PLAN): Review of Two Major Sakai, Taketoshi Ono, and Hisao Nishijo
Neurodegeneration with Brain Iron Accumula-
Acupuncture Point Specificity
tion (NBIA) Phenotypes
Jing-jing Xing, Bai-Yun Zeng, Juan Li, Yi Zhuang,
Manju A. Kurian and Susan J. Hayflick
and Fan-rong Liang
Mitochondrial Membrane Protein-Associated
Acupuncture Stimulation Induces Neurogenesis
Neurodegeneration (MPAN)
in Adult Brain
Monika Hartig, Holger Prokisch, Thomas Meitinger,
Min-Ho Nam, Kwang Seok Ahn, and Seung-Hoon
and Thomas Klopstock
Choi
BPAN: The Only X-Linked Dominant NBIA
Acupuncture and Neurotrophin Modulation
Disorder
Marzia Soligo, Stefania Lucia Nori, Virginia Protto,
T.B. Haack, P. Hogarth, A. Gregory, P. Prokisch,
Fulvio Florenzano, and Luigi Manni
and S.J. Hayflick
Acupuncture Stimulation and Neuroendocrine
Neuroferritinopathy
Regulation
M.J. Keogh, C.M. Morris, and P.F. Chinnery
Jung-Sheng Yu, Bai-Yun Zeng, and
Aceruloplasminemia: An Update Ching-Liang Hsieh
Satoshi Kono
Current Development of Acupuncture Research
Therapeutic Advances in Neurodegeneration with in Parkinson’s Disease
Brain Iron Accumulation Bai-Yun Zeng, Sarah Salvage, and
Giovanna Zorzi and Nardo Nardocci Peter Jenner
Contents of Recent Volumes 369