ArticlePDF AvailableLiterature Review

Maternal Immune Activation and Autism Spectrum Disorder: Interleukin-6 Signaling as a Key Mechanistic Pathway

Authors:

Abstract

An emerging area of research in autism spectrum disorder (ASD) is the role of prenatal exposure to inflammatory mediators during critical developmental periods. Epidemiological data has highlighted this relationship showing significant correlations between prenatal exposure to pathogens, including influenza, and the occurrence of ASD. Although there has not been a definitive molecular mechanism established, researchers have begun to investigate this relationship as animal models of maternal infection have support- ed epidemiological findings. Several groups utilizing these animal models have found that activation of the maternal immune system, termed maternal immune activation (MIA), and more specifically the exposure of the developing fetus to maternal cytokines precipitate the neurological, immunological and behavioral abnormalities observed in the offspring of these animals. These abnormalities have correlated with clinical findings of immune dysregulation, neurological and behavioral abnormalities in some autistic individuals. Additionally, researchers have observed genetic variations in these models in genes which regulate neurological and immunological development, similar to what is observed clinically in ASD. Altogether, the role of MIA and cytokine dysregulation, as a key mediator in the neuropathological, behavioral and possibly genetic irregularities observed clinically in autism are important factors that warrant further investigation.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Neurosignals 2010;18:113–128
DOI: 10.1159/000319828
Maternal Immune Activation and Autism
Spectrum Disorder: Interleukin-6
Signaling as a Key Mechanistic Pathway
E.CarlaParker-Athill a–c JunTan a–c
a Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center,
b Department of Psychiatry
and Behavioral Medicine,
c College of Medicine, University of South Florida, Tampa, Fla. , USA
in these models in genes which regulate neurological and
immunological development, similar to what is observed
clin ically in ASD. Altogether, the role of MIA and cytokine
dysregulation, as a key mediator in the neuropathological,
behavioral and possibly genetic irregularities observed clin-
ically in autism are important factors that warrant further in-
vestigation. Copyright © 2010 S. Karger AG, Base l
Introduction
Originally adapted form the Greek ‘autos’ or ‘self’, the
term autism was first used in the early 1900s by Swiss
psychiatrist Eugen Bleuler to describe a cluster of symp-
toms in schizophrenic individuals which included with-
drawal or ‘self-isolation’ [1] . The term would later be used
by Kanner in the 1940s, to describe a similar group of
symptoms in children [1] . These early clinicians believed
autism and the manifestations of social withdrawal to be
the result of ‘cold’ unemotional parents. As a result, treat-
ments for autism centered on medications such as lysergic
acid diethylamide (LSD), electric shock and behavior
change techniques, which encompassed pain and pun-
ishment therapy. As the understanding of the disorder
Key Words
Autism spectrum disorder Fetal immune response
syndrome Maternal immune activation Interleukin-6
Abstract
An emerging area of research in autism spectrum disorder
(ASD) is the role of prenatal exposure to inflammatory me-
diators during critical developmental periods. Epidemiolog-
ical data has highlighted this relationship showing signifi-
cant correlations between prenatal exposure to pathogens,
including influenza, and the occurrence of ASD. Although
there has not been a definitive molecular mechanism estab-
lished, researchers have begun to investigate this relation-
ship as animal models of maternal infection have support-
ed epidemiological findings. Several groups utilizing these
animal models have found that activation of the maternal
immune system, termed maternal immune activation (MIA),
and more specifically the exposure of the developing fetus
to maternal cytokines precipitate the neurological, immuno -
logical and behavioral abnormalities observed in the off-
spring of these animals. These abnormalities have correlated
with clinical findings of immune dysregulation, neurological
and behavioral abnormalities in some autistic individuals.
Additionally, researchers have observed genetic variations
Receive d: May 31, 2010
Accepted af ter revision: July 30, 2010
Publishe d online: October 2, 2010
Dr. Jun Tan
Department of Psychiatr y, Silver Ch ild Development Center
Universit y of South Florida, 3515 E. Fletc her Ave.
Tampa, FL 33613 (USA)
Tel. +1 813 974 9326, Fax +1 813 974 3223, E-Ma il jtan
@ health.usf.edu
© 2010 S. Karger AG, Basel
Accessible online at:
www.karger.com/nsg
Parker-Athill /Tan
Neurosignals 2010;18:113–128
114
evolved largely during the late 1990s, the role of behav-
ioral therapy centered on the use of highly controlled
learning environments, a form of therapy which has re-
mained a key feature of autism therapy today. With an
increased understanding of the disorder, clinicians began
to characterize key phenotypes to aid in the assessment
of individuals. With this came the inclusion of autism in
the Diagnostic and Statistical Manual of Mental Disorders
(DSM), as a behaviorally defined developmental disorder.
Today, the term autism is still used to describe the
cluster of symptoms manifested in children displaying
withdrawn or ‘isolated’ social behaviors, although clas-
sification and diagnostic criteria have evolved. Usually
diagnosed during the first 3 years, early autistic patients
often presented with a narrow range of symptoms, which
often included moderate-to-severe mental retardation,
although patients now present with symptoms that vary
in both severity and combination with almost 30% of per-
sons now having normal verbal expressions and IQ scores
[2, 3] . These changes in clinical observations have led to
the classification of autism as a spectrum disorder, autism
spectrum disorder (ASD), which describes a group of de-
velopmental disorders characterized by impairments in
reciprocal social interaction and verbal and nonverbal
communication skills compounded by symptoms of re-
strictive and repetitive behaviors and/or stereotyped pat-
terns of interest that are abnormal in their intensity or
focus [4–7] . Included in this spectrum isclassical au-
tism, which usually involves the stereotypical social isola-
tion, impaired verbal communication and repetitive be-
haviors; Asperger syndrome, often described as a milder
form of autism, lacking the components of intense repet-
itive behaviors and social withdrawal; and pervasive de-
velopmental disorder, which usually encompasses disor-
ders that cannot otherwise be classified [2, 7–9] .
These diagnostic changes in the clinical community,
coupled with increased education and awareness among
the clinical, research and general community, have also
led to important changes in the epidemiological study of
ASD. Previous epidemiological surveys, including those
conducted prior to the classification of autism as a spec-
trum disorder, often used strict diagnostic criteria which
resulted in the exclusion of individuals who would today
be classified as autistic. These changes have led to in-
creased research into the etiology of autism through the
establishment of autism registries and the utilization of
cohort studies. Although the exact etiology of the disor-
der has not been identified, potential environmental
‘triggers’, such as prenatal viral exposure, and genetic
vulnerabilities, including mutations and polymorphisms
in critical genes, have been among the epidemiological
findings [10, 11] . Additionally, these studies have provid-
ed important information for the scientific community,
which has begun to utilize animal models to investigate
the role of environmental insults, such as prenatal viral
exposure, in the etiology of autism. Several researchers
have had promising results in this area not only confirm-
ing the potential for prenatal viral exposure to induce ab-
errant behavioral outcomes in offspring, but also the
identification of the maternal cytokine response to viral
pathogens, referred to as maternal immune activation
(MIA) by Smith et al. [12] , as a possible mechanism in the
precipitation of these aberrant behaviors.
Despite this evidence, there is still disagreement with-
in the research community concerning the environmen-
tal etiology of autism. Nevertheless, epidemiological and
scientific observations supporting this role cannot be ig-
nored, leading several researchers to conclude that cer-
tain ‘environmental insults’ may act as triggers, precipi-
tating the development of autism. Furthermore, the lack
of a single gene candidate and the observed genetic diver-
sity w it hin the autistic population have also led research-
ers to theorize that in genetically vulnerable individuals,
environmental insults, such as prenatal viral exposure
and resulting MIA, may explain the diverse behavioral
phenotypes presented clinically in this spectrum disor-
der, as different genetic polymorphisms may differential-
ly precipitate certain behavioral outcomes.
Environmental Theory of ASD: Epidemiological
Perspective
Epidemiological studies have long been used in the
investigation of diseases and disorders to deduce their
etiology through the examination of trends, risk factors
and commonalities in affected populations. Their ap-
plication to neurodevelopmental disorders such as ASD
has however had several obstacles including small sam-
ple sizes and changes in diagnostic criteria and classifi-
cation systems. Early autism studies, for example, often
utilized Kanner’s autism criteria which employed strict
diagnostic criteria encompassing narrow phenotypic
ranges. The results were smaller sample sizes and lower
observations of prevalence [13] . Today, with increased
education and utilization of the DSM and the Interna-
tional Classification of Diseases criteria, researchers are
more aware of the various phenotypic presentations
seen within the autistic population, including more
atypical types of autism. As a result, today’s studies of-
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
115
ten encompass larger sample populations allowing for
the collection of more statistically relevant data, al-
though they have also resulted in perceived higher rates
of prevalence than previous studies [7, 14–16] . In a com-
parative analysis by Kielinen et al. [17] , for example, the
Kanner and DSM criteria were used to assess the preva-
lence of autism within the same sample population,
wh ich reveale d an i ncrease in preva len ce w hen the DSM
criteria was used. As eluded to by Kielinen, these chang-
es in prevalence may be attributable to methodological
changes in these studies; changes in diagnostic, classifi-
cation and inclusion criteria; and increased awareness
and education among the clinical and general commu-
nities [17–19] . It is important to note that although an
upward trend in prevalence has been observed, consis-
tencies have remained in gender disparities as a higher
rate of autism is reported in males throughout these
studies [17, 20] .
Epidemiological Studies: Genetic Predictors of ASD
Although changes within the field of ASD research
have altered the way epidemiological surveys are con-
ducted making chronological analysis of prevalence dif-
ficult, they have allowed for increased utilization of preg-
nancy/birth cohorts and other registries previously un-
derutilized in the study of ASD. Additionally, dedicated
autism registries now provide not only diagnostic and
survey information, but often include biological speci-
mens, such as serum samples, which can be utilized for
biological assays and genetic analysis for commonalities
within this population as well as divergences from con-
trol populations – all necessary information for the elu-
cidation of the etiology of this disorder.
Genetic high-risk cohorts have been another resource
utilized to examine the role of genetics in the etiology of
ASD. These studies have differed from other areas, how-
ever, by utilizing sibling data rather than offspring.
Through these studies, the genetic component of ASD has
been shown as the recurrence rate in siblings, estimated at
3–8%, 60–90% for identical twins, show not only a strong
genetic component but also a potentially significant role
for epigenetic factors, including maternal inheritance and
imprinting. Despite these findings, there has not been a
‘single gene’ candidate discovered for ASD, although sev-
eral genes have been identified as abnormally expressed
or polymorphic at a higher rate within the autistic popu-
lation. Among these genes have been those involved in
neuronal and synaptic formation and function, including
reelin, neurogenin-1 and neuroligin-4, as well as those in-
volved in immune regulation, such as cytokine receptor
glycoprotein 130 (gp-130), although this gene product has
been shown to possess several biological functions [21–25] .
Epidemiological Studies: Environmental Predictors of
ASD
The genetic diversity within the autistic population, in
addition to the phenotypic variability observed clinically
has posed a significant obstacle in identifying the etiol-
ogy of ASD; however, it has led many researchers to ex-
amine alternative causative agents that may contribute to
the neuropathology of this disorder. Researchers have
now begun to utilize exposure-based high-risk cohorts,
which are used to examine the effects of environmental
risk factors on the etiology of disorders, to elucidate po-
tential environmental etiologies of ASD. These cohorts
not only examine environmental etiologies of disorders,
but also how these environmental factors can interact
with genes to precipitate these disorders, often creating
diverse phenotypic presentations within a given disorder
such as variations in symptom severity.
The utilization of these cohort studies, including the
exposure risk cohort design, in the field of schizophrenia
has identified several factors, including genetic polymor-
phisms and prenatal viral exposure, as significant risk
factors in the etiology of this disorder [26–30] . Similarly,
recent studies in ASD research, such as those examining
prenatal exposure to rubella and other viral agents, have
also alluded to a possible environmental etiology of ASD,
and several researchers have seen significant correlations
between prenatal exposure and the occurrence of this
disorder in offspring [31–33] . It is important to note, how-
ever, that there have been several studies, such as the in-
vestigation of intrauterine human parvovirus infection
by Anlar et al. [34] , which have found no significant cor-
relation between prenatal viral exposure and the occur-
rence of autism [34 –37] . Nevertheless, several studies
have shown significant correlations, primarily with pre-
natal rubella exposure and the development of autism.
Most notable have been several studies by Chess [38, 39]
who has found significant correlations with congenital
rubella and the development of ASD. Other researchers
have observed similar correlations with other viral patho-
gens including, herpes simplex, cytomegalovirus and
varicella zoster [4042] . Interestingly, Deykin and Mac-
Mahon [35] , in a study which reported no association be-
tween prenatal viral exposure and the development of au-
tism, actually reported an increased frequency of expo-
sure in a ut is ti c s ubjects i n com pa ri so n t o c ont ro l s ubject s.
It is therefore plausible to theorize that an environmental
insult, such as prenatal viral exposure, can act as a trigger
Parker-Athill /Tan
Neurosignals 2010;18:113–128
116
precipitating the aberrant behavioral phenotypes ob-
served in ASD. This is not to say that an environmental
etiology is the only contributor to the development of
ASD. On the contrary, several researchers believe that
these environmental triggers may exacerbate genetic vul-
nerabi litie s in some ind ividuals , or may themselves cause
alterations in gene and/or protein expression, precipitat-
ing the abnormal phenotypes observed in autistic indi-
viduals [11, 43, 44] .
Evidence from Fetal Inflammatory Response
Syndrome
The idea of an ‘environmental insult’ precipitating
neurodevelopmental disorders is not a novel one as sev-
eral compounds, including toxins and heavy metals, have
been shown to cause neurological and congenital abnor-
malities in children exposed prenatally [4548] . The link
between inflammation and disruption in fetal develop-
ment is also not a novel idea as several researchers have
observed correlations between maternal infections, pre-
term births and neurological disorders in these preterm
infants [11, 4 957] . Fetal inflammatory response syn-
drome (FIRS), commonly observed in spontaneous pre-
term labor and considered the fetal counterpart of sys-
temic inflammatory response syndrome, is one example
of how maternal infection can disrupt fetal development.
[54, 5860] . Researchers, such as Madsen-Bouterse et al.
[58] , have found that intrauterine infection is often a sig-
nif icant contributor to t his syndrome with int ra-a mniot-
ic infection and/or inflammation being present in one
third of the patients with preterm labor. Usually the result
of maternal bacterial infections, FIRS has been strongly
associated with complications in preterm infants, includ-
ing neurological disruption such as cerebral palsy and
systemic FIRS in some instances [61–64] . Similarly, ASD
has also been linked to maternal infection, as epidemio-
logical studies have noted a strong correlation between
maternal infection, particularly with pathogenic agents,
and the occurrence of ASD and other developmental and
psychiatric disorders in resultant offspring [65– 69] . This
research, compounded with clinical findings of immu-
nological disruptions and increased serum cytokine lev-
els in some autistic patients have driven the development
of an ima l mo de ls to in vest iga te th e rol e of ma te rna l i nfec-
tion in the etiology of ASD [70–74] . These studies have
yielded several major findings: (1) maternal infection
triggers an activation of the maternal immune system,
termed MIA; (2) the resulting production of inflamma-
tory cytokines can traverse the blood-placental barrier
and activate the fetal immune system; and (3) this activa-
tion can lead to neurological and immunological distur-
bances that may precipitate the behavioral phenotypes
observed clinically. These studies have also identified
interleukin (IL)-6 as a key cytokine precipitating these
events [12, 75 –78] .
The observations in ASD and models of MIA have
been analogous to those seen in FIRS, as one of the diag-
nostic markers is an increase in IL-6 concentration in t he
umbilical cord plasma and/or the presence of funisitis,
which is an inflammation of the connective tissue of the
umbilical cord, as well as elevations in several other cy-
tokines [12, 54, 58] . The parallels between these two dis-
orders do not end at the neurological and immunological
disruptions mediated by maternal infection and fetal ex-
posure to maternally derived cytokines as several inves-
tigators have also observed genetic alterations, primarily
in genes associated with immune regulation in FIRS in-
fants, a feature also observed in autistic patients [11, 4 3 ,
58] . Altogether, these findings support a probable role of
maternal infection in ASD and further suggest that ma-
ternally derived inf lammatory cytokines can have delete-
rious and persistent effects on the developing fetus not
only precipitating neurological and immunological ab-
normalities, but potentially altering the expression of
genes important in immune regulation and neurodevel-
opment.
Prenatal Viral Exposure, MIA, Genetics and ASD
Genetic dysregulation has long been an area of interest
in the study of ASD as there is a high degree of heritabil-
ity observed in this disorder evidenced by the increased
occurrence in siblings compared to the general popula-
tion, estimated at 3–8% [79– 82] . In comparison to other
genetic disorders, this probability is still low, making the
development of pedigrees difficult; however, a significant
increase in the occurrence of the disorder in identical
twins with a probability estimated at 60–92% has led
many researchers and clinicians to utilize twin studies to
investigate the role of genetics in ASD [80, 83, 84] . Inter-
estingly, although the probability of the occurrence in
fraternal twins is significantly lower than that of identical
twins as expected, it has been shown to be greater than
that observed in other sibling relationships, estimated at
approximately 10%, suggesting a potential role for mater-
nal and environmental inf luences [80, 84, 85] .
In addition to familial inheritance, a strong occur-
rence of ‘autistic-like’ behaviors with genetic disorders,
such as tuberous sclerosis and fragile X syndrome, has
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
117
strengthened the theory of a genetic mode of transmis-
sion. Approximately 18–33% of children with fragile X
syndrome and an estimated 25–50% of children with tu-
berous sclerosis have some degree of ASD, although, like
previously referenced sibling data, these estimates often
vary as a result of differences in diagnostic criteria [2 4,
86–90] . Despite this high degree of comorbidity, only a
small percentage of diagnosed ASD cases are attributable
to these genetic disorders, with 6% of diagnosed autism
cases due to fragile X syndrome and 1–4% to tuberous
sclerosis [87, 90] . Together, these genetic disorders ac-
count for only 10% of diagnosed ASD cases, with the re-
maining 90% still of unknown etiology, although several
other genes have been implicated as polymorphisms and
abnormal expression patterns have been seen in several
autistic individuals [80, 81, 87, 91] . Among those genes
characterized are those involved in the development and
function of the central nervous system (CNS), particu-
larly those regulating neural differentiation, migration,
axonal pathfinding and synapse formation. For example,
the neuroligins, cell adhesion molecules involved in
synaptic maturation and function, have been linked
to autism as mutations and deletions in this gene have
been observed within the autistic population [92]. Simi-
larly, mutations in contactin-associated protein-like 2
(CASPR2), which interacts with neuroligins to promote
synap tic f un ct ion , ha ve a ls o been obse rv ed w it hi n t he a u-
tistic population [92]. Another prominent gene linked to
autism is reelin. Involved in neuronal migration, poly-
morphisms in this gene have been linked to autism, a
finding largely supported by correlations between animal
models of reelin mutations and postmortem analysis of
autistic brains [92]. Additionally, genes involved in im-
mune regulation, including IL-6, and genes important
in several signal transduction pathways have also been
shown to be differentially expressed in autistic individu-
als [93–98] .
MIA: Cytokines and Genetics
Interestingly, researchers have observed neuropatho-
logical and genetic abnormalities similar to those ob-
served in ASD in models of MIA and FIRS patients, sug-
gesting early exposure to inflammatory cytokines may
cause the abnormal expression of some genes (primarily
those involved in immune and CNS regulation) [11, 1 2 ,
43, 58, 72, 99] . In FIRS, for example, genes involved in
MCH antigen presentation, leukocyte adhesion and che-
motaxis are differentially expressed in comparison to
control patients, as are other immune regulatory factors
including cytokines such as IL-6 [58] . In models of MIA,
similar dysregulations have been noted for genes involved
in immune regulation [11, 43, 68] . Utilizing the MIA
model, researchers have also noted dysregulation in genes
involved in the regulation of CNS development and func-
tion, including those involved in neural differentiation
and migration, axonal pathfinding and synapse forma-
tion, maintenance and function, and neurotransmitter
synthesis – genes which are also dysregulated in autistic
individuals [11, 43, 68] .
These observations have mirrored clinical findings in
ASD; however, it is still unclear how these genetic abnor-
malities contribute to etiologic factors. Although de-
scribed as a spectrum disorder, the range of phenotypes
observed clinically do not correlate to the considerable
genetic variations observed. Several researchers now hy-
pothesize that certain genetic polymorphisms, which oc-
cur normally in the population and often have no patho-
logical attributes, can in some individuals, when coupled
with an environmental insult such as maternal infection
and maternal cytokine exposure, lead to the development
of different autistic phenoty pes or ASD [10 0] . This theory
has been used to explain the diverse genetic and pheno-
typic variations seen in autistic individuals, and it has
been suggested that the risk of developing ASD as a result
of maternal infection may be 3- to 7-fold higher in ‘ge-
netically susceptible’ individuals. What this theory does
not explain is the role played by maternal infection, or
more specifically maternally derived inflammatory cyto-
kines in altering the expression of certain genes (primar-
ily those involved in regulation of immune and CNS de-
velopment and function). Although research in FIRS and
MIA models have shown that exposure to maternally de-
rived inflammatory cytokines is sufficient to alter the ex-
pression of several immune related genes, the mechanism
by which this is accomplished is still largely unknown.
IL-6 and Gene Expression
The answer to this question may lie in the innate abil-
it y o f m a ny c ytok in e s t o r eg ul at e g en e e x pr es s io n t h ro ug h
the activation of signal transduction pathways that ulti-
mately activate transcription factors [101] . Cytokines
such as IL-6, for example, activate Janus tyrosine kinase,
mitogen-activated protein and/or other kinases, which in
turn activate transcription factors such as signal trans-
ducer and activator of transcription (STAT) [101, 102] .
These transcription factors in turn regulate the tran-
scription of a range of gene products, binding to the cor-
responding gene promoter regions to induce or repress
gene transcription [102] . During inf lammation for exam-
ple, IL-6-mediated STAT3 activation leads to the in-
Parker-Athill /Tan
Neurosignals 2010;18:113–128
118
creased expression of other cytokines and immune regu-
latory gene s, a nd u lti mately protei ns [103–105] . In neuro-
logical disorders characterized by increased inf lamma-
tion such as Alzheimer’s disease, this increased activity
leads to a persistent inflammatory state with correspond-
ing morphological changes, including increased glial cell
activity [10 6, 107] .
These observations may explain the changes in gene
expression seen in FIRS and ASD patients and animal
models of MIA. In the fetal brain, exposure to maternal-
ly derived inflammatory cytokines may have effects sim-
ilar to what is seen in these neurological disorders, pre-
cipitating the abnormal expression of fetal genes involved
in the development and function of the immune system,
such as what is obser ved in FIRS [78, 106, 108] . Similarly,
cytokines have also been implicated in the regulation of
genes involved in the development and function of the
CNS, analogous to the way they regulate immunological
genes [44, 108] . Cytokines such as IL-6, for example, can
initiate the transcription of neural regulatory genes, im-
portant in the proliferation and differentiation of neural
stem cells (NSC) through the activation of STAT3 and
other transcription factors [44, 109] . In cases where ma-
ternally derived cytokines are present, this can lead to an
increased activation and aberrant expression of these
genes and the neurological and behavioral abnormalities
observed clinically in ASD and models of MIA [11, 12, 50,
68] . It is therefore important not only to examine how
certain genetic polymorphisms can predispose individu-
als to the development of ASD, but also how maternal
cytokine exposure can act as a trigger for this disorder in
susceptible individuals. It is equally important to exam-
ine how this exposure to maternal cytokines can disrupt
the expression of important regulatory genes, and how
these events affect the etiology and phenotypes of ASD,
namely the variations in severity and combinations of
signs and symptoms manifested clinically.
MIA, Neurodevelopment and ASD
Although the genetic component of ASD cannot be
ignored, it has been accepted that maternal infection, or
MIA, may be a significant contributor to the pathology of
developmental disorders (e.g. ASD) and maternal cyto-
kine expression and migration across the placenta, the
main precipitator of the neurological, immunological
and behavioral disruptions observed clinically [51, 54,
110–112] . Furthermore, this exposure to maternal cyto-
kines has been shown to cause behavioral disruptions in
offspring reminiscent of the phenotypes observed clini-
cally in ASD, such as impaired social interaction [12 , 5 0 ,
113] . In examining the potential mechanisms underlying
this phenomenon, it is important to note the physiologi-
cal significance of these molecules during key neurode-
velopmental periods. Under normal physiological condi-
tions, cytokines such as IL-6 are an important part of the
neurodevelopmental signaling cascades involved in sur-
vival, proliferation, differentiation and phenotypic main-
tenance; regulation of neuronal migration and axonal
pathfinding; and synapse formation and elimination [44,
101, 109, 112 , 114–116] . Research has confirmed these
physiological roles showing the regulation of NSC prolif-
eration, survival and differentiation by the gp-130 family,
BMP and members of the interleukin and interferon fam-
ilies [44, 101, 108, 109, 114, 117] .
Cytokines and Neurodevelopment
During normal neurodevelopment, cytokines have
been shown to be present and functional in the fetal
br ain, w it h receptor s id ent if ied on b oth n euronal a nd gli-
al cells [118] . Although present at minimal levels, these
cytokines, such as IL-6 and its receptor, have been de-
tected in the rat cortex as early as embryonic day 18, and
in the human brain as early as 8 weeks [118121] . Simi-
larly, IL-1 has been detected in the embryonic rat brain,
peaking during embryonic days 18–20 and again at post-
natal day 7 [122] . Although the physiological role of these
cytokines in the developing CNS is not fully understood,
studies have confirmed that they function as important
regulatory molecules and are involved in all phases of
CNS development [44, 121, 123] . Their minimal concen-
trations also suggest that their regulatory activities may
be closely related to circulating concentration, making
the maintenance of homeostasis critical in maintaining
the balance between their physiological and pathological
activities. Several researchers have now begun to exam-
ine the physiological roles of these cytokines in an effort
to better understand their pathological roles in ASD.
Among those examined have been tumor necrosis factor
(TNF), the interferons and interleukins; most notably
IL-6 and other members of the gp-130 family, primarily
due to their observed pleiotropic characteristics. Many of
these cytokines, although originally defined as proin-
flammatory, have now been shown to also possess neuro-
tropic properties regulating cell survival and prolifera-
tion in addition to differentiation and axonal guidance
[44, 114, 117, 122, 124] . IL-6 for example has been shown
to induce the differentiation of NSC into astrocytes and
to a lesser extent neurons and oligodendrocytes while
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
119
promoting the survival of postnatal mesencephalic cate-
choloaminergic neurons and spinal cord cholinergic neu-
rons in culture [125 128] . TNF-
and IL-1 h ave also bee n
shown to have similar effects, specifically through IL-1
modulating the survival and growth of neuronal and gli-
al cells, and TNF- inducing synthesis of IL-6 from as-
trocytes and enhancing nerve growth factor release [1 22 ,
129, 130] .
The activities of these cytokines observed in vitro are
believed to mimic their physiological roles, although the
mechanisms by which they exert their effects are still not
fully understood. Contributing to this lack of under-
standing is the observed overlapping function of several
of these cytokines, often able to elicit the same events,
granted through different mechanisms. During CNS
development, for example, cardiotrophin-1 appears to be
the key cytokine inducing glial differentiation, although
other members of the gp-130 family have been shown to
be capable of accomplishing this [44, 114] . These redun-
dant functions may explain why IL-6 has emerged as a
key cytokine in disorders linked to maternal infection
such as FIRS and ASD.
MIA and ASD: Potential Mechanisms
There has been tremendous resea rch showing that sev-
eral cytokines play an integral role in CNS development.
There has also been research suggesting that even minute
changes in the physiological concentrations of these cy-
tokines (e.g. from maternal infections) can be deleterious
to the developing fetus, leading to aberrant gene and pro-
tein expression as well as neurological, immunological
and behavioral abnormalities. It is still unclear, however,
how these effects are mediated as there remains disagree-
ment concerning whether or not these maternal cyto-
kines can traverse the placental barrier to induce re-
sponses in the fetal CNS. Several studies have yielded
conflicting results as some suggest that only negligible
levels of some cytokines pass this barrier, while others
suggest that this does not occur. Whatever the mecha-
nism, it is obvious that MIA somehow triggers activation
of the fetal immune system as studies have shown in-
creases in fetal cytokine expression in the CNS following
exposure to maternal cytokines.
Cytokines and the Placental Barrier
Although there is disagreement as to whether or not
cytokines can traverse the placental barrier, it has been
well accepted that they are ubiquitously expressed in the
placenta, where they serve an important role in the regu-
lation of the maternal-fetal immune interface, aiding in
preventing maternal rejection of the fetus through the
regulation of the maternal immune system [1 31–133] .
During pregnancy, the fetus, which produces alloanti-
gens encoded by paternal genes, can illicit an immune
response from the maternal immune system. Several re-
searchers have hypothesized that instances of spontane-
ous abortions, aberrant fetal growth and recurrent mis-
carriages may be the result of the inability of the placenta
to fully suppress this maternal response to these fetal al-
loantigens, allowing the maternal immune system to at-
tack the developing fetus similar to what is seen in graft-
versus-host rejections and other classical immunological
responses to foreign invaders, such as pathogens [63, 134 –
136] .
Therefore, the placenta functions as an important im-
mu ne ‘org an’, c oord in ating mate rna l-feta l i mmu ne i nte r-
actions in addition to furnishing the metabolic needs of
the developing fetus. Among the resident immunological
molecules in the placenta are maternal immune factors,
including regulatory T cells (T
reg ), human leukocyte an-
tigen-G (HLA-G, a member of the MHC class 1 mole-
cules) and T cell costimulatory molecules in addition to
cytokines and other immune regulatory molecules that
play an important role in regulating the maternal im-
mune system, preventing the rejection of the developing
fetus while still maintaining the capacity to defend the
mother against pathogenic invaders [133, 135, 137] . Un-
der normal conditions, there is an increase in the number
of immu nosuppressive T
reg preceding and after implanta-
tion of the fetus, which prevents the proliferation and ac-
tivation of proinflammatory Th17 cells [13 8 –14 2] . Sev-
eral animal and clinical studies have confirmed this in-
terplay, with observations of T
reg activity, correlated to
increased uterine weight [141, 143, 144] . The role of resi-
dent cytokines becomes increasingly important during
this process as several have been shown to be potent mod-
ulators of T cell regulations capable of shifting the bal-
ance between T
reg and Th17s, recruiting and maintaining
the T
reg population in the placenta [14 5] .
MIA and the Placental Barrier
The role of MIA in disrupting the balance between
regulatory and proinf lammatory T cells remains the fo-
cus of several studies as it is still unknown whether this
event contributes to the phenotypes manifested in autis-
tic patients. What has been shown is the role of cytokines,
particularly IL-6, in maintaining the balance between
T
reg and Th17s. IL-6, an important regulator in the tran-
Parker-Athill /Tan
Neurosignals 2010;18:113–128
120
sition from innate and adaptive immunity, is also a potent
modulator of T cell function, shifting the balance from
T
reg to Th17 cells, downregulating T
reg while promoting
the differentiation of naive T cells into Th17 cells [146
148] . This relationship becomes particularly important
when examined in the context of placental function and
MIA. As mentioned previously, T
reg
play an important
role in maintaining tolerance during pregnancy while
suppressing the activity of inflammatory Th17 cells,
wh ich have been impl icated in autoi mmu ne t issue i njury.
Here it can be appreciated that IL-6 expression, the result
of MIA, can promote the expression of Th17 cells while
downregulating T
reg
expression compromising placental
function while promoting a potential autoimmune re-
sponse to the developing fetus [14 4 , 149 151] . Observa-
tions of an increase in maternal antibodies against neu-
ronal and lymphocytic markers in the serum of mothers
of autistic children further strengthen this hypothesis,
suggesting the involvement of a maternal immune re-
sponse in precipitating this disorder [71, 110] . Addition-
ally, potentially more significant have been observations
of autoimmune disorders in a subset of autistic patients,
ter me d A utoi mmu ne Aut is ti c Disor de r (A A D). T hese pa -
tients exhibit increased proinflammatory cytokines and
other immunological disruptions in Th1/Th2 ratios as
well as auto-antibodies to brain myelin basic protein,
phenomenons researchers have contributed to increases
in Th17 cells potentially precipitated by prenatal viral in-
fection [67, 71, 72, 152, 153] . Equally interesting are ob-
servations made in FIRS, including altered expression of
genes associated with this arm of the immune system,
including MHC antigen presentation, and leukocyte ad-
hesion and chemotaxis [58] . Although more research is
needed to make a direct correlation between these events
and ASD, it is evident that maternal infection may impair
the ability of the placenta to regulate the maternal im-
mune system. It is also evident that resultant impairment
of placental function may allow the passage of maternal
cytokines, and/or elicit the induction of fetal inflamma-
tory cytokines, which in turn disrupts important devel-
opmental pathways, primarily those involved in the de-
velopment and function of CNS and immune system.
Despite these studies, there is still disagreement as to
whether or not these maternal cytokines, produced dur-
ing maternal infection, can traverse the placental barrier
and disrupt normal fetal development. Research in FIRS
has shown that in instances of maternal infection, the
placenta can become inflamed, producing cytokines
such as IL-6 which is a key biological marker in FIRS of-
ten detected in the amniotic fluid and maternal, fetal and
umbilical cord serum [54, 58, 154, 155] . Nevertheless, re-
search focusing on the placental migration of cytokines,
following maternal infection continues to yield contra-
dictory results as some researchers have observed mini-
mal cytokine migration while others note that the levels
that cross the placenta are too negligible to exert any ef-
fect. These contradictory findings may be attributed to
variations in experimental design as comparative studies
have shown that the ability of maternal cytokines to tra-
verse the placental barrier may be highly dependent on
the time of immune challenge. Independent work by sev-
eral groups has shown that IL-6 can traverse the rat pla-
centa to the fetus early in mid-gestation, approximately
on embryonic days 11–13, while during embryonic days
17–19, this is not seen [15 6 , 1 5 7] . This observation may
explain epidemiological findings which suggest 2nd-tri-
mester maternal infection pose the greatest risk for the
development of ASD [158] .
It is worthwhile to note, however, that even with no
observable cytokine migration during late gestation, ma-
ternal infection during this period still results in an in-
crease in cytokine levels in the fetal CNS as there have
been obser vations of increased cytokine mRNA a nd pro-
tein levels, as well as other immunological disruptions in
FIRS infants [54, 59, 78, 154] . Additionally, observations
in models of MIA mimic FIRS findings of increased in-
flammatory cytokines and other immunological events
in the fetal CNS in response to maternal injection, includ-
ing the induction of monocyte chemoattractant protein-1
and increased glial cell reactivity [156, 157] . As LPS does
not traverse the placental barrier, researchers have sug-
gested the presence of intermediary mechanisms, possi-
bly in the maternal-fetal interface, the placenta, that trig-
ger these immunological responses in the fetal CNS.
Whatever the exact mechanism, it is evident that mater-
nal infection and MIA results in increased inf lammation
in the fetal CNS as well as changes in the transcription,
expression and activity of factors that regulate the devel-
opment and function of the CNS and immune system.
IL-6 and STAT3 and ASD
Although several cytokines have been found to be el-
evated in the fetal brain following exposure to maternal
inflammatory cytokines, it has been proposed that IL-6
is the key cytokine responsible for precipitating the path-
ological effects observed in disorders such as FIRS and
ASD. There have been several key pieces of evidence
which have led to the identification of IL-6, including
findings of increased IL-6 levels in the amniotic fluid of
FIRS infants and its aberrant expression and/or function
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
121
in autoimmune disorders, some of which have been as-
sociated w ith ASD, as well as neurological disorders hall-
marked by increased inflammation such as Alzheimer’s
disease [154 , 1 59, 16 0] . Additionally, animal studies in-
volving aberrant expression of IL-6 resulted in neurolog-
ical disorders, with behavioral abnormalities analogous
to clinical observations to disorders such as ASD [10 7,
161, 162] . However, models of MIA which have isolated
IL-6 by selective inhibition of cytokines known to be el-
evated following maternal infection have been more con-
vincing. In one model proposed by Smith et al. [12] , single
maternal injections of several cytokines were coadminis-
tered with their corresponding blocking peptide. Of these
cytokines, only inhibition of IL-6 was sufficient to atten-
uate the behavioral, immunological and neurological ab-
normalities observed in offspring as a result of MIA. This
study showed improvements in measures of social inter-
action, prepulse inhibition and latent inhibition, behav-
ioral measures abnormal in ASD in adult offspring which
were comparable to observations made in control ani-
mals [12] .
The role of IL-6 as a key contributor to these patholo-
gies is further strengthened when its physiological role is
examined. As mentioned previously, IL-6 and other
members of the gp-130 family maintain the proliferation
and survival of NSC through activation of the STAT3
pathway. Additionally, these cytokines regulate neuronal
and axonal pathfinding and the formation and mainte-
nance of functional synapses. In vitro experiments and
comparative in vivo analysis have demonstrated that
many of these cytokines share redundant pathways and
functions, activating analogous pathways to accomplish
the same endpoint. Although an evolutionary favorable
characteristic, it can be seen how pathological expression
of these cytokines can lead to the dysregulation of these
pathways. IL-6 for example has been shown to maintain
the survival of and promote the proliferation of NSC in
vitro, although in vivo experiments have shown that this
may be accomplished by LIF and other members of the
gp-130 family [4 4] . Furthermore, IL-6 has been shown to
promote the differentiation of NSC in vitro, primarily
promoting a glial phenotype although it has been shown
to induce neuronal dif ferentiation in NSC, and mimic the
ac tion of ner ve grow th fa ctor in PC12 cel ls [109, 163, 164] .
In vivo, however, the induction of gliogenesis is attribut -
ed to gp-130 family member, cardiotrophin-1 although
studies have confirmed the presence of IL-6 receptors
and IL-6 during early embryonic time points [44, 108] . It
i
s plausible to hypothesize that during maternal infec-
tion, maternally derived IL-6 can traverse the placenta to
act on the fetal brain, inducing the synthesis of fetal IL-6
and/or activating common pathways such as the STAT3
pathway as several studies have shown negligible migra-
tion of IL-6 preferentially to other cytokines [165, 16 6] .
Once elevated, IL-6 can induce actions similar to those
seen in vitro, such as increased cell survival and prolif-
eration. Although in isolation or in conditions of neuro-
logical injury, these events may seem innocuous or even
beneficial; during development, these events may lead to
abnormal increases in neuronal and/or glial populations,
as the negligible expression of cytokines, such as IL-6 in
the fetal CNS, suggest that these processes are closely reg-
ulated by circulating concentrations of these cytokines.
This situation can be put into perspective when we exam-
ine clinical observations of ASD, such as increased neu-
ronal numbers in certain areas of the brain, which is be-
lieved to be a contributor to the behavioral abnormalities
manifested in these patients.
T he rol e of IL -6 in ind ucing di ff ere ntia ti on, or r eg ul at-
ing axonal guidance and synapse formation, can also be
examined in the context of clinical observations. During
normal CNS development, induction of differentiation is
highly controlled, with neurogenesis preceding gliogen-
esis. These events are not only controlled by cytokine-
induced activation of necessary pathways, but also by in-
hibition of transcription factors and methylation of gene
loci [4 4] . Duri ng neurogenesis for example, STAT3, which
has been shown to be a key transcription factor in the ac-
tivation of gliogenic genes is inhibited, by protein modi-
fication in addition to other mechanisms. Similarly, glio-
genic genes are methylated, preventing the binding of
STAT3 and these genes [44] . As neurogenesis proceeds,
proteins ass ociated wit h embryonic neurons, such a s car-
diotrophin-1 accumulates, activating the STAT3 pathway
[16 7] . This event coincides with the demethylation of
gliogenic genes as well as the inhibition of neurogenic
transcription factors. With pathological IL-6 expression,
such as that derived from maternal sources, premature
differentiation can mean disruption in neuronal or glial
populations. These events can also lead to disruption of
other important processes such as neuronal migration,
axonal pathfinding and synapse formation. Several re-
searchers hypothesize that these neurological events pre-
cipitate resultant behavioral abnormalities observed clin-
ically in autistic patients. These findings not only impli-
cate a potential ‘trigger’ for the development of ASD, but
also identify potential biological markers which may be
utilized to diagnose this disorder at earlier time points
similarly to clinical diagnosis of FIRS. Early diagnosis
does not necessarily mean early intervention, but the
Parker-Athill /Tan
Neurosignals 2010;18:113–128
122
identification of a biological marker may enable the use
of pharmacological intervention in the management of
this disorder.
Viral Pathogens, MIA and ASD
To date several researchers have cited a viral-induced
activation of the maternal immune system, MIA, as the
primary contributor to the behavioral abnormalities ob-
served in animal models of MIA and in ASD [68] . Smith
et al. [1 2 ] , utiliz ing an anima l model of MIA have strength-
ened this hypothesis, finding that the activation of the
maternal im mune system in response to viral e xposu re is
a key mechanism in precipitating the behavioral abnor-
ma lities in M IA offspr ing. Furthermore, t his group iden-
tified IL-6 as a key cytokine in this mechanism as inhib-
iting this cytokine attenuated the behavioral abnormali-
ties observed in offspring exposed to MIA prenatally [1 2] .
Other groups, however, have cited a potential alternative
mechanism to the changes seen in these offspring, citing
the actions of the viral pathogen itself as a key precipita-
tor in ASD neuropathologies. In a study in which preg-
nant mice were injected with human influenza at embry-
onic day 9, Fatemi et al. [11] observed changes in protein
expression in several key proteins involved in neurode-
velopment, most notable of which was connexin 43. In
another study by the same group, alternations in expres-
sion of several genes including those regulating myelin
expression were also observed following prenatal viral
infection [43, 168, 169] .
It is important to note that although Smith et al. [1 2]
and others cited increases in maternal cytokine levels,
most notably IL-6, as a key factor in these disruptions,
they also noted changes in gene expression following vi-
ral exposure. Additionally, in cases of FIRS, although im-
munological disruptions such as increased IL-6 levels
were noted as one of the most significant characteristics,
changes in gene expression were also observed in genes
involved in immune regulation [58] . Fatemi’s work does
not preclude the role of the maternal immune response in
precipitating the changes observed in models of MIA and
clinically in ASD, but rather suggest the importance of
examining the mechanistic aspects of viral activation of
the immune system and the potential downstream ef fects
on gene transcription and protein expression. It can be
argued that either mechanism may be a precipitating fac-
tor as it is the initial viral exposure that initiates the acti-
vation of the maternal immune system and resultant in-
flammatory cascade. Although it is unclear whether the
changes we observed in MIA and clinically in ASD can
be attributed solely to either mechanism, or even wheth-
er these two can be separated, it is evident that prenatal
viral exposure leads to an increase in maternal cytokine
exposure and IL-6 levels and altered gene expression, and
that these factors have the potential to precipitate the
behavioral and neuropathologies observed clinically in
ASD.
Current Therapeutic Approaches
Currently there is no cure for ASD and therapeutic in-
tervention focuses primarily on the management of
symptoms, utilizing behavioral therapy to control the ab-
norma l behavioral phenotypes associated with the disor-
der. Although pharmacological intervention is not com-
mo nl y u se d i n t he ma na geme nt of A SD, it is of te n ut il iz ed
in the treatment of comorbid disorders, which frequently
occur with ASD, including seizures, increased aggression
and other pharmaceutically responsive conditions. The
use of palliative care in the management of development
disorders is not uncommon, as many, especially those of
genetic origin are incurable, making the management of
symptoms the only possible therapeutic intervention.
Recent research in ASD has highlighted the role of ma-
ternal infection in the etiology of ASD. More specifically,
they have identified exposure to maternal cytokines, pri-
marily IL-6, as a key environmental ‘trigger’ precipitating
the development of ASD in some individuals. These find-
ings are significant not only in understanding the etiology
of ASD, but the identification of IL-6 may provide a po-
tential biological marker enabling the early diagnosis of
the disorder and earlier therapeutic intervention. The
identification of IL-6 has also provided a potential thera-
peu ti c t ar ge t a s model s o f M IA ha ve s ho wn th at in hi biti ng
IL-6 during maternal infection can attenuate the behav-
ioral abnormalities observed in MIA offspring. In studies
by Smith et al. [12] , coad ministration of poly(I:C), used to
induce MIA and IL-6 neutralizing antibody, attenuated
the behavioral abnormalities observed in the offspring, as
the y obs er ved imp roveme nt i n prepu ls e inhibit ion a nd la-
tent inhibition comparable to observations in control an-
imals. Similarly, research by our group has also conf irmed
IL-6 as a potential therapeutic target, sufficient to attenu-
ate the symptoms associated with ASD. Furthermore, we
discovered that inhibition of IL-6-induced activation of
STAT3 was also able to attenuate the behavioral and im-
munological abnormalities observed in MIA offspring. In
vitro experiments utilizing an LPS-microglial model to
induce inf lammation showed that biof lavonoid pretreat-
ment or cotreatment was sufficient to inhibit the produc-
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
123
tion of IL-6 and TNF- , while IL-6 and bioflavonoid co-
treatment of neuronal cultures showed t hat biof lavonoids
could also inhibit IL-6-induced activation of STAT3 [113] .
These results, which support previous findings by our
group and others, suggest that bioflavonoid treatment not
only has the potential to inhibit pathological cytokine
production, but also to inhibit cytokine-induced activa-
tion of key signaling molecules such as STAT3 [17 0 , 171] .
Furthermore, in vivo experiments have shown similar re-
sults, with prophylactic bioflavonoid treatment reducing
cytokine expression and STAT3 activity in MIA offspring
[113] . These observations suggest that it is not only pos-
sible to attenuate the pathological increase in cytokine ex-
pression resulting from maternal infection, but therapeu-
tic intervention to attenuate the resultant activation of
critical signaling pathways may also be possible. These
observations further suggest that prophylactic interven-
tion may serve a protective role, especially in instances of
known maternal infection. Similarly to how folic acid has
been shown to be essential in the prevention of cephalic
disorders, inhibition of pathological maternal cytokine
expression may have therapeutic potential. Furthermore,
postnatal bioflavonoid treatment to attenuate the activi-
ties of IL-6, such as pathological activation of STAT3
pathways, may prove to be of potential therapeutic value
by providing more than a palliative treatment for ASD.
Although these results are preliminary, and there are still
many unanswered questions concerning the role mater-
nal infection plays in precipitating ASD, it is evident that
the IL-6 plays a key role in precipitating this disorder,
making it a key target for therapeutic intervention. It is
necessary not only to determine the mechanism by which
maternal infection and IL-6 precipitate this disorder and
how therapeutic intervention can aid in managing the
symptoms associated with ASD, but also to attenuate the
symptoms of this disorder.
Discussion
Since its first use in the early 1900s to describe a clus-
ter of abnormal symptoms observed in a group of schizo-
phrenics, and later similar symptoms in children, there
have been significant advances in the understanding and
treatment of ASD. There are still, however, many unan-
swered questions concerning the etiology of ASD as ge-
netic variability among autistic individuals coupled with
diverse phenotypic presentations have complicated the
identification of a single causative agent. Nevertheless,
researchers have made significant progress and have
identified several genes differentially expressed in autis-
tic individuals. Although these genes are not sufficient to
cause ASD, they may provide a way to identify geneti-
cally susceptible individuals, or aid in the diagnosis of
this disorder. Recently, maternal infection and fetal ex-
posure to maternal cytokines, primarily IL-6, have been
identified as potential environmental ‘trigger’, which,
when coupled with certain genetic polymorphism, may
lead to the development of ASD. Although the exact
mechanism by which IL-6 elicits these events is un-
known, its identification provides a potential diagnostic
marker and therapeutic intervention point for ASD.
These advancements are promising; however, several
unanswered questions still remain, prompting the need
for more research. The identification of IL-6 as a potential
trigger, for example, has raised questions concerning the
ability of cytokines to traverse the placental barrier, and
whether or not this passage is confined to certain devel-
opmental time points. Also unknown is whether or not
the placenta itself contributes to the increase in fetal cy-
tokine expression, as this has been observed in the ab-
sence of maternal cytokine migration. Researchers are
also faced with the question concerning the mechanism
by which IL-6 induces the abnormalities observed clini-
cally in autism and whether or not inhibition of patho-
logical IL-6 is sufficient to attenuate these abnormalities.
It is also unclear as to whether or not maternally derived
IL-6 is sufficient to promote the development of ASD or
whether certain genetic polymorphisms are needed. Al-
though animal models of MIA suggest that IL-6 is suffi-
cient to induce the behavioral, immunological and neu-
rological abnormalities observed in ASD, it is still unclear
whether these findings completely mirror clinical obser-
vations. The role of genetics in the etiology of ASD also
poses several questions including why some people, ex-
posed prenatally to maternal cytokines may develop ASD
while others do not.
As we investigate the etiology of this disorder, there
are also important questions that arise concerning the
diagnosis of this disorder. Although it is well accepted
that autism is a part of a spectrum of disorders collec-
tively referred to as ASD, there are some within the re-
search and clinical community that would classify it as a
syndrome, a singular manifestation of different disor-
ders. This approach may explain the difficulty in deter-
mining the exact etiology of the disorder as well as the
diversity of phenotypes presented clinically. Proponents
of the syndrome approach argue that the term spectrum
sug gests a si ng ul ar di sor de r w it h c li n ica l s ym pt om s v ar y-
ing within a given phenotypic range or spectrum. As a
Parker-Athill /Tan
Neurosignals 2010;18:113–128
124
syndrome, researchers such as Mary Coleman and Chris-
topher Gillberg suggest that autism is a singular pheno-
typic presentation of different diseases with varying eti-
ologies [17 2] . This approach suggests that autism may be
a phenotype manifested as a result of different diseases
and etiologies acting singularly or synergistically to pro-
duce the behavioral symptoms manifested clinically. Al-
though Coleman’s and Gillberg’s suggestions differ from
current norms within the ASD community, they do ac-
knowledge the diagnostic importance of investigating
autism as a spect rum disorder as it is necessa ry in t he i ni-
tial diagnosis. It is for the investigation of etiologies and
the provision of therapeutics that these authors propose
newer approaches, as the treatment of autism as a product
of its etiology may be advantageous in providing more
patient-tailored treatments. Whether a syndrome or a
spectrum, it is evident that autism or ASD is the result of
several etiologies. It is also evident that environmental
triggers, when coupled with genetic polymorphisms, may
lead to diverse phenotypic presentations. What is not
known is how these factors, genetics and environment,
interact in a given individual to precipitate this disorder.
References
1 Rodier PM: The early origins of autism. Sci
Am 2000;
282: 56–63.
2 Chakrabarti S, Fombonne E: Pervasive de-
velopmental d isorders in preschool c hildren.
JAMA 2001;
285: 3093–3099.
3 Gillberg C: Neurodevelopmental processes
and psychological functioning in autism.
Dev Psychopathol 1999;
11: 567–587.
4 Trevarthen C: Autism as a neurodevelop-
mental disorder affecting communication
and lear ning in early chi ldhood: prenatal or-
igins, post-natal course and effective educa-
tional support. Prostaglandins Leukot Es-
sent Fatty Acids 2000;
63: 41–46.
5 Zwaigenbau m L, Bryson S , Rogers T, Roberts
W, Brian J, Szatmari P: Behavioral manifes-
tations of autism in the f irst year of life. Int J
Dev Neurosci 2005;
23: 143–152 .
6 Koenig K, Scahill L: Assessment of children
with pervasive developmental disorders. J
Child Adolesc Psychiatr Nurs 2001;
14: 159–
166 .
7 America n Psychiatric As sociation: Diag nos-
tic and Statistical Manual of Mental Disor-
ders. Arl ington, Americ an Psychiatric A sso-
ciation, 1994.
8 Allen DA, Steinberg M, Dunn M, Fein D,
Feinstein C, Waterhouse L, Rapin I: Autistic
disorder ver sus other perva sive developmen-
tal disorders in young children: same or dif-
ferent? Eur Child Adolesc Psychiatry 2001;
10: 67–78.
9 Charman T, Baron-Cohen I, Bai rd G, Cox A,
Wheelwright S, Swettenham J, Drew A:
Commentary: the modif ied checklist for au-
tism in toddlers. J Autism Dev Disord 2001;
31: 145–148 , d iscus sion 149 –151.
10 Hertz-Picciotto I, Croen LA, Hansen R,
Jones CR, van de Water J, Pessah IN: The
CHARGE study: an epidemiologic investiga-
tion of genetic and environmental factors
contributing to autism. Environ Health Per-
spect 2006;
114: 1119–11 25.
11 Fatem i SH, Reutiman TJ, Fol som TD, Huang
H, Oishi K, Mori S, Smee DF, Pearce DA,
Winter C, Sohr R, Juckel G: Maternal infec-
tion leads to abnormal gene regulation and
brain atrophy in mouse offspring: implica-
tions for genesis of neurodevelopmental dis-
orders. Schizophr Res 2008;
99: 56–70.
12 Smith SE, Li J, Garbett K, Mirnics K, Patter-
son PH: Maternal immune activation alters
fetal brain development through interleu-
kin-6. J Neurosci 2007;
27: 10695 –10702.
13 Kanner L: Autistic disturbances of affective
contact. Acta Paedopsychiatr 1968;
35: 100
136.
14 World Health Organization: International
Classification of Diseases. Geneva, World
Health Organization, 1992.
15 Wi ng L, Yeates SR, Brierley LM, Gou ld J: The
prevalence of early childhood autism: com-
parison of administrat ive and epidemiologi-
cal studies. Psychol Med 1976;
6: 89–100.
16 Fombon ne E: Epidemiologica l surveys of au-
tism and ot her pervasive developmenta l dis-
orders: an upd ate. J Autism Dev Dis ord 2003;
33: 365–382.
17 K iel ine n M, L in na SL , Mo ila nen I: Au tis m in
Northern Finland. Eur Child Adolesc Psy-
chiatry 2000;
9: 162–167.
18 Chen CY, Liu CY, Su WC, Huang SL, Lin
KM: Factors associated wit h the diagnosis of
neurodevelopmental disorders: a popula-
tion-based longitudinal study. Pediatrics
2007;
119:e4 35 –e 443.
19 Honda H, Shimizu Y, Imai M, Nitto Y: Cu-
mulative incidence of childhood autism: a
total popu lation study of be tter accurac y and
precision. Dev Med Child Neurol 2005;
47:
10–18.
20 Wing L: Sex ratios in early chi ldhood autism
and related conditions. Psychiatry Res 1981;
5: 129–137.
21 Resch B, Radinger A, Mannhalter C, Binder
A, Haas J, Muller WD: Interleukin-6 g(–174)
c polymorphism is associated with mental
retardation in cystic periventricular leuco-
malacia in preterm infants. Arch Dis Child
Fetal Neonatal Ed 2009;
94:F304–F306.
22 Bolton PF, Dennis NR, Browne CE, Thomas
NS, Veltman MW, Thompson RJ, Jacobs P:
The phenoty pic manifest ations of interstit ial
duplications of proximal 15q with special
reference to the autistic spectrum disorders.
Am J Med Genet 2001;
105: 675–685.
These are just a few of the questions facing researchers
today, and although significant advancements have been
made, these advancements have also created more ques-
tions. Nevertheless, the identification of IL-6, among
other discoveries, has paved the way for improved diag-
nostic criteria in addition to new therapeutic intervention
points. The potential for IL-6 as an intervention point is
also promising as it provides a measurable diagnostic
marker in addition to potential therapeutic target. With
these advancements, early therapeutic intervention, with
the goal of significantly attenuating or alleviating the
symptoms of ASD, may eventually become a treatment
option as opposed to strictly palliative therapy.
Acknowledgments
This work was supported by the Silver Endowment and the
NIH/NIMH (R21MH087849, J.T.). J.T. holds the Silver Chair in
Developmental Neurobiology. We thank Demian Obregon (Uni-
versity of South Florida) for his helpful advice.
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
125
23 Boyar FZ, Whitney MM, Lossie AC, Gray
BA, Keller KL , Stalker HJ, Zori RT, Geffken
G, Mutch J, Edge PJ, Voeller KS, Williams
CA, Driscoll DJ: A family with a grand-ma-
ternal ly derived interstitial duplication of
proximal 15q. Clin Genet 2001;
60: 421430.
24 Demark JL , Feldman MA, Holden JJ: Behav-
ioral relat ionship between aut ism and frag ile
X syndrome. Am J Ment Retard 2003;
108:
314–326.
25 Marui T, Funatogawa I, Koi shi S, Yamamoto
K, Matsumoto H, Hashimoto O, Nanba E,
Nishida H, Sugiyama T, Kasai K, Watanabe
K, Kano Y, Sasaki T, Kato N: Association
of the neurona l cell adhesion molecule
(NRCAM) gene variants with autism. Int J
Neuropsychopharmacol 20 09;
12: 1–10.
26 Brown AS, Schaefer CA, Wyatt RJ, Goetz R,
Begg MD, Gorman JM, Susser ES: Maternal
exposure to respirator y infections and adult
schizophrenia spectrum disorders: a pro-
spective birth cohort study. Schizophr Bu ll
2000;
26: 287–295.
27 Schaefer CA, Brown AS, Wyatt RJ, Kline J,
Begg MD, Bresnahan MA, Susser ES: Mater-
nal prepregnant body mass and risk of
schizophrenia in adult offspring. Schizophr
Bull 2000;
26: 275–286.
28 Susser ES, Schaefer CA, Brown AS, Begg
MD, Wyatt RJ: The design of the prena-
tal determinants of schizophrenia study.
Schizophr Bull 2000;
26: 257–273.
29 Susser ES , Lin SP: Schiz ophrenia aft er prena-
tal exposure to the Dutch Hunger Winter of
1944 –1945. Arch Gen Psychiatry 1992;
49:
983–988.
30 Li Q, Cheung C, Wei R, Hui ES, Feldon J,
Meyer U, Chung S, Chua SE, Sham PC, Wu
EX, McAlonan GM: Prenatal immune
challenge is an environmental risk factor
for brain and behavior change relevant to
schizophrenia: evidence from MRI in a
mouse model. PLoS One 2009;
4:e6354.
31 Hwang SJ, Chen YS: Congenital rubella syn-
drome wit h autistic dis order. J Chin Med As-
soc;73:
104–107.
32 Assumpcao FB Jr, Kuczy nski E: Autism, bi-
polar disorder and menta l retardation in a
ma l e a d ol es c en t w it h co ng e ni ta l r ub el l a: c as e
report (in Portuguese). Arq Neuropsiquiatr
2002;
60: 324–327.
33 Libbey JE, Sweeten TL, McMa hon WM, Fu-
jinami RS: Autistic disorder and viral infec-
tions. J Neurovirol 2005;
11: 1–10.
34 Anlar B, Oktem F, Torok T: Human parvo-
virus b19 antibodies in infantile autism. J
Child Neurol 1994;
9: 104–105.
35 Deykin EY, MacMahon B: Viral exposure and
autism. Am J Epidemiol 1979;
109: 628–638.
36 Chen W, Landau S, Sham P, Fombonne E: No
evidence for l inks between autism, MM R and
measles v irus. Psychol Med 20 04;
34: 543–553.
37 Da ssa D, Takei N, Sha m PC, Murray RM : No
association between prenatal exposure to in-
fluenza and autism. Acta Psychiatr Scand
1995;
92: 145 –149.
38 Chess S: Follow-up report on autism in con-
genital rubella. J Autism Child Schizophr
1977;
7: 69–81.
39 Chess S: Autism in children with congenital
rubella. J Autism Child Schizophr 1971;
1:
33–47.
40 Atladottir HO, Thorsen P, Ostergaard L,
Schen del DE , Lemcke S, Ab dal lah M , Par ner
ET: Maternal infection requiring hospital-
ization during pregnancy and autism spec-
trum disorders. J Autism Dev Disord 2010,
E-pub ahead of print.
41 Markowitz PI: Autism in a child with con-
genital c ytomega lovirus infection. J Autism
Dev Disord 1983;
13: 249–253.
42 Ritvo ER, Mason-Brothers A, Freeman BJ,
Pingree C, Jenson WR, McMahon WM, Pe-
tersen PB, Jorde LB, Mo A, Ritvo A: The
UCLA-University of Utah epidemiologic
survey of autism: the etiologic role of rare
diseases. Am J Psychiatry 1990;
147: 1614–
1621.
43 Fatemi SH, Folsom TD, Reutiman TJ, Abu-
Odeh D, Mori S, Huang H, Oishi K: Abnor-
mal expression of myelination genes and
alterations in white matter fractional a niso-
tropy following prenatal vira l inf luenza in-
fection at e16 in mice. Schizophr Res 2009;
112: 46–53.
44 Deverman BE, Patterson PH: Cytokines and
CNS development. Neuron 2009;
64: 61–78.
45 Geier DA, King PG, Sykes LK, Geier MR: A
comprehensive review of mercur y provoked
autism. Indian J Med Res 2008;
128: 383–411.
46 Herbert MR: Contributions of the environ-
ment and environmenta lly vulnerable physi-
ology to autism spectrum disorders. Curr
Opin Neurol;23:
103–110.
47 Yor bik O, K ur t I, H as im i A, Ozt ur k O: Ch ro-
mium, cadmium, and lead levels in urine of
children with autism and typically develop-
ing controls. Biol Trace Elem Res;135:
10–15.
48 Adams JB, Holloway CE, George F, Quig D:
Analyses of toxic metals and essential min-
erals i n the hair of Ar izona children w ith au-
tism and associated conditions, and their
mothers. Biol Trace Elem Res 2006;
110: 193–
209.
49 Dietert RR, Dietert JM: Possible role for ear-
ly-life immune insult including develop-
mental immunotoxicity in chronic fatigue
syndrome (CFS) or mya lgic encephalomyel i-
tis (ME). Toxicology 2008;
247: 61–72.
50 Fatemi SH, Earle J, Kanodia R, Kist D,
Emami an ES, Patterson PH, Sh i L, Sidwell R:
Prenatal viral infection leads to py ramidal
cell atrophy and macrocephaly in adult-
hood: implications for genesis of autism and
schizophrenia. Cell Mol Neurobiol 2002;
22:
25–33.
51 Fuksman RB, Mazzitelli NG: Second-tri-
mester histopathological placenta l findings
in maternal-fetal inflammatory response
syndrome. Pediatr Dev Pat hol 2009;
12: 42–
46.
52 Go lan H, K ashtuts ky I, Hal lak M, So roki n Y,
Huleihel M: Maternal hypoxia during preg-
nancy delays the development of motor re-
flexes in newborn mice. Dev Neurosci 2004;
26: 24–29.
53 Golan H, Kashtuzki I, Hallak M, Sorokin Y,
Huleihel M: Maternal hypoxia during preg-
nancy induces fetal neurodevelopmental
brain damage: partia l protection by magne-
sium sulfate. J Neurosci Res 2004;
78: 430–
441.
54 Gomez R, Romero R, Ghezzi F, Yoon BH,
Mazor M, B erry SM: The feta l infla mmatory
response sy ndrome. Am J Obstet Gynecol
1998;
179: 194–202.
5 5 Hu l ei he l M , G o la n H , H a ll a k M : I nt r aut e ri ne
infection/inflammation during pregnancy
and offs pring brain d amages: Possible mech-
anisms involved. Reprod Biol Endocrinol
2004;
2: 17.
56 Lahra MM, Beeby PJ, Jeffery HE: Intrauter-
ine inf lammation, neonatal sepsis, and
chronic lung disease: a 13-year hospita l co-
hort study. Pediatrics 2009;
123: 1314–1319.
57 Minkoff H: Prematurity: infection as an eti-
ologic factor. Obstet Gynecol 1983;
62: 137–
144 .
58 Madsen-Bouterse SA, Romero R, Tarca AL,
Kusanovic JP, Espinoza J, Kim CJ, Kim JS,
Edwin SS, Gomez R, Draghici S: The tran-
scriptome of the fetal inflammator y re-
sponse syndrome. Am J Reprod Immu-
nol;63:
73–92.
59 Romero R, Maymon E, Pacora P, Gomez R,
Mazor M, Yoon BH, Berr y SM: Further ob-
servations on the fetal inf lammatory re-
sponse syndrome: a potential homeostatic
role for the soluble rece ptors of tumor necro-
sis factor alpha. Am J Obstet Gynecol 2000;
183: 1070–1077.
60 Ornoy A, Diav-Citrin O: Fetal effects of pri-
mary and secondary cy tomegalovirus infec-
tion in pregnancy. Reprod Toxicol 2006;
21:
399–409.
61 Girard S, Kadhim H, Roy M, Lavoie K, Bro-
chu ME, Larouche A, Sebire G: Role of peri-
natal inflammation in cerebral palsy. Pediatr
Neurol 2009;
40: 168–174 .
62 Garnier Y, Coumans AB, Jensen A, Hasaart
TH, Berger R: Infection-related perinatal
brain i njury: the pat hogenic role of impaired
fetal cardiovascular control. J Soc Gynecol
Investig 2003;
10: 450–459.
63 Romero R, Gotsch F, Pineles B, Kusanovic
JP: Inf lammation in pregnancy: its roles in
reproductive physiology, obstetrical compli-
cations, and fetal injury. Nutr Rev 2007;
65:S194–S202.
64 Pleasure D, Soulika A, Singh SK, Gallo V,
Bannerman P: Inflammation in white mat-
ter: clinical and pathophysiological aspects.
Ment Retard Dev Disabil Res Rev 2006;
12:
141–14 6.
65 Benvenuto A, Moavero R, Alessandrelli R,
Manzi B, Curatolo P: Syndromic autism:
causes and pathogenetic pathways. World J
Pediatr 20 09;
5: 169–176.
Parker-Athill /Tan
Neurosignals 2010;18:113–128
126
66 Blaylock RL: A possible central mechanism
in aut ism s pect rum disord ers, part 2: im mu-
noexcitotoxicity. Altern Ther Health Med
2009;
15: 60–67.
67 Li X, Chauhan A, Sheikh AM, Pati l S, Chau-
han V, Li XM, Ji L, Brow n T, Ma lik M: Elevat-
ed immune response in the brain of autistic
patients. J Neuroimmu nol 2009;
207: 111–116.
68 Patterson PH: Immune involvement in
schizophrenia and autism: etiology, pathol-
ogy and animal models. Behav Brain Res
2009;
204: 313–321.
69 Singer HS, Morris C, Gause C, Pollard M,
Zimmerman AW, Pletnikov M: Prenatal ex-
posure to antibodies from mothers of chil-
dren with autism produces neurobehavioral
alterations: a pregnant dam mouse model. J
Neuroimmunol 2009;
211: 39–48.
70 Jyonouchi H, Geng L, Ruby A, Zimmerman-
Bier B: Dysregulated innate immune re-
sponses in young children with autism
spectrum disorders: their relationship to gas-
trointestinal symptoms and dietary inter-
vention. Neurop sychobiology 2005 ;
51: 77–85.
71 Cohly H H, Panja A: Immunolog ical find ings
in autism. Int Rev Neurobiol 2005;
71: 317–
341.
72 Croonenberghs J, Bosmans E, Deboutte D,
Kenis G, Maes M: Activation of the inflam-
matory response system in autism. Neuro-
psychobiology 2002;
45: 1–6.
73 Vargas DL, Nascimbene C, Krishnan C,
Zimmerman AW, Pardo CA: Neuroglial ac-
tivation a nd neuroinf lammat ion in the brai n
of patients w ith autism. An n Neurol 2005;
57:
67–81.
74 DeFelice ML, Ruchelli ED, Markowitz JE,
Strogatz M, Reddy KP, Kadivar K, Mulberg
AE, Brown KA: Intestinal cytokines in chil-
dren with pervasive developmental disor-
ders. Am J Gastroenterol 2 003;
98: 1777–1782.
75 Meyer U, Engler A, Weber L , Schedlowski M,
Feldon J: Prelim inary ev idence for a modula-
tion of fetal dopaminergic development by
maternal immune activation during preg-
nancy. Neuroscience 2008;
154: 701–709.
76 Meyer U, Feldon J: Prenatal exposure to in-
fection: a primar y mechanism for abnorma l
dopaminergic development in schizophre-
nia. Psychopharmacology (Berl) 2009;
206:
587–602.
77 Meyer U, Nyffeler M, Yee BK, Knuesel I, Fel-
don J: Adult brain and behav ioral pathologi-
cal markers of prenatal immune challenge
during early/middle and late fetal develop-
ment in mice. Brain Behav Immun 2008;
22:
469–486.
78 Ash down H, D umont Y, Ng M, Poole S , Bok-
sa P, Luheshi GN: The role of cy tokines in
mediating effects of prenatal infection on the
fetus: implications for schizophrenia. Mol
Psychiatry 2006;
11: 47–55.
79 Lauritsen MB, Pedersen CB, Mortensen PB:
Effects of fami lial risk factors and place of
birth on the risk of autism: a nat ionwide reg-
ister-based study. J Child Psychol Psychiat ry
2005;
46: 963–971.
80 Mu hle R, Trentacoste SV, Rapin I: The genet-
ics of autism. Pediatrics 2004;
113:e472–4 86 .
81 Smalley SL: Genetic influences in autism.
Psychiatr Clin North Am 1991;
14: 125–139.
82 Sumi S, Taniai H, Miyachi T, Tanemura M:
Sibling risk of pervasive developmental dis-
order estimated by means of an epidemio-
logic survey in Nagoya, Japan. J Hum Genet
2006;
51: 518–522 .
83 Virkud YV, Todd RD, Abbacchi AM, Zhang
Y, Constantino JN: Familial aggregation of
quantitative autistic traits in multiplex ver-
sus simplex autism. Am J Med Genet B Neu-
ropsychiatr Genet 2009;
150B:328–334 .
84 Bai ley A, Le Couteur A, Gottesma n I, Bolton
P, Simonoff E, Yuzda E, Rutter M: Autism as
a strongly genetic disorder: evidence from a
British t win study. Psychol Med 1995;
25: 63–
77.
85 Badcock C, Crespi B: Imbalanced genomic
imprinting in brain development: an evolu-
tionary basis for the aetiology of autism. J
Evol Biol 2006;
19: 1007–1032.
86 Sma lley SL, Tanguay PE , Smith M, Gutierrez
G: Autism and tuberous sclerosis. J Autism
Dev Disord 1992;
22: 339–355.
87 Wiz nitzer M: Autism and t uberous sclerosis.
J Child Neurol 2004;
19: 675–679.
88 Roge rs SJ, Wehner DE, Ha german R: The be-
havioral phenotype in fragile X: symptoms
of a uti sm in very young c hil dre n wit h fr agi le
x syndrome , idiopathic autism , and other de-
velopmental disorders. J Dev Behav Pediatr
2001;
22: 409–417.
89 Moss J, Howlin P: Autism spectrum disor-
ders in genetic syndromes: implications for
diagnosis, intervention and understanding
the wider autism spectrum disorder popula-
tion. J Intel lect Disabil Res 2009;
53: 852–873.
90 Belmonte MK, Bourgeron T: Fragile X syn-
drome and autism at the intersection of ge-
netic and neural net works. Nat Neurosci
2006;
9: 1221–1225.
91 Feinstein C, Reiss AL: Autism: The point of
view from fragile X studies. J Autism Dev
Disord 1998;
28: 393–405.
92 Losh M, Sullivan PF, Trembath D, Piven J:
Current developments in the genetics of au-
tism: from phenome to genome. J Neuor-
pathol Exp Neurol 2008;67:829–837.
93 Bernardet M, Crusio WE: Fmr1 KO mice as
a possible model of autistic features. Scienti-
ficWorldJournal 2006;
6: 1164 –1176.
94 Blundel l J, Blaiss CA, Et herton MR, Es pinosa
F, Tabuchi K, Walz C, Bolliger MF, Sudhof
TC, Powell CM: Neuroligin-1 deletion re-
sults in impaired spatial memory and in-
creased repetitive behavior. J Neurosci
2010;30:
2115–2129.
95 Dahlhaus R, El-Husseini A: Altered neuroli-
g in e xp re ss ion is in vol ve d i n so ci al de fi ci ts in
a mouse model of the fragile X syndrome.
Behav Brain Res 2010;
208: 96–105.
96 Kantojarvi K, Onkamo P, Vanhala R, Alen
R, Hedman M, Sajantila A, Nieminen-von
Wendt T, Jarvela I: Analysis of 9p24 and
11p12–13 regions in autism spectrum dis-
orders: rs1340513 in the J MJD2C gene is as-
so ciat ed wi th AS Ds in Finni sh sa mple . Psy-
chiatr Genet 2010;
20: 102–108 .
97 Ritvo ER, Spence M A, Freeman BJ, Mason-
Brothers A, Mo A, Marazita ML: Evidence
for autosomal recessive inheritance in 46
fa m il ie s w i th mu lt ip le in ci de nce s o f a ut is m.
Am J Psychiatry 1985;
142: 187–192.
9 8 Na kashi ma N, Ya maga ta T, Mori M, K uwa-
jima M, Suwa K, Momoi MY: Expression
analysis and mutation detection of DLX5
and DLX6 in autism. Brain Dev 2010;
32:
98–104.
99 Tetreault NA, Williams BA, Hasenstaub A,
Hakeem AY, Liu M, Abelin ACT, Wold BJ,
Allman JM: RNA-seq Studies of Gene Ex-
pression in Fronto-Insular (FI) Cortex in
Autistic and Control Subjects Reveal Gene
Networks Related to Inflammation and
Synaptic Function. Chicago, Society for
Neuroscience, 2009.
100 Holst D, Garnier Y: Preterm birth and in-
flammation-the role of genetic polymor-
phisms. Eur J Obstet Gynecol Reprod Biol
2008;
141: 3–9.
101 A kira S: Il- 6-regulat ed transcr iption factors.
Int J Biochem Cell Biol 1997;
29: 1401–1418.
102 Heinrich PC, Behrmann I, Muller-Newen
G, Schaper F, Graeve L: Interleukin-6-type
cytokine signalling through the gp130/Jak/
STAT pathway. Biochem J 1998;
334: 297–
314.
103 Bromberg J, Wang TC: Inf lammation and
cancer: Il-6 and STAT3 complete the link.
Cancer Cell 2009;
15: 79–80.
104 Gabay C: Interleukin-6 and chronic in-
flammation. Arthritis Res Ther 2006;
8(Suppl 2):S3.
105 Heinrich PC, Horn F, Graeve L, Dittrich E,
Kerr I, Muller-Newen G, Grotzinger J,
Wollmer A: Interleu kin-6 and related cyto-
kines: effect on the acute phase reaction. Z
Ernahrungswiss 1998;
37(Suppl 1):43–49.
106 Rojo LE, Fernandez JA, Maccioni AA, Ji-
menez JM, Maccioni RB: Neuroinf lamma-
tion: implications for the pathogenesis and
molecula r diagnosis of A lzheimer’s diseas e.
Arch Med Res 2008;
39: 1–16.
107 Campbell IL, Abraham CR, Masliah E,
Kemper P, Inglis JD, Oldstone MB, Mucke
L: Neurologic disease induced in transgen-
ic mice by cerebral overexpression of inter-
leukin 6. Proc Natl Acad Sci USA 1993;
90:
10061–10065.
108 Bauer S, Kerr BJ, Patterson PH: The neuro-
poietic cy tokine family in development,
plasticity, disease and injur y. Nature Rev
2007;
8: 221–232.
109 I slam O, Gong X, Rose-John S, Hee se K: In-
terleukin-6 and neural stem cells: more
than gliogenesis. Mol Biol Cell 2009;
20:
188–199.
Maternal Immune Activation and Autism
Spectrum Disorder
Neurosignals 2010;18:113–128
127
110 Brau nschweig D, Ashwood P, Krakowia k P,
Hertz-Picciotto I, Hansen R, Croen LA,
Pessah IN, Van de Water J: Autism: mater-
nally derived antibodies specif ic for fetal
brain proteins. Neurotoxicology 2008;
29:
226–231.
111 Jonakait GM: The effects of materna l in-
flammation on neuronal development:
possible mechanisms. Int J Dev Neurosci
2007;
25: 415–4 25.
112 Zhao B, Schwartz JP: Involvement of cyto-
kines in normal CNS development and
neurological diseases: recent progress and
perspectives. J Neurosci Res 1998;
52: 7–16.
113 Parker-Athill E, Luo D, Bailey A, Giunta B,
Tian J, Shytle RD, Murphy T, Legradi G,
Tan J: Flavonoids, a prenata l prophylax is
via targeting JAK 2/STAT3 signaling to op-
pose IL- 6/MIA associated autism. J Neuro-
immunol 2009;
217: 20–27.
114 Nakanishi M, Niidome T, Matsuda S,
Akaike A, Kihara T, Sugimoto H: Microg-
lia-der ived interleuki n-6 and leuk aemia in-
hibitory factor promote astroc ytic differen-
tiation of neural stem/progenitor cells. Eur
J Neurosci 2007;
25: 649–658.
115 Rothwell NJ, Luheshi G, Toulmond S: Cy-
tokines and their receptors in the central
nervous s ystem: physiology, pharm acology,
and patholog y. Pharmacol Ther 1996;
69:
85–95.
116 Bromberg KD, Iyengar R, He JC: Regula-
tion of neurite outgrowt h by G(i/o) signal-
ing pathways. Front Biosci 2008;
13: 4544–
4557.
117 He F, Ge W, Martinowich K, Becker-Cata-
nia S, Coskun V, Zhu W, Wu H, Castro D,
Guillemot F, Fan G, de Vellis J, Sun YE: A
positive autoregulatory loop of Jak-STAT
signaling controls the onset of astroglio-
genesis. Nat Neurosci 2005;
8: 616–625.
118 Dame JB, Juul SE: The distribution of re-
ceptors for the pro-inf lammatory cyto-
kines interleukin (IL)-6 and IL-8 in the de-
veloping human fetus. Early Hum Dev
2000;
58: 25–39.
119 Ulfig N, Friese K: Interleukin-6 receptor is
highly expressed in the ga nglionic emi-
nence of the human fetal brain. Biol Neo-
nate 1999;
76: 320 –324.
120 Pousset F: Developmenta l expression of cy-
tokine genes in the cortex and hippocam-
pus of the rat central ner vous system. Brain
Res 1994;
81: 143–14 6.
121 Burns TM, Clough JA, Klein RM, Wood
GW, Berman NE: Developmental regula-
tion of cytokine expression in the mouse
brain. Growth Factors 1993;
9: 253–258.
122 Giulian D, Young DG, Woodward J, Brown
DC, Lac hman LB: Interleu kin-1 is an astro-
glial growt h factor in the developing brain.
J Neurosci 1988;
8: 709–714.
123 Pousset F: Cy tokines as mediators in the
centra l nervous syste m. Biomed Pharmaco -
ther 1994;
48: 425–431.
124 Golan H, Levav T, Mendelsohn A, Huleihel
M: Involvement of tu mor necrosis factor al-
pha in hippocampal development and
function. Cereb Cortex 2004;
14: 97–105.
125 Hama T, Kushima Y, Miyamoto M, Kubota
M, Takei N, Hatana ka H: Interleuki n-6 im-
proves the survival of mesencephalic cate-
cholaminergic and septal cholinergic neu-
rons from postnatal, two-week-old rats in
cultures. Neuroscience 1991;
40: 445–452.
12 6 K ush ima Y, Ha ma T, H ata nak a H: Int erle u-
kin-6 as a neurotrophic factor for promot-
ing the survival of cultured catecholamin-
ergic neurons in a chemically def ined
medium from fetal and postnatal rat mid-
brains. Neurosci Res 1992;
13: 267–280.
127 Kushim a Y, Hat anaka H: Interle ukin-6 and
leukemia inhibitory factor promote the
survival of acetylcholinesterase-positive
neurons in culture from embryonic rat spi-
nal cord. Neurosci Lett 1992;
143: 110 –114.
128 Nakafuku M, Satoh T, Kaziro Y: Differen-
tiation factors, including nerve growth fac-
tor, fibroblast growth factor, and interleu-
kin-6, induce an accumulation of an active
Ras.GTP complex in rat pheochromocyto-
ma PC12 cells. J Biol Chem 1992;
267:
1944 8–19454.
129 Ajmone-Cat MA, Cacci E, Ragazzoni Y,
Minghetti L, Biagioni S: Pro-gliogenic ef-
fect of IL-1alpha in the dif ferentiation of
embryonic neural precursor cells in vitro. J
Neurochem 2010;
113: 1060–1072.
130 Cacci E, Ajmone- Cat MA, Anelli T, Biagio -
ni S, Mi nghetti L: In v itro neuronal and g li-
al differentiation from embryonic or adult
neura l precursor cell s are different ly affect-
ed by c hron ic o r ac ute act iv ati on of mic rog -
lia. Glia 2008;
56: 412–425.
131 Poole JA, Claman HN: Immunolog y of
pregnancy. Implications for the mother.
Clin Rev Allerg y Immunol 2004;
26: 161–
170.
132 Hunt JS, Petroff MG, Morales P, Sedlmayr
P, Geraghty DE, Ober C: HLA-G in repro-
duction: studies on the maternal-fetal in-
terface. Hum Immunol 2000;
61: 1113 –1117.
133 Petroff MG: Immune interactions at the
maternal-fetal interface. J Reprod Immunol
2005;
68: 1–13.
134 Torry D S, Mukherjea D, Ar royo J, Torr y RJ:
Expres sion and funct ion of placenta grow th
factor: implications for abnormal placenta-
tion. J Soc Gynecol Investig 2003;
10: 178–
188.
135 Kanellopoulos-Langev in C, Caucheteux
SM, Verbeke P, Ojcius DM: Toleranc e of the
fetus by the maternal immune system: role
of inf lammatory mediators at the feto-ma-
ternal interface. Reprod Biol Endocrinol
2003;
1: 121.
136 Sasaki Y, Sakai M, Miyazaki S, Higuma S,
Shiozaki A, Saito S: Decidual and periph-
eral blood CD4+CD25+ regulatory T cells
in early pregnancy subjects and spontane-
ous abort ion cases. Mol Hum Reprod 2 004;
10: 347–353.
137 Hunt JS, Petroff MG, McIntire R H, Ober C:
HLA-G and immune tolerance in preg nan-
cy. FASEB J 2005;
19: 681–693.
138 Zenclussen AC: Regulatory T cells in preg-
nancy. Springer Semin Immunopathol
2006;
28: 31–39.
139 Zenclussen AC, Gerlof K, Zenclussen ML,
R it sc h el S, Z am b on Be r to ja A, Fe s t S , H on t su
S, Ueha S, Mat sushima K, Leber J, Volk HD:
Regulatory T cel ls induce a privileged toler-
ant microenvironment at the fetal-maternal
interface. Eur J Immunol 2006;
36: 82–94.
140 Maloy KJ, Powrie F: Regulator y T cells in
the control of immune pathology. Nat Im-
munol 2001;
2: 816– 822.
141 Le Bouteiller P, Legrand-Abravanel F, So-
lie r C: Solu ble HLA-G1 a t the ma terno-foe -
tal interface – a rev iew. Placenta 2003;
24(Suppl A):S10–S15.
142 Saito S, Sasaki Y, Sakai M: CD4(+)CD-
25high reg ulatory T cells in human preg-
nancy. J Reprod I mmunol 2005;
65: 111–120 .
143 Hunt JS, Jadhav L, Chu W, Geraghty DE,
Ober C: Soluble HLA-G circulates in ma-
ternal blood during pregnancy. Am J Ob-
stet Gynecol 2000;
183: 682–688.
144 Heikkinen J, Mottonen M, Alanen A, Las-
sila O: Phenotypic characterization of reg-
ulatory T cells in the human decidua. Clin
Exp Immunol 2004;
136: 373–378.
145 Ka llikourd is M, Betz AG: Period ic accumu-
lation of regulatory T cells in t he uterus:
preparation for the implantation of a semi-
allogeneic fetus? PLoS One 2007;
2:e382.
146 Veldhoen M, Moncrieffe H, Hocking RJ,
Atkins CJ, Stock inger B: Modu lation of
dendrit ic cell function by naive and reg ula-
tory CD4+ T cells. J Immunol 2006;
176:
6202–6210.
147 Bettelli E , Carrier Y, Gao W, Korn T, Strom
TB, Ouk ka M, Weiner HL, Kuchroo VK:
Reciprocal developmental pathways for the
ge ne ra ti on of p at ho ge ni c e ff ec to r T H17 an d
regulatory T cells. Nature 2006;
441: 235–
238.
148 Veldhoen M, Stockinger B: TGFbeta1, a
‘Jack of al l trades’: the lin k with pro-inf lam-
matory IL -17-producing T cel ls. Trends Im-
munol 2006;
27: 358–361.
149 Higuma-Myojo S, Sasaki Y, Miyazaki S,
Sakai M, Siozaki A, Miwa N, Saito S: Cyto-
kine profile of natural killer cells in early
human pregnancy. Am J Reprod Immunol
2005;
54: 21–29.
150 Weaver CT, Harrington LE, Mangan PR,
Gavrieli M, Murphy KM: Th17: an effector
CD4 T cell lineage with regulatory T cell
ties. Immunity 2006;
24: 677–688.
151 Roberts CT, White CA, Wiemer NG, Ram-
say A, Robertson SA: Altered placental de-
velopment in interleukin-10 null mutant
mice. Placenta 2003;
24(Suppl A):S94–S99.
152 Jyonouchi H, Geng L, Cushing-Ruby A,
Quraishi H: Impact of innate immunity in
a subset of children with autism spectrum
disorders: a case control study. J Neuroin-
flammation 2008;
5: 52.
Parker-Athill /Tan
Neurosignals 2010;18:113–128
128
153 Singh VK: Phenotypic expression of auto-
immune autistic disorder (AAD): a major
subset of aut ism. Ann Cli n Psychiatry 2 009;
21: 148–161.
154 Nishimaki S, Sato M, An H, Shima Y,
Akaike T, Yokoyama U, Yokota S: Compa r-
ison of markers for fetal inflammatory re-
sponse sy ndrome: fetal blood interleuki n-6
and neonatal urinary beta(2)-microglobu-
lin. J Obstet Gynaecol Res 2009;
35: 472–
476.
155 Fidel PL Jr., Romero R, Wolf N, Cutright J,
Ramirez M, Araneda H, Cotton DB: Sys-
temic and local cy tokine prof iles in endo-
toxin-indu ced preterm part urition in mic e.
Am J Obstet Gy necol 1994;
170: 146 7–1475.
156 Bell MJ, Hallenbeck JM, Gallo V: Deter-
mining the fetal inf lammatory response in
an experimental model of intrauterine in-
flammation in rats. Pediatr Res 2004;
56:
541–546.
157 L iverman CS, Ka ftan HA, Cu i L, Hersperg-
er SG, Taboada E, Klein RM, Berma n NE:
Altered expression of pro-inflammator y
and developmental genes in the fetal brain
in a mouse model of maternal infection.
Neurosci Lett 2006;
399: 220–225.
158 Meyer U, Nyffeler M, Engler A, Urwyler A,
Schedlowski M, Knuesel I, Yee BK, Feldon
J: The time of prenatal immune challenge
determines the specificity of inflamma-
tion-mediate d brain and behav ioral pathol-
ogy. J Neurosci 2006;
26: 4752–4762.
159 Keul R, Heinrich PC, Muller-Newen G,
Muller K, Woo P: A possi ble role for soluble
I L- 6 r ec e pt or in th e p at h og en es is of sy s te m-
ic onset juveni le chronic ar thritis . Cytokine
1998;
10: 729–734.
160 Carey R, Jurickova I, Ballard E, Bonkowski
E, Han X, Xu H, Denson LA: Activation of
an IL-6: STAT3-dependent transcriptome
in pediatric-onset inflammatory bowel dis-
ease. Inflamm Bowel Dis 2008;
14: 446–457.
161 Armario A, Herna ndez J, Bluethmann H,
Hidal go J: IL-6 deficie ncy leads to increa sed
emotionality in mice : evidence in transgen-
ic mice carrying a null mutation for IL- 6. J
Neuroimmunol 1998;
92: 160 –169.
162 Samuelsson AM, Jennische E, Hansson
HA, Holmang A: Prenata l exposure to in-
terleukin-6 results in inf lammatory neuro-
degeneration i n hippoca mpus with N MDA/
GABA(A) dysregulation and impaired spa-
tial learning. Am J Physiol Regul Integr
Comp Physiol 2006;
290:R1345–1356.
163 Wu YY, Bradshaw RA: Sy nergistic induc-
tion of neurite outgrowt h by nerve growth
factor or epidermal grow th factor and in-
terleuk in-6 in PC12 cells . J Biol Chem 1996;
271: 13033–13039.
164 Wu Y Y, Bra dshaw RA: Induct ion of neurite
outgrowt h by interleuki n-6 is accompan ied
by activation of STAT3 signaling pathway
in a variant PC12 cell (E 2) line. J Biol Chem
1996;
271: 13023–13032.
165 Aaltonen R, Heikkinen T, Hakala K, Laine
K, Alanen A: Transfer of proinflammatory
cytok ines across term placenta. Obstet Gy-
necol 2005;
106: 802–807.
166 Za retsky MV, Alexander JM, Byrd W, Baw-
don RE: Transfer of inflammator y cyto-
kines across the placenta. Obstet Gynecol
2004;
103: 546–550.
167 Ba rnabe-Heider F, Wasylnk a JA, Fernandes
KJ, Porsche C, Sendtner M, Kaplan DR,
Miller FD: Evidence that embryonic neu-
rons regulate the onset of cortical gliogen-
esis via cardiotrophin-1. Neuron 2005;
48:
253–265.
168 Fatemi SH, Cuadra AE, El-Fakahany EE,
Sidwell RW, Thuras P: Prenatal vira l infec-
tion causes alterations in nNOS expression
in developing mouse brains. Neuroreport
2000;
11: 1493 –1496.
169 Fatemi SH, Pearce DA, Brooks AI, Sidwell
RW: Prenatal viral infection in mouse
causes differential expression of genes in
brains of mouse progeny: a potential ani-
mal model for schizophrenia and autism.
Synapse 2005;
57: 91–99.
170 Rezai-Zadeh K, Ehrhart J, Bai Y, Sanberg
PR, Bickford P, Tan J, Shytle RD: Apigenin
and luteolin modulate microglial act ivation
via inhibition of STAT1-induced CD40 ex-
pression. J Neuroinf lammation 2008;
5: 41.
171 Hi rano T, Arimitsu J, Higa S, Nak a T, Oga-
ta A, Shima Y, Fujimoto M, Yamadori T,
Ohkawara T, Kuwabara Y, Kawai M, Ka-
wase I, Tanaka T: Luteolin, a flavonoid, in-
hibits CD40 ligand expression by activated
human basophils. Int Arch Allergy Immu-
nol 2006;
140 : 150–156.
172 Coleman M, Gi llberg C: The Biology of Au-
tistic Sy ndromes, ed 3. London, Mac Keith
Press, 2000.
... Triggered by various stimuli such as viral or bacterial infections, exposure to environmental toxins, and maternal autoimmune conditions, MIA initiates a cascade of inflammatory responses, releasing pro-inflammatory cytokines, chemokines, and other immune signaling molecules [17][18][19][20]. These mediators, notably interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and IL-1β, play pivotal roles in immune regulation but, when overproduced during pregnancy due to MIA, may adversely impact fetal neurodevelopment [21][22][23] The developing fetal brain, particularly vulnerable during critical periods of neurogenesis and synaptogenesis, faces potential disruptions in the intricate processes governing neural circuit formation. MIA has been associated with an elevated risk of neurodevelopmental disorders, including ASD and schizophrenia, although the precise mechanisms remain under investigation. ...
Article
Full-text available
This review examines the intricate association between maternal immune activation (MIA) and autism spectrum disorder (ASD), emphasizing the impact of maternal infections during pregnancy. Epidemiological studies link viral and bacterial infections to an elevated risk of ASD, revealing the complex interplay between environmental factors and neurodevelopmental outcomes. Immunological mechanisms, including cytokine dysregulation and neuroinflammation, involve key players such as interleukin-6 and tumor necrosis factor-alpha, influencing fetal brain development and ASD risk. Genetic and environmental interactions contribute to individual susceptibility, with specific variants influencing MIA's impact on ASD risk. Epigenetic modifications provide a molecular link between environmental exposures, including MIA, and enduring neurodevelopmental changes. Recognizing critical periods during fetal neurodevelopment susceptible to MIA is crucial. Long-term studies highlight enduring consequences on behavior and cognition into childhood and adolescence. Exploring potential therapeutic interventions, including immunomodulatory strategies during pregnancy, offers hope for mitigating MIA's impact on ASD outcomes. Despite progress, knowledge gaps persist, motivating future research guided by emerging technologies and interdisciplinary approaches to unravel the intricate MIA-ASD relationship.
... Prenatal IL-6 exposure in rats induced inflammatory neurodegeneration in the hippocampus and impaired spatial learning in adulthood [14]. Thus, IL-6 signaling has been proposed as a key mechanism of maternal immune activation that may be associated with ASD [15]. Inflammation triggers the release of many cytokines and signaling molecules, some of which have been found to predispose to FS and alter long-term synaptic plasticity in the hippocampus [16,17]. ...
Article
Full-text available
Febrile seizure (FS), which occurs as a response to fever, is the most common seizure that occurs in infants and young children. FS is usually accompanied by diverse neuropsychiatric symptoms, including impaired social behaviors; however, research on neuropsychiatric disorders and hippocampal inflammatory changes following febrile seizure occurrences is very limited. Here, we provide evidence linking FS occurrence with ASD pathogenesis in rats. We developed an FS juvenile rats model and found ASD-like abnormal behaviors including deficits in social novelty, repetitive behaviors, and hyperlocomotion. In addition, FS model juvenile rats showed enhanced levels of gliosis and inflammation in the hippocampal CA2 region and cerebellum. Furthermore, abnormal levels of social and repetitive behaviors persisted in adults FS model rats. These findings suggest that the inflammatory response triggered by febrile seizures in young children could potentially serve as a mediator of social cognitive impairments.
... Although most research has focused on autoimmune and infectious triggers, it has been proposed that other conditions such as maternal stress, environmental pollutants, as well as allergies and asthma can also contribute to the risk for ASD by activating maternal immune response [9,20]. Recent preclinical experiments have demonstrated that a maternal Maternal asthma and allergy condition, more common in male children with ASD comparing to female children with ASD (p = 0.009) asthma-allergy model is causative of autism-related behaviors in mouse offspring [45,46]; and cytokines, particularly interleukin (IL)-6, have been proposed to be key mediators of this relationship [47] (see Fig. 2). Asthma, like many other inflammatory diseases, increases interleukin (IL)-6 levels [48], which is the only proinflammatory cytokine known to pass from maternal to fetal circulation via the placenta [19,49,50]. ...
... Although most research has focused on autoimmune and infectious triggers, it has been proposed that other conditions such as maternal stress, environmental pollutants, as well as allergies and asthma can also contribute to the risk for ASD by activating maternal immune response [9,20]. Recent preclinical experiments have demonstrated that a maternal Maternal asthma and allergy condition, more common in male children with ASD comparing to female children with ASD (p = 0.009) asthma-allergy model is causative of autism-related behaviors in mouse offspring [45,46]; and cytokines, particularly interleukin (IL)-6, have been proposed to be key mediators of this relationship [47] (see Fig. 2). Asthma, like many other inflammatory diseases, increases interleukin (IL)-6 levels [48], which is the only proinflammatory cytokine known to pass from maternal to fetal circulation via the placenta [19,49,50]. ...
Article
Full-text available
Autism spectrum disorder (ASD) is a disabling neurodevelopmental condition with complex etiology. Emerging evidence has pointed to maternal atopy as a possible risk factor. It is hypothesized that maternal atopic disease during pregnancy can lead to increased levels of inflammatory cytokines in fetal circulation via placental transfer or increased production. These cytokines can then pass through the immature blood–brain barrier, causing aberrant neurodevelopment via mechanisms including premature microglial activation. The objective of this study is to systematically review observational studies that investigate whether a maternal history of atopic disease (asthma, allergy, or eczema/atopic dermatitis) is associated with a diagnosis of ASD in offspring. A search was conducted in Ovid MEDLINE, PsycINFO, and Embase databases for relevant articles up to November 2021; this was later updated in January 2022. Observational studies published in peer-reviewed journals were included. Data were synthesized and qualitatively analyzed according to the specific atopic condition. Quality assessment was done using the Newcastle–Ottawa Scale. Nine articles were identified, with all including asthma as an exposure, alongside four each for allergy and eczema. Findings were inconsistent regarding the association between a maternal diagnosis of either asthma, allergy, or eczema, and ASD in offspring, with variations in methodology contributing to the inconclusiveness. More consistent associations were demonstrated regarding maternal asthma that was treated or diagnosed during pregnancy. Evidence suggests that symptomatic maternal asthma during pregnancy could be associated with ASD in offspring, underscoring the importance of effective management of atopic conditions during pregnancy. Further research is needed, particularly longitudinal studies that use gold-standard assessment tools and correlate clinical outcomes with laboratory and treatment data. PROSPERO Registration Number and Date: CRD42018116656, 26.11.2018.
... Prenatal IL-6 exposure in rats induced inflammatory neurodegeneration in the hippocampus and impaired spatial learning in adulthood [12]. Thus, IL-6 signaling has been proposed as a key mechanism of maternal immune activation that may be associated with ASD [13]. Inflammation triggers the release of many cytokines and signaling molecules, some of which have been found to predispose to FS and alter long-term synaptic plasticity in the hippocampus [14,15]. ...
Preprint
Full-text available
Febrile seizure (FS), which occurs as a response to fever, is the most common seizure that occurs in infants and young children. FS is usually accompanied by diverse neuropsychiatric symptoms including impaired social behaviors; however, no causative link between FS and autism spectrum disorder (ASD) has yet been established. Here, we provide evidence linking FS occurrence with ASD pathogenesis in rats. We developed an FS juvenile rats model and found ASD-like abnormal behaviors including deficits in social novelty, repetitive behaviors, and hyperlocomotion. In addition, FS model juvenile rats showed enhanced levels of gliosis and inflammation in the hippocampal CA2 region and cerebellum. Furthermore, abnormal levels of social and repetitive behaviors persisted in adults FS model rats. These findings suggest that occurrence of FS in infants can cause behavioral endophenotypes of ASD.
... Prenatal IL-6 exposure in rats induced in ammatory neurodegeneration in the hippocampus and impaired spatial learning in adulthood [29]. Thus, IL-6 signaling has been proposed as a key mechanism of maternal immune activation (MIA) that may be associated with ASD [30]. In ammation triggers the release of many cytokines and signaling molecules, some of which have been found to predispose to FS and alter long-term synaptic plasticity in the hippocampus [31][32][33][34][35]. ...
Preprint
Full-text available
Background Febrile seizure (FS), which occurs as a response to fever, is the most common seizure that occurs in infants and young children. FS is usually accompanied by diverse neuropsychiatric symptoms including impaired social behaviors; however, no causative link between FS and autism spectrum disorder (ASD) has yet been established. Here, we provide evidence linking FS occurrence with ASD pathogenesis in rats. Methods We inducted febrile seizures through a mercury vapor lamp using infantile rats (postnatal 11 days). At 3 and 12 weeks after FS induction, we examined behavioral tests and immunostaining to identify the association of ASD-like phenotype development with inflammatory damage. Results We developed an FS juvenile rats model and found ASD-like abnormal behaviors including deficits in social novelty, repetitive behaviors, and hyperlocomotion. In addition, FS model juvenile rats showed enhanced levels of gliosis and inflammation in the hippocampal CA2 region and cerebellum. Furthermore, abnormal levels of social and repetitive behaviors persisted in adults FS model rats. Conclusion These findings suggest that occurrence of FS in infants can cause behavioral endophenotypes of ASD.
Preprint
Full-text available
The functional alteration of microglia emerges in the brains exposed to extrinsic stress insults during early development. Pathophysiological findings of psychiatric disorders such as schizophrenia and autism implied a localized deficit of the cerebellum. However, the link between microglia reactivity and cerebellar dysfunction is missing. Here, we investigated the developmental immune environment of translational mouse models that combine two risk factors: maternal infection and repeated social defeat stress ( i.e. , 2HIT). We found synergy of inflammatory stress insults produced reactive microglia, specifically in the cerebellum in both sexes. The microglial proliferation correlated with the Purkinje neuron loss in 2HIT mice. Highly multiplexed imaging-mass-cytometry demonstrated a TREM2-positive population of stress-associated microglia in the cerebellum. Single-cell-proteomic clustering revealed IL-6- and TGFβ-signaling association with microglial cell transition. Purkinje cells reduced excitability, cerebellum-involved brain-wide functional dysconnectivity, and behavioral anomalies underpin the cerebellar cognitive dysfunctions in 2HIT animals, ameliorated by cerebellum-specific microglia replacement.
Article
Full-text available
Autism, also known as an autism spectrum disorder, is a complex neurodevelop-mental disorder usually diagnosed in the first three years of a child's life. A range of symptoms characterizes it and can be diagnosed at any age, including adolescence and adulthood. However, early diagnosis is crucial for effective management, prognosis, and care. Unfortunately, there are no established fetal, prenatal, or newborn screening programs for autism, making early detection difficult. This review aims to shed light on the early detection of autism prenatally, natally, and early in life, during a stage we call as "pre-autism" when typical symptoms are not yet apparent. Some fetal, neonatal, and infant biomarkers may predict an increased risk of autism in the coming baby. By developing a biomarker array, we can create an objective diagnostic tool to diagnose and rank the severity of autism for each patient. These biomarkers could be genetic, immunological, hormonal, metabolic, amino acids, acute phase reactants, neonatal brainstem function biophysical activity, behavioral profile, body measurements, or radiological markers. However, every biomarker has its accuracy and limitations. Several factors can make early detection of autism a real challenge. To improve early detection, we need to overcome various challenges, such as raising community awareness of early signs of autism, improving access to diagnostic tools, reducing the stigma attached to the diagnosis of autism, and addressing various culturally sensitive concepts related to the disorder.
Article
Full-text available
Autism spectrum disorder (ASD) is a common neurological disorder. Although the etiologies of ASD have been widely speculated, evidence also supports the pathogenic role of cutaneous inflammation in autism. The prevalence of ASD is higher in individuals with inflammatory dermatoses than in those without inflammatory diseases. Anti-inflammation therapy alleviates symptoms of ASD. Recent studies suggest a link between epidermal dysfunction and ASD. In the murine model, mice with ASD display epidermal dysfunction, accompanied by increased expression levels of proinflammatory cytokines in both the skin and the brain. Children with ASD, which develops in their early lifetime, also exhibit altered epidermal function. Interestingly, improvement in epidermal function alleviates some symptoms of ASD. This line of evidence suggests a pathogenic role of cutaneous dysfunction in ASD. Either an improvement in epidermal function or effective treatment of inflammatory dermatoses can be an alternative approach to the management of ASD. We summarize here the current evidence of the association between the skin and ASD.
Article
The hypothesis that direct exposure to or clinical illness with measles, rubella, mumps or chickenpox may play a causal role in autism has been evaluated. The study subjects were 163 cases of autism and their 355 unaffected siblings. Medical data relating to prenatal life and early infancy were obtained by interviewing the parents, and from medical records. Medical records were found for only about half the instances In which a pertinent illness or exposure was reported in the home Interview. However, there was no evidence of systematic bias In recall on the part of parents when reporting on an index case. When medical records and parental statements were both available they were relatively consonant. Increased frequency of combined illness and exposure was seen for the cases relative to their sibling controls with regard to prenatal measles, rubella and mumps and to postnatal mumps. Yet the proportion of cases with histories of viral experience during the periods of interest was small, indicating that these four viruses were unlikely to have played a major role In any substantial proportion of the cases. Some associations were noted with parental histories of other infections during gestation or infancy. Again, however, the proportion of cases that could be accounted for by any of these associations, even if causal, is small.
Article
Neuropoietic cytokines are well known for their role in the control of neuronal, glial and immune responses to injury or disease. Since this discovery, it has emerged that several of these proteins are also involved in nervous system development, in particular in the regulation of neurogenesis and stem cell fate. Recent data indicate that these proteins have yet more functions, as key modulators of synaptic plasticity and of various behaviours. In addition, neuropoietic cytokines might be a factor in the aetiology of psychiatric disorders.
Article
• We tested the hypothesis that first-trimester exposure to acute food deprivation is a risk factor for schizophrenia. A sharp and time-limited decline in the food intake of the Dutch population following a Nazi blockade in 1944 to 1945 created a unique if tragic natural experiment to test this hypothesis in three regions of Holland (west, north, and south). In the west, or famine region, birth cohorts exposed to severe food deprivation (an average daily ration under 4200 kJ) during the first trimester showed a substantial increase in hospitalized schizophrenia for women but not for men. Relative risks for women were 2.17 for "broad" and 2.56 for "restricted" schizophrenia. Moderate food deprivation during the first trimester (average daily ration under 6300 kJ) was not associated with increased risk of schizophrenia in the famine region. In the north and south regions, numbers were smaller and there was no exposure to severe famine. Birth cohorts exposed to moderate food deprivation during the first trimester showed a trend toward increased risk of schizophrenia for women. These findings give plausibility to the proposition that early prenatal nutrition can have a gender-specific effect on the risk of schizophrenia.
Article
Tumor necrosis factor alpha (TNFα) is a cytokine produced mainly by cells of the immune system. It is also expressed by brain neurons and glia. The physiological role of TNFα in the brain is not yet fully clear. Using TNFα-deficient mice, we have examined its role in hippocampal development and function. We report here that TNFα is involved in the regulation of morphological development in the hippocampus. TNFα-deficient mice exhibited an accelerated maturation of the dentate gyrus region and smaller dendritic trees in CA1 and CA3 regions in young mouse. In addition to its involvement in hippocampal morphogenesis, TNFα deficiency specifically improved performance of affected mice in behavioral tasks related to spatial memory. Moreover, lack of TNFα increased the expression of nerve growth factor (NGF), but not brain-derived neurotrophic factor (BDNF), following performance of the learning task. Our results suggest that TNFα actively influences hippocampal development and function. In adult mice, TNFα may interfere with memory consolidation, perhaps by regulating NGF levels.
Article
A relationship between autism and gastrointestinal (GI) immune dysregulation has been postulated based on incidence of GI complaints as well as macroscopically observed lymphonodular hyperplasia and microscopically determined enterocolitis in pediatric patients with autism. To evaluate GI immunity, we quantitatively assessed levels of proinflammatory cytokines, interleukin (IL)-6, IL-8, and IL-1β, produced by intestinal biopsies of children with pervasive developmental disorders.