You are on page 1of 16

MENTAL RETARDATION AND DEVELOPMENTAL DISABILITIES

RESEARCH REVIEWS 10: 303–317 (2004)

TOWARD A DEVELOPMENTAL NEUROBIOLOGY


OF AUTISM

Franck Polleux1* and Jean M. Lauder2


1
Department of Pharmacology–Neuroscience Center, School of Medicine, University of North Carolina–Chapel Hill,
Chapel Hill, North Carolina
2
Department of Cell and Developmental Biology, School of Medicine, University of North Carolina–Chapel Hill,
Chapel Hill, North Carolina

Autism is a complex, behaviorally defined, developmental brain dis- The etiology of autism is poorly understood [Piven,
order with an estimated prevalence of 1 in 1,000. It is now clear that autism 1997]. However, the amount and quality of research performed
is not a disease, but a syndrome with a strong genetic component. The
etiology of autism is poorly defined both at the cellular and the molecular in several fields, including epidemiology, early brain imaging,
levels. Based on the fact that seizure activity is frequently associated with and genetic identification of candidate chromosomal regions,
autism and that abnormal evoked potentials have been observed in autistic has led to new hypotheses regarding possible etiologies [Stoks-
individuals in response to tasks that require attention, several investigators tad, 2001; Rubenstein and Merzenich, 2003; Belmonte et al.,
have recently proposed that autism might be caused by an imbalance be-
tween excitation and inhibition in key neural systems including the cortex.
2004]. Interestingly, upon more detailed clinical examination,
Despite considerable ongoing effort toward the identification of chromo- approximately 10% of autistic cases reveal association with other
some regions affected in autism and the characterization of many potential genetic neuropathologies, such as fragile X; tuberous sclerosis,
gene candidates, only a few genes have been reproducibly shown to display and Rett syndrome. Several other clinical features are frequently
specific mutations that segregate with autism, likely because of the complex associated with autism, such as mental retardation [roughly 70%
polygenic nature of this syndrome. Among those, several candidate genes
have been shown to control the early patterning and/or the late synaptic
of cases have an intelligence quotient (IQ) ⬍ 70], epileptic
maturation of specific neuronal subpopulations controlling the balance seizures (30%), and macrocephaly (larger head circumference)
between excitation and inhibition in the developing cortex and cerebellum. and megencephaly (larger brain volume) [Tuchman, 2003]. Ap-
In the present article, we review our current understanding of the develop- proximately one-third of autistic individuals develop clinically
mental mechanisms patterning the balance between excitation and inhibi- apparent seizures and, of those, more than 50% develop “sharp-
tion in the context of the neurobiology of autism. © 2004 Wiley-Liss, Inc.

MRDD Research Reviews 2004;10:303–317. spike” activity during sleep when recorded by EEG or magne-
toencephalography [Lewine et al., 1999; Ballaban-Gil and
Tuchman, 2000]. Taken together, these observations have re-
Key Words: autism; development; cortex; mouse genetics; serotonin; do- cently led several investigators to hypothesize that the cortex of
pamine; norepinephrine; glutamate; neurotransmitters; GABA; interneu- autistic individuals is characterized by an imbalance between
rons; patterning; gene expression; neuroligin; neuronal migration; dendrite; excitation and inhibition, leading to hyperexcitability and an
synaptogenesis
unstable activity of cortical networks following normal sensory
stimulation [Hussman, 2001; Rubenstein and Merzenich, 2003;
Belmonte et al., 2004].
Most interestingly, recent advances in functional imaging

A
utism is a developmental brain disorder characterized by in humans, but also in nonhuman primates, have revealed that
a general inability to form reciprocal social interactions, rhythmic synchronization of neural discharges in the gamma
severe impairment in verbal and nonverbal communica- frequency band (20 – 60 Hz) may provide the necessary spatial
tion, and a markedly restricted repertoire of activities and inter- and temporal links that bind together the processing in different
ests. The incidence of autism is currently estimated at 1 in 1,000 brain areas to build a coherent percept [Tallon-Baudry and
children [Folstein and Rosen-Sheidley, 2001]. Familial recur-
rence of the disorder is 100-fold higher than in the general
population, and the concordance rate among monozygotic Grant sponsor: NIMH; Grant number: U54 MH66418 (to JML). Grant sponsor:
NIEHS; Grant number: P30ES10126 (to JML). Grant sponsor: NIH/NIDCR; Grant
twins is estimated at between 70 and 90%, but close to 0% in number R01 DE13314 – 05 (to JML). Grant sponsor: NIH-NINDS; Grant number: 1
dizygotic twins, thus indicating a strong genetic component to RO1 NS047701– 01. (to FP). Grant sponsor: March of Dimes Birth Defects Founda-
tion (to FP, JML).
the disease [reviewed in Veenstra-Vanderweele et al., 2003; *Correspondence to: F. Polleux. E-mail: polleux@med.unc.edu or Jean Lauder
Veenstra-VanderWeele and Cook, 2004; see also Wassink et al., E-mail: unclau@med.unc.edu
this issue]. Furthermore, the prevalence of autism and Asperger Received 17 November 2004; Accepted 22 November 2004
Published online in Wiley InterScience (www.interscience.wiley.com).
syndrome are, respectively, 4 and 8 times higher in males than DOI: 10.1002/mrdd.20044
females, strongly suggesting an X-linked genetic component.
© 2004 Wiley-Liss, Inc.
Bertrand, 1999]. These results suggest tractive to describe the pathophysiology of Hippocampus and Amygdala
that one particular type of gamma oscil- the autistic brain in light of recent progress An isolated report has revealed a
lation resulting from synchronized neu- made in understanding the generation, mi- decreased level of dendritic branching in
ronal activity at the cortical and subcor- gration, and differentiation of glutamatergic the CA1 and CA4 regions of the hip-
tical levels not only plays an essential role and GABAergic neurons in the cortex. We pocampus of two autistic patients com-
in sensory perception, but also in atten- will also discuss the development of neu- pared to two control cases [Raymond et
tion-based cognitive tasks. Importantly, romodulatory systems well known to con- al., 1996]. Other studies have reported an
neuronal synchrony in hippocampal, trol the global levels of neuronal excitability increased neuronal packing density in the
cortical, and thalamic networks has been in the forebrain, including the serotonin hippocampus, subiculum, mammilary
shown to be critically dependent on the inputs that have been suspected for a long bodies, entorhinal cortex, medial septal
integrity of the discharge of interneurons time to be altered in the autistic brain. nuclei, and several nuclei of the amygdala
in relation to the activity of the pyramidal Finally, we review the classes of genes that [reviewed by Courchesne, 1997]; the
neuron. In the hippocampus, synchro- have been linked to autism in recent ge- neuronal networks playing a critical role
nous gamma oscillation is considered to netic studies and discuss several candidate in the formation, maintenance, and re-
occur through the GABAA receptor–me- genes in the context of this neurodevelop- trieval of memory. However, other stud-
diated mutual inhibition among inter- mental hypothesis. ies have failed to reproduce the observed
neurons [Buhl et al., 1994; Cobb et al., differences in the packing density of
1995; Freund and Buzsaki, 1996]. In the CA1–CA4 pyramidal neurons [Bailey et
neocortex; it is known that a class of NEUROANATOMICAL al., 1998].
pyramidal neurons termed fast rhythmic ABNORMALITIES IN AUTISM Interestingly, a recent analysis of
bursting cells or “chattering” neurons A few studies using either neuro- brain morphometric features was per-
synchronize their activities at gamma fre- anatomical examination of autopsied formed in a large sample of carefully di-
quencies and are thought to be necessary brains of autistic patients or, more re- agnosed 3- to 4-year-old children with
for selective attention or binding process- cently, functional and structural MRI autism spectrum disorder (ASD) com-
ing in object recognition [Aoyagi et al., studies, have revealed three main types of pared with age-matched control groups
2003]. Therefore, a slight disruption of defects in (1) the brainstem and cerebel- of typically developing (TD) children
the balance between excitation and inhi- lum, (2) the limbic system (amygdala and and developmentally delayed (DD) chil-
bition in the cortex could have dramatic hippocampus), and (3) the cortex dren [Sparks et al., 2002]. This analysis
consequences on the function of the [Courchesne, 1997]. revealed increased cortical and cerebellar
neuronal networks underlying percep- volume and also an increased volume of
tion and attention. the amygdala in autistic children that
Brain Stem and Cerebellum
In a very recent study, the brain cannot be accounted for by a simple in-
An early anatomical study first re-
activation of a group of high-functioning creased cortical volume. This and an-
vealed that several brainstem nuclei were
autistic individuals was measured using other recent analysis [Schumann et al.,
either missing or displayed strong neuro-
functional MRI during sentence com- 2004] suggest abnormal brain develop-
nal loss, such as the facial motor nuclei or
prehension and the results were com- mental processes affecting the amydgala
the superior olive nuclei [Rodier et al.,
pared with those of a verbal IQ–matched early in the clinical course of autism.
control group [Just et al., 2004]. Their 1996]. A longitudinal study performed
results show that the functional connec- on a large set of cases using quantitative Brain Enlargement in Autism
tivity; i.e., the degree of synchroniza- structural imaging also revealed hypopla- Although the defining behavioral
tion/correlation of brain activation was sia of brainstem structures [Hashimoto et features of autism are present from the
consistently lower for the autistic indi- al., 1995]; however, this set of data prob- earliest ages and change over time with
vidual than for the control participants in ably needs to be replicated to evaluate its age, perhaps the most compelling biolog-
the main cortical areas involved in lan- validity in light of the progress made in ical argument for autism being a disorder
guage processing [Just et al., 2004]. These diagnosis of autism. of abnormal brain development comes
findings suggest that the neural basis of One of the most reproducible neu- from studies showing increased brain
disordered language in autism entails a roanatomical findings in autistic brain is volume in this disorder. Kanner in his
lower degree of information integration the paucity of Purkinje neurons in the first descriptions of autism [Kanner,
and synchronization across the large-scale cerebellum [from 35 to 90%; reviewed 1943] noted that 5 of 11 children he
cortical network for language processing by Courchesne, 1997]. An early MRI described had no obvious dysmorphic
[Brock et al., 2002; Just et al., 2004]. study reported a significantly smaller area features, but did show enlarged head size.
Taken together, these results suggest of the cerebellar vermis in lobules Over the years others described this an-
that the autistic brain might be character- VIII–X [Courchesne et al., 1988]. Re- ecdotally in studies of dysmorphology
ized by a synchronization deficit during the cent MRI studies have also revealed a near [Steg and Rapoport, 1975]. In 1992,
activation of cortical networks involved in 40% increase in cerebellar white matter Piven et al. reported enlarged midsagittal
language processing (and maybe in other volume in autistic children compared to brain area [Piven et al., 1992]. A system-
sensory modalities or attention) and that age-matched controls [Courchesne et al., atic study of 35 autistic adults and ado-
this synchronization deficit could be the 2001]. Subsequent independent reports, lescents versus 36 controls, to follow up
result of an imbalance between excitation however, have failed to replicate the orig- on this initial report, similarly reported
and inhibition [Rubenstein and Mer- inal observation of hypoplasia of the neo- enlarged brain volume and brain tissue
zenich, 2003; Belmonte et al., 2004]. Al- cerebellar vermis [Piven et al., 1997] and volume [Piven et al., 1995]. More re-
though still speculative, this hypothesis is additional studies of the volume of cerebel- cently Courchesne et al. [2001] exam-
attractive because it is based on functional lar cortical volume will be necessary before ined head circumference in a sample of
studies. The present review will discuss final conclusions can be drawn about the 60 boys compared to a nonmentally re-
why this new hypothesis is especially at- size of this structure in autism. tarded group of controls and reported
304 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
enlargement of cortical gray and white pathogenesis. Importantly, this early dif- drites reaching layer 4B (Fig. 1A). The
matter limited to those in the 2- to ference in brain size is not found in adult second important component of mini-
4-year-old age group [Courchesne et al., autistic patients compared to age- columns is GABAergic interneurons pro-
2001], with an actual decrease in cortical matched control brains [Courchesne et viding powerful and specific inhibitory
white matter in the older 6- to 16-year- al., 2003], suggesting that the initially control in cortical neuronal networks
old group. Increased white matter in the accelerated rate of cortical growth is fol- (Fig. 1B). Cortical interneurons can be
cerebellum was also noted in this lowed by an abnormally slower period of classified based on morphological and
younger age group (2– 4 years) with de- growth [see Piven et al., this issue]. As we functional criteria into four main classes,
creased gray matter in the older age will discuss later, this abnormally rapid each characterized by a precise synaptic
group. Findings by Sparks et al.[2002] growth rate detected in cortical grey and targeting of a specific part of the pyrami-
support the finding of increased brain white matter during the first years of life dal neuron: (1) large and small basket
volume in 45 3- to 4-year olds with in the frontal, temporal, and parietal cells, respectively, making their synaptic
either autism or pervasive developmental lobes could reflect (1) an increase in den- contacts onto the cell soma and proximal
disorder–not otherwise specified (PDD- dritic branching/outgrowth and a corre- part of the apical dendrite of pyramidal
NOS) compared to both typically devel- spondingly increased rate of synaptogen- neurons within the same layer; (2) double
oping and mentally retarded controls, esis (grey matter) accompanied by an bouquet cells making their synaptic con-
noting increased volume of both the ce- accelerated rate of axon myelination tacts onto more distal parts of the basal
rebral cortex and cerebellum as well as (white matter) and/or (2) a reduced dendrites of pyramidal neurons and (3)
increased volume of the amygdalae elimination of aberrant connections (re- Chandelier cells making synapses along
[Sparks et al., 2002]. Finally; increased duced dendritic/synaptic pruning). Re- the initial axonal segment of pyramidal
brain volume was found in a large sample cent work has demonstrated that, during neurons; and (4) Cajal-Retzius interneu-
of high-functioning (nonmentally re- development, there is a causal relation- rons found in layer I making synapses
tarded) autistic individuals under 12 years ship between the number of active pre- onto the distal part of apical dendrites of
of age; but not in those over 12 years of synaptic afferent inputs and the size and cortical pyramidal neurons [reviewed in
age [Aylward et al., 2002]. Others have level of branching of the corresponding DeFelipe et al., 1990]. Based on this spe-
reported evidence to suggest enlargement dendritic field harboring postsynaptic cialized synaptic targeting, each of these
of the caudate nucleus [Sears et al., 1999], densities [reviewed in Cline, 2001; see interneuronal subpopulations is thought
amygdalae [Schumann et al., 2004], and below]. Therefore, one attractive hy- to play a specific functional role in the
hippocampus [Schumann et al., 2004], pothesis for the pathogenesis of autism, control of synaptic integration, and
with a decrease in cross-sectional area of and potentially other developmental dis- therefore, on the propagation of infor-
the corpus callosum [Piven et al., 1997; abilities, could be a disregulation of the mation through cortical networks
Manes et al., 1999; Hardan et al., 2000]. developmental mechanisms patterning [Freund, 2003]. Disorganization of mini-
The findings from brain MRI studies are axonal outgrowth and/or dendritic ar- columns could reflect distinct aspects of
also consistent with systematic large scale borizations/synaptic contacts between an abnormal cortical cytoarchitecture,
surveys showing that approximately 20% excitatory and inhibitory neurons in the including: (1) a disrupted laminar distri-
of autistic individuals have macrocephaly cortex [Zoghbi, 2003]. The behavioral bution of a specific subclass of interneu-
(greater than the 98th percentile for head consequences of an abnormal balance be- rons and/or (2) a numerical imbalance
circumference) that appears not to be tween excitatory and inhibitory neuro- between pyramidal neurons and inter-
present at birth [Lainhart et al., 1997; transmission in the cortex are only start- neurons and/or (3) a disrupted level of
Stevenson, 1997]. More recent prelimi- ing to be explored experimentally branching of the axons and dendrites of
nary evidence suggests that the increase [Powell et al., 2003] and are likely to be both pyramidal neurons and interneu-
in head circumference may have its ori- specific and complex, especially if a sub- rons. Future investigations are required
gins in the latter part of the first year of population of cortical interneurons is to characterize the number, laminar dis-
postnatal life [Courchesne et al., 2001]. specifically reduced or absent as it as been tribution, and morphology of specific
In summary these reports provide proposed for schizophrenia [Lewis and classes of interneurons in the autistic cor-
firm support for the idea that the brain in Levitt, 2002]. tex to assess whether this disorder could
autism is enlarged. There is evidence that A recent study by Casanova et al. be due to the abnormal patterning of a
the enlargement occurs in both gray and [2002] has provided evidence for abnor- subpopulation of GABAergic neurons.
white matter and occurs in selected re- mal patterning of minicolumns in the Interestingly, an early study by
gions and structures in the brain (with a frontal and temporal lobes of autistic pa- Bailey et al. [1998] carefully assessing the
decrease in the size of some other struc- tients. Interestingly, in humans, cortical clinicopathology of six carefully diag-
tures). There is also evidence to suggest minicolumns are the cellular manifesta- nosed autistic individuals revealed several
that this enlargement occurs predomi- tion of the basic columnar organization abnormalities that suggest abnormal mi-
nantly during early postnatal brain devel- of excitatory and inhibitory neurons into gration of pyramidal neurons and/or pat-
opment. More specific conclusions about canonical functional units (Fig. 1; [DeFe- terning of their dendrite outgrowth. In
the nature of this phenomenon, includ- lipe et al., 1990; Peters, 1994; Mount- four of six cases, postmortem examina-
ing the exact timing or window of brain castle, 1997; Jones, 2000]). Functional tion of the frontal cortex revealed (1) the
enlargment, patterns over time in brain and anatomical investigations have en- presence of ectopic neurons in the white
structures, regions, and tissues, and clin- abled an accurate definition of the cellu- matter and in layer 1 as well as (2) mi-
ical correlates, are not yet known. lar composition of these minicolumns. soriented pyramidal neurons in layer 5
Each column contains an array of pyra- with their apical dendrite not oriented
Cortical Columnar Architecture midal neurons in layers 5 and 2/3, char- toward layer 1 as in normal cortex and (3)
The finding of brain enlargement acterized by bundled apical dendrites a disorganized cellular organization in su-
occurring in the early postnatal period reaching layer 1 and pyramidal neurons pragranular layers of areas located in the
suggests a range of possibilities about its in layer 6 with their bundled apical den- superior temporal gyrus [Bailey et al.,
MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 305
1998]. To our knowledge, this finding
has never been reexamined or replicated
but suggests the interesting possibility of a
neuronal migration defect and/or den-
drite patterning defect in the cortex of
some autistic patients. This finding could
also explain partially the reduced regular-
ity of minicolumn spacing because, as
explained above (Fig. 1), the spacing be-
tween the cell-body column is formed by
tight fascicles of apical dendrites ascend-
ing to layer 1. Therefore, a defect in the
orientation of apical dendrites might re-
sult in a disorganized columnar organiza-
tion. Future investigations should be di-
rected at testing how reproducible these
findings are.

MULTIPLE
NEUROTRANSMITTER
SYSTEMS CONTROLLING
EXCITATION/INHIBITION
RATIO MAY BE ABNORMAL IN
AUTISM
Neurochemical features of autism
include evidence of disrupted development
of multiple neurotransmitter systems from
functional brain imaging, neurochemistry,
and clinical pharmacological studies [re-
viewed by Trottier et al., 1999; Korvatska
et al., 2002]. So far, the strongest evidence
implicates the glutamatergic and GABAer-
gic and serotonergic systems, although
weaker evidence also exists for changes in
catecholaminergic, peptidergic, and cho-
linergic systems.

Glutamate
Contrasting hypotheses have been
proposed to underlie autism; one current
hypothesis is that autism is a hypogluta-
matergic disorder [Purcell et al., 2001;
Fig. 1. Origin and functional circuitry of glutamatergic and GABAergic in the mammalian cortex. Jamain et al., 2002; Serajee et al., 2003],
(A) In mammals the precursors that are competent to give rise to pyramidal glutamatergic neurons whereas the other suggested a decreased
in the cortex are located in the dorsal telencephalon (red cell in 1). Upon cell-cycle exit, pyramidal
neurons migrate radially to reach their final layer where they will terminally differentiate. By contrast,
level of inhibition, i.e., [Rubenstein and
the majority of GABAergic cortical interneurons are generated in the ventral part of the telenceph- Merzenich, 2003; Belmonte et al., 2004].
alon, the medial ganglionic eminence (MGE), and migrate tangentially (green cells in 2) to reach the It is difficult to reconcile these two op-
cortex and establish synaptic contacts with glutamatergic pyramidal neurons. (B) Schematic view of posite models, but both have strengths
a cortical minicolumn, the canonical microcircuitry thought to underlie the function of cortical and weaknesses with regard to the neu-
networks in primates. Pyramidal neurons in layers VIA, V have their apical dendrites organized in
bundles directed toward layer I, making the center of the column relatively cell sparse as seen on robiology of autism reflecting the need
Nissl-stained sections of human and nonhuman primates [see Casanova et al., 2002]. The roman for more accurate and direct investiga-
numerals indicate cortical layers. Thalamocortical afferents are depicted in yellow. Each cortical layer tion of the cortical cytoarchitecture in
is composed of a precise number of different subtypes of interneurons (orange and green cells). This autism using cellular and molecular tech-
numerical balance between different types of interneurons and pyramidal neurons in each layer is
species- and area-specific. (C) Simplified view of the different subtypes of cortical interneurons
niques. As mentioned previously, the hy-
illustrating the remarkably precise connectivity they establish with pyramidal neurons. Chandelier pothesis of a decreased GABAergic inhi-
cells (orange) express the calcium-binding protein (CBP) Parvalbumin (PV) and their axon precisely bition in specific cortical regions is
targets the axon hillock of pyramidal neurons (1). Large basket cells also express PV but their axon attractive to explain the hyperexcitability
targets the perisomatic region of the cell body of pyramidal neurons (2). Double bouquet cells of cortical tissue frequently associated
coexpress another set of CBPs called calbindin 28kD (CB) and Calretinin (CR) and their axons target
the distal portion of the basal dendrites of pyramidal neurons (3). Another class of Basket cells with autism. Importantly, these two hy-
display small to medium cell body sizes and express PV, CB, and CR. Interestingly, unlike large basket pothesis are not totally incompatible:
cells, their axons target the spines along the apical dendrite of pyramidal neurons (4). Finally, specific cortical regions could be charac-
Cajal-Retzius cells are expressing a large secreted protein called reelin and their axons target the terized by an increased excitatory/inhib-
apical branches of pyramidal neurons in layer I. Interestingly, Cajal-Retzius neurons are the primary
synaptic target of afferent serotonin (5HT) axons from the raphe nucleus, which plays a critical role
itory ratio whereas some other regions
in the modulation of cortical function (see text for details). B and C were redrawn and modified from could show the reverse trend. The be-
Peters and Sethares [1996]; DeFelipe and Fajinas [1992]; and Jones [2000]. havioral consequence of such a regional
306 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
imbalance will be important to assess us- and double knockout mice for Ngn1/2 vious section, much work needs to be
ing new genetic animal models [Levitt et show a striking phenotype, where the done to explore (1) whether specific
al., 2004]. vast majority of cortical neurons express GABAergic neuron subpopulations are
The reduction in global levels of GABA as a neurotransmitter instead of misplaced or absent in specific cortical
glutamate signaling might occur by over- glutamate [Fode et al., 2000; Schuurmans regions, (2) whether there is a global
activation of excitatory receptors on cor- et al., 2004]. numerical decrease of the number of
tical GABA interneurons, such as those Interestingly, in other parts of the GABAergic interneuron per unit of cor-
of the 5-HT2A receptor subtype, leading nervous system, for example, the spinal tical microcolumns, or (3) whether the
to pronounced depression of the excita- cord, expression of a specific neurotrans- number and localization of cortical inter-
tory glutamatergic circuitry [Carlsson, mitter is partially determined by a combi- neurons is normal in autism but charac-
1998]. This possibility is supported by the natorial expression of transcription factors terized by a functional deficit in GABA
relative efficacy of combined treatment [reviewed in Jessell, 2000]. However, a re- expression at the synaptic level as recently
with partial glutamate agonists and cent report demonstrates that the level of shown in another neurodevelopmental
5-HT2A receptor antagonists (e.g., ris- spontaneous activity and the frequency of pathology, schizophrenia [Lewis and
peridone) in autism as well as in hypo- intracellular Ca2⫹ elevations in developing Levitt, 2002].
glutamatergic animal models [Carlsson et neural circuitry can have dramatic long-
al., 1998, 1999; Nilsson et al., 2001]. term consequences on the number of neu- Developmental Origin of
Recent evidence also suggests involve- rons expressing a given type of neurotrans- GABAergic Cortical Interneurons
ment of polymorphisms in genes encod- mitter [Borodinsky et al., 2004]. This In rodents, the vast majority of
ing both metabotropic and ionotropic important finding raises the possibility that cortical interneurons are generated in ap-
glutamate receptors in autism [Jamain et transcription factors restrict the type of proximately the same time window as
al., 2002; Serajee et al., 2003]. Recently, neurotransmitter that a neuronal precursor cortical pyramidal neurons [E11 to E17
a postmortem study has reported elevated can express but that the level of spontane- in the mouse; E13 to E19 in the rat;
expression of the glutamate transporter in ous activity may play an important role in Miller, 1985, 1986]. However, cortical
autistic brain [Purcell et al., 2001]. fine tuning the final number of neurons interneurons are generated by a distinct
expressing a given neurotransmitter pheno- set of precursors located in the ventral
Developmental Origin of type [Spitzer et al., 2004]. Therefore, the telencephalon, more specifically in the
Glutamatergic Cortical Neurons balance between the number of neurons medial and caudal parts of the ganglionic
In mammals, the vast majority of expressing glutamate and GABA is proba- eminence [Fig. 1A; MGE and CGE, re-
glutamatergic pyramidal neurons are bly the results of complex interactions be- spectively; Anderson et al., 1997; Tama-
generated in the dorsal telencephalon by tween intrinsic (transcription factor expres- maki et al., 1997; Lavdas et al., 1999;
precursors in the ventricular and subven- sion) and extrinsic (spontaneous activity Wichterle et al., 2001; Nery et al., 2002;
tricular zones during embryonic devel- and calcium signaling) activity during de- Polleux et al., 2002]. At the genetic level,
opment. In rodents, these neurons are velopment. the generation of GABAergic cortical in-
generated during the second week of ges- terneurons is under the control of distinct
tation (E11 to E17 in mouse or E13 to GABA transcription factors: the bHLH tran-
E19 in rat). In nonhuman primates, cor- As discussed earlier, another neu- scription factor Mash1 and the homeodo-
tical neurons are generated from E40 and rochemical abnormality hypothesized to main-containing transcription factors
E100 approximately [Rakic, 1972]. In explain the pathophisiology of autism is Dlx1 and Dlx2 [Anderson et al., 1997;
humans, the generation of cortical neu- suppressed GABAergic inhibition [Hus- Casarosa et al., 1999]. Interestingly, in-
rons is thought to occur between the 6th sman, 2001; Rubenstein and Merzenich, terneurons generated in the ventral tel-
and the 13th week of gestation based on 2003; Belmonte et al., 2004], which may encephalon have to undergo a “long
histological appearance of VZ and SVZ be due, at least in part, to reduced ex- journey” to reach their dorsal destination
mitotic figures [reviewed in Sidman and pression of GAD65 and GAD67 [Fatemi in the cortex through a tangential mode
Rakic, 1973]. Upon their last mitosis in et al., 2002], the two isoforms of the of migration, which, by definition, does
the germinal pseudoepithelium lining the GABA biosynthetic enzyme glutamate not involve fasciculation along the radial
ventricles, glutamatergic neurons migrate decarboxylase. Reduced GABAergic glial scaffold until they reach their final
along the radial glial scaffold to accumu- neurotransmission may also be caused by destination [Marin and Rubenstein,
late in the cortical plate in an inside-first deletional mutations in chromosome 2001].
outside-last manner, so that neurons in 15q11-q13, the locus of genes encoding Recently, experiments performed
infragranular layers 6 and 5 are generated subunits of the GABAA receptor in human telencephalic tissue have
first, before neurons populating more su- (GABRB3, GABRA5, and GABRG3). shown that, unlike in rodents, a specific
perficial layers 4, 3, and 2 [Rakic, 1972]. In Angelman’s syndrome (AS), such de- subset of dorsal progenitors seems to be
Recent progress in the analysis of letions lead to reduced expression of competent to produce a subpopulation of
the genetic mechanisms underlying the GABAA/BZD receptors [Holopainen et cortical interneurons in a Mash1-depen-
generation of cortical neurons has re- al., 2001a, 2001b; Luddens et al., 1995]. dent manner [Letinic et al., 2002]. This
vealed that cortical glutamatergic neu- AS patients exhibit a behavioral pheno- suggests that, in humans, a subpopulation
rons are generated by dorsal precursors type consistent with compromised of cortical dividing precursors are directly
under the control of bHLH transcription GABAergic inhibitory neurotransmis- involved in the generation of more than
factors, Neurogenin1 and 2 (Ngn1, Ngn2) sion, including hyperactivity, hyperkine- 60% of cortical GABAergic interneurons.
and as well as Pax6 and Emx2 [reviewed sis, seizures, and sleep disturbances as well Although some differences might exist
in Schuurmans and Guillemot, 2002]. In- as severe mental retardation, lack of mo- between the mode of generation of cor-
terestingly, Ngn1 and Ngn2 are involved tor coordination, craniofacial abnormali- tical interneurons in rodents and pri-
in the specification of the glutamatergic ties, and autism [Holopainen et al., mates, the important concept emerging is
neurotransmitter phenotype and single 2001a, 2001b]. As discussed in the pre- that (1) specific sets of precursors along
MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 307
the ventro-dorsal axis of the telencepha- plasma levels reflect a change in central Continued interest in the seroto-
lon are specialized with regard to their brain levels remains to be explored. In- nergic system comes from the relative
competence to generate glutamatergic or terestingly, polymorphisms in the 5-HT efficacy of serotonergic drugs in the
GABAergic cortical neurons and (2) they transporter (5-HTT) promoter have symptomatic treatment of some cases of
undergo a drastically different migration been proposed as an underlying cause of autism [Volkmar, 2001]. Pharmacologi-
pathway to invade the cortex and to dif- autism [Cook et al., 1997; Klauck et al., cal treatment studies in autism are com-
ferentiate in the appropriate layer. 1997]. This is presently a controversial plicated by various factors, including a
Interestingly, several extracellular topic, as several studies have recently tremendous range of syndrome expres-
cues have been found to control the mo- provided conflicting evidence of a poten- sion, a lack of robust animal models of
tility of cortical interneurons migrating tial link between 5-HTT and autism the disorder, and various methodological
from the basal forebrain to the cortex. [Anderson et al., 1990; Maestrini et al., problems. Theories have tended to fol-
Among them are two types of ligands 1999; Persico et al., 2000; Yirmiya et al., low treatments, and various neurochem-
that activate receptor tyrosine kinases 2001; Betancur et al., 2002; Persico et al., ical systems have been the focus of study.
(RTK) expressed by migrating interneu- 2002; Coutinho et al., 2004]. Interest- Treatments developed are effective rela-
rons: hepatocyte growth factor (HGF), ingly, finding evidence for transmission tive to certain disabling symptoms, but
which activates the MET RTK, and of polymorphic alleles of the 5-HTT “core” problems (e.g., deficits in social
NT4, which binds TRK [Powell et al., may depend on the extent of social and relatedness and communication) appear
2001; Polleux et al., 2002]. Mice carrying communication deficits in the autism less responsive to medications. Among
a targeted mutation of the gene encoding proband, suggesting that the alleles may these pharmacotherapies, especially in
urokinase plasminogen activator (uPAR; modify severity of autistic traits rather children and adolescents, include selec-
a component of HGF activation) and than conveying risk for autism per se tive serotonin reuptake inhibitors (SS-
knockout mice for trkB both show a [Tordjman et al., 2001]. Further support RIs), 5-HT2A receptor antagonists, tricy-
reduced number of cortical interneurons for 5-HTT comes from functional MRI clic antidepressants, or mixed dopamine/
compared to controls at late embryonic/ studies showing that individuals with one 5-HT receptor antagonists [Carlsson,
early postnatal stages [Powell et al., 2001; or two copies of a short allele of the gene 1998; Carlsson et al., 1999; McDougle
Polleux et al., 2002]. Interestingly, had greater neuronal activity than con- and Posey, 2002; Veenstra-VanderWeele
uPAR knockout mice display a regional trols in the amygdala, a brain region im- et al., 2000]. Although the reasons un-
defect in which approximately 50% of plicated in autism [Hariri et al., 2002; derlying the efficacy of these drugs is
calbindin-positive cortical interneurons Hariri and Weinberger, 2003]. In addi- poorly understood, it is possible that such
are absent from frontal and parietal cor- tion, multiple single polymorphisms in treatments may counteract developmen-
tical areas [reviewed in Levitt et al., tryptophan 2,3 diooxygenase (TDO2), tal defects in serotonergic pathways
2004]. The uPAR knockout mice display the rate-limiting enzyme in L-tryptophan and/or abnormal dynamics of 5-HT syn-
behavioral deficits ranging from scattered catabolism, have been associated with au- thesis, catabolism, or transport.
and frequent seizures and, more interest- tism. Such mutations could increase lev- Dysregulation of the developing
ingly, increased anxiety-like behaviors els of 5-HT throughout the body and serotonergic system could occur by var-
[Powell et al., 2003]. The extent to brain [Nabi et al., 2004]. ious mechanisms, including mutations in
which this mouse model recapitulates The developing serotonergic sys- genes encoding transcription factors in-
some behavioral traits characterizing au- tem may be dysregulated in autism. Brain volved in specification and patterning of
tism remains to be explored, but this at 5-HT synthesis is normally high in young 5-HT receptors or neurons (discussed
least provides evidence that a mutation in children, followed by a gradual decline to below). Another possibility is that altered
a single gene controlling cortical inter- adulthood. This dynamic is disrupted in expression of genes regulating cholesterol
neuron migration and differentiation can autism, such that 5-HT levels are initially biosynthetic or metabolic pathways
have complex consequences on mouse lower than normal, but gradually increase could compromise functioning of key
behavior [Levitt et al., 2004]. In sum- to a greater extent than adult levels by 2 players in the serotonergic system, such as
mary, another important concept regard- to 15 years of age [Chugani et al., 1999; the 5-HTT [Scanlon et al., 2001],
ing the link between interneuronal mi- Chugani, 2002]. Positron emission to- VMAT2, MAO-A, or 5-HT receptors
gration and the potential etiology of mography (PET) imaging with radiola- [Hillbrand et al., 2000]. This possibility is
autism (and other neurodevelopmental belled L-tryptophan has demonstrated supported by hypertrophic development
disorders potentially) is the fact that this asymmetric 5-HT synthesis in the den- of raphe 5-HT neurons in mice with
long-range migration of cortical inter- tate-thalamo-cortical pathway of autistic targeted disruption of Dhcr7, a gene en-
neurons (several millimeters in humans) boys [Chugani et al., 1997; Chugani, coding the last enzyme in cholesterol
might be more vulnerable than radial mi- 2002]. Consequences of elevated levels of biosynthesis, which is also mutated in
gration of pyramidal neurons to genetic 5-HT in the developing somatosensory Smith-Lemli-Opitz Syndrome [Waage-
or environmental alterations during early system have been analyzed in a variety of Baudet et al., 2003].
development because of the relative animal models, providing evidence for
complexity of the cell– cell interactions disruption in the formation of thalamo- Catecholamines
involved in tangential migration [Marin cortical sensory circuits [reviewed by Luo Evidence for possible involvement
and Rubenstein, 2001; Levitt et al., et al., 2003]. This is not all that surpris- of dopamine and norepinephrine in au-
2004]. ing, since accumulated evidence shows tism comes from evidence of decreased
5-HT to be a critical regulator of key activity of dopamine ␤-hydroxylase
Serotonin events in neural and glial development, (DBH) and increased levels of norepi-
One feature of autism is high levels including cell proliferation, differentia- nephrine in serum from autistic children
of serotonin (5-HT) in blood platelets tion, migration, apoptosis, and synapto- and their parents [Lake et al., 1977] and
[hyperserotonemia; Anderson et al., genesis [reviewed by Lauder, 1990, 1993; changes in catecholamine metabolites in
1990]. To what extent such elevated Whitaker-Azmitia, 2001]. such children [Martineau et al., 1994].
308 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
Recently, the DBH gene on chromo- transmitter system has received little atten- a susceptibility locus for autistic spectrum
some 9q34 has been investigated as a tion in the field of autism. Recently, disorder [Gharani et al., 2004]. The highly
possible candidate locus using an affected however, the forebrain cholinergic system conserved homeodomain-proteins EN-
sib-pair method. Although no association has come under closer scrutiny, with sev- GRAILED-1 and ENGRAILED-2 are
was found for polymorphic alleles in sib- eral studies utilizing neurochemical, immu- specifically expressed in the MHB region
pairs, there was a significantly higher fre- nocytochemical, or molecular biological during distinct periods of embryogenesis.
quency in mothers, raising the possibility techniques, providing evidence of abnor- This spatio-temporal pattern of engrailed
that polymorphisms in DBH could in- mal expression of cholinergic receptors in gene expression coincides with the gener-
crease the risk for having an autistic child cortex and cerebellum in autism. The first ation of cerebellar precursor cells, which
[Robinson et al., 2001]. Recent evidence study reported lower levels of muscarinic has led to the suggestion that they may be
for maternal modifier effects (where the and nicotinic cholinergic receptors in pari- important for establishing correct cell num-
maternal genotype affects the fetal phe- etal and frontal cortices, together with ele- bers in the cerebellum [Kuemerle et al.,
notype) at DBH and MAO-A loci [Jones vated levels of the trophic factor BDNF in 1997; Baader et al., 1998]. In support of
et al., 2004] provides support for this idea basal forebrain [Perry et al., 2001]. A sub- this view; disruption of En-1 in mice results
and suggests changes in the interuterine sequent study found evidence for decreased in a deletion of the cerebellar anlage and
environment as a possible risk factor. ligand binding to ␣3/␣4/␤2 nicotinic re- midbrain [Wurst et al., 1994] and perinatal
This is consistent with evidence that both ceptors in cerebellum [Lee et al., 2002]. death of these mice. En2 knockout mice
tyrosine hydroxylase (TH) and DBH are Recently; reduced expression of ␣4␤2 nic- are viable but show reduced cerebellar size
required for prenatal development of otinic receptor subunits and receptor bind- and mildly abnormal foliation [Millen et al.,
mouse embryos [Thomas et al., 1995, ing in parietal cortex of autistic brains, to- 1994]. Given the likely functional redun-
1998; Roffler-Tarlov and Rios, 2001; gether with increased ␣7 receptor binding dancy of the two engrailed genes, En1 may
Portbury et al., 2003]. in the cerebellum, was reported [Martin- compensate for the loss of En2, thus ex-
Ruiz et al., 2004]. Because the level of plaining the mild phenotypic changes in
Developmental Origin of evidence implicating the cholinergic cir- En2 knockout mice [Joyner et al., 1991],
Monoamine Neurons cuitry in autism is still poor at the present especially since En2 is expressed in cerebel-
In the vertebrate embryo, midbrain time, we will not review in detail the lar Purkinje cells (PCs) throughout embry-
dopamine (DA) neurons, hindbrain mechanisms patterning development of onic development but is down-regulated in
5-HT neurons of the raphe nuclei, and this neurotransmitter system. these cells after birth.
noradrenergic (NA) neurons of the locus
coeruleus (LC) are generated adjacent to CASE REVIEW OF SPECIFIC Engrailed and differentiation of dopaminergic
the midhindbrain boundary (MHB) or CANDIDATE GENES FOR neurons
isthmus, an organizing center induced by AUTISM It has recently been shown that
networks of transcription factors and se- The concept that emerges from analyzing En1 and En2 are essential for the main-
creted signals [Ye et al., 2001]. DA neu- the current literature is that autism obvi- tenance of DA neurons in the substantia
rons are generated rostral to the MHB, in ously constitutes a very heterogeneous nigra (SN) and ventral tegmentum (VT)
response to intersecting signals, sonic neuropathology at the genetic level, [Simon et al., 2003]. In the mouse, both
hedgehog (Shh), from the floorplate, and which probably means that there are engrailed genes are expressed in mesen-
Fgf8 from the MHB. 5-HT neurons many gene mutations that could inde- cephalic DA (mDA) neurons from E11
form caudal to the MHB in response to pendently lead to an “autistic” brain at to adulthood. In engrailed 1/2 double
the same two signals, but preceded by an the functional level. In Table 1A–C, we mutants, mDA neurons are normally in-
earlier signal, Fgf4, from the primitive summarize all of the existing evidence duced and express several differentiation
streak [Ye et al., 1998; Hynes and from gene linkage and association studies markers, but then fail to mature and are
Rosenthal, 1999]. NA neurons of the LC for genes associated with autism. These lost by E14.
are also generated caudal to the MHB, in tables represent candidate genes identi- The association study reporting
response to signaling by Shh, Fgf8, and fied as confirmed targets in familial forms En2 as a susceptibility locus for autism
BMPs [Crossley et al., 1996; Morin et al., of autism as well as other genes that are spectrum disorder [Gharani et al., 2004],
1997; Guo et al., 1999; Vogel-Hopker simply candidates within genetic loci im- together with the phenotype observed in
and Rohrer, 2002; Jaszai et al., 2003; plicated in autism through linkage and En1/2 knockout mice [Simon et al.,
Lam et al., 2003; Eddison et al., 2004]. As association studies. In the following sec- 2003], correlates well with the well-doc-
discussed below, specification of neuro- tions we perform a case review of several umented cerebellar phenotype in autism,
transmitter phenotypes of MA neurons of the most interesting candidates classi- including cerebellar hypoplasia and de-
requires a complex signaling through fied into two categories: (1) genes in- creased number of PCs (see Neuroana-
networks of transcription factors, which volved in the early patterning of specific tomical Abnormalities in Autism). Fur-
is only beginning to be elucidated regions of the CNS or specific neuronal thermore, the role of Engrailed in the
[Briscoe et al., 1999; Pattyn et al., 1999, subpopulations and (2) genes involved in patterning of dopaminergic neurons is
2000, 2004]. However, it is already clear the synapses assembly of specific neuronal also compatible with some findings im-
that these transcriptional networks must circuits. plicating a dopaminergic defect in autism
include En1, En2, Wnt1, and Lmx1b [Ernst et al., 1997].
[Smidt et al., 2003]. Genes Patterning the Central
Nervous System (CNS) Wnt is a patterning gene also involved in
Acetylcholine activity-dependent dendritic development
Since an early study reported evi- Engrailed and cerebellar neurodevelopmental Several reports have provided evi-
dence of neuropathology in basal forebrain defects dence for Wnt2 as a susceptibility gene
of autistics, the location of cholinergic neu- Recently, association studies have for autism [Wassink et al., 2001; how-
rons innervating the cortex, this neuro- provided evidence for Engrailed-2 (En2) as ever, see also McCoy et al., 2002]. Sev-
MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 309
Table 1A. Autism Candidate Genes
Chromo-
somes X–2

Marker Symbol Description Chromosome References

GDB:191610 MAOA monoamine oxidase A Xp11.4-p11.3 Cohen et al., 2003


Hs.438877 NLGN3 Neuroligin 3 Xq13.1 Jamain et al., 2003
KIAA1260 NLGN4 Neuroligin 4 Xp22.33 Jamain et al., 2003
PMC153509P1 AGTR2 angiotensin II receptor, type 2 Xq22-q23 Thomas et al., 1999;
Vervoort et al.,
2002
DXS7069 MECP2 methyl CpG binding protein 2 (Rett syndrome) Xq28 Muhle et al., 2004;
Longo et al., 2004
DXS1047 MRXS11 mental retardation, X-linked, syndromic 11 Xq26-q27 Liu et al., 2001; Shao
et al., 2003
DXS6789 PTOS2 Ptosis, hereditary congenital 2 Xq24-q27.1 Liu et al., 2001; Shao
et al., 2003
DXS6789 CHDS3 Coronary heart disease, susceptibility to, 3 Xq23-q26 Liu et al., 2001; Shao
et al., 2003
DXS6789 MTBS Mycobacterium tuberculosis, susceptibility to infection by Xq Liu et al., 2001; Shao
et al., 2003
D1S1675 GGAA20F08 NA (moderate effect) 1p12 Risch et al., 1999
D1S1656 SCZD9 schizophrenia disorder 9 1q21-1q22 Buxbaum et al.,
D1S534 2004; Risch et al.,
1999
D1S2141 KCNK2 potassium channel, subfamily K, member 2 1q41 Buxbaum et al., 2004
G65049 DISC1 disrupted in schizophrenia 1 1q42.1 Buxbaum et al., 2004
D1S3462 SLEB1 systemic lupus erythematosus susceptibility 1 1q41-q42 Buxbaum et al., 2004
D1S547 KMO kynurenine 3-monooxygenase (kynurenine 3-hydroxylase) 1q42-q44 Buxbaum et al., 2004
WI-20654 KCNK1 potassium channel, subfamily K, member 1 1q42-q43 Buxbaum et al., 2004
606058 cAMP-GEFII cAMP-regulated guanine nucleotide exchange factor 1 2q21-q33 Bacchelli et al., 2003
(also mutations in TBR-1; GAD1; DLX1; DLX2; CHN1;
ATF2; HOXD1, NEUROD1)
D2S142 PSDA Phrase speech delay, autism-related 2q Buxbaum et al., 2004
SHGC-82894 SCN2A2 sodium channel, voltage-gated, type II, alpha 2 2q23-q24 Weiss et al., 2003
RH79252 SCN3A sodium channel, voltage-gated, type III, alpha 2q24 Weiss et al., 2003
A006O14 SCN1A sodium channel, voltage-gated, type I, alpha 2q24.3 Weiss et al., 2003
D2S364 MPRM Myopathy, proximal, with early respiratory muscle involvement 2q24-q31 Buxbaum et al., 2004
(Edstr
D2S2188 MMDK Mesomelic dysplasia, Kantaputra type 2q24-q32 IMGSAC, 2001
D2S2716 CHRNA1 cholinergic receptor, nicotinic, alpha polypeptide 1 (muscle) 2q24-q32 Agulhon et al., 1999;
Martin-Ruiz, et
al., 2004
G54269 AGC1 mitochondrial aspartate-glutamate carrier protein 2q24-q33 Ramoz et al., 2004
G16421 INPP1 inositol polyphosphate-1-phosphatase 2q32 Serajee et al., 2003
RH68995 ERBB4 receptor for NDF/heregulin 2q33.3-q34 Pescucci et al., 2003
RH66377 ADAM23 disintegrin and metalloproteinase domain 23 2q33 Pescucci et al., 2003
RH15615 KLF7 Kruppel-like factor 7 2q33.3-q34 Pescucci et al., 2003
RH65625 GPR1 G protein-coupled receptor 1 2q33.3 Pescucci et al., 2003
PMC16008P2 FZD5 frizzled homolog 5 2q33-q34 Pescucci et al., 2003
G62667 MAP2 microtubule-associated protein 2 2q34-q35 Pescucci et al., 2003
D2S2128 NRP2 neuropilin 2 2q33.3 Pescucci et al., 2003
D2S2634 CREB1 cAMP responsive element binding protein 1 2q34 Pescucci et al., 2003

eral mutations affecting the Wnt2 coding teins (sFRPs) as one of several unrelated 2002], while loss-of-function mutations in
sequence were initially found to segre- protein families that negatively regulate re- individual Wnts cause deletions or malfor-
gate with autism in families containing ceptor binding by Wnts. mations of distinct brain regions [Brault et
multiple affected individuals [Wassink et In the developing mammalian brain, al., 2001; Lyuksyutova et al., 2003; Zhou
al., 2001]. Wnts have been implicated in the pattern- et al., 2004]. Spatial and temporal expres-
The Wingless-Int (Wnt) genes encode ing of the CNS along the antero-posterior sion patterns of WNT genes show striking
a large family of cysteine-rich secreted gly- axis, including patterning of the telenceph- similarity between human and mouse, sug-
coproteins that regulate diverse cell behav- alon, diencephalon, and mesencephalon. gesting that the developmental roles of
iors during embryonic development [re- The signaling mechanisms underlying Wnt these genes may be highly conserved
viewed in Wodarz and Nusse, 1998]. function during CNS patterning include [Grove et al., 1998; Abu-Khalil et al.,
Nineteen Wnt genes have been identified many cytoplasmic proteins, including Di- 2004].
in the murine and human genome. The shevelled and ␤-catenin. Specifically, over- Recent investigation of the role
secreted Wnt signals positively through the expression of ␤-catenin increases cortical of Wnt signaling during neuronal dif-
Frizzled family of receptors and can be an- size by positively regulating the generation ferentiation has revealed a surprising
tagonized by secreted Frizzled-related pro- of neural precursors [Chenn and Walsh, late role in the regulation of activity-
310 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
Table 1B. Autism Candidate Genes
Chromosomes
3–13

Marker Symbol Description Chromosome References

D3S3680 KIAA0121 KIAA0121 gene product 3p25.2 IMGSAC 2001


D3S3037 AUTS2 Autism, susceptibility to, 2 3q25-27 Auranen et al. 2002
D3S3037 AUTS3 Autism, susceptibility to, 3 3q25.1-3q25.2 Auranen et al. 2002
D3S2418 DFNA44; FGF12 deafness, autosomal dominant; fibro- 3q28-29 Risch et al. 1999
blast growth factor 12
D4S2366 HLN2 Huntington-like neurodegenerative 4p15.3 Risch et al. 1999
disorder 2
D4S2366 SLEB3 systemic lupus erythematosus suscepti- 4p16-15.2 Risch et al. 1999
bility 3
D4S2366 PPP2R2C protein phosphatase 2 (2A), regulatory 4p16.1 Risch et al. 1999
subunit B (PR 52), gamma isoform
SHGC-50349 GLRB glycine receptor, beta 4q31.3 Ramanthan et al. 2004
PMC310777P9 NPY5R neuropeptide Y receptor Y5 4q31-q32 Ramanthan et al. 2004
NPY1R_956 NPY1R neuropeptide Y receptor Y1 4q31.3-q32 Ramanthan et al. 2004
D4S3293 TDO2 tryptophan 2,3-dioxygenase 4q31-q32 Nabi et al. 2004
SHGC-67298 GRIA2 glutamate receptor, ionotropic, 4q32-q33 Samanthan et al. 2004
AMPA 2
GDB:373727 GRIK2 glutamate receptor, ionotropic, kai- 6q16.3-q21 Jamain et al. 2002
nate 2 (GluR6 kainate receptor)
D6S283 IDDM15 insulin-dependent diabetes mellitus 15 6q21 Philippe et al. 1999
D7S640 AUTS1 autism susceptibility to, 1 7q Ashley-Koch et al. 1999;
Barrett et al. 1999;
IMGSAC 2001
KIAA0442 AUTS2 autism, susceptibility to, 2 7q11.22- Sultana et al. 2002
q11.23
D7S2204 CMT2F Charcot-Marie-Tooth disease, axonal, 7q11-q21 IMGSAC 1998
F
D7S3120 RELN reelin 7q22 Hutcheson et al. 2003
D7S501-D7S2847 FOXP2, SPCH1; WNT2; RAY1; NRCAM; 7q22.1-q33 Hutcheson et al. 2003;
SMO; CAV1,2; GPR22; CFTR; MEST/ IMGSAC 2001;
PEG1; MEK2; PAX4 Adres 2002
sWSS3754 GMR8 glutamate receptor, metabotropic 8 7q31-q33 Serajee et al. 2003
STS-X83368 PIK3CG phosphoinositide-3-kinase, catalytic, 7q22.3 Serajee et al. 2003
gamma polypeptide
D7S23 WNT2 wingless-type MMTV integration site 7q31 Wassinck et al. 2001
family member 2
D7S684 CHDM Chordoma (malignant tumors derived 7q33 IMGSAC 1998
from notochordal remnants)
D7S495 OTSC2 otosclerosis 2 (sclerosis of labyrinthine 7q34-q36 Shao et al. 2003
capsule; hearing impairment)
RH102827 EN2 engrailed homolog 2 7q36 Gharani et al. 2004
D9S1826 SPG19 spastic paraplegia 19 (autosomal domi- 9q IMGSAC 2001
nant)
GDB:551079 DBH dopamine beta-hydroxylase 9q34 Robinson et al. 2001
D9S158 JBTS1 Joubert syndrome 1 9q34.3 IMGSAC 2001
D9S1826 DFNB33 deafness, autosomal recessive 33 9q34-qtel IMGSAC 2001
D11S2371 SCZD2 schizophrenia disorder 2 11q14-q21 Risch et al. 1999
D12S1901 AVPR1A arginine vasopressin receptor 1A 12q14-q15 Wassink et al. 2004
608049 AUTS3 Autism, susceptibility to, 3 13q14-q22 Bradford et al. 2001
HTR2A-112F HTR2A-2 5-hydroxytryptamine (serotonin) re- 13q14-q21 Andres et al. 2002;
ceptor 2A Veenstra-Vander-
Weele et al. 2002
D13S779 SCZD7 schizophrenia disorder 7 13q32 Risch et al. 1999

dependent arborization of dendrites in Malenka, 2003]. This study also re- ity-dependent dendritic differentiation
hippocampal pyramidal neurons. Yu vealed that the release of Wnt, which [Yu and Malenka, 2003].
and Malenka [2003] have shown that occurs during normal neuronal devel- Interestingly, inactivation of Di-
overexpression of ␤-catenin (and other opment, is enhanced by manipulations shevelled-1 (Dvl1) in the mouse leads to
members of the cadherin/catenin com- that mimic increased activity and that abnormal development of social behav-
plex) enhances dendritic arborization, Wnt contributes to the effects of neural iors, such as differences in whisker trim-
whereas sequestering endogenous activity on dendritic arborization. ming, deficits in nest building, reduced
␤-catenin causes a decrease in dendritic These results show that ␤-catenin is an huddling contact during cage sleeping,
branch tip number and prevents the important mediator of dendritic mor- and subordinate responses in a social
enhancement of dendritic morphogen- phogenesis and that Wnt/-catenin sig- dominance test [Lijam et al., 1997]. No-
esis caused by neural activity [Yu and naling is likely to be important for activ- tably, sensorimotor gating was abnormal
MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 311
Table 1C. Autism Candidate Genes
Chromosomes
15–20

Marker Symbol Description Chromosome References

D15S652 OTSC1; LCS1 otosclerosis 1; lymphedema-cholestasis syndrome 1 15q25-q26 Risch et al. 1999
D7S530 AUTS1 autism susceptibility 1 15q11-q13 IMGSAC 1998
PMC311424P2 HERC2 hect domain and RLD 2 15q13 Smith et al. 2000
STS-L08485 GABRA5 gamma-aminobutyric acid (GABA) A receptor, al- 15q11.2-q12 Shao et al. 2003; Longo et
pha 5 al. 200
RH11160 GABRB3 gamma-aminobutyric acid (GABA) A receptor, beta 15q11.2-q12 Shao et al. 2003; Longo et
3 al. 200
PMC24540P1 NDN NECDIN [necdin (mouse) homolog] 15q11-q13 Chibuk et al. 2001
D15S10 UBE3A ubiquitin protein ligase E3A (human papilloma vi- 15q11-q13 Nurmi et al. 2003
rus E6-associated protein, Angelman syndron
GDB:593257 GABRG3 gamma-aminobutyric acid (GABA) A receptor, 15q11-q13 Shao et al. 2003; Long et
gamma 3 al. 2004
RH44910 PWCR1 Prader-Willi syndrome chromosome region 1 15q11.2 Veltman et al. 2004
RH121309 SNURF SNRPN upstream reading frame 15q12 Mann and Bartolomei 1999
GDB:626138 SNRPN small nuclear ribonucleoprotein polypeptide N 15q12 Mann and Bartolomei 1999
PMC64493P3 NDNL2/ necdin-like 2/Melanoma-associated antigen F1 15q13.1 Chibuk et al. 2001
MAGE-G
G10644 PTPN9 protein tyrosine phosphatase, non-receptor type 9 15q23 Smith et al. 2000
G06258 SLP-1, hUNC-24 stomatin (EPB72)-like 1 15q24-q25 Smith et al. 2000
D16S403 RP22 retinitis pigmentosa 22 (autosomal recessive) 16p12.3-p12.1 Risch et al. 1999
GDB:378419 TSC2; PKD1* tuberous sclerosis 2; polycystic kidney disease 1 16p13.3 Serajee et al. 2003
D16S407 GRIN2A glutamate receptor, ionotropic, N-methyl D-aspar- 16p13.2 IMGSAC 1998
tate 2A
G67509 SSTR5 somatostatin receptor 5 16p13.3 Lauritsen et al. 2003
SLC6A4 HTT, HTTINT2 Solute carrier family 6 (neurotransmitter transporter, 17q11.1-q12 IMGSAC 2001
serotonin), member 4; brain 5-HT transpo
D17S1298 FIMG1 myasthenia gravis, familial infantile, 1 17p13 Risch et al. 1999
D19S587 HSCRS3 Hirschsprung disease, short-segment, 3 19q12 Liu et al. 2001
PMC310963P4 APOE apolipoprotein E 19q13.2 Persico et al. 2004
D20S482 CHED2 comeal endothelial dystrophy 2 (autosomal reces- 20p13 Risch et al. 1999
sive)

in these mice, as assessed by prepulse demonstrated that this gene controls ap- tinct mutations in ARX may cause di-
inhibition of acoustic and tactile startle propriate proliferation of dorsal and ven- verse forms of mental retardation,
reflexes. This interesting behavioral phe- tral forebrain neural precursors, but also epilepsy, or autism in males [Sherr,
notype has been suggested as a potential the migration and differentiation of 2003].
model for aspects of several human psy- GABAergic interneurons from the me-
chiatric disorders, including autism. dial ganglionic eminence to the cortex
Genes Controlling Synaptic
Given the developmental mechanisms [Kitamura et al., 2002]. These findings
Assembly and Dendritic
underlying this interesting behavioral prompted these investigators to ask
Development
phenotype, it is tempting to speculate whether any human diseases are caused
that the Wnt/Dvl1 pathway might con- by ARX inactivation. Their search led
trol patterning and synaptic assembly and them to several individuals suffering from Neuroligin (NLGN) 3 and 4
dendritic branching in key cortical and X-linked lissencephaly with abnormal Recently, the identification of a
mesencephalic neuronal networks con- genitalia (XLAG), who turn out to have frameshift mutation (1186insT) in the
trolling complex social behaviors im- mutations in ARX. Interestingly, consis- NLGN4 gene as well as a cysteine to
paired in autism. Surprisingly, to date, tent with the findings in mice, pheno- threonine substitution in position 451 of
very few studies have explored this pos- type/genotype studies in humans suggest NLGN3 have been reported to segregate
sibility, and future investigations should that truncating mutations cause X-linked with autism in two independent Swedish
test this hypothesis. lissencephaly with abnormal genitalia, families [Jamain et al., 2003]. Subse-
and insertion/missense mutations result quently, another team has confirmed this
Transcription factors specifying cortical in epilepsy and mental retardation with- finding and has found a 2-base-pair de-
interneuron phenotype out cortical dysplasia [Sherr, 2003]. letion in the NLGN4 gene, leading to a
Aristaless-related proteins (includ- Therefore, mutations in the homeobox premature stop codon, in both autistic
ing Arx) belong to the Pax family of gene, ARX, cause a diverse spectrum of and nonautistic mentally retarded males
homeobox transcription factors, several disorders, including cognitive impair- [Laumonnier et al., 2004]. This strongly
members of which are known to be in- ment, epilepsy, and, in another group of suggests that NLGN4 might not only be
volved in genetic diseases. A recent study patients, severe cortical malformations. involved in autism, but also in mental
using a gene-targeting approach to iden- Although the precise prevalence of ARX retardation, suggesting that autism and
tify a pivotal role for the Arx transcrip- mutations is unclear, further investiga- mental retardation might have common
tion factor during brain development has tions will undoubtedly test whether dis- genetic origins.
312 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
The Neuroligin gene family was ligands) or NT3 (trkC ligands) are un- by the Diagnostic and Statistical Manual
initially identified as a component of changed compared to controls. Of course of Mental Disorders, Fourth Edition
postsynaptic glutamatergic synapses, one should be careful about translating (DSM-IV) and the International Classifi-
which have been shown to play a role in increased plasma levels with increased cation of Diseases, Tenth Revision. Re-
the trans-neuronal signaling that controls CNS levels, but these findings suggest cent studies indicate that at least 80% of
synapse differentiation through binding that, during early infancy, trkB ligands Rett disorder cases are caused by muta-
of neurexin-␤ on the presynaptic side might specifically be expressed and/or tions in the MeCP2 gene. Since there is
[Ichtchenko et al., 1995; Song et al., secreted at higher levels in the CNS of some phenotypic overlap between autis-
1999]. As we were writing this review, autistic or mentally retarded children tic disorder and Rett disorder [Muhle et
Chih and coworkers reported conse- from a source that remains to be deter- al., 2004], a recent study aimed at ana-
quences of these disease-associated muta- mined (central or peripheral nervous sys- lyzing large groups of females clinically
tions on neuroligin function in vitro tem?). The effect of BDNF and NT4 on diagnosed with autistic disorder for the
[Chih et al., 2004]. These authors dem- activity-dependent dendritic outgrowth presence of mutations in the MeCP2
onstrated that a point mutation in the and branching is well established during gene. Two females (4%) presenting au-
arginine residue in position 451 and a development [McAllister et al., 1996, tistic disorder were found to have de
nonsense mutation in aspartate 396 of 1997, 1995] and therefore could be cor- novo mutations in MeCP2. These data
NLGN3 and NLGN4, respectively, re- related with the transient, early increase provide additional evidence of variable
sulted in intracellular retention of the of brain growth reported in young autis- expression in the Rett disorder pheno-
mutant proteins. Overexpression of tic babies [Lainhart et al., 1997; type and, taken together with the role of
wildtype NLGN3 and NLGN4 proteins Courchesne et al., 2001, 2003]. Future MeCP2 in the control of BDNF tran-
in hippocampal neurons stimulated the investigations should test whether scription mentioned above, these finding
formation of presynaptic terminals, BDNF-mediated signaling is increased in suggest a potential mechanism whereby
whereas the disease-associated mutations the early period of postnatal brain devel- MeCP2 mutations might represent a risk
resulted in loss of this synaptogenic func- opment in autism and whether this cor- factor for the appearance of autism
tion. These findings suggest that the pre- relates with a premature increase in den- through regulating BDNF expression
viously identified mutations in Neuroligin dritic outgrowth of pyramidal neurons, and potentially dendritic differentiation
genes are likely to be relevant for the which could in turn have profound con- in the cortex.
neurodevelopmental defects in autism sequences on the establishment of corti-
spectrum disorders and mental retarda- cal networks. CONCLUSION
tion, since they impair the function of What could cause this increased The concept that emerges from an-
synaptic cell adhesion molecules [Chih et level of BDNF expression and/or secre- alyzing the current literature is that au-
al., 2004]. tion? An interesting possibility lies in the tism has a strong genetic component, but
analysis of the role of MeCP2 in the obviously constitutes a very heteroge-
BDNF and MeCP2 control of BDNF transcription [Chen et neous neuropathology at the genetic
Neurotrophins have multiple func- al., 2003]. Mutations in methyl-CpG- level (see Wassink et al., this issue; see
tions during peripheral and CNS devel- binding protein 2 (MeCP2), which en- also Table 1A–C). In light of current
opment, such as controlling neuronal codes a protein that has been proposed to evidence, it seems likely that the etiology
survival, target innervation, and synapto- function as a global transcriptional re- of autism involves complex interactions
genesis. Neurotrophins are secreted li- pressor, are the cause of Rett syndrome, between environmental factors and genetic
gands that exert their biological functions an X-linked progressive neurological dis- mutations controlling either (1) the pat-
by binding to a high-affinity receptor, order. Interestingly; Rett syndrome is terning of neuronal populations critical
the Trk tyrosine kinase receptor. A sometime associated with autism. Al- for the control of inhibition/excitation in
prominent member of this family of neu- though the selective inactivation of the cortex and/or (2) the synaptic assem-
rotrophins is brain-derived neurotrophic MeCP2 in neurons is sufficient to confer bly of these excitatory and inhibitory
factor (BDNF), which plays multiple a Rett-like phenotype in mice [Shahba- neuronal networks and/or the neuronal
roles during neuronal differentiation, in- zian et al., 2002], the specific functions of networks involved in large-scale cortical
cluding neuronal survival, activity-de- MeCP2 in postmitotic neurons are not neuromodulation.
pendent dendritic and axonal out- known. Chen and coworkers have re- An important concept in develop-
growth/branching, synapse formation, cently shown that MeCP2 binds selec- mental neurobiology is that genes in-
and neuronal plasticity underlying learn- tively to BDNF promoter III and func- volved in the development of the CNS
ing and memory [reviewed in Shieh and tions to repress expression of the BDNF are extremely pleiotropic: i.e., several
Ghosh, 1997; Kaplan and Miller, 2000]. gene [Shahbazian et al., 2002]. Mem- important genes involved in the early
In two recent studies monitoring brane depolarization mimicking sus- patterning and specification of neuronal
the plasma levels of different neurotro- tained levels of neuronal activity triggers subpopulations also act later in develop-
phins on large samples of randomly the calcium-dependent phosphorylation ment to regulate the proper synaptic as-
picked children retrospectively diagnosed and release of MeCP2 from BDNF pro- sembly of the same or other neuronal
with autism spectrum disorder or mental moter III, thereby facilitating transcrip- populations. Therefore, a mutation in
retardation without autism, cerebral palsy tion. These studies indicate that MeCP2 one of the genes reviewed above will
or age-matched controls revealed that plays a key role in the control of neuronal undoubtedly have many complex, non-
both NT4 and BDNF (the two trkB activity– dependent gene regulation and redundant functions during development
ligands) are significantly increased in au- suggest that the deregulation of this pro- of the CNS that could lead to a divergent
tistic and mentally retarded patients com- cess may underlie the pathology of Rett and complex neuropathology such as au-
pared to controls [Nelson et al., 2001; syndrome. tism.
Miyazaki et al., 2004]. Interestingly, Rett disorder and autism are per- We hope that after reading this re-
other neurotrophins such as NGF (trkA vasive developmental disorders as defined view the reader will realize that, although
MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 313
much work needs to be done in the Belmonte MK, Cook EH, Anderson GM, et al. thalamocortical pathway in autistic boys. Ann
exploration of the neurodevelopmental 2004. Autism as a disorder of neural informa- Neurol 42:666 – 669.
tion processing: Directions for research and Cline HT. 2001. Dendritic arbor development and
mechanisms that could underlie autism, targets for therapy. Mol Psychiatry 9:646 – synaptogenesis. Curr Opin Neurobiol 11:118 –
we now have new directions to explore. 663. 126.
A new hypothesis such as the imbalance Betancur C, Corbex M, Spielewoy C, et al. 2002. Cobb SR, Buhl EH, Halasy K, et al. 1995. Syn-
between excitation and inhibition in the Serotonin transporter gene polymorphisms chronization of neuronal activity in hip-
and hyperserotonemia in autistic disorder. pocampus by individual GABAergic inter-
cortex is attractive because it is based on neurons. Nature 378:75–78.
Mol Psychiatry 7:67–71.
functional evidence characterizing the Borodinsky LN, Root CM, Cronin JA, et al. 2004. Cohen I, Liu X, Schutz C, et al. 2003. Association
autistic brain. The conceptual framework Activity-dependent homeostatic specification of autism severity with a monoamine oxidase
and the molecular tools developed in this of transmitter expression in embryonic neu- A functional polymorphism. Clin Genet 64:
rons. Nature 429:523–530. 190 –197.
emerging field will undoubtedly allow Cook EH Jr., Courchesne R, Lord C, et al. 1997.
Brault V, Moore R, Kutsch S, et al. 2001. Inacti-
rapid progress in the characterization of vation of the beta-catenin gene by Wnt1- Evidence of linkage between the serotonin
the neuronal networks underlying this Cre-mediated deletion results in dramatic transporter and autistic disorder. Mol Psychi-
devastating developmental neuropathol- brain malformation and failure of craniofacial atry 2:247–250.
ogy. f development. Development 128:1253–1264. Courchesne E. 1997. Brainstem, cerebellar and lim-
Briscoe J, Sussel L, Serup P, et al. 1999. Homeobox bic neuroanatomical abnormalities in autism.
gene Nkx2.2 and specification of neuronal Curr Opin Neurobiol 7:269 –278.
REFERENCES identity by graded Sonic hedgehog signalling. Courchesne E, Carper R, Akshoomoff N. 2003.
Nature 398:622– 627. Evidence of brain overgrowth in the first year
Abu-Khalil A, Fu L, Grove EA, et al. 2004. Wnt
Brock J, Brown CC, Boucher J, et al. 2002. The of life in autism. JAMA 290:337–344.
genes define distinct boundaries in the devel-
temporal binding deficit hypothesis of autism. Courchesne E, Karns CM, Davis HR, et al. 2001.
oping human brain: Implications for human
Dev Psychopathol 14:209 –224. Unusual brain growth patterns in early life in
forebrain patterning. J Comp Neurol 474:
Buhl EH, Halasy K, Somogyi P. 1994. Diverse patients with autistic disorder: An MRI study.
276 –288.
sources of hippocampal unitary inhibitory Neurology 57:245–254.
Agulhon C, Abitbol M, Bertrand D, Malafosse, A.
postsynaptic potentials and the number of Courchesne E, Yeung-Courchesne R, Press G, et
1999. Localization of mRNA for CHRNA7
synaptic release sites. Nature 368:823– 828. al. 1988. Hypoplasia of cerebellar vermal lob-
in human fetal brain. Neuroreport 10:2223– ules VI and VII in autism. N Engl J Med
2227 Buxbaum JD, Silverman J, Keddache M, et al.
2004. Linkage analysis for autism in subset 318:1349 –1354.
Anderson GM, Horne WC, Chatterjee D, et al. Coutinho AM, Oliveira G, Morgadinho T, et al.
1990. The hyperserotonemia of autism. Ann families with obsessive-compulsive behaviors:
evidence for an autism susceptibility gene on 2004. Variants of the serotonin transporter
N Y Acad Sci 600:331–340; discussion 341– gene (SLC6A4) significantly contribute to hy-
332. chromosome 1 and further support for sus-
perserotonemia in autism. Mol Psychiatry
Anderson SA, Eisenstat DD, Shi L, et al. 1997. ceptibility genes on chromosome 6 and 19.
9:264 –271.
Interneuron migration from basal forebrain to Mol Psychiatry 9:144 –150.
Crossley PH, Martinez S, Martin GR. 1996. Mid-
neocortex: Dependence on Dlx genes. Sci- Carlsson ML. 1998. Hypothesis: Is infantile autism
brain development induced by FGF8 in the
ence 278:474 – 476. a hypoglutamatergic disorder? Relevance of
chick embryo. Nature 380:66 – 68.
Andres C. 2002. Molecuar genetics and animal glutamate–serotonin interactions for pharma-
DeFelipe J, Farinas I. 1992. The pyramidal neuron
models in autistic disorder. Brain Res Bull cotherapy. J Neural Transm 105:525–535.
of the cerebral cortex: Morphological and
57:109 –119 Carlsson ML, Martin P, Nilsson M, et al. 1999. The
chemical characteristics of the synaptic inputs.
Aoyagi T, Takekawa T, Fukai T. 2003. Gamma 5-HT2A receptor antagonist M100907 is
Prog Neurobiol 39:563– 607.
rhythmic bursts: Coherence control in net- more effective in counteracting NMDA an-
DeFelipe J, Hendry SH, Hashikawa T, et al. 1990.
works of cortical pyramidal neurons. Neural tagonist than dopamine agonist–induced hy- A microcolumnar structure of monkey cere-
Comput 15:1035–1061. peractivity in mice. J Neural Transm 106: bral cortex revealed by immunocytochemical
Ashley-Koch A, Wolpert CM, Menold MM, et al. 123–129. studies of double bouquet cell axons. Neuro-
1999. Genetic studies of autistic disorder and Casanova MF, Buxhoeveden DP, Switala AE, et al. science 37:655– 673.
chromosome 7. Genomics 61:227–236. 2002. Minicolumnar pathology in autism. Eddison M, Toole L, Bell E, et al. 2004. Segmental
Auranen M, Vanhala R, Varilo T, et al. 2002. A Neurology 58:428 – 432. identity and cerebellar granule cell induction
genomewide screen for autism-spectrum dis- Casarosa S, Fode C, Guillemot F. 1999. Mash1 in rhombomere 1. BMC Biol 2:14.
orders: evidence for a major susceptibility lo- regulates neurogenesis in the ventral telen- Ernst M, Zametkin AJ, Matochik JA, et al. 1997.
cus on chromosome 3q25-27. Am J Hum cephalon. Development 126:525–534. Low medial prefrontal dopaminergic activity
Genet 71:777–790. Chen WG, Chang Q, Lin Y, et al. 2003. Derepres- in autistic children. Lancet 350:638.
Aylward EH, Minshew NJ, Field K, et al. 2002. sion of BDNF transcription involves calcium- Fatemi SH, Halt AR, Stary JM, et al. 2002. Glu-
Effects of age on brain volume and head cir- dependent phosphorylation of MeCP2. Sci- tamic acid decarboxylase 65 and 67 kDa pro-
cumference in autism. Neurology 59:175–183. ence 302:885– 889. teins are reduced in autistic parietal and cer-
Baader SL, Sanlioglu S, Berrebi AS, et al. 1998. Chenn A, Walsh CA. 2002. Regulation of cerebral ebellar cortices. Biol Psychiatry 52:805– 810.
Ectopic overexpression of engrailed-2 in cer- cortical size by control of cell cycle exit in Fode C, Ma Q, Casarosa S, et al. 2000. A role for
ebellar Purkinje cells causes restricted cell loss neural precursors. Science 297(5580):365– neural determination genes in specifying the
and retarded external germinal layer develop- 369. dorsoventral identity of telencephalic neu-
ment at lobule junctions. J Neurosci 18:1763– Chibuk TK, Bischof JM, Wevrick R. 2001. A rons. Genes Dev 14:67– 80.
1773. necdin/MAGE-like gene in the chromosome Folstein SE, Rosen-Sheidley B. 2001. Genetics of
Bacchelli E, Blasi F, Biondolillo M, et al. 2003. 15 autism susceptibility region: expression, autism: Complex aetiology for a heteroge-
Screening of nine candidate genes for autism imprinting, and mapping of the human and neous disorder. Nat Rev Genet 2:943–955.
on chromosome 2q reveals rare nonsynony- mouse orthologues. BMC Genet 2:22. Freund TF. 2003. Interneuron diversity series:
mous variants in the cAMP-GEFII gene. Mol Chih B, Afridi SK, Clark L, et al. 2004. Disorder- Rhythm and mood in perisomatic inhibition.
Psychiatry 8:916 –924. associated mutations lead to functional inac- Trends Neurosci 26:489 – 495.
Bailey A, Luthert P, Dean A, et al. 1998. A clini- tivation of neuroligins. Hum Mol Genet 13: Freund TF, Buzsaki G. 1996. Interneurons of the
copathological study of autism. Brain 121: 1471–1477. hippocampus. Hippocampus 6:347– 470.
889 –905. Chugani DC. 2002. Role of altered brain serotonin Gharani N, Benayed R, Mancuso V, et al. 2004.
Ballaban-Gil K, Tuchman R. 2000. Epilepsy and mechanisms in autism. Mol Psychiatry 7 Association of the homeobox transcription
epileptiform EEG: Association with autism Suppl 2:S16 –S17. factor, ENGRAILED 2, 3, with autism spec-
and language disorders. Ment Retard Dev Chugani DC, Muzik O, Behen M, et al. 1999. trum disorder. Mol Psychiatry 9:474 – 484.
Disabil Res Rev 6:300 –308. Developmental changes in brain serotonin Grove EA, Tole S, Limon J, et al. 1998. The hem
Barrett S, Beck JC, Bernier R, et al. 1999. An synthesis capacity in autistic and nonautistic of the embryonic cerebral cortex is defined by
autosomal genomic screen for autism. Collab- children. Ann Neurol 45:287–295. the expression of multiple Wnt genes and is
orative linkage study of autism. Am J Med Chugani DC, Muzik O, Rothermel R et al. 1997. compromised in Gli3-deficient mice. Devel-
Genet 88:609 – 615. Altered serotonin synthesis in the dentato- opment 125:2315–2325.

314 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
Guo S, Brush J, Teraoka H, et al. 1999. Develop- Just MA, Cherkassky VL, Keller TA, et al. 2004. Lewis DA, Levitt P. 2002. Schizophrenia as a dis-
ment of noradrenergic neurons in the ze- Cortical activation and synchronization dur- order of neurodevelopment. Annu Rev Neu-
brafish hindbrain requires BMP, FGF8, and ing sentence comprehension in high-func- rosci 25:409 – 432.
the homeodomain protein soulless/Phox2a. tioning autism: Evidence of underconnectiv- Lijam N, Paylor R, McDonald MP, et al. 1997.
Neuron 24:555–566. ity. Brain 127(Pt 8):1811–1821. Social interaction and sensorimotor gating ab-
Hardan AY, Minshew NJ, Keshavan MS. 2000. Kanner, L. 1943. Autistic disturbances of affective normalities in mice lacking Dvl1. Cell 90:
Corpus callosum size in autism. Neurology contact. Nervous Child. 2:217–250. 895–905.
55(7):1033–1036. Kaplan DR, Miller FD. 2000. Neurotrophin signal Liu J, Nyholt DR, Magnussen P, et al. 2001. A
Hariri AR, Mattay VS, Tessitore A, et al. 2002. transduction in the nervous system. Curr genomewide screen for autism susceptibility
Serotonin transporter genetic variation and Opin Neurobiol 10:381–391. loci. Am J Hum Genet 69:327–340.
the response of the human amygdala. Science Kitamura K, Yanazawa M, Sugiyama N, et al. 2002. Longo I, Russo L, Meloni I, et al. 2004. Three Rett
297:400 – 403. Mutation of ARX causes abnormal develop- patients with both MECP2 mutation and
Hariri AR, Weinberger DR. 2003. Functional ment of forebrain and testes in mice and X- 15q11-13 rearrangements. Eur J Hum Genet
neuroimaging of genetic variation in seroto- linked lissencephaly with abnormal genitalia 12:682– 685.
nergic neurotransmission. Genes Brain Behav in humans. Nat Genet 32:359 –369. Luddens H, Korpi ER, Seeburg PH. 1995.
2:341–349. Klauck SM, Poustka F, Benner A, et al. 1997. GABAA/benzodiazepine receptor heteroge-
Hashimoto T, Tayama M, Murakawa K, et al. Serotonin transporter (5-HTT) gene variants neity: Neurophysiological implications. [Re-
1995. Development of the brainstem and cer- view]. Neuropharmacology 34:245–254.
associated with autism?. Hum Mol Genet
ebellum in autistic patients. J Autism Dev Luo X, Persico A, Lauder J. 2003. Serotonergic
6:2233–2238.
Disord 25:1–18. regulation of somatosensory cortical develop-
Korvatska E, Van de Water J, Anders TF, et al.
Hillbrand M, Waite BM, Miller DS, et al. 2000. ment: Lessons from genetic mouse models.
2002. Genetic and immunologic consider-
Serum cholesterol concentrations and mood Dev Neurosci 25:173–183.
ations in autism. Neurobiol Dis 9:107–125.
states in violent psychiatric patients: An expe- Lyuksyutova AI, Lu CC, Milanesio N, et al. 2003.
rience sampling study. J Behav Med 23:519 – Kuemerle B, Zanjani H, Joyner A, et al. 1997.
Pattern deformities and cell loss in En- Anterior-posterior guidance of commissural
529. axons by Wnt-frizzled signaling. Science 302:
Holopainen IE, Kokkonen H, Korpi ER. 2001a. grailed-2 mutant mice suggest two separate
patterning events during cerebellar develop- 1984 –1988.
Angelman syndrome: From the phenotype of Maestrini E, Lai C, Marlow A, et al. 1999. Sero-
a developmental disorder to the genes. Duo- ment. J Neurosci 17:7881–7889.
Lainhart JE, Piven J, Wzorek M, et al. 1997. Mac- tonin transporter (5-HTT) and gamma-ami-
decim 117:383–387. nobutyric acid receptor subunit beta3
Holopainen IE, Metsahonkala EL, Kokkonen H, et rocephaly in children and adults with autism.
J Am Acad Child Adolesc Psychiatry 36:282– (GABRB3) gene polymorphisms are not as-
al. 2001b. Decreased binding of [11C]fluma- sociated with autism in the IMGSA families.
zenil in Angelman syndrome patients with 290.
Lake CR, Ziegler MG, Murphy DL. 1977. In- The International Molecular Genetic Study of
GABA(A) receptor beta3 subunit deletions. Autism Consortium. Am J Med Genet 88:
Ann Neurol 49:110 –113. creased norepinephrine levels and decreased
dopamine-beta-hydroxylase activity in pri- 492– 496.
Hussman JP. 2001. Suppressed GABAergic inhibi-
mary autism. Arch Gen Psychiatry 34:553– Manes F, Piven J, Vrancic D, et al. 1999. An MRI
tion as a common factor in suspected etiolo-
study of the corpus callosum and cerebellum
gies of autism. J Autism Dev Disord 31:247– 556.
in mentally retarded autistic individuals. J
248. Lam CS, Sleptsova-Friedrich I, Munro AD, et al.
Neuropsychiatry Clin Neurosci 11(4):470 –
Hynes M, Rosenthal A. 1999. Specification of do- 2003. SHH and FGF8 play distinct roles dur-
474.
paminergic and serotonergic neurons in the ing development of noradrenergic neurons in
Mann MR, Bartolomei MS. 1999. Towards a mo-
vertebrate CNS. Curr Opin Neurobiol 9:26 – the locus coeruleus of the zebrafish. Mol Cell
lecular understanding of Prader-Willi and
36. Neurosci 22:501–515.
Angelman syndromes. Hum Mol Genet
IMGSAC. 1998. A full genome screen for autism Lauder JM. 1993. Neurotransmitters as growth reg-
8:1867–1873.
with evidence for linkage to a region on ulatory signals: Role of receptors and second
chromsome 7q. International Molecular Ge- Marin O, Rubenstein JL. 2001. A long, remarkable
messengers. Trends Neurosci 16:233–240.
netic Study of Autism Consortium. Hum Mol journey: Tangential migration in the telen-
Lauder JM. 1990. Ontogeny of the serotonergic cephalon. Nat Rev Neurosci 2:780 –790.
Genet 7:571–578. system in the rat: Serotonin as a developmen-
IMGSAC. 2001. A genomewide screen for autism: Martineau J, Herault J, Petit E, et al. 1994. Cat-
tal signal. Ann N Y Acad Sci 600:297–314. echolaminergic metabolism and autism. Dev
strong evidence for linkage to chromosomes Lauritsen MB, Nyegaard M, Betancur C, et al.
2q, 7q, and 16p. Am J Hum Genet 69:570 – Med Child Neurol 36:688 – 697.
2003. Analysis of transmission of novel poly- Martin-Ruiz CM, Lee M, Perry RH, et al. 2004.
581. morphisms in the somatostatin receptor 5
Ichtchenko K, Hata Y, Nguyen T, et al. 1995. Molecular analysis of nicotinic receptor ex-
(SSTR5) gene in patients with autism. Am J pression in autism. Brain Res Mol Brain Res
Neuroligin 1: A splice site-specific ligand for Med Genet 121B:100 –104.
beta-neurexins. Cell 81:435– 443. 123:81–90.
Laumonnier F, Bonnet-Brilhault F, Gomot M, et McAllister AK, Katz LC, Lo DC. 1996. Neurotro-
Jamain S, Betancur C, Quach H, et al. 2002. Link- al. 2004. X-linked mental retardation and au-
age and association of the glutamate receptor phin regulation of cortical dendritic growth
tism are associated with a mutation in the requires activity. Neuron 17:1057–1064.
6 gene with autism. Mol Psychiatry 7:302–
NLGN4 gene, a member of the neuroligin McAllister AK, Katz LC, Lo DC. 1997. Opposing
310.
family. Am J Hum Genet 74:552–557. roles for endogenous BDNF and NT-3 in
Jamain S, Quach H, Betancur C, et al. 2003. Mu-
Lavdas AA, Grigoriou M, Pachnis V, et al. 1999. The regulating cortical dendritic growth. Neuron
tations of the X-linked genes encoding neu-
medial ganglionic eminence gives rise to a pop- 18:767–778.
roligins NLGN3 and NLGN4 are associated
ulation of early neurons in the developing ce- McAllister AK, Lo DC, Katz LC. 1995. Neurotro-
with autism. Nat Genet 34:27–29.
Jaszai J, Reifers F, Picker A, et al. 2003. Isthmus- rebral cortex. J Neurosci 19:7881–7888. phins regulate dendritic growth in developing
to-midbrain transformation in the absence of Lee M, Martin-Ruiz C, Graham A, et al. 2002. visual cortex. Neuron 15:791– 803.
midbrain-hindbrain organizer activity. Devel- Nicotinic receptor abnormalities in the cere- McCoy PA, Shao Y, Wolpert CM, et al. 2002. No
opment 130:6611– 6623. bellar cortex in autism. Brain 125(Pt 7):1483– association between the WNT2 gene and au-
Jessell TM. 2000. Neuronal specification in the 1495. tistic disorder. Am J Med Genet 114:106 –
spinal cord: Inductive signals and transcrip- Letinic K, Zoncu R, Rakic P. 2002. Origin of 109.
tional codes. Nat Rev Genet 1:20 –29. GABAergic neurons in the human neocortex. McDougle CJ, Posey D. 2002. Genetics of child-
Jones EG. 2000. Microcolumns in the cerebral cor- Nature 417:645– 649. hood disorders: XLIV. Autism, part 3: Psy-
tex. Proc Natl Acad Sci U S A 97:5019 –5021. Levitt P, Eagleson KL, Powell EM. 2004. Regula- chopharmacology of autism. J Am Acad
Jones MB, Palmour RM, Zwaigenbaum L, et al. tion of neocortical interneuron development Child Adolesc Psychiatry 41:1380 –1383.
2004. Modifier effects in autism at the and the implications for neurodevelopmental Millen KJ, Wurst W, Herrup K, et al. 1994. Ab-
MAO-A and DBH loci. Am J Med Genet disorders. Trends Neurosci 27:400 – 406. normal embryonic cerebellar development
126B:58 – 65. Lewine JD, Andrews R, Chez M, et al. 1999. and patterning of postnatal foliation in two
Joyner AL, Herrup K, Auerbach BA, et al. 1991. Magnetoencephalographic patterns of epilep- mouse Engrailed-2 mutants. Development
Subtle cerebellar phenotype in mice homozy- tiform activity in children with regressive au- 120:695–706.
gous for a targeted deletion of the En-2 ho- tism spectrum disorders. Pediatrics 104(3 Pt Miller MW. 1985. Cogeneration of retrogradely
meobox. Science 251:1239 –1243. 1):405– 418. labeled corticocortical projection and GABA-

MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 315
immunoreactive local circuit neurons in ce- Peters AR. 1994. Primary visual cortex of primates. Rodier PM, Ingram JL, Tisdale B, et al. 1996.
rebral cortex. Brain Res 355:187–192. In: Peters AR, Rockland K. S., editor. Cere- Embryological origin for autism: Develop-
Miller MW. 1986. The migration and neurochem- bral Cortex. New York: Plenum. p 1–36. mental anomalies of the cranial nerve motor
ical differentiation of gamma-aminobutyric Peters A, Sethares C. 1996. Myelinated axons and nuclei. J Comp Neurol 370:247–261.
acid (GABA)-immunoreactive neurons in rat the pyramidal cell modules in monkey pri- Roffler-Tarlov S, Rios M. 2001. Catecholamines
visual cortex as demonstrated by a combined mary visual cortex. J Comp Neurol 365:232– before nervous sctivity. In: Kalverboer AF,
immunocytochemical-autoradiographic tech- 255. Gramsbergen A, editors. Handbook of Brain
nique. Brain Res 393:41– 46. Philippe A, Martinez M, Guilloud-Bataille M, et al. and Human Development. Dordrecht/Bos-
Miyazaki K, Narita N, Sakuta R, et al. 2004. Serum 1999. Genome-wide scan for autism suscep- ton/London: Kluwer Academic Publishers. p
neurotrophin concentrations in autism and tibility genes. Human Molecular Genetics 165–184.
mental retardation: A pilot study. Brain Dev 8:805– 812. Rubenstein JL, Merzenich MM. 2003. Model of
26:292–295. Piven J. 1997. The biological basis of autism. Curr autism: Increased ratio of excitation/inhibi-
Morin X, Cremer H, Hirsch MR, et al. 1997. Opin Neurobiol 7:708 –712. tion in key neural systems. Genes Brain Behav
Defects in sensory and autonomic ganglia and Piven J. 2004. Preface. Ment Retard Dev Disabil 2:255–267.
absence of locus coeruleus in mice deficient Res Rev 10:219 –220 Scanlon SM, Williams DC, Schloss P. 2001. Mem-
for the homeobox gene Phox2a. Neuron 18: Piven J, Arndt S, Bailey J, et al. 1995. An MRI brane cholesterol modulates serotonin trans-
411– 423. study of brain size in autism. Am J Psychiatry porter activity. Biochemistry 40:10507–10513.
152:1145–1149. Schumann CM, Hamstra J, Goodlin-Jones BL, et
Mountcastle VB. 1997. The columnar organization
Piven J, Nehme E, Simon J, et al. 1992. Magnetic al. 2004. The amygdala is enlarged in children
of the neocortex. Brain 120(Pt 4):701–722.
resonance imaging in autism: Measurement of but not adolescents with autism; the hip-
Muhle R, Trentacoste SV, Rapin I. 2004. The
the cerebellum, pons, and fourth ventricle. pocampus is enlarged at all ages. J Neurosci
genetics of autism. Pediatrics 113:e472– e486.
Biol Psychiatry 31:491–504. 24:6392– 6401.
Nabi R, Serajee FJ, Chugani DC, et al. 2004.
Piven J, Saliba K, Bailey J, et al. 1997. An MRI Schuurmans C, Armant O, Nieto M, et al. 2004.
Association of tryptophan 2,3 dioxygenase Sequential phases of cortical specification in-
gene polymorphism with autism. Am J Med study of autism: The cerebellum revisited.
Neurology 49:546 –551. volve Neurogenin-dependent and -indepen-
Genet 125B:63– 68. dent pathways. Embo J 23:2892–2902.
Nelson KB, Grether JK, Croen LA, et al. 2001. Polleux F, Whitford KL, Dijkhuizen PA, et al.
2002. Control of cortical interneuron migra- Schuurmans C, Guillemot F. 2002. Molecular
Neuropeptides and neurotrophins in neonatal mechanisms underlying cell fate specification
blood of children with autism or mental re- tion by neurotrophins and PI3-kinase signal-
ing. Development 129:3147–3160. in the developing telencephalon. Curr Opin
tardation. Ann Neurol 49:597– 606. Neurobiol 12:26 –34.
Nery S, Fishell G, Corbin JG. 2002. The caudal Portbury AL, Chandra R, Groelle M, et al. 2003.
Catecholamines act via a beta-adrenergic re- Sears LL, Vest C, Mohamed S, et al. 1999. An MRI
ganglionic eminence is a source of distinct study of the basal ganglia in autism. Prog
cortical and subcortical cell populations. Nat ceptor to maintain fetal heart rate and sur-
vival. Am J Physiol Heart Circ Physiol 284: Neuropsychopharmacol Biol Psychiatry 23:
Neurosci 5:1279 –1287. 613– 624.
Nilsson M, Waters S, Waters N, et al. 2001. A H2069 –H2077.
Serajee FJ, Zhong H, Nabi R, et al. 2003. The
behavioural pattern analysis of hypoglutama- Powell EM, Campbell DB, Stanwood GD, et al.
metabotropic glutamate receptor 8 gene at
2003. Genetic disruption of cortical interneu-
tergic mice: Effects of four different antipsy- 7q31: Partial duplication and possible associ-
ron development causes region- and GABA
chotic agents. J Neural Transm 108:1181– ation with autism. J Med Genet 40:e42.
cell type-specific deficits, epilepsy, and be-
1196. Shahbazian M, Young J, Yuva-Paylor L, et al.
havioral dysfunction. J Neurosci 23:622– 631.
Nurmi EL, Amin T, Olson LM, et al. 2003. Dense 2002. Mice with truncated MeCP2 recapitu-
Powell EM, Mars WM, Levitt P. 2001. Hepatocyte
linkage disequilibrium mapping in the 15q11- late many Rett syndrome features and display
growth factor/scatter factor is a motogen for
q13 maternal expression domain yields evi- hyperacetylation of histone H3. Neuron 35:
interneurons migrating from the ventral to
dence for association in autism. Mol Psychi- 243–254.
dorsal telencephalon. Neuron 30:79 – 89.
atry 8:570. Shao Y, Cuccaro ML, Hauser ER, et al. 2003. Fine
Purcell AE, Jeon OH, Zimmerman AW, et al. mapping of autistic disorder to chromosome
Pattyn A, Goridis C, Brunet JF. 2000. Specification
2001. Postmortem brain abnormalities of the 15q11-q13 by use of phenotypic subtypes.
of the central noradrenergic phenotype by the glutamate neurotransmitter system in autism.
homeobox gene Phox2b. Mol Cell Neurosci Am J Hum Genet 72:539 –548.
Neurology 57:1618 –1628. Sherr EH. 2003. The ARX story (epilepsy, mental
15:235–243. Rakic P. 1972. Mode of cell migration to the
Pattyn A, Morin X, Cremer H, et al. 1999. The retardation, autism, and cerebral malforma-
superficial layers of fetal monkey neocortex. tions): One gene leads to many phenotypes.
homeobox gene Phox2b is essential for the J Comp Neurol 145:61– 83.
development of autonomic neural crest deriv- Curr Opin Pediatr 15:567–571.
Ramanathan S, Woodroffe A, Flodman PL, et al. Shieh PB, Ghosh A. 1997. Neurotrophins: New
atives. Nature 399:366 –370. 2004. A case of autism with an interstitial
Pattyn A, Simplicio N, Van Doorninck JH, et al. roles for a seasoned cast. Curr Biol 7:R627–
deletion on 4q leading to hemizygosity for R630.
2004. Ascl1/Mash1 is required for the devel- genes encoding for glutamine and glycine
opment of central serotonergic neurons. Nat Sidman RL, Rakic P. 1973. Neuronal migration,
neurotransmitter receptor sub-units with special reference to developing human
Neurosci 7:589 –595. (AMPA2, GLRA3, GLRB) and neuropeptide brain: A review. Brain Res 62:1–35.
Perry EK, Lee ML, Martin-Ruiz CM, et al. 2001. receptors NPY1R, NPY5R. BMC Med Simon HH, Bhatt L, Gherbassi D, et al. 2003.
Cholinergic activity in autism: Abnormalities Genet 5:10. Midbrain dopaminergic neurons: Determina-
in the cerebral cortex and basal forebrain. Ramoz N, Reichert JG, Smith CJ, et al. 2004. tion of their developmental fate by transcrip-
Am J Psychiatry 158:1058 –1066. Linkage and association of the mitochondrial tion factors. Ann N Y Acad Sci 991:36 – 47.
Persico AM, D’Agruma L, Zelante L, et al. 2004. aspartate/glutamate carrier SLC25A12 gene Smidt MP, Smits SM, Burbach JP. 2003. Molecular
Enhanced APOE2 transmission rates in fam- with autism. Am J Psychiatry 161:662– 669. mechanisms underlying midbrain dopamine
ilies with autistic probands. Psychiatr Genet Raymond GV, Bauman ML, Kemper TL. 1996. neuron development and function. Eur
14:73– 82. Hippocampus in autism: A Golgi analysis. J Pharmacol 480:75– 88.
Persico AM, Militerni R, Bravaccio C, et al. 2000. Acta Neuropathol (Berl) 91:117–119. Roscj Smith M, Filipek PA, Wu C, et al. 2000. Analysis
Lack of association between serotonin trans- M. Spiker D, Lotspeich L, et al. 1999. A of a 1-megabase deletion in 15q22-q23 in an
porter gene promoter variants and autistic dis- genomic screen of autism: evidence for a mul- autistic patient: identification of candidate
order in two ethnically distinct samples. Am J tilocus etiiology. Am J Hum Genet 65:493– genes for autism and of homologous DNA
Med Genet 96:123–127. 507. segments in 15q22-q23 and 15q11-q13. Am J
Persico AM, Pascucci T, Puglisi-Allegra S, et al. Risch N, Spiker D, Lotspeich L, et al. 1999. A Med Genet 96:765–770.
2002. Serotonin transporter gene promoter genomic screen of autism: evidence for a mul- Song JY, Ichtchenko K, Sudhof TC, et al. 1999.
variants do not explain the hyperserotonine- tilocus etiology. Am J Hum Genet 65:493– Neuroligin 1 is a postsynaptic cell-adhesion
mia in autistic children. Mol Psychiatry 507. molecule of excitatory synapses. Proc Natl
7:795– 800. Robinson PD, Schutz CK, Macciardi F, et al. 2001. Acad Sci U S A 96:1100 –1105.
Pescucci C, Meloni I, Bruttini M, et al. 2003. Genetically determined low maternal serum Sparks BF, Friedman SD, Shaw DW, et al. 2002.
Chromosome 2 deletion encompassing the dopamine beta-hydroxylase levels and the eti- Brain structural abnormalities in young chil-
MAP2 gene in a patient with autism and ology of autism spectrum disorders. Am J dren with autism spectrum disorder. Neurol-
Rett-like features. Clin Genet 64:497–501. Med Genet 100:30 –36. ogy 59:184 –192.

316 MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER
Spitzer NC, Root CM, Borodinsky LN. 2004. Tuchman R. 2003. Autism. Neurol Clin 21:915– Wassink TH, Piven J, Vieland VJ, et al. 2001.
Orchestrating neuronal differentiation: Pat- 932, viii. Evidence supporting WNT2 as an autism sus-
terns of Ca(2⫹) spikes specify transmitter Veenstra-VanderWeele J, Anderson GM, Cook ceptibility gene. Am J Med Genet 105:406 –
choice. Trends Neurosci 27:415– 421. EH Jr. 2000. Pharmacogenetics and the sero- 413.
Steg JP, Rapoport JL. 1975. Minor physical anom- tonin system: Initial studies and future direc- Whitaker-Azmitia PM. 2001. Serotonin and brain
alies in normal, neurotic, learning disabled, tions. Eur J Pharmacol 410:165–181. development: Role in human developmental
and severely disturbed children. J Autism Veenstra-Vanderweele J, Cook E Jr., Lombroso PJ. diseases. Brain Res Bull 56:479 – 485.
Child Schizophr 5:299 –307. 2003. Genetics of childhood disorders: XLVI. Wichterle H, Turnbull DH, Nery S, et al. 2001. In
Stevenson RE, Schroer RJ, Skinner C, et al. 1997. Autism, part 5: Genetics of autism. J Am Acad utero fate mapping reveals distinct migratory
Autism and macrocephaly. Lancet 349(9067): Child Adolesc Psychiatry 42:116 –118. pathways and fates of neurons born in the
1744 –1745. Veenstra-VanderWeele J, Cook EH. 2004. Molec- mammalian basal forebrain. Development
Stokstad E. 2001. Development: New hints into ular genetics of autism spectrum disorder. 128:3759 –3771.
the biological basis of autism. Science 294: Mol Psychiatry 9:819 – 832. Wodarz A, Nusse R. 1998. Mechanisms of Wnt
34 –37. Veenstra-VanderWeele J, Kim SJ, Lord C, et al. signaling in development. Annu Rev Cell
Sultana R, Yu CE, Yu J, et al. 2002. Identification 2002. Transmission disequilibrium studies of Dev Biol 14:59 – 88.
of a novel gene on chromosome 7q11.2 in- the serotonin 5-HT2A receptor gene Wurst W, Auerbach AB, Joyner AL. 1994. Multi-
terrupted by a translocation breakpoint in a (HTR2A) in autism. Am J Med Genet 114: ple developmental defects in Engrailed-1 mu-
pair of autistic twins, Genomics 80:129 –134. 277–283. tant mice: An early mid-hindbrain deletion
Tallon-Baudry C, Bertrand O. 1999. Oscillatory
Veltman MW, Thompson RJ, Roberts SE, et al. and patterning defects in forelimbs and ster-
gamma activity in humans and its role in object
2004. Prader-Willi Syndrome—a study com- num. Development 120:2065–2075.
representation. Trends Cogn Sci 3:151–162.
paring deletion and uniparental disomy cases Ye W, Bouchard M, Stone D, et al. 2001. Distinct
Tamamaki N, Fujimori KE, Takauji R. 1997. Or-
with reference to autism spectrum disorders. regulators control the expression of the mid-
igin and route of tangentially migrating neu-
Eur Child Adolesc Psychiatry 13:42–50. hindbrain organizer signal FGF8. Nat Neuro-
rons in the developing neocortical intermedi-
ate zone. J Neurosci 17:8313– 8323. Vervoort VS, Beachem MA, Edwards PS, et al. sci 4:1175–1181.
Thomas SA, Marck BT, Palmiter RD, et al. 1998. 2002. AGTR2 mutations in X-linked mental Ye W, Shimamura K, Rubenstein JL, et al. 1998.
Restoration of norepinephrine and reversal of retardation. Science 296:2401–2403. FGF and Shh signals control dopaminergic
phenotypes in mice lacking dopamine beta- Vogel-Hopker A, Rohrer H. 2002. The specifica- and serotonergic cell fate in the anterior neu-
hydroxylase. J Neurochem 70:2468 –2476. tion of noradrenergic locus coeruleus (LC) ral plate. Cell 93:755–766.
Thomas SA, Matsumoto AM, Palmiter RD. 1995. neurones depends on bone morphogenetic Yirmiya N, Pilowsky T, Nemanov L, et al. 2001.
Noradrenaline is essential for mouse fetal de- proteins (BMPs). Development 129:983–991. Evidence for an association with the serotonin
velopment. Nature 374:643– 646. Volkmar FR. 2001. Pharmacological interventions transporter promoter region polymorphism
Thomas NS, Sharp AJ, Browne CE, et al. 1999. Xp in autism: Theoretical and practical issues. and autism. Am J Med Genet 105:381–386.
deletions associated with autism in three fe- J Clin Child Psychol 30:80 – 87. Yu X, Malenka RC. 2003. Beta-catenin is critical
males. Hum Genet 104:43– 48. Waage-Baudet H, Lauder JM, Dehart DB, et al. for dendritic morphogenesis. Nat Neurosci
Tordjman S, Gutknecht L, Carlier M, et al. 2001. 2003. Abnormal serotonergic development in 6:1169 –1177.
Role of the serotonin transporter gene in the a mouse model for the Smith-Lemli-Opitz Zhou CJ, Zhao C, Pleasure SJ. 2004. Wnt signaling
behavioral expression of autism. Mol Psychi- syndrome: Implications for autism. Int J Dev mutants have decreased dentate granule cell
atry 6:434 – 439. Neurosci 21:451– 459. production and radial glial scaffolding abnor-
Trottier G, Srivastava L, Walker CD. 1999. Etiol- Wassink TH, Brzustowicz LM, Bartlett CW, Szat- malities. J Neurosci 24:121–126.
ogy of infantile autism: A review of recent mari P. 2004. The search for autism disease Zoghbi HY. 2003. Postnatal neurodevelopmental
advances in genetic and neurobiological re- genes. Ment Retard Dev Disabil Res Rev disorders: Meeting at the synapse? Science
search. J Psychiatry Neurosci 24:103–115. 10:272–283. 302:826 – 830.

MRDD RESEARCH REVIEWS ● DEVELOPMENTAL NEUROBIOLOGY OF AUTISM ● POLLEUX AND LAUDER 317

You might also like