Sie sind auf Seite 1von 10

Clinical Review & Education

JAMA Psychiatry | Review

The Emerging Clinical Neuroscience


of Autism Spectrum Disorder
A Review
Rebecca A. Muhle, MD, PhD; Hannah E. Reed, MD; Katharine A. Stratigos, MD; Jeremy Veenstra-VanderWeele, MD

Related article
IMPORTANCE Autism spectrum disorder (ASD) is a highly prevalent disorder, and community
psychiatrists are likely to treat many individuals with ASD during their clinical practice. This
clinical case challenge describes a routine evaluation of irritability and self-injury in a
preschool-aged child who meets the criteria for ASD. The case also illustrates the importance
of known risk factors for ASD, such as chromosomal deletion and prematurity. This clinical
neuroscience article seeks to educate the clinician of current avenues of research that can
inform and may already affect clinical practice for this patient, while providing a preview of
research that may yield biological treatments for ASD within the next decade.

OBSERVATIONS A diagnosis of ASD is defined behaviorally; therefore, many genetic and


environmental risk factors, working singly or in concert, are linked to ASD. The prenatal
period of brain development is particularly sensitive to risk factors such as gene mutation or
drug exposure that affect brain development and circuitry formation. Currently,
neuroimaging researchers can detect changes in brain connectivity of children with ASD as
young as 6 months, followed by an atypical trajectory of brain development through
preschool age and ongoing connectivity inefficiencies across the lifespan. Animal and cellular
Author Affiliations: Child Study
model systems have provided a means for defining the molecular and cellular changes
Center, Yale School of Medicine, New
associated with risk factors for ASD. The ability to connect specific treatments to particular Haven, Connecticut (Muhle);
subgroups of people with ASD is the defining hope of precision medicine initiatives. Department of Psychiatry, Columbia
University, New York, New York
(Reed, Stratigos,
CONCLUSIONS AND RELEVANCE The advent of next-generation sequencing technology,
Veenstra-VanderWeele); New York
advanced imaging techniques, and cutting-edge molecular techniques for modeling ASD has State Psychiatric Institute, New York
allowed researchers to define ASD risk-related biological pathways and circuits that may, for (Reed, Stratigos,
the first time, unify the effects of disparate risk factors into common neurobiological Veenstra-VanderWeele); Center for
Autism and the Developing Brain,
mechanisms. The path from these mechanisms to biological treatments that improve the NewYork-Presbyterian Hospital, New
lives of individuals with autism remains unclear, but the cumulative output of multiple lines of York, New York (Reed, Veenstra-
research suggests that subtyping by genetic risk factors may be a particularly tractable way to VanderWeele).
capitalize on individual differences amenable to specific treatments. Corresponding Author: Jeremy
Veenstra-VanderWeele, MD,
Department of Psychiatry, Columbia
JAMA Psychiatry. doi:10.1001/jamapsychiatry.2017.4685 University, 1051 Riverside Dr, Mail
Published online March 28, 2018. Unit 78, New York, NY 10032 (jeremy
.veenstra@nyspi.columbia.edu).

T
he Clinical Challenge in this issue of JAMA Psychiatry occurringsymptoms.ThiscaseillustrateswhereweareinthearcofASD
describes a young boy who presented with irritability and assessment and care: at the cusp of incorporating an understanding
self-injury in the context of developmental delay, social of autism neurobiology into treatment, but not quite there yet.
deficits, and restricted, repetitive behaviors.1 Evaluation confirmed Parallelingrapidadvancesinautismresearch,wehaveseenasurge
a diagnosis of autism spectrum disorder (ASD), along with intellectual in the recognition and diagnosis of ASD, and current prevalence rates
and language impairments per DSM-5 criteria (Table 1). 16p11.2 Dele- are estimated at 1% to 2%, with 4 to 5 males receiving a diagnosis for
tion syndrome can also be added to the diagnosis, to indicate relevant every female who receives a diagnosis of ASD. This surge, coupled with
neurodevelopmental and medical co-occurring conditions. However, fraudulent research, led to concerns that vaccines could contribute to
this molecular genetic diagnosis did not change the process of evalu- the risk of ASD, which has clearly been disproven.2 Most data instead
ating him in the psychiatric clinic, and there are currently no pharma- point to increased awareness and resources for screening, diminished
cologic treatment recommendations based on genotype. Research- stigma, and changes in diagnostic criteria as explanations of today’s
ers are just beginning to test treatments for core symptoms based on higher recognition of ASD. A few known risk factors, such as advanced
biologicalfindingsinsubgroupsofchildrenwithASD,andcurrenttreat- parental age and surviving extreme preterm birth, are also increasing
ments therefore remain either behavioral or applied only to co- in parallel with ASD prevalence.

jamapsychiatry.com (Reprinted) JAMA Psychiatry Published online March 28, 2018 E1

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


Clinical Review & Education Review The Emerging Clinical Neuroscience of Autism Spectrum Disorder

cations for screening. Third, we describe the use of model systems,


Table 1. DSM-5 Criteria for Autism Spectrum Disorder (299.00 [F84.0])
from fish to mice to human cells, to illuminate the neurobiology that
Restricted Interests, Repetitive
Social Communication Domain Behavior Domain
connects genes to behavioral outcomes. Finally, we highlight the
Manifested by all of the following Manifested by at least 2 of the promise of precision medicine that could emerge from an under-
(currently or by history): following (currently or by history): standing of neurobiology.
Deficits in social-emotional Stereotyped or repetitive motor
reciprocity movements, use of objects, or
speech
Deficits in nonverbal communicative Insistence on sameness,
behaviors used for social interaction inflexible adherence to routines, Theme 1: Risk Factors for ASD
or ritualized patterns of verbal
or nonverbal behavior In1943,thefirstdescriptionbyKanner3 ofautisticdisturbancesincluded
Deficits in developing, maintaining, Highly restricted, fixated speculationaboutgeneticinheritance,butearlypsychoanalytichypoth-
and understanding relationships interests that are abnormal in
intensity or focus eses focused on the early social environment as a cause for autism, in-
Hyper- or hyporeactivity to cludingthefallacyofthe“refrigeratormother.”4 Inthe1970s,twinstud-
sensory input or unusual ies identified genetics as a major contributor to the risk of autism.5
interest in sensory aspects of
the environment Within the heterogeneous spectrum of ASD, data point to both genetic
Level 3: requiring very substantial and environmental risk factors that contribute to risk. In the described
support case, the patient has a genetic diagnosis of 16p11.2 microdeletion and
Severe deficits in verbal and Inflexibility of behavior, extreme
nonverbal social communication skills difficulty coping with change, or
a birth history of prematurity, risk factors that both likely contribute to
cause severe impairments in other restricted or repetitive his presentation and diagnosis of ASD.
functioning, very limited initiation of behaviors markedly interfere with
social interactions, and minimal functioning in all spheres; great
response to social overtures from distress or difficulty changing focus Genetic Risk Factors
others or action
Familystudiesindicatethatsharedgeneticriskfactorsincreasethelike-
Level 2: requiring substantial support
lihoodofautism.Identicaltwinshaveautismconcordanceratesof50%
Marked deficits in verbal and Inflexibility of behavior, difficulty
nonverbal social communication coping with change, or other to80%inmoststudies,indicatingstrongheritabilitybutalsothatother
skills, social impairments apparent restricted and repetitive behaviors factors determine whether ASD develops.6-9 Fraternal twins have up
even with supports in place, limited appear frequently enough to be
initiation of social interactions, and obvious to the casual observer and to 30% concordance,6,7,10 whereas the overall rate of recurrence be-
reduced or abnormal responses to interfere with functioning in a tween siblings is approximately 25%.7,8,11 Sibling recurrence increases
social overtures from others variety of contexts; distress and/or
difficulty changing focus or action to 50% if there are 2 or more siblings with ASD or if any affected sib-
Level 1: requiring support ling is female.12,13
Without supports in place, deficits in Inflexibility of behavior causes Common variants (those present in >1% of the population) con-
social communication cause significant interference with tribute the majority of ASD risk at a population level,14,15 but individual
noticeable impairments; difficulty functioning in 1 or more contexts;
initiating social interactions and clear difficulty switching between commonvariantscontributeonlyasmallportionoftheoverallrisk,sug-
examples of atypical or unsuccessful activities; problems of organization gesting that they must act in concert. Genome-wide association is a
responses to social overtures of and planning hamper independence
others; may appear to have decreased powerful method to link common variation in specific genetic regions
interest in social interactions
to the risk of ASD, but these studies require large sample sizes to re-
Symptoms cause clinically significant impairment in social, occupational, or
other important areas of current functioning producibly link risk to specific common variants.16 Additional research
Symptoms must be present in the early developmental period will be needed to understand how multiple common variants interact
These disturbances are not better explained by intellectual disability with each other and with unique environmental exposures to result in
(intellectual developmental disorder) or global developmental delay; to make an outcome of ASD.
comorbid diagnoses of autism spectrum disorder and intellectual disability,
social communication should be below that expected for general Rapidprogresshasbeenmadeinthediscoveryofraregeneticvari-
developmental level ants (present in <1% of the population) that are associated with the risk
Specifiers of ASD (Figure 1). These genetic variants are likely rare owing to det-
Intellectual impairment rimental biological effects that affect the carrier’s reproductive fitness.
Language impairment Rare genetic syndromes (eg, fragile X syndrome and tuberous sclero-
Known genetic, medical, or sis) have long been known to contribute to the risk of ASD17 and can
environmental factor
arise spontaneously (de novo) in the germline. Researchers can now
Associated with another
neurodevelopmental, mental, or compare the genetic information from a child with ASD with the ge-
behavioral disorder netic information from his or her unaffected family members using se-
Catatonia quencing and microarray techniques that survey the entire genome to
identify de novo variants at large scale. De novo variants present only
Here, we focus on a few themes in ASD research that are either in the child with ASD are more likely to contribute to the ASD diagno-
already affecting clinical practice or are likely to affect clinical prac- sis and can be more confidently associated with ASD risk. Research-
tice within the next 5 to 10 years, with an emphasis on research rel- ers first assessed the entire genome for rare de novo microdeletions
evant to 16p11.2 deletion syndrome. First, we discuss the growing and microduplications (copy number variants [CNVs]) using chromo-
understanding of the risk of ASD, both genetic and environmental, somal microarrays. De novo CNVs were detected at a higher frequency
and how identification of risk factors can guide clinical care. Sec- in participants with ASD than in their unaffected siblings, and several
ond, we outline emerging data on neurodevelopmental changes as- recurrent CNVs have been linked to ASD risk,18-22 including 16p11.2
sociated with ASD that support the early onset of ASD, with impli- (Table 220,23-49).

E2 JAMA Psychiatry Published online March 28, 2018 (Reprinted) jamapsychiatry.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


The Emerging Clinical Neuroscience of Autism Spectrum Disorder Review Clinical Review & Education

Figure 1. Genetic Risk Variants Associated With the Development of ASD

A Inherited genetic risk variants De novo genetic risk variants

ASD ASD ASD

Common and/or rare variants De novo variant

Genetic variants associated with autism spectrum disorder (ASD) Clinical examples Genetic test

Aneuploidy Trisomy Trisomy 21 Cytogenetics


(karyotyping)
Normal Sex chromosome
aneuploidy Chromosomal
Monosomy (XXY, XXYY) microarray

Copy number variant (CNV) 16p11.2 Deletion Chromosomal


syndrome microarray
Deletion Normal Duplication
16p11.2 Duplication Fluorescence in situ
syndrome hybridization (FISH)

(see Table 2 for


other examples)

Coding Fragile X syndrome PCR fragment


Trinucleotide repeat expansion Untranslated region region Intron
FMR1 gene length analysis
Normal <50 CGG repeats Fragile XE syndrome

Premutation carrier 50-200 CGG repeats

Affected >200 methylated CGG repeats

Nucleotide insertion and/or deletion (indel) Single-nucleotide variant (SNV) ASD associated with Whole exome
CHD8 mutation sequencing
Normal Normal
ASD with Targeted gene
DNA DNA macrocephaly sequencing by PCR
associated with
Protein Protein
PTEN mutation

Insertion Indel Substitution SNV (see Table 3 for


other examples)
DNA DNA

Protein STOP Protein STOP


Premature stop codon Premature stop codon

A, Pedigrees of people affected with autism spectrum disorder (ASD) and their families. Left, pedigree of a family demonstrating multiple inherited genetic risk factors in
the affected children of 2 nonaffected parents. Right, pedigree of a family showing the development of a new mutation in the offspring that contributed to the develop-
ment of ASD in that child. B, Large- and small-scale genetic changes associated with increased risk of ASD. The different classes of genetic variants are presented by size,
from large-scale alterations (entire chromosomes or large regions missing or duplicated) at the top, to substitutions of just 1 DNA nucleotide at the bottom. Examples of
DNA sequence changes in the coding region of the CHD8 gene associated with ASD are presented in the bottom panel. Sequence corresponds to chromosome 14 (hg19),
21 878 138 to 21 878 130 base pairs (left) and 21 870 179 to 21 870 160 base pairs (right). Variants may be inherited from a carrier parent or develop de novo in the person
with ASD. Variants that are more damaging to biological function are less likely to be inherited and may interact with other genetic, environmental, and developmental risk
factors to yield a clinical diagnosis of ASD. Single-nucleotide polymorphisms (SNPs) (not depicted) are common variants that contribute to ASD risk, but at this time, indi-
vidual SNPs have not been linked to risk for ASD to justify clinical screening. Whole-genome sequencing detects copy number and nucleotide sequence variants, but it is not
yet clinically available for molecular genetic diagnostic testing owing to increased expense and data storage limitations. PCR indicates polymerase chain reaction.

jamapsychiatry.com (Reprinted) JAMA Psychiatry Published online March 28, 2018 E3

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


Clinical Review & Education Review The Emerging Clinical Neuroscience of Autism Spectrum Disorder

Table 2. Recurrent De Novo Copy Number Variants (CNVs) Associated With Autism Spectrum Disordera

CNV Locus Copy Number Change Other Associated Conditions Syndrome Name
1q21.1 Duplication ID, SCZ, ADHD, seizures, macrocephaly, CHD, scoliosis, and short 1q21.1 Duplication syndrome
stature23,24
2p16.3 Deletion ID, speech delay, SCZ, seizures, macrocephaly, dysmorphology, CHD, and 2p16.3 Deletion syndrome
hypotonia25-27
3q29 Deletion ID, speech delay, SCZ, anxiety disorder, dysmorphology, dental 3q29 Microdeletion syndrome
abnormalities, FTT, gastrointestinal problems, CHD, and recurrent ear
infections28,29
7q11.23 Duplication ID, speech delay, ADHD, anxiety disorder, seizures, macrocephaly, Williams-Beuren syndrome
dysmorphology, CHD, and renal anomalies30-32
15q11.2-q13.1 Duplication ID, ADHD, seizures, hypotonia, CHD, and short stature33-38 15q11.2-q13.1 Duplication syndrome
15q13.2-q13.3 Deletion ID and ADHD33,39 15q13.3 Microdeletion syndrome
15q13.2-q13.3 Duplication ID, speech delay, epilepsy, urogenital anomalies, and recurrent 15q11-q13 Duplication syndrome
infections33,36
16p11.2 Deletion ID, ADHD, DCD, seizures, macrocephaly, obesity, hypotonia, and 16p11.2 Deletion syndrome
articulation abnormalities40-42
16p11.2 Duplication ID, SCZ, seizures, microcephaly, hypotonia, tremor, and articulation 16p11.2 Duplication syndrome
abnormalities40,41,43
22q11.2 Deletion ID, learning disorder, speech delay, SCZ, ADHD, anxiety disorder, Velocardiofacial syndrome, DiGeorge
dysmorphology, palate abnormalities, CHD, renal anomalies, hypocalcemia, syndrome
and recurrent infections44
22q11.2 Duplication ID, ADHD, learning disorder, hypotonia, hearing loss, palate abnormalities, 22q11.2 Microduplication syndrome
CHD, urogenital anomalies, and growth retardation45-47
22q13.3 Deletion ID, seizures, macrocephaly, dysmorphology, strabismus, CHD, renal Phelan-McDermid syndrome
anomalies, hypotonia, and scoliosis48,49
a
Abbreviations: ADHD, attention-deficit/hyperactivity disorder; CHD, congenital Copy number variants associated with autism spectrum disorder sourced from
heart disease; DCD, developmental coordination disorder; FTT, failure to thrive; Sanders et al.20
ID, intellectual disability; SCZ, schizophrenia.

More recently, geneticists have examined the protein-coding re- The patient carried a microdeletion of 16p11.2, a region that en-
gions of the genome (the exome) using whole-exome sequencing, compasses 29 genes. There are now several large clinical cohorts with
which allows detection of single-nucleotide variants. Compared with 16p11.2 deletions (del16p11.2) and duplications (dup16p11.2), and as-
their unaffected siblings, children with ASD had an elevated rate of sociations have been reported with ASD, schizophrenia, intellec-
rare, de novo, single-nucleotide variants and insertions and/or de- tual disability, and seizures.20,58-60 People carrying 16p11.2 CNVs ex-
letions that likely disrupt genes (Figure 1).20,50-53 Combined analy- hibit mirror phenotypes: deletion is associated with obesity and
sis of multiple family cohorts found that individual genes could be macrocephaly,60-63 while duplication is associated with lower weight
confidently assigned ASD risk based on being disrupted in affected and microcephaly.19,59,64 Some people with a 16p11.2 CNV do not
individuals more frequently than could be expected by chance have a clinical disorder, but performance IQ and general function-
(Table 3).20 These genes cluster into the following 2 groups: 1 en- ing may be lower compared with the general population.65,66 There
riched for proteins involved in brain function (eg, synaptic function- are likely additional genetic and environmental modifiers that ulti-
ing and neuron projection) and the other enriched for gene regula- mately dictate whether the carrier has a particular disorder and to
tion (eg, chromatin organization and transcription). Using what degree.67
computational approaches incorporating a gene expression atlas of
the human brain across the lifespan, researchers identified en- Environmental Risk Factors
riched coexpression of ASD risk genes in specific cortical regions from As noted previously, twin studies suggest that both genetic and en-
early gestation to midgestation.54,55 vironmental risk factors are associated with ASD (Figure 2). Poten-
As remarkable as progress has been, the known variants are just tial nongenetic risk factors include maternal conditions, complica-
the tip of the iceberg. Modeling predicts that approximately 200 de tions of pregnancy, medications, and exposure to toxic materials.
novo CNVs and hundreds of genes contribute to ASD risk,20,53 likely Most of these risk factors act on brain development during the pre-
interacting with environmental risk factors to result in ASD. Al- natal and perinatal stages. Establishing causal roles for environmen-
though each of these variants may be individually rare, collectively tal factors is complicated by retrospective reporting and overlap-
they are found in a large minority of the clinical population with ASD. ping risk factors, but several nongenetic risk factors for ASD have
Currently, medical associations recommend molecular diagnostic now been established.
testing using chromosomal microarray and fragile X testing in all The most well-supported environmental risk factor for ASD is
people diagnosed as having ASD, and adjunctive clinical whole- advanced parental age,68-71 with most evidence suggesting that this
exome sequencing may be helpful in those with congenital abnor- contributes to genetic risk. Maternal and paternal age appear to be
malities or unexplained co-occurring medical conditions.56,57 The independent risk factors, leading to an 18% (for mothers) and 21%
identification of a molecular diagnosis can provide clinical care guide- (for fathers) increase in ASD risk for every 10-year increase in the age
lines and may warn of potential comorbidities that would other- of the parent.71 The risk of chromosomal abnormalities is well known
wise go unmonitored. in mothers of advanced maternal age, and prenatal genetic screen-

E4 JAMA Psychiatry Published online March 28, 2018 (Reprinted) jamapsychiatry.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


The Emerging Clinical Neuroscience of Autism Spectrum Disorder Review Clinical Review & Education

Table 3. High-Confidence Autism Spectrum Disorder Risk Genesa


Gene OMIM Cytogenetic Molecular Function or
Symbol Identifier Location Biological Processb Associated Syndromesc
ADNP 611386 20q13.13 Transcription Helsmoortel-van der Aa syndrome
ANK2 106410 4q25-q26 Protein transport Cardiac arrhythmia, ankyrin-B–related
ARID1B 614556 6q25.3 Transcription Coffin Siris syndrome 1
ASH1L 607999 1q22 Transcription Autosomal dominant mental retardation 52
ASXL3 615115 18q12.1 Transcription Bainbridge-Ropers syndrome
a
CHD8 610528 14q11.2 Transcription Autism, susceptibility to, 18 Genes harboring multiple, rare,
CUL3 603136 2q36.2 Transport Pseudohypoaldosteronism, type IIE single-gene mutations that are
scored as 1 (high confidence) by a
DSCAM 602523 21q22.2 Cell adhesion None Simons Foundation Autism
DYRK1A 600855 21q22.13 Protein kinase Mental retardation, autosomal dominant 7 Research Initiative gene scoring
GRIN2B 138252 12p13.1 Ion transport Early infantile epileptic encephalopathy 27 advisory panel (https://gene.sfari
.org). This is an iteratively curated
KATNAL2 614697 18q21.1 Hydrolase None list of >800 human genes with
KMT2A 159555 11q23.3 Transcription Wiedemann-Steiner syndrome genetic, clinical, or functional
KMT5B 610881 11q13.2 Transcription Mental retardation, autosomal dominant 51 evidence of association with risk of
autism spectrum disorder, with
MYT1L 613084 2p25.3 Transcription Mental retardation, autosomal dominant 39 highest ranking reserved for genes
NAA15 608000 4q31.1 Transcription Mental retardation, autosomal dominant 50 with independently verified,
POGZ 614787 1q21.3 Cell cycle White-Sutton syndrome genome-wide statistical significance
between cases and controls.
PTEN 601728 10q23.31 Apoptosis Cowden syndrome I, macrocephaly/autism syndrome b
Molecular function or biological
RELN 600514 7q22.1 Cell adhesion Lissencephaly 2 (Norman-Roberts type) process keywords were retrieved
SCN2A 182390 2q24.3 Ion transport Epileptic encephalopathy, early infantile, 11 for each gene from Uniprot (http:
//www.uniprot.org).
SETD5 615743 3p25.3 Transcription Mental retardation, autosomal dominant 23
c
Associated syndromes were
SHANK3 606230 22q13.33 Axon guidance Phelan-McDermid syndrome
retrieved for each gene from OMIM
SYNGAP1 603384 6p21.32 Dendrite development Mental retardation, autosomal dominant 5 (http://omim.org). Mental
TBR1 604616 2q24.2 Transcription None retardation is a retired term that is
now currently described as
TRIP12 604506 2q36.3 DNA repair Mental retardation, autosomal dominant 49
intellectual disability.

ing is routinely offered in this population. Older fathers contribute Exogenous risk factors, such as medication exposure during preg-
more de novo mutations as they age.50,52,72 The total elevated risk nancy, also play a role in the risk of ASD. The prenatal use of anti-
is likely owing to other factors as well, including an increase in com- convulsants has been associated with an increased risk of ASD, with
plications of pregnancy, epigenetic modifications of the genome, and valproic acid use during pregnancy showing the clearest
social factors that may contribute to having children later in life.71 association.70,77 Asthma medications, particularly β2-adrenergic re-
Prematurity, even up to 37 weeks, is another well-established ceptor agonists, have also been linked to autism risk.70 Although still
risk factor for ASD.70,73 The preterm brain has vulnerability to cer- controversial, there is emerging evidence to support prenatal ex-
tain forms of gray and white matter brain injury that affect subcor- posure to air pollution as a risk factor for ASD.70,71
tical and cortical connectivity (theme 2); however, preterm birth is Environmental risk factors for ASD have significant implica-
associated with ASD even in the absence of overt neuronal injury.74 tions for public policy and prenatal health care. Discerning the mecha-
The elevated risk of ASD is likely compounded by prematurity- nisms behind environmental risks may be particularly difficult, how-
associated complications, such as anoxia and hypoglycemia. The risk ever, because they may act directly on neurons and supporting cells
for ASD is greatest in extremely preterm infants. In those with very or may act indirectly by influencing hormones or inflammatory re-
low birth weight and extreme preterm birth, ASD is more likely to sponses. Alternatively, environmental risks may act by altering the
be associated with major cognitive and motor impairments.75 In the expression of relevant genes that affect ASD risk. Despite the diver-
case presented, the patient was born at a gestational age of 32 weeks, sity of genetic and environmental risk factors in ASD, it is important
a time when the brain is still in the migration phase and has begun to understand how they may interact and lead to downstream ef-
synaptic pruning and myelination (Figure 2). This child therefore ap- fects on brain development, and thereby indicate common path-
pears to have 2 independent risk factors for ASD. ways underlying the core deficits of ASD.
Dozens of other environmental factors have been linked with
ASD, although further evidence is needed to consider them as es-
tablished risk factors for ASD. These risk factors can generally be cat-
Theme 2: Altered Brain Development in ASD
egorized as either endogenous, originating from within the maternal-
fetal unit, or exogenous, originating from outside of the mother.76 Neuroimaging studies increasingly support a picture of early atypi-
The endogenous risk factors with the best support, outside of pre- cal brain development and widespread alterations in neural con-
maturity, are short-interval pregnancies, defined as pregnancies oc- nectivity in individuals with ASD. Some of the most promising find-
curring fewer than 12 months apart70; birth complications involv- ings are in infants and toddlers who have an older sibling with ASD,
ing hypoxia and ischemia or trauma; maternal obesity; and diabetes.71 and are therefore at increased risk for ASD. Prospective neuroim-

jamapsychiatry.com (Reprinted) JAMA Psychiatry Published online March 28, 2018 E5

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


Clinical Review & Education Review The Emerging Clinical Neuroscience of Autism Spectrum Disorder

Figure 2. Risk Factors in ASD and Timeline of Brain Development

A Risk factors associated with autism spectrum disorder (ASD)

Advanced parental age Maternal obesity and diabetes Premature birth


Short-interval pregnancy Prenatal exposure to certain medications Birth complications
(<12 months apart) (anticonvulsants; β2-adrenergic receptor agonists) (hypoxia; trauma)
Mutations in ASD risk genes (see Figure 1)

B Timeline of brain development and alterations in ASD


Prenatal Postnatal
Normal brain development A, The majority of known risk factors
Neurulation for autism spectrum disorder (ASD)
Neural proliferation occur primarily before conception or
during prenatal development. B, The
Neural apoptosis
earliest published data on brain
Neural migration
development in ASD are at 6 months
Synaptogenesis of age. These data indicate that, by at
Synaptic pruning least that time point, both brain
Myelination volume and connectivity are affected.
It is presumed that these changes
Peak expression of ASD risk genes begin earlier in development,
Genes involved in chromatin organization although the exact timing and
and transcription regulation mechanisms are unknown. Changes
Genes involved in synaptic function in brain development in ASD likely
and neuronal projection begin during the prenatal period,
when many ASD risk genes have peak
Alterations in brain development in ASD
expression levels. It is further
Abnormal neural apoptosis (hypothesized) ? hypothesized that brain development
Abnormal synaptic pruning ? in ASD may involve abnormal
(hypothesized) synaptic pruning and/or neural
Brain volume enlargement
apoptosis during this time. If
Atypical brain connectivity appropriate pruning and/or apoptosis
fail to occur early in development,
First trimester Second trimester Third trimester Year 1 Year 2 Year 3 Year 4 Year 5 this may manifest later as atypical
Birth Early behavioral Median age at connectivity and brain volume
changes ASD diagnosis enlargement, particularly once
myelination begins.

aging studies consistently point to abnormal trajectories of brain de- owing to a surplus of neurons that did not undergo typical apoptosis
velopment, rather than static changes or localized lesions. Effec- and pruning earlier in development.
tive brain development depends on the balance between cellular and From the ages of 2 to 4 years, brain volumes in children with ASD
synaptic growth and appropriately timed pruning of neurons and syn- remain enlarged compared with those of their peers.85,86 The amyg-
apses. In at least some children with ASD, this balance appears to dala, frontal cortex, and temporal cortex, which are important for
be disrupted. social cognition, language, and regulation of emotions, are particu-
Neuroimaging studies find that changes occur well before the larly affected.79,85 By school age, brain growth has slowed, with brain
onset of clear symptoms of ASD. The association of risk with pre- volumes of typically developing children catching up to those in chil-
natal exposures suggests that many of these disruptions begin dur- dren with ASD.85,86 Connectivity studies find ongoing disruptions
ing the prenatal period. Recent neuroimaging studies have exam- in how brain regions communicate throughout school age, adoles-
ined brain development in high-risk infants from 6 months of age, cence, and adulthood. A large pooled analysis of resting-state data
suggesting that infants who will later develop ASD already exhibit from functional magnetic resonance imaging performed in individu-
abnormal connectivity by this time.78-81 Connections involved in low- als with ASD found a predominance of hypoconnectivity of long-
level sensory processing appear particularly affected at this early range cortical-cortical and interhemispheric projections compared
stage.81 In some studies, the degree of atypical connectivity at 6 with age-matched typical controls.87 In contrast, subcortical re-
months correlates with future symptom severity.79,81 gions exhibited hyperconnectivity of local connections. As a whole,
Starting between the ages of 6 and 12 months, children who will these data suggest that higher-order brain functions requiring com-
go on to have ASD show an expansion of cortical surface area, starting munication between separate brain regions are depressed in favor
insensorydomainsunderlyingauditoryandvisualprocessing,followed of local circuits that may be overactive and difficult to disrupt.
by more global overgrowth from the ages of 12 to 24 months.82,83 This The 16p11.2 microdeletion illustrates the strong influence that
trajectory mirrors typical development, but in an exaggerated manner genetic variants can have on brain development. Individuals with
(Figure 2). In typical development, brain growth during this period is a deletion (1 copy of this region) show significantly larger brain
largely attributed to the expansion of glia and myelination.84 Similarly, volumes than do unaffected individuals who carry 2 copies.88
in individuals with ASD, this observed overgrowth likely does not rep- Similar to results from general studies of ASD, both gray matter
resent new neuron development, although it could reflect overgrowth and white matter are affected by 16p11.2 deletion, particularly in

E6 JAMA Psychiatry Published online March 28, 2018 (Reprinted) jamapsychiatry.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


The Emerging Clinical Neuroscience of Autism Spectrum Disorder Review Clinical Review & Education

regions implicated in reward, language, and social cognition.89 exhibit hyperactivity, repetitive behaviors, and learning deficits remi-
More important, these associations are seen at a group level, with niscent of the human phenotype.96-100
considerable variability across individuals. Individuals with a Animal models are also crucial in the development of poten-
duplication (3 copies) of 16p11.2 tend to have microcephaly, which tial treatments. For example, mice with del16p11.2 showed dimin-
is also associated with other ASD risk factors (eg, fetal alcohol ished long-term potentiation, similar to a mouse model of fragile
syndrome and DYRK1A [OMIM 600855] disruption) (Table 3). X syndrome.98,101 These animals were unable to acquire or extin-
This dichotomy demonstrates the heterogeneity underlying ASD. guish a memory of an unpleasant stimulus, which could be res-
Although neuroimaging research is advancing our under- cued by treatment with a glutamate receptor mGluR5 modulator.
standing of the biology of ASD, there is no evidence to support More recently, researchers used R-baclofen, a γ-aminobutyric
routine neuroimaging in autistic individuals.90,91 In 16p11.2 dele- acid–B receptor agonist, to rescue deficits in object or spatial rec-
tion syndrome, the recommendation is the same, with the caveat ognition in mice with del16p11.2.102
that individuals with symptoms suggestive of Chiari I malforma- In addition to animal models, new techniques using human
tion or syringomyelia, conditions that are more common in this cell lines can illuminate the molecular consequences of genetic
syndrome, should undergo neuroimaging.92 More research is risk variants. For example, using lymphocyte cell lines obtained
needed to better understand and standardize the trajectory of from carriers of 16p11.2 CNVs, researchers found that expression
brain development in ASD before neuroimaging could be consid- of genes within this region rose or fell with duplications or dele-
ered for a clinical role. These findings do emphasize the utility of tions, as expected, but some genes outside of the region were
early diagnostic screening, however, and early behavioral inter- also affected, potentially pointing to critical pathways that medi-
vention is critical to improve social and communication function- ate the risk of ASD.103,104 Alternatively, reprogramming cells into
ing. Fine-tuned behavioral screening, such as the use of eye track- induced pluripotent stem cells and differentiating them into indi-
ing to monitor attention to social scenes, could eventually be vidual neuronal lineages may provide better consistency with
used to identify children at risk of ASD before overt symptoms brain cells.105 These cultures have thus far been limited to early
emerge, but this finding has not yet been demonstrated in clinical stages of brain development and manifest fewer specific cell
settings.93 types, but if deficits are identified in cells derived from patients,
therapeutic interventions could be evaluated and provide test-
able targets and hypotheses for clinical research trials.106
Theme 3: Model Systems of ASD
Computational, cellular, and animal model systems can help to
dissect the molecules, cells, and circuits that connect risk factors
Future Directions and the Promise
for ASD to biology. Model systems are particularly important for
of Precision Medicine
understanding how and when these risk factors affect brain
development and lead to downstream changes in behavior. Ani- Based on available evidence, a diagnosis of ASD should be followed as
mal models provide a developmentally informed context in which soon as possible by implementation of early, intensive behavioral in-
to study the consequences of a genetic variant on brain struc- tervention,whichimprovescognitiveandadaptiveoutcomesformany
tures and cell types. Even relatively simple animals, such as (but not all) preschoolers with ASD.107 Behavioral and educational in-
zebrafish, have substantial conservation of ASD risk genes. For terventions for school-age children are tailored to individual needs but
example, of the 29 genes in the 16p11.2 CNV in humans, 24 genes have much less research support.107 Evidence-based pharmacologic
are conserved in zebrafish.94 Rodent and primate models have treatments in ASD are limited to co-occurring symptoms, such as agi-
more biological similarity to humans and may exhibit behaviors tationorhyperactivity.108 Somecliniciansandparentsalsochoosefrom
that may be more analogous to human social or repetitive behav- apanoplyoftreatmentswithnoevidence,drivenbyadesiretoaddress
ior. Researchers recently created several monkeys with disruption the root causes of ASD.109 This use of treatments with no evidence is
of the Rett syndrome gene, MECP2 [OMIM 300005], which understandable when none of our available biological treatments are
exhibited decreased social contact and more stereotyped treating the core neurobiological determinants of the disorder.
behaviors.95 Although primate models are closest in behavior and Thediscoveryofraresingle-genevariantsandCNVsraisesthepos-
biology to humans, ethical issues and their high cost have limited sibility of ASD treatments rooted in biology, rather than in behavioral
their widespread use. principlesorpharmacologicobservations.Thepresenceofaknownge-
The mouse brain is less complex than the human brain, but netic risk variant in the described child is quite likely to have tipped the
mouse models that recapitulate a human gene variant can be cost- scales toward an ASD outcome. This motivates investigations for treat-
efficient and time-saving while still providing a developmental con- ments targeted to molecular genetic diagnoses rather than to an ASD
text to observe the effect of risk factors on specific cells, tissues, and diagnosis per se. With the advance of designer gene editing technol-
organs. For example, several altered cellular mechanisms and path- ogy, it is possible that interventions could be targeted to specific ge-
ways have been identified in mice with the 16p11.2 deletion, includ- netic loci (to repair a genetic sequence or to correct aberrant gene
ing premature exiting of neuronal progenitor cells from the cell cycle activity),110 althoughthepossibilityofunintendedgeneticchanges(off-
and increased dopamine receptor expression on striatal γ-amino- target effects) is currently too large to suggest such interventions for
butyric acid–ergic neurons.96,97 While it must be stated that behav- all but the most devastating syndromes.111
ioral tests in mice may not accurately reflect symptom domains of Precision medicines in ASD-related genomic disorders will not
ASD (or human social cognition more generally), these animals did be as straightforward to test as in cancer, in which clinical response

jamapsychiatry.com (Reprinted) JAMA Psychiatry Published online March 28, 2018 E7

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


Clinical Review & Education Review The Emerging Clinical Neuroscience of Autism Spectrum Disorder

can be measured by tumor regression or normalization of cell counts. ture. Our neuroscience themes are focused on future clinical effect,
Tests of neurobiological hypotheses in developmental disorders re- rather than what makes a difference for treatment right now. For the
quire careful consideration of our standard protocols. We almost cer- boy and his family described in the Clinical Challenge,1 knowing his ge-
tainly need to study treatments at an earlier point in childhood de- nomic risk variant allows for family planning, provides insight into one
velopment, rather than binding ourselves to the standard timeline cause of his disorder, and connects the family to a network of others
of efficacy studies in adults, then in adolescents, and finally in withthesamegenomicfinding.Theidentificationofawell-definedbio-
children.112 We also need to define and refine outcome measures that logical risk factor can relieve parental self-blame and reduce the pres-
are more sensitive to change and that are informed by the experi- sure to pursue alternative treatments based on false promises to ad-
ences and voices of the autistic community.113 dress the causes of ASD. This scenario represents benefit to the patient
The DSM-5 defines ASD by behavioral symptoms and man- and his family right now, even while we wait for the development and
dates the diagnosis of separate disorders to indicate language im- implementation of precision medicines.
pairment or inattention. Unlike the DSM-5 approach, with well-
defined splitting between categories, we do not imagine that
independent biological pathways separately lead to ASD symp-
Conclusions
toms, language impairment, and inattention. Instead, it is likely that
neurodevelopmental consequences cascade from risk factors to mul- Autism spectrum disorder is not a singular disorder, but rather de-
tiple domains of difficulty, depending on the genetic or environ- scribes a common set of behavioral features that unite a very hetero-
mental insult and when it is incurred. Here, we have largely de- geneous population reflecting diverse genetic and environmental
scribed ASD as a disorder, but ASD is instead made up of many risk factors. Genetic testing, including a chromosomal microarray and
different disorders that have core ASD symptoms in common but fragile X testing, is recommended for all individuals with ASD. While
many disparate symptoms as well. we await precision medicine treatments based on neurobiology, cli-
Simultaneously,whilewethinkthatallpsychiatristsneedtobepre- nicians should use behavioral approaches for core symptoms and
pared for a future in which biological insights lead to transformative consider medications only for co-occurring symptoms in addition to
treatments, we must acknowledge that we do not yet live in that fu- an ongoing behavioral program.

ARTICLE INFORMATION 3. Kanner L. Autistic disturbances of affective stoppage from large UK ASD research family
Accepted for Publication: December 21, 2017. contact. Nerv Child. 1943;2:217-250. databases. Autism Res. 2015;8(1):73-81.

Published Online: March 28, 2018. 4. Bettelheim B. The Empty Fortress: Infantile 14. Gaugler T, Klei L, Sanders SJ, et al. Most genetic
doi:10.1001/jamapsychiatry.2017.4685 Autism and the Birth of the Self. New York, NY: risk for autism resides with common variation. Nat
The Free Press; 1967. Genet. 2014;46(8):881-885.
Author Contributions: Drs Muhle and Reed were
co-first authors and contributed equally to this 5. Folstein S, Rutter M. Infantile autism: a genetic 15. Klei L, Sanders SJ, Murtha MT, et al. Common
work. Drs Muhle and Reed had full access to all the study of 21 twin pairs. J Child Psychol Psychiatry. genetic variants, acting additively, are a major
data in the study and takes responsibility for the 1977;18(4):297-321. source of risk for autism. Mol Autism. 2012;3(1):9.
integrity of the data and the accuracy of the data 6. Bailey A, Le Couteur A, Gottesman I, et al. 16. Anney R, Klei L, Pinto D, et al. Individual
analysis. Autism as a strongly genetic disorder: evidence common variants exert weak effects on the risk for
Study concept and design: All authors. from a British twin study. Psychol Med. 1995; autism spectrum disorders. Hum Mol Genet. 2012;
Acquisition, analysis, or interpretation of data: 25(1):63-77. 21(21):4781-4792.
Muhle, Reed. 7. Ritvo ER, Freeman BJ, Mason-Brothers A, Mo A, 17. Betancur C. Etiological heterogeneity in autism
Drafting of the manuscript: All authors. Ritvo AM. Concordance for the syndrome of autism spectrum disorders: more than 100 genetic and
Critical revision of the manuscript for important in 40 pairs of afflicted twins. Am J Psychiatry. genomic disorders and still counting. Brain Res.
intellectual content: Muhle, Reed, Veenstra- 1985;142(1):74-77. 2011;1380:42-77.
VanderWeele.
Administrative, technical, or material support: 8. Sandin S, Lichtenstein P, Kuja-Halkola R, Larsson 18. Levy D, Ronemus M, Yamrom B, et al. Rare de
Muhle, Reed. H, Hultman CM, Reichenberg A. The familial risk of novo and transmitted copy-number variation in
Study supervision: Veenstra-VanderWeele. autism. JAMA. 2014;311(17):1770-1777. autistic spectrum disorders. Neuron. 2011;70(5):
9. Steffenburg S, Gillberg C, Hellgren L, et al. A twin 886-897.
Conflict of Interest Disclosures: Dr Veenstra-
VanderWeele reported serving on advisory boards study of autism in Denmark, Finland, Iceland, 19. Sanders SJ, Ercan-Sencicek AG, Hus V, et al.
or as a consultant for Roche, Novartis, and Norway and Sweden. J Child Psychol Psychiatry. Multiple recurrent de novo CNVs, including
SynapDx; receiving research funding from Roche, 1989;30(3):405-416. duplications of the 7q11.23 Williams syndrome
Novartis, Seaside Therapeutics, and Forest; and 10. Rosenberg RE, Law JK, Yenokyan G, McGready region, are strongly associated with autism. Neuron.
receiving editorial stipends from Springer and J, Kaufmann WE, Law PA. Characteristics and 2011;70(5):863-885.
Wiley. No other conflicts were reported. concordance of autism spectrum disorders among 20. Sanders SJ, He X, Willsey AJ, et al; Autism
277 twin pairs. Arch Pediatr Adolesc Med. 2009; Sequencing Consortium. Insights into autism
REFERENCES 163(10):907-914. spectrum disorder genomic architecture and
1. Muhle RA, Reed HE, Aguayo P, 11. Constantino JN, Todorov A, Hilton C, et al. biology from 71 risk loci. Neuron. 2015;87(6):
Veenstra-VanderWeele J. Evaluation and Autism recurrence in half siblings: strong support 1215-1233.
management of self-injury in a child with a rare for genetic mechanisms of transmission in ASD. Mol 21. Pinto D, Delaby E, Merico D, et al. Convergence
genomic variant [published online March 28, 2018]. Psychiatry. 2013;18(2):137-138. of genes and cellular pathways dysregulated in
JAMA Psychiatry. doi:10.1001/jamapsychiatry.2017 12. Werling DM, Geschwind DH. Recurrence rates autism spectrum disorders. Am J Hum Genet. 2014;
.4677 provide evidence for sex-differential, familial 94(5):677-694.
2. Taylor LE, Swerdfeger AL, Eslick GD. Vaccines are genetic liability for autism spectrum disorders in 22. Pinto D, Pagnamenta AT, Klei L, et al. Functional
not associated with autism: an evidence-based multiplex families and twins. Mol Autism. 2015;6:27. impact of global rare copy number variation in
meta-analysis of case-control and cohort studies. 13. Wood CL, Warnell F, Johnson M, et al. Evidence autism spectrum disorders. Nature. 2010;
Vaccine. 2014;32(29):3623-3629. for ASD recurrence rates and reproductive 466(7304):368-372.

E8 JAMA Psychiatry Published online March 28, 2018 (Reprinted) jamapsychiatry.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


The Emerging Clinical Neuroscience of Autism Spectrum Disorder Review Clinical Review & Education

23. Bernier R, Steinman KJ, Reilly B, et al; Simons individuals with features of autism and other 55. Parikshak NN, Luo R, Zhang A, et al. Integrative
VIP consortium. Clinical phenotype of the recurrent neuropsychiatric disorders. J Med Genet. 2009;46 functional genomic analyses implicate specific
1q21.1 copy-number variant. Genet Med. 2016;18 (4):242-248. molecular pathways and circuits in autism. Cell.
(4):341-349. 40. D’Angelo D, Lebon S, Chen Q, et al; Cardiff 2013;155(5):1008-1021.
24. Dolcetti A, Silversides CK, Marshall CR, et al. University Experiences of Children With Copy 56. Jeste SS, Geschwind DH. Disentangling the
1q21.1 Microduplication expression in adults. Genet Number Variants (ECHO) Study; 16p11.2 European heterogeneity of autism spectrum disorder through
Med. 2013;15(4):282-289. Consortium; Simons Variation in Individuals Project genetic findings. Nat Rev Neurol. 2014;10(2):74-81.
25. Dabell MP, Rosenfeld JA, Bader P, et al. (VIP) Consortium. Defining the effect of the 16p11.2 57. Tammimies K, Marshall CR, Walker S, et al.
Investigation of NRXN1 deletions: clinical and duplication on cognition, behavior, and medical Molecular diagnostic yield of chromosomal
molecular characterization. Am J Med Genet A. comorbidities. JAMA Psychiatry. 2016;73(1):20-30. microarray analysis and whole-exome sequencing
2013;161A(4):717-731. 41. Steinman KJ, Spence SJ, Ramocki MB, et al; in children with autism spectrum disorder. JAMA.
26. Rujescu D, Ingason A, Cichon S, et al; GROUP Simons VIP Consortium. 16p11.2 deletion and 2015;314(9):895-903.
Investigators. Disruption of the neurexin 1 gene is duplication: Characterizing neurologic phenotypes 58. Cooper GM, Coe BP, Girirajan S, et al. A copy
associated with schizophrenia. Hum Mol Genet. in a large clinically ascertained cohort. Am J Med number variation morbidity map of developmental
2009;18(5):988-996. Genet A. 2016;170(11):2943-2955. delay. Nat Genet. 2011;43(9):838-846.
27. Ching MS, Shen Y, Tan WH, et al; Children’s 42. Hanson E, Bernier R, Porche K, et al; Simons 59. Shinawi M, Liu P, Kang SH, et al. Recurrent
Hospital Boston Genotype Phenotype Study Group. Variation in Individuals Project Consortium. The reciprocal 16p11.2 rearrangements associated with
Deletions of NRXN1 (neurexin-1) predispose to a cognitive and behavioral phenotype of the 16p11.2 global developmental delay, behavioural problems,
wide spectrum of developmental disorders. Am J deletion in a clinically ascertained population. Biol dysmorphism, epilepsy, and abnormal head size.
Med Genet B Neuropsychiatr Genet. 2010;153B(4): Psychiatry. 2015;77(9):785-793. J Med Genet. 2010;47(5):332-341.
937-947. 43. McCarthy SE, Makarov V, Kirov G, et al; 60. Weiss LA, Shen Y, Korn JM, et al; Autism
28. Mulle JG, Gambello MJ, Cook EH, Rutkowski Wellcome Trust Case Control Consortium. Consortium. Association between microdeletion
TP, Glassford M. 3q29 Recurrent deletion. Microduplications of 16p11.2 are associated with and microduplication at 16p11.2 and autism. N Engl J
GeneReviews. https://www.ncbi.nlm.nih.gov/books schizophrenia. Nat Genet. 2009;41(11):1223-1227. Med. 2008;358(7):667-675.
/NBK385289/. Updated October 19, 2017. Accessed 44. McDonald-McGinn DM, Sullivan KE, Marino B, 61. Fernandez BA, Roberts W, Chung B, et al.
January 17, 2017. et al. 22q11.2 deletion syndrome. Nat Rev Dis Primers. Phenotypic spectrum associated with de novo and
29. Glassford MR, Rosenfeld JA, Freedman AA, 2015;1:15071. inherited deletions and duplications at 16p11.2 in
Zwick ME, Mulle JG; Unique Rare Chromosome 45. Firth HV. 22q11.2 Duplication. GeneReviews. individuals ascertained for diagnosis of autism
Disorder Support Group. Novel features of 3q29 https://www.ncbi.nlm.nih.gov/books/NBK3823/. spectrum disorder. J Med Genet. 2010;47(3):195-203.
deletion syndrome: Results from the 3q29 registry. Updated November 21, 2013. Accessed January 17, 62. Walters RG, Jacquemont S, Valsesia A, et al.
Am J Med Genet A. 2016;170A(4):999-1006. 2017. A new highly penetrant form of obesity due to
30. Mervis CB, Morris CA, Klein-Tasman BP, 46. Van Campenhout S, Devriendt K, Breckpot J, deletions on chromosome 16p11.2. Nature. 2010;
Velleman SL, Osborne LR. 7q11.23 Duplication et al. Microduplication 22q11.2: a description of the 463(7281):671-675.
syndrome. GeneReviews. https://www.ncbi.nlm.nih clinical, developmental and behavioral 63. Zufferey F, Sherr EH, Beckmann ND, et al;
.gov/books/NBK327268/. Published November 25, characteristics during childhood. Genet Couns. Simons VIP Consortium; 16p11.2 European
2015. Accessed January 17, 2017. 2012;23(2):135-148. Consortium. A 600 kb deletion syndrome at 16p11.2
31. Van der Aa N, Rooms L, Vandeweyer G, et al. 47. Portnoï MF. Microduplication 22q11.2: a new leads to energy imbalance and neuropsychiatric
Fourteen new cases contribute to the chromosomal syndrome. Eur J Med Genet. 2009; disorders. J Med Genet. 2012;49(10):660-668.
characterization of the 7q11.23 microduplication 52(2-3):88-93. 64. Jacquemont S, Reymond A, Zufferey F, et al.
syndrome. Eur J Med Genet. 2009;52(2-3):94-100. 48. Soorya L, Kolevzon A, Zweifach J, et al. Mirror extreme BMI phenotypes associated with
32. Morris CA, Mervis CB, Paciorkowski AP, et al. Prospective investigation of autism and gene dosage at the chromosome 16p11.2 locus.
7q11.23 Duplication syndrome: Physical genotype-phenotype correlations in 22q13 deletion Nature. 2011;478(7367):97-102.
characteristics and natural history. Am J Med Genet syndrome and SHANK3 deficiency. Mol Autism. 65. Stefansson H, Meyer-Lindenberg A, Steinberg
A. 2015;167A(12):2916-2935. 2013;4(1):18. S, et al. CNVs conferring risk of autism or
33. Vorstman JAS, Parr JR, Moreno-De-Luca D, 49. Phelan K, McDermid HE. The 22q13.3 deletion schizophrenia affect cognition in controls. Nature.
Anney RJL, Nurnberger JI Jr, Hallmayer JF. Autism syndrome (Phelan-McDermid syndrome). Mol 2014;505(7483):361-366.
genetics: opportunities and challenges for clinical Syndromol. 2012;2(3-5):186-201. 66. Moreno-De-Luca A, Evans DW, Boomer KB,
translation. Nat Rev Genet. 2017;18(6):362-376. 50. Neale BM, Kou Y, Liu L, et al. Patterns and rates et al. The role of parental cognitive, behavioral, and
34. Battaglia A. The inv dup (15) or idic (15) of exonic de novo mutations in autism spectrum motor profiles in clinical variability in individuals
syndrome (Tetrasomy 15q). Orphanet J Rare Dis. disorders. Nature. 2012;485(7397):242-245. with chromosome 16p11.2 deletions. JAMA Psychiatry.
2008;3:30. 51. O’Roak BJ, Vives L, Girirajan S, et al. Sporadic 2015;72(2):119-126.
35. Thomas JA, Johnson J, Peterson Kraai TL, et al. autism exomes reveal a highly interconnected 67. Girirajan S, Rosenfeld JA, Coe BP, et al.
Genetic and clinical characterization of patients protein network of de novo mutations. Nature. Phenotypic heterogeneity of genomic disorders
with an interstitial duplication 15q11-q13, 2012;485(7397):246-250. and rare copy-number variants. N Engl J Med. 2012;
emphasizing behavioral phenotype and response to 52. Sanders SJ, Murtha MT, Gupta AR, et al. 367(14):1321-1331.
treatment. Am J Med Genet A. 2003;119A(2):111-120. De novo mutations revealed by whole-exome 68. Hultman CM, Sandin S, Levine SZ, Lichtenstein
36. Berg JS, Potocki L, Bacino CA. Common sequencing are strongly associated with autism. P, Reichenberg A. Advancing paternal age and risk
recurrent microduplication syndromes: diagnosis Nature. 2012;485(7397):237-241. of autism: new evidence from a population-based
and management in clinical practice. Am J Med 53. De Rubeis S, He X, Goldberg AP, et al; DDD study and a meta-analysis of epidemiological
Genet A. 2010;152A(5):1066-1078. Study; Homozygosity Mapping Collaborative for studies. Mol Psychiatry. 2011;16(12):1203-1212.
37. Battaglia A. The inv dup(15) or idic(15) Autism; UK10K Consortium. Synaptic, 69. Sandin S, Hultman CM, Kolevzon A, Gross R,
syndrome: a clinically recognisable neurogenetic transcriptional and chromatin genes disrupted in MacCabe JH, Reichenberg A. Advancing maternal
disorder. Brain Dev. 2005;27(5):365-369. autism. Nature. 2014;515(7526):209-215. age is associated with increasing risk for autism:
38. Kalsner L, Chamberlain SJ. Prader-Willi, 54. Willsey AJ, Sanders SJ, Li M, et al. Coexpression a review and meta-analysis. J Am Acad Child
Angelman, and 15q11-q13 duplication syndromes. networks implicate human midfetal deep cortical Adolesc Psychiatry. 2012;51(5):477-486.e1.
Pediatr Clin North Am. 2015;62(3):587-606. projection neurons in the pathogenesis of autism. 70. Lyall K, Croen L, Daniels J, et al. The changing
39. Miller DT, Shen Y, Weiss LA, et al. Cell. 2013;155(5):997-1007. epidemiology of autism spectrum disorders. Annu
Microdeletion/duplication at 15q13.2q13.3 among Rev Public Health. 2017;38(1):81-102.

jamapsychiatry.com (Reprinted) JAMA Psychiatry Published online March 28, 2018 E9

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018


Clinical Review & Education Review The Emerging Clinical Neuroscience of Autism Spectrum Disorder

71. Modabbernia A, Velthorst E, Reichenberg A. 86. Redcay E, Courchesne E. When is the brain phenotypes in mouse models of 16p11.2 deletion
Environmental risk factors for autism: an enlarged in autism? a meta-analysis of all brain size and duplication syndromes. PLoS Genet. 2016;12(2):
evidence-based review of systematic reviews and reports. Biol Psychiatry. 2005;58(1):1-9. e1005709.
meta-analyses. Mol Autism. 2017;8:13. 87. Di Martino A, Yan C-G, Li Q, et al. The autism 101. Michalon A, Sidorov M, Ballard TM, et al.
72. Kong SW, Lee IH, Leshchiner I, et al; MedSeq brain imaging data exchange: towards a large-scale Chronic pharmacological mGlu5 inhibition corrects
Project. Summarizing polygenic risks for complex evaluation of the intrinsic brain architecture in fragile X in adult mice. Neuron. 2012;74(1):49-56.
diseases in a clinical whole-genome report. Genet autism. Mol Psychiatry. 2014;19(6):659-667. 102. Stoppel LJ, Kazdoba TM, Schaffler MD, et al.
Med. 2015;17(7):536-544. 88. Qureshi AY, Mueller S, Snyder AZ, et al; Simons R-Baclofen reverses cognitive deficits and improves
73. Guy A, Seaton SE, Boyle EM, et al. Infants born VIP Consortium. Opposing brain differences in social interactions in two lines of 16p11.2 deletion
late/moderately preterm are at increased risk for a 16p11.2 deletion and duplication carriers. J Neurosci. mice. Neuropsychopharmacology. 2018;43(3):
positive autism screen at 2 years of age. J Pediatr. 2014;34(34):11199-11211. 513-524.
2015;166(2):269-275.e3. 89. Maillard AM, Ruef A, Pizzagalli F, et al; 16p11.2 103. Blumenthal I, Ragavendran A, Erdin S, et al.
74. Padilla N, Eklöf E, Mårtensson GE, Bölte S, European Consortium. The 16p11.2 locus modulates Transcriptional consequences of 16p11.2 deletion
Lagercrantz H, Ådén U. Poor brain growth in brain structures common to autism, schizophrenia and duplication in mouse cortex and multiplex
extremely preterm neonates long before the onset and obesity. Mol Psychiatry. 2015;20(1):140-147. autism families. Am J Hum Genet. 2014;94(6):
of autism spectrum disorder symptoms. Cereb Cortex. 90. Johnson CP, Myers SM; American Academy of 870-883.
2017;27(2):1245-1252. Pediatrics Council on Children With Disabilities. 104. Migliavacca E, Golzio C, Männik K, et al;
75. Movsas TZ, Pinto-Martin JA, Whitaker AH, et al. Identification and evaluation of children with 16p11.2 European Consortium. A Potential
Autism spectrum disorder is associated with autism spectrum disorders. Pediatrics. 2007;120(5): contributory role for ciliary dysfunction in the
ventricular enlargement in a low birth weight 1183-1215. 16p11.2 600 kb BP4-BP5 pathology. Am J Hum Genet.
population. J Pediatr. 2013;163(1):73-78. 91. Filipek PA, Accardo PJ, Ashwal S, et al. Practice 2015;96(5):784-796.
76. Mandy W, Lai M-C. Annual research review: the parameter: screening and diagnosis of autism: 105. Paşca AM, Sloan SA, Clarke LE, et al.
role of the environment in the developmental report of the Quality Standards Subcommittee of Functional cortical neurons and astrocytes from
psychopathology of autism spectrum condition. the American Academy of Neurology and the Child human pluripotent stem cells in 3D culture. Nat
J Child Psychol Psychiatry. 2016;57(3):271-292. Neurology Society. Neurology. 2000;55(4):468-479. Methods. 2015;12(7):671-678.
77. Christensen J, Grønborg TK, Sørensen MJ, et al. 92. Miller DT, Chung W, Nasir R, et al. 16p11.2 106. Paşca SP, Portmann T, Voineagu I, et al. Using
Prenatal valproate exposure and risk of autism Recurrent microdeletion. GeneReviews. iPSC-derived neurons to uncover cellular
spectrum disorders and childhood autism. JAMA. https://www.ncbi.nlm.nih.gov/books/NBK11167/. phenotypes associated with Timothy syndrome.
2013;309(16):1696-1703. Updated December 10, 2015. Accessed January 17, Nat Med. 2011;17(12):1657-1662.
78. Emerson RW, Adams C, Nishino T, et al; IBIS 2017. 107. Weitlauf AS, McPheeters ML, Peters B, et al.
Network. Functional neuroimaging of high-risk 93. Jones W, Klin A. Attention to eyes is present Therapies for children with autism spectrum
6-month-old infants predicts a diagnosis of autism but in decline in 2-6-month-old infants later disorder: behavioral interventions update:
at 24 months of age. Sci Transl Med. 2017;9(393): diagnosed with autism. Nature. 2013;504(7480): comparative effectiveness review no. 137.
eaag2882. 427-431. (Prepared by the Vanderbilt Evidence-based
79. Wolff JJ, Jacob S, Elison JT. The journey to 94. Golzio C, Willer J, Talkowski ME, et al. KCTD13 Practice Center under Contract No.
autism: insights from neuroimaging studies of is a major driver of mirrored neuroanatomical 290-2012-00009-I.) AHRQ Publication No.
infants and toddlers. Dev Psychopathol. 2017:1-17. phenotypes of the 16p11.2 copy number variant. 14-EHC036-EF. Rockville, MD: Agency for
Nature. 2012;485(7398):363-367. Healthcare Research and Quality; 2014.
80. Wolff JJ, Gu H, Gerig G, et al; IBIS Network.
Differences in white matter fiber tract development 95. Chen Y, Yu J, Niu Y, et al. Modeling Rett 108. Fung LK, Mahajan R, Nozzolillo A, et al.
present from 6 to 24 months in infants with autism. syndrome using TALEN-edited MECP2 mutant Pharmacologic treatment of severe irritability and
Am J Psychiatry. 2012;169(6):589-600. cynomolgus monkeys. Cell. 2017;169(5):945-955.e10. problem behaviors in autism: a systematic review
and meta-analysis. Pediatrics. 2016;137(S2)
81. Lewis JD, Evans AC, Pruett JR Jr, et al; Infant 96. Portmann T, Yang M, Mao R, et al. Behavioral (suppl 2):S124-S135.
Brain Imaging Study Network. The emergence of abnormalities and circuit defects in the basal
network inefficiencies in infants with autism ganglia of a mouse model of 16p11.2 deletion 109. Levy SE, Hyman SL. Complementary and
spectrum disorder. Biol Psychiatry. 2017;82(3): syndrome. Cell Rep. 2014;7(4):1077-1092. alternative medicine treatments for children with
176-185. autism spectrum disorders. Child Adolesc Psychiatr
97. Pucilowska J, Vithayathil J, Tavares EJ, Kelly C, Clin N Am. 2015;24(1):117-143.
82. Hazlett HC, Gu H, Munsell BC, et al; IBIS Karlo JC, Landreth GE. The 16p11.2 deletion mouse
Network; Clinical Sites; Data Coordinating Center; model of autism exhibits altered cortical progenitor 110. Hsu PD, Lander ES, Zhang F. Development and
Image Processing Core; Statistical Analysis. Early proliferation and brain cytoarchitecture linked to applications of CRISPR-Cas9 for genome
brain development in infants at high risk for autism the ERK MAPK pathway. J Neurosci. 2015;35(7): engineering. Cell. 2014;157(6):1262-1278.
spectrum disorder. Nature. 2017;542(7641):348-351. 3190-3200. 111. Peng R, Lin G, Li J. Potential pitfalls of
83. Shen MD, Nordahl CW, Young GS, et al. Early 98. Tian D, Stoppel LJ, Heynen AJ, et al. CRISPR/Cas9-mediated genome editing. FEBS J.
brain enlargement and elevated extra-axial fluid in Contribution of mGluR5 to pathophysiology in a 2016;283(7):1218-1231.
infants who develop autism spectrum disorder. Brain. mouse model of human chromosome 16p11.2 112. Erickson CA, Davenport MH, Schaefer TL, et al.
2013;136(pt 9):2825-2835. microdeletion. Nat Neurosci. 2015;18(2):182-184. Fragile X targeted pharmacotherapy: lessons
84. Tau GZ, Peterson BS. Normal development of 99. Horev G, Ellegood J, Lerch JP, et al. learned and future directions. J Neurodev Disord.
brain circuits. Neuropsychopharmacology. 2010; Dosage-dependent phenotypes in models of 2017;9:7.
35(1):147-168. 16p11.2 lesions found in autism. Proc Natl Acad Sci 113. Budimirovic DB, Berry-Kravis E, Erickson CA,
85. Courchesne E, Campbell K, Solso S. Brain U S A. 2011;108(41):17076-17081. et al. Updated report on tools to measure outcomes
growth across the life span in autism: age-specific 100. Arbogast T, Ouagazzal AM, Chevalier C, et al. of clinical trials in fragile X syndrome. J Neurodev
changes in anatomical pathology. Brain Res. 2011; Reciprocal effects on neurocognitive and metabolic Disord. 2017;9:14.
1380:138-145.

E10 JAMA Psychiatry Published online March 28, 2018 (Reprinted) jamapsychiatry.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a University of Toledo Libraries User on 03/28/2018

Das könnte Ihnen auch gefallen