Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 9.
Published in final edited form as: Curr Opin Investig Drugs. 2009 May;10(5):463–473.

Autoimmunity in autism

Amanda M Enstrom 1, Judy A Van de Water 2, Paul Ashwood 1,*
PMCID: PMC6952169  NIHMSID: NIHMS1063770  PMID: 19431079

Abstract

Autism spectrum disorders is a heterogenous group of neurodevelopmental disorders, the etiology or etiologies of which remain unknown. Increasing evidence of autoimmune phenomena in individuals with autism could represent the presence of altered or inappropriate immune responses in this disorder, and this immune system dysfunction may represent novel targets for treatment. Furthermore, in recent studies, antibodies directed against the fetal brain have been detected in some mothers of children with autism; these antibodies have the ability to alter behavioral outcomes in the offspring of animal models. A better understanding of the involvement of the immune response in early brain development, with respect to autism, may have important therapeutic implications.

Keywords: Autism spectrum disorder, autoantibody, autoimmunity, immune, inflammation, neuroinflammation

Introduction

Autism spectrum disorder (ASD) is a group of heterogeneous, neurodevelopmental disorders that include autistic disorder, pervasive developmental disorder-not otherwise specified (PDD-NOS) and Asperger syndrome, which, along with Rett syndrome and childhood disintegrative disorder, comprise the pervasive developmental disorders [1]. ASD typically is diagnosed within the first 36 months of life and is characterized by impairments in social reciprocity, verbal and nonverbal communication, and repetitive and stereotypical behaviors [1]. The prevalence of ASD is increasing, with some estimates as high as 8 cases per 1000 births [2,3].

The exact etiology of ASD remains unknown; however, due to the heterogeneity of symptoms within ASD, such as the pattern of symptom onset, severity of disabilities, epilepsy, sleep problems, gastrointestinal disturbances, and deficits in IQ, distinct phenotypes may exist within ASD and may represent separate etiologies or combinations of susceptibility and environmental factors [46]. As with most complex disorders, it is likely that interactions between multiple causative factors, including genetics, epigenetics, pre- and postnatal environments, exist that may contribute to neurodevelopment changes and may lead to features that are characteristic of ASD. As such, the factors contributing to the development of ASD may vary between individuals. Recent research has highlighted a potential role for the immune system in ASD, including immunogenetic abnormalities and the inappropriate response of the immune system to environmental challenges. Evidence of abnormal immune activation has included repeated observations of increased autoreactive antibodies in children with ASD and their mothers [729]. In this review, the complex relationship that may exist between autoimmunity and ASD, and the possible treatment implications for ASD are discussed.

Evidence of immune system abnormalities in autism spectrum disorder

Genetics and the immune system

Multiple genetic association studies have implicated different genes with the development of ASD, several of which are involved with the function of the immune system. The HLA alleles A2, DR4 and DR11 have been linked with susceptibility to ASD [2427], as well as decreased lymphocyte responsiveness [25]. Within the HLA class III region, there is a complement C4B null allele, resulting from duplications of C4A, that confers a relative risk of 4.3 for the development of ASD [28,30,31]. In addition, several studies have implicated the MET (met proto-oncogene) tyrosine kinase pathway with ASD [3234]. Signaling via the MET receptor is necessary for immune regulation in APCs, including dendritic cells and monocytes [35]. Similarly, the serine and threonine kinase C gene PRKCB1, which is involved in both B-cell activation and neuronal function, has been linked to ASD in some studies [36,37], but not in others [38,39], perhaps reflective of population differences. Peripheral blood RNA expression studies demonstrated an upregulation of genes involved in innate immune activation through the NK pathway in individuals with ASD [40], a finding that was confirmed in functional studies using NK-cells [4143]. Functional polymorphisms of macrophage inhibitory factor (MIF), which has several effects on innate and adaptive immune responses, have also been associated with individuals with ASD, but has not been detected in unaffected siblings [29]. Interestingly, increased sera concentrations of MIF correlated with worsening behavioral assessments in individuals with ASD compared with their unaffected siblings [29]. Moreover, genes that can affect immune responses, such as PTEN (which may be involved in T regulatory cell development) and reelin, have been associated with ASD [2429,33,4446]. An epidemiological study in Portugal identified mitochondrial respiratory-chain disease in 4.2% of children with ASD [47]; conditions such as mitochondrial respiratory-chain disease could have widespread effects on many body systems, including the activity of the immune system. Overall, these studies suggest that multiple susceptibility genes related to innate immune activation and/or the loss of adaptive immune regulation may be involved in the etiology of ASD.

Evidence of inflammatory cytokine responses in ASD

Neuroimmune interactions begin early in development, and the health of one system is contingent on the health of the other. Both systems share common signaling molecules and receptors, such as cytokines and neuropeptides/neurotransmitters. Aberrant responses from immune cells present in the CNS, including microglial cells in the brain, have been implicated in neuronal cell death [4851], which is partly mediated through the actions of inflammatory cytokines and neuropeptides. The inflammatory cytokines IL-1, IL-6 and TNFα can directly affect the CNS, can alter neurodevelopment and, subsequently, may have an impact on behavior [5254]. For example, neuropoietic cytokines, such as IL-6, can have direct effects on neurons and glia in vitro, including changes in proliferation, survival, death, neurite outgrowth and gene expression [54,55]; while in vivo, the peripheral administration of IL-6 to gestating mice has been demonstrated to affect behavior and neurodevelopment of offspring [10]. In addition, cytokines such as TNFα have been linked to oligodendrocyte toxicity, as well as aberrant neurite growth [5658]. While a cytokine-mediated mechanism has yet to be conclusively established in neurodevelopmental disorders such as ASD or schizophrenia, evidence of adverse immunological functioning in these disorders suggests that neuroimmune interactions may be altered and may affect neurodevelopment and early brain development [5962].

In studies of post-mortem brain specimens, evidence of elevated cytokine responses, including IL-6 and TNFα, has been observed in the brains of individuals with ASD compared with controls [63,64]. These cytokines appear to have been derived from microglial and astroglial cells of the innate immune system [64]. In the temporal cortex of the brain, transcriptome expression patterns from individuals with ASD also demonstrated that enhanced immune responses occurred in the brains of individuals with ASD compared with controls, and were consistent with increased autoimmune responses and increased inflammatory cytokine signaling [34]. Moreover, several researchers have suggested that there is an increased presence of proinflammatory cytokines and a skewing of the ratio of Th1-associated cytokines compared with Th2-associated cytokines in the brain and periphery of individuals with ASD (Table 1) [9,11,12,6568]. Increased IFNγ has been detected in brain tissue, CSF, plasma, serum, cultured PBMCs and cultured NK-cells from individuals with ASD [11,41,63,64,66,67]. However, a separate study failed to demonstrate any differences in plasma IFNγ levels [69], and another study demonstrated decreased IFNγ responses to phorbol myristate acetate (PMA)/ionomycin stimulation in PBMCs in individuals with ASD compared with controls [65]. These discrepancies may be due to the patient population, for example, whether individuals had a full autism diagnosis or were classified on the broader ASD spectrum, patient age and gender, as well as the analytical techniques, power of the statistical analysis and tissue/ specimens used. Another factor that often complicates studies of ASD has been the use of siblings of children with ASD as controls. Importantly, as autism is highly heritable, similar findings in analyses of the immune system in individuals with ASD compared with their siblings may reflect that there are shared immune susceptibilities that are not the result of brain pathology or neurodevelopmental changes. While most studies have demonstrated increased proinflammatory cytokine production in individuals with ASD [9,11,12,6568], some studies have reported a decreased production of regulatory cytokines, including IL-10 and TGFβ1, in individuals with ASD [9,12,68,70,71]. As both IL-10 and TGFβ1 have a role in regulating the immune response, these data suggest that immune regulation may be impaired in ASD; this impaired immune regulation could lead to a breakdown in immunological tolerance.

Table 1.

A summary of studies of cytokine levels in individuals with autism spectrum disorder.

Cytokine/
chemokine tested
Change detected in
cytokine/chemokine levels
in patients with ASD
Tissue source Method Reference
Th1-associated cytokines
IFNγ Increased CSF, brain tissue, whole blood, plasma and cultured PBMCs Luminex and ELISA [11,63,64,66,67]
IFNγ No change Plasma ELISA [69]
IFNγ + T-cells Decreased Cultured PBMCs Flow cytometry [65]
IL-12 Increased Plasma ELISA [11]
IL-2 Increased Serum and cultured PBMCs ELISA [8,66]
IL-2 No Change Brain tissue Luminex [63]
IL-2 + T-cells Decreased Cultured PBMCs Flow cytometry [65]
Acute inflammatory cytokines
IL-1β Increased Brain tissue and stimulated PBMCs Luminex and ELISA [63,115]
IL-1β No change Plasma ELISA [69]
IL-6 Increased CSF, brain tissue and cultured PBMCs Luminex and ELISA [63,64,67,115]
IL-6 No change Plasma, serum and cultured PBMCs ELISA and flow cytometry [7,11,65]
TNFα Increased Brain tissue and cultured PBMCs Luminex and ELISA [63,67,115]
TNFα No change Plasma ELISA [11,69]
TNFRI Increased Stimulated PBMCs ELISA [115]
TNFRII Increased Stimulated PBMCs and serum ELISA [7,115]
IL-8 Increased CSF and brain tissue Luminex [63,64]
MCP-1 Increased CSF and brain tissue Luminex and ELISA [64]
MIP-1β Increased CSF Luminex [64]
TGFβ Increased Brain tissue Luminex and ELISA [64]
TGFβ Decreased Plasma ELISA [70,71]
GM-CSF Increased Brain tissue Luminex [63]
Th2-associated cytokines
IL-4 Increased Cultured PBMCs ELISA and flow cytometry [66,65]
IL-4 No change Brain tissue Luminex [63]
IL-5 Increased Cultured PBMCs ELISA [63,66]
IL-5 No change Brain tissue Luminex [63]
IL-10 No change Brain tissue and cultured PBMCs Luminex, ELISA and flow cytometry [63,65,66]
IL-13 Increased Cultured PBMCs ELISA [66]

Four studies were excluded from this table because children with autism spectrum disorder (ASD) that were recruited in these excluded studies experienced gastrointestinal symptoms, which may confound cytokine results [9,12,68,116].

MCP-1 monocyte chemotactic protein 1, MIP-1β macrophage inflammatory protein 1β, TNFRI TNF receptor 1, TNFRII TNF receptor II

The potential role of perinatal or chronic infection in ASD

Environmental factors, such as congenital rubella infection, have previously been reported to be associated with ASD [72,73]. Perinatal Haemophilus influenzae and CMV infections have been associated with ASD through significant damage to the immature brain [74]. Associations between ASD diagnosis and increased ear infections [75], antibiotic use [75] and infections in the first 30 days of life (and lower rates compared with controls thereafter) [76] have also been observed. However, many of these association studies have not been replicated and, to date, no infectious agent has been definitively causally linked with ASD. Furthermore, because many different infectious agents have been implicated in ASD, one possible hypothesis is that an inappropriate immune response or immune dysfunction in response to a variety of possible infectious agents, which occurs at critical periods in neurodevelopment, may have more of an impact on neurodevelopment than any one specific causative agent.

Evidence of a role for autoimmunity in ASD

Brain autoantibodies in individuals with ASD

Evidence of a loss of tolerance that could lead to autoimmunity in ASD includes the demonstration of antibody reactivity to several regions of the brain, such as the human frontal cortex [15,77], temporal cortex [78], cerebellum [15,21,79], hypothalamus and thalamus [19], and brain endothelial cells [18], as well as the presence of anti-nuclear antibodies [78] (Table 2). Antibodies with reactivity to the caudate nucleus and cerebellum in rats, but not toward the hippocampus or brainstem, have also been described [80]. As several different CNS targets have been observed in individuals with ASD, these data may suggest that there are multiple mechanisms whereby CNS reactivity has developed in ASD. In addition, it is unclear whether these antibodies are produced as a primary event that may affect neurodevelopment or as a result of early brain inflammation. Moreover, the increased presence of antibodies against CNS targets does not address the possible function these antibodies have, specifically whether the antibodies block cellular function, act as agonists or antagonists on receptor systems, or bind to self-protein and result in tissue damage. Previous disparate reports of autoantibody reactivity to myelin basic protein (MBP) in ASD underscore the difficulty in accessing autoreactive immune responses in a highly heterogenous disorder. Singh and colleagues reported that sera samples from approximately 58% of children with ASD (≤ 10 years old), compared with 9% of controls (p ≤ 0.0001), reacted to MBP in a protein-immunoblotting assay [14], a finding that was replicated in further studies by these researchers [81,82], and was independently demonstrated in studies by Connolly and colleagues [18] and by Vojdani and colleagues [83]. In the studies by Singh and colleagues, Western immunoblotting was used to confirm MBP reactivity; this assay was conducted by separating MBP on a reducing (denaturing) gel, transferring the MBP to a nitrocellulose membrane, and incubating the denatured protein in the presence of sera with a detergent. The reactivity of the sera to the denatured MBP then was confirmed using secondary anti-human Ig antibody. One advantage of this method is that only reactivity to the protein of interest (eg, MBP) was identified; however, false positives due to the use of non-human MBP or poor blocking may have occurred. Vojdani and colleagues [83] and Connolly and colleagues [18] used ELISA to determine MBP reactivity in order to limit reactivity to intact non-denatured protein; in addition, individuals with multiple sclerosis, who produce antibodies to MBP during active phases of disease, were included as disease controls in the study by Vojdani and colleagues [83]. In contrast, Silva and colleagues examined plasma reactivity to homogenized human brain and human MBP protein by Western immunoblotting in children with ASD (n = 171) and pediatric controls (n = 54), and were able to identify antibody reactivity to a protein at approximately 20 kDa; this reactivity could be used to discriminate between children with ASD and controls (p = 0.00046, Mann-Whitney U-test) [84]. Although the putative antigen was a similar molecular weight to MBP, it was not an MBP isoform [84]. The brain extracts and MBP proteins were separated on the same denaturing gels, which minimizes inter-assay variability. One limiting factor of this study is that reactivity to whole protein was not determined. Furthermore, Libbey and colleagues reported similar titers of antibodies against MBP, as assessed by ELISA, in the plasma of individuals with ASD (either individuals with autism symptoms that appeared early in life or those that met normal developmental milestones, but then underwent regression and developed ASD) when compared with controls [85]. Moreover, it was demonstrated that MBP-reactive antibodies positively correlated with increased age, therefore age matching is critical in these experiments, and disparate ages of cases compared with controls could have affected previously published reports [85]. Similarly, contradictory results have been reported from examinations of sera for the presence of autoantibodies against glial fibrillary acidic protein (GFAP) in individuals with autism and individuals with Tourette syndrome, compared with controls [86]. There are, however, some findings that suggest antibodies to neurofilaments and brain-derived neurotrophic factor may be increased in individuals with ASD compared with controls (Table 2) [18,20,79,83], although these findings have not been replicated. Overall, these data suggest that antibodies reactive against CNS and brain proteins exist in individuals with ASD; however, the exact antigen to which these antibodies bind has not been identified. These data also suggest that not all individuals with ASD have the same autoantibody repertoire, and that antibody specificity may vary among individuals with ASD.

Table 2.

Studies investigating the presence of autoreactive antibodies in individuals with ASD.

Sample tested Positive or
negative finding
Findings Reference
Brain
Whole brain extract Positive Samples from individuals with ASD reacted to a 20 kDa protein. [84]
Cerebral cortex Positive 18% of samples from individuals with ASD vs 0% controls had antibodies against rat cerebral cortex. [80]
Prefrontal cortex Positive Samples from individuals with ASD reacted to a 100 kDa protein. [15]
Negative No reactivity to frontal cortex proteins was detected. [77,79]
Temporal cortex Positive 27% of samples from individuals with ASD vs 2% controls had antibodies against proteins in the temporal cortex. [78]
Brodmann’s area 10 Positive Increased density of a 160 kDa protein band in samples from individuals with ASD. [15]
Caudate nucleus Positive 49% individuals with ASD vs 0% controls reacted to bands at 49, 115 and 160 kDa from rat caudate nucleus. [80]
Positive Reactivity towards 100 kDa in ASD compared with controls. [15]
Cingulate gyrus Positive Increased density of a 73 kDa protein band in samples from individuals with ASD. [15]
Putamen Positive Increased density of a 100 kDa protein band in samples from individuals with ASD was detected. [15]
Thalamus Positive 29% of samples from individuals with ASD vs 8% of controls reacted to a 52 kDa protein. [19]
Hypothalamus Positive 37% of samples from individuals with ASD vs 11% of controls reacted with 42, 46 and 48 kDa proteins. [19]
Positive 30% of samples from individuals with ASD vs 11% controls reacted to a 52 kDa protein. [19]
Cerebellum Positive 9% of samples from individuals with ASD vs 0% of controls reacted to rat cerebellum proteins. [80]
Positive Samples from individuals with ASD reacted with a 73 kDa protein. [15]
Positive Reactivity to cerebellum proteins in 88% of samples from individuals with ASD vs 23% controls. [79]
Positive 21% of samples from individuals with ASD vs 2% controls reacted to an approximately 52 kDa protein. [21]
CNS proteins
MBP Positive 35% of samples from individuals with ASD vs 5% controls reacted to MBP and other ligands simulaneously1. [83]
Positive 58% of samples from individuals with ASD vs 9% controls reacted with MBP. [14]
Positive 60% of samples from individuals with ASD vs 0% controls reacted with MBP. [81]
Positive 64% of samples from individuals with ASD vs 0% controls reacted with MBP. [82]
Positive Increased levels of MBP autoantibodies in individuals with ASD vs controls (p < 0.001). [18]
Negative No reactivity to myelin basic protein was detected. [84]
Neurofilaments Positive Anti-neurofilament IgG detected in 41% of samples from individuals with ASD vs 7% controls; IgM detected in 53% ASD vs 22% controls. [79]
Positive 35% of samples from individuals with ASD vs 5% of controls reacted to neurofilament protein1. [83]
Positive 57% of samples from individuals with ASD vs 0% of controls reacted to neurofilament protein. [82]
Positive 55% of samples from individuals with ASD vs 27% of controls reacted to neurofilament protein. [20]
Glial fibrillary acidic protein (GFAP) Positive 32% of samples from individuals with ASD vs 9% of controls reacted to GFAP. [20]
Negative No reactivity to GFAP was detected. [86]
Brain-derived neurotrophic factor (BDNF) Positive Increased BDNF autoantibodies present in individuals with ASD vs healthy controls (p < 0.001). [18]
Other
Anti-nuclear antibodies (ANAs) Positive 20% of samples from individuals with ASD vs 3% of controls had ANAs; levels correlated to disease severity. [117]
Positive 27% of samples from individuals with ASD vs 0% of controls had ANAs. [78]
Negative No ANAs or laminin antibodies were detected. [118]
Hsps Positive Hsp90 autoantibodies detected in 19% of samples from individuals with ASD vs 0% of controls. [119]
Positive Hsp60 autoantibody levels in individuals with ASD were similar to those in autoimmune adults. [120]
Tubulin Positive Autoantibodies detected in 35% of samples from individuals with ASD vs 5% of controls1. [83]
Metallothionein-1 Positive Autoantibodies detected in 35% of samples from individuals with ASD and their families vs 9% of controls and their families; no difference between individuals with ASD and their family members was observed. [121]
Negative No reactivity to metallothionein-1 was detected. [122]

Increased self-reactive antibodies have been detected in individuals with autism spectrum disorder (ASD) compared with controls, using human brain extract unless otherwise indicated.

1

35% of ASD and 5% of control individuals had simultaneous IgG reactivity to myelin basic protein (MBP), myelin-associated glycoprotein, neurofilament protein, ganglioside, sulfatide, α,β-crystallin, chondroitin sulfate, myelin oligodendrocyte glycoprotein and tubulin.

Predisposition to autoimmunity in families with children with ASD

Several studies have demonstrated increased familial histories of autoimmune diseases including type I diabetes, rheumatoid arthritis (RA), hypothyroidism, psoriasis and systemic lupus erythematosus in first degree relatives of individuals with ASD [8791]. Interestingly, 70% of children that had a developmental regression associated with ASD diagnosis had a 1st or 2nd degree relative with an autoimmune disease, compared with 55% of children who had an early onset of ASD symptoms [89]. Of the familial autoimmune disorders linked to ASD, an RA diagnosis was observed in greater than one-third of relatives (34 to 46%) [87], and an increased frequency of hyperthyroidism has also been reported (36% in families with an individual with ASD compared with 14% in control families studied) [92]. In addition, a maternal diagnosis of psoriasis in the 4 years surrounding pregnancy, or allergy/asthma in the second trimester of pregnancy conferred a 2-fold increased risk of ASD diagnosis in offspring [88]. These data suggest that inappropriate maternal immune responses may alter the course of neurodevelopment. It should be noted, however, that many of these reports rely on medical chart reviews or interviews, which can underreport or overreport incidences, respectively, and as such the role of familial autoimmunity in the etiology of ASD requires further investigation.

The presence of maternal anti-fetal antibodies

The breakdown of maternal tolerance to the fetal brain and the generation of antibodies directed toward fetal brain tissue has been detected in mothers of children with ASD (Table 3) [13,16,22,23,93,94]. Braunschweig and colleagues identified the presence of antibodies that were reactive to human fetal brain proteins that were 37 kDa and 73 kDa in molecular weight in 12% of mothers of children with autism [16]. Another study by Croen and colleagues detected the presence of antibodies to human fetal brain proteins of 39 kDa and 73 kDa in mothers of children with autism [23]. Interestingly, the pattern of 37 kDa and 73 kDa staining was more prevalent in children who had autism symptom regression [16], and the 39 kDa and 73 kDa pattern was present more often in children with early onset autism symptoms [23]. In both studies, the specificity of the antibody staining patterns was high, compared with mothers of typically developing children or mothers of children with developmental delay, who did not demonstrate these antibody staining patterns. Notably, Singer and colleagues also reported that sera from mothers of children with ASD was reactive to a 73 kDa embryonic brain protein from rats [13], and to proteins of approximately 36 kDa and 39 kDa derived from human fetal brains at 17 weeks of gestation [13]. At present, the significance of these bands is difficult to ascertain without the identification of moieties to which these bands correspond, which could potentially be ubiquitous, tissue specific, or only expressed during specific development stages. In addition, with the exception of the 73 kDa bands, which were also identified in children with ASD [15], the bands found in the sera of mothers of children with ASD do not correspond to those of their children. To explore the possible pathogenic roles of these antibodies, the effect of these individual antibodies on developing fetuses must be investigated in animal models.

Table 3.

Studies investigating the presence of maternal fetal-reactive antibodies in ASD.

Maternal fetal-reactive antibodies
Study investigators (year) Target of maternal antibodies Tissue source Reference
Warren et al(1990) ~ 54% (6/11) of sera from MAC reacted to lymphocytes from children with ASD. Peripheral blood from children [94]
Dalton et al (2003) Sera from a mother of two children with ASD reacted against murine cerebellar Purkinje cells and brainstem neurons. Mouse postnatal day 1 and adult brain [93]
Zimmerman et al (2007) (i) Of 11 MAC, sera samples from 5 (45%) MAC reacted to a 30 kDa protein band. The remaining 6 (55%) MAC reacted to a > 250 kDa protein band compared with 0% (0/10) of MTDC.
(ii) Reactivity to gestational day 18 fetal brain proteins, but not postnatal or adult rat brain proteins, was detected in sera from MAC, children with autism, and children with other developmental disorders.
Fetal, postnatal and adult rat brain extracts [22]
Croen et al (2008) (i) Reactivity to a 39 kDa protein in 7% (6/84) of samples from MAC compared with 2% (3/152) of MTDC was detected.
(ii) Reactivity to a 73 kDa protein in 13% of samples from MAC and 7% of MTDC was detected.
(iii) Reactivity to both proteins was detected in 37.5% (3/84) of samples from MAC with early onset ASD, but both bands were not present in samples from mothers with children with developmental regression ASD.
(iv) Reactivity to a 37 kDa protein was detected in MAC compared with MTDC, but these results were not significant.
Human fetal brain extract [23]
Braunschweig et al (2008) 37 kDa and 73 kDa protein bands were detected in 11.5% (7/61) of samples from MAC compared with 0% (0/40) MTDC in fetal brain extracts, but not adult brain extracts; these findings were associated with developmental regression ASD. Human fetal and adult brain extract [16]
Singer et al (2008) (i) Reactivity at 36 and 39 kDa in human fetal brain extract.
(ii) Greater band specificity in 100 MAC compared with 100 MTDC at 73 kDa to rat embryo, but not in adult rat, adult human or human fetal brain.
(iii) Reactivity to a 100 kDa protein toward adult human brain region samples.
Adult and fetal rat and human brain extracts [13]
Transfer of human maternal fetal-reactive antibodies to pregnant animals
Animal model Results Reference
Mice Decreased exploratory behavior and decreased ability to orientate was observed. [93]
Rhesus macaques Whole body stereotypies, pacing during mother-preference test and increased nonsocial activity were observed. [95]

ASD autism spectrum disorders, MAC mothers of children with ASD, MTDC mothers of typically developing children

Maternal brain-reactive antibodies and models of neurodevelopment

Two studies have investigated the effect of transferring antibodies from mothers of children with ASD to pregnant animals to determine the mechanism of influence of the antibodies on neurodevelopment [93,95]. The first study transferred sera demonstrated to contain neuronal specific antibodies from a mother of three children, one of whom had autism and another with a language disorder, into pregnant mice by intraperitoneal injection. The offspring of the mice injected with sera from mothers of children with ASD were tested using a battery of behavioral assessments, and demonstrated several deficits in social behavior and motor skills [93]. In addition, examinations of brain specimens from these offspring demonstrated evidence of cerebellar abnormalities [93]. The offspring of mice injected with sera from mothers of healthy, typically developing children did not demonstrate any abnormalities in behaviors or neuropathology [93].

In the second study, human IgG was isolated from mothers of children with ASD or mothers of typically developing children and injected into rhesus macaques [95]. Purified IgG was pooled from the sera of mothers of children with ASD (n = 12) that had demonstrated IgG reactivity to fetal brain extract and intravenously injected into rhesus macaques (n = 4) at gestation days 27, 41 and 55. Similarly, pooled IgG from mothers (n = 7) of typically developing children who had no evidence of reactivity to fetal brain extract were used as controls and injected into pregnant rhesus monkeys (n = 4) [95]. The offspring from both groups were observed, and behavioral testing was conducted from 1 month to 1 year of age. Following weaning, animals exposed to IgG from mothers of children with ASD displayed more social irregularities, including less social behavior and increased whole body stereotypic behaviors, including increased activity, pacing and back-flipping, compared with animals exposed to IgG from mothers of typically developing children, who did not demonstrate these behaviors [95]. These data suggest that the transfer of IgG from mothers of children with ASD can lead to abnormalities in behavioral outcomes. Also, antibodies to neuronal proteins that are contained within the IgG fraction may likely be responsible for such changes; however, further experiments are needed to confirm these findings. In addition, as the antibodies were not selected based on specific reactivity, it is difficult to determine what these antibodies may be reactive to and whether it is one antibody or a combination of several antibodies with different specificities. Moreover, it is unclear whether these antibodies cause neuronal damage, or inhibit functions that lead to behavioral changes.

Therapeutics targeting the immune response in ASD

Pharmaceutical treatments for ASD currently focus on behavioral function, including irritability and stereotypies; however, it is unknown whether these medications significantly affect the immune system. SSRIs and risperidone are common clinical interventions for managing behavior in ASD; these treatments have been reviewed in previous publications (see references [9698]). While risperidone is the only FDA-approved treatment for irritability in ASD, typical and atypical antipsychotics, including clozapine and olanzapine, as well as psychotropics, such as divalproex sodium and lithium, have also been used to treat other behavioral aspects of ASD [96]. The immunological effects of several of these pharmaceuticals have been evaluated in another neurodevelopmental disorder, schizophrenia, in which improved symptoms correlated with reductions in proinflammatory cytokines, and support the potential involvement of cytokine abnormalities in behavioral symptoms [99102]. These findings raise the possibility that in some individuals with ASD, behavior may be altered in part through changes of the immune response, as well as through direct or indirect effects of drugs on the immune response, such as through serotonin receptors on immune cells. It is also possible that the immune profile of a patient prior to treatment may predict responsiveness to specific pharmaceutical interventions. There have been no studies conducted to date specifically examining this relationship in autism. Furthermore, there have been few placebo-controlled clinical trials of anti-inflammatory or immunomodulatory agents to evaluate whether these drugs beneficially alter behavioral responses in individuals with ASD. Thus far, contradictory evidence has been reported in the limited pilot clinical trials undertaken for immunomodulatory therapies, such as steroid [103105] or IVIG treatment [106109], most likely due to patient selection and the highly heterogeneous nature of ASD. Future studies that either demonstrate immune or autoimmune dysfunction in ASD are warranted and may help to identify patient groups that would benefit from certain treatments. In contrast to conventional pharmaceutical treatments, recent studies concluded that more than half of children with ASD were undergoing complementary and alternative medicinal (CAM) therapy to treat specific ASD symptoms [110114], with many treatments aimed at addressing immune dysfunctions. While there is little or conflicting evidence to support many CAM therapeutics, these therapies remain popular within the autism community.

Conclusion

A large amount of evidence supports widespread immune dysregulation in individuals with ASD. Immune dysfunction, increased cytokine production, decreased regulation and the presence of brain reactive antibodies suggest that autoimmune responses may occur, or have previously occurred, in individuals with ASD. Moreover, researchers have demonstrated that purified IgG containing fetal brain-reactive antibodies from the mothers of children with ASD can induce abnormalities in behavioral symptoms in animal models. These findings suggest that neural reactive antibodies may be causative rather than secondary to abnormal brain development in ASD. Taken together, these results suggest that therapeutic immunomodulation, which can lead to more balanced immune responses, may be beneficial. Future therapeutic strategies targeting the immune abnormalities observed in ASD may be warranted. However, it must be noted that the exact role of any immune dysfunction in ASD and whether this dysfunction has a critical role in the core features of ASD or contributes to secondary features, such as epilepsy, sleep disturbances or gastrointestinal symptoms, is not fully understood. Further research into the nature of the immune dysfunction in ASD and the specific targets of CNS/brain-reactive antibodies in individuals with ASD is needed. Such studies will enhance the understanding of the underlying mechanisms of both immune anomalies and altered neurodevelopment in the etiology of ASD, and may reveal targets for future therapeutic interventions.

References

•• of outstanding interest

• of special interest

  • 1.American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders DSM-IV-TR: 4th edition American Psychiatric Association Publishing Inc, Washington DC, USA: (2000). [Google Scholar]
  • 2.Hertz-Picciotto I, Delwiche L: The rise in autism and the role of age at diagnosis. Epidemiology (2009) 20(1):84–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brachlow AE, Ness KK, McPheeters ML, Gurney JG: Comparison of indicators for a primary care medical home between children with autism or asthma and other special health care needs: National Survey of Children’s Health. Arch Pediatr Adolesc Med 161(4):399–405. [DOI] [PubMed] [Google Scholar]
  • 4.Lord C, Leventhal BL, Cook EH Jr: Quantifying the phenotype in autism spectrum disorders. Am J Med Genet (2001) 105(1):36–38. [PubMed] [Google Scholar]
  • 5.Rogers SJ: Developmental regression in autism spectrum disorders. Ment Retard Dev Disabil Res Rev (2004) 10(2):139–143. [DOI] [PubMed] [Google Scholar]
  • 6.Xue M, Brimacombe M, Chaaban J, Zimmerman-Bier B, Wagner GC: Autism spectrum disorders: Concurrent clinical disorders. J Child Neurol (2008) 23(1):6–13. [DOI] [PubMed] [Google Scholar]
  • 7.Zimmerman AW, Jyonouchi H, Comi AM, Connors SL, Milstien S, Varsou A, Heyes MP: Cerebrospinal fluid and serum markers of inflammation in autism. Pediatr Neurol (2005) 33(3):195–201. [DOI] [PubMed] [Google Scholar]
  • 8.Singh VK, Warren RP, Odell JD, Cole P: Changes of soluble interleukin-2, interleukin-2 receptor, T8 antigen, and interleukin-1 in the serum of autistic children. Clin Immunol Immunopathol (1991) 61(3):448–455. [DOI] [PubMed] [Google Scholar]
  • 9.Ashwood P, Wakefield AJ: Immune activation of peripheral blood and mucosal CD3+ lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms. J Neuroimmunol (2006) 173(1–2):126–134. [DOI] [PubMed] [Google Scholar]
  • 10.Smith SE, Li J, Garbett K, Mirnics K, Patterson PH: Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci (2007) 27(40):10695–10702.•• Demonstrated that peripherally administered inflammatory cytokine IL-6 can result in altered neurodevelopment and behavior of offspring in a murine model of gestational maternal infection, and that blocking the cytokine cascade using an antibody against IL-6, even in the presence of a viral trigger, can prevent these changes.
  • 11.Singh VK: Plasma increase of interleukin-12 and interferon-γ. Pathological significance in autism. J Neuroimmunol (1996) 66(1–2):143–145. [DOI] [PubMed] [Google Scholar]
  • 12.Ashwood P, Anthony A, Torrente F, Wakefield AJ: Spontaneous mucosal lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms: Mucosal immune activation and reduced counter regulatory interleukin-10. J Clin Immunol (2004) 24(6):664–673. [DOI] [PubMed] [Google Scholar]
  • 13.Singer HS, Morris CM, Gause CD, Gillin PK, Crawford S, Zimmerman AW: Antibodies against fetal brain in sera of mothers with autistic children. J Neuroimmunol (2008) 194(1–2):165–172.• Investigated the anti-fetal brain antibodies from mothers of children with autism and control mothers (n = 100/group) by Western immunoblotting. Increased reactivity to proteins of 36 and 39 kDa molecular weight in human fetal brain specimens was observed. No reactivity to MBP or GFAP was detected.
  • 14.Singh VK, Warren RP, Odell JD, Warren WL, Cole P: Antibodies to myelin basic protein in children with autistic behavior. Brain Behav Immun (1993) 7(1):97–103. [DOI] [PubMed] [Google Scholar]
  • 15.Singer HS, Morris CM, Williams PN, Yoon DY, Hong JJ, Zimmerman AW: Antibrain antibodies in children with autism and their unaffected siblings. J Neuroimmunol (2006) 178(1–2):149–155.• Investigated the presence of anti-brain antibodies in children with autism and their siblings compared with controls using Western immunoblotting, which demonstrated specific patterns of antibody recognition to several regions of the brain, including recognition of a 100 kDa protein in the caudate and prefrontal cortex, and reactivity to a 73 kDa protein in the cerebellum and cingulate gyrus.
  • 16.Braunschweig D, Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Croen LA, Pessah IN, Van de Water J: Autism: Maternally derived antibodies specific for fetal brain proteins. Neurotoxicology (2008) 29(2):226–231.•• Identified specific anti-fetal brain antibodies that corresponded to putative reactivities to proteins of 37 kDa and 73 kDa in a subset of mothers of children with autism. These patterns were not observed in control mothers. Furthermore, band reactivity appeared to be increased in mothers whose children had a regressive form of autism.
  • 17.Wills S, Cabanlit M, Bennett J, Ashwood P, Amaral D, Van de Water J: Autoantibodies in autism spectrum disorders (ASD). Ann NY Acad Sci (2007) 1107:79–91. [DOI] [PubMed] [Google Scholar]
  • 18.Connolly AM, Chez M, Streif EM, Keeling RM, Golumbek PT, Kwon JM, Riviello JJ, Robinson RG, Neuman RJ, Deuel RM: Brain-derived neurotrophic factor and autoantibodies to neural antigens in sera of children with autistic spectrum disorders, Landau-Kleffner syndrome, and epilepsy. Biol Psychiatry (2006) 59(4):354–363. [DOI] [PubMed] [Google Scholar]
  • 19.Cabanlit M, Wills S, Goines P, Ashwood P, Van de Water J: Brain-specific autoantibodies in the plasma of subjects with autistic spectrum disorder. Ann NY Acad Sci (2007) 1107:92–103. [DOI] [PubMed] [Google Scholar]
  • 20.Singh VK, Warren R, Averett R, Ghaziuddin M: Circulating autoantibodies to neuronal and glial filament proteins in autism. Pediatr Neurol (1997) 17(1):88–90. [DOI] [PubMed] [Google Scholar]
  • 21.Wills S, Cabanlit M, Bennett J, Ashwood P, Amaral DG, Van de Water J: Detection of autoantibodies to neural cells of the cerebellum in the plasma of subjects with autism spectrum disorders. Brain Behav Immun (2009) 23(1):64–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zimmerman AW, Connors SL, Matteson KJ, Lee LC, Singer HS, Castaneda JA, Pearce DA: Maternal antibrain antibodies in autism. Brain Behav Immun (2007) 21(3):351–357.• Demonstrated that auto-reactive antibodies in mothers of children with autism are directed to prenatal, but not postnatal, brain targets and are not observed in mothers of typically developing children.
  • 23.Croen LA, Braunschweig D, Haapanen L, Yoshida CK, Fireman B, Grether JK, Kharrazi M, Hansen RL, Ashwood P, Van de Water J: Maternal mid-pregnancy autoantibodies to fetal brain protein: The early markers for autism study. Biol Psychiatry (2008) 64(7):583–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Daniels WW, Warren RP, Odell JD, Maciulis A, Burger RA, Warren WL, Torres AR : Increased frequency of the extended or ancestral haplotype B44-SC30-DR4 in autism. Neuropsychobiology (1995) 32(3):120–123. [DOI] [PubMed] [Google Scholar]
  • 25.Ferrante P, Saresella M, Guerini FR, Marzorati M, Musetti MC, Cazzullo AG: Significant association of HLA A2-DR11 with CD4 naive decrease in autistic children. Biomed Pharmacother (2003) 57(8):372–374. [DOI] [PubMed] [Google Scholar]
  • 26.Lee LC, Zachary AA, Leffell MS, Newschaffer CJ, Matteson KJ, Tyler JD, Zimmerman AW: HLA-DR4 in families with autism. Pediatr Neurol (2006) 35(5):303–307. [DOI] [PubMed] [Google Scholar]
  • 27.Torres AR, Sweeten TL, Cutler A, Bedke BJ, Fillmore M, Stubbs EG, Odell D: The association and linkage of the HLA-A2 class I allele with autism. Hum Immunol (2006) 67(4–5):346–351. [DOI] [PubMed] [Google Scholar]
  • 28.Warren RP, Yonk J, Burger RW, Odell D, Warren WL: DR-positive T cells in autism: Association with decreased plasma levels of the complement C4B protein. Neuropsychobiology (1995) 31(2):53–57. [DOI] [PubMed] [Google Scholar]
  • 29.Grigorenko EL, Han SS, Yrigollen CM, Leng L, Mizue Y, Anderson GM, Mulder EJ, de Bildt A, Minderaa RB, Volkmar FR, Chang JT, Bucala R: Macrophage migration inhibitory factor and autism spectrum disorders. Pediatrics (2008) 122(2): e438–e445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Odell D, Maciulis A, Cutler A, Warren L, McMahon WM, Coon H, Stubbs G, Henley K, Torres A: Confirmation of the association of the C4B null allelle in autism. Hum Immunol (2005) 66(2): 140–145. [DOI] [PubMed] [Google Scholar]
  • 31.Warren RP, Singh VK, Cole P, Odell JD, Pingree CB, Warren WL, White E: Increased frequency of the null allele at the complement C4B locus in autism. Clin Exp Immunol (1991) 83(3):438–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Campbell DB, D’Oronzio R, Garbett K, Ebert PJ, Mirnics K, Levitt P, Persico AM: Disruption of cerebral cortex MET signaling in autism spectrum disorder. Ann Neurol (2007) 62(3):243–250. [DOI] [PubMed] [Google Scholar]
  • 33.Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R, Bravaccio C, Trillo S, Elia M, Schneider C, Melmed R, Sacco R, Persico AM et al. : A genetic variant that disrupts MET transcription is associated with autism. Proc Natl Acad Sci USA (2006) 103(45):16834–16839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Garbett K, Ebert PJ, Mitchell A, Lintas C, Manzi B, Mirnics K, Persico AM: Immune transcriptome alterations in the temporal cortex of subjects with autism. Neurobiol Dis (2008) 30(3):303–311.• Demonstrated differential gene expression profiles of the temporal gyrus from individuals with autism compared with age-matched controls (n = 6 per group). A total of 130 genes were upregulated and 22 were downregulated in the brains of individuals with autism. Significant changes in gene pathways related to immune function/activation were observed, particularly in late-phase recovery autoimmune responses.
  • 35.Okunishi K, Dohi M, Nakagome K, Tanaka R, Mizuno S, Matsumoto K, Miyazaki J, Nakamura T, Yamamoto K: A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J Immunol (2005) 175(7):4745–4753. [DOI] [PubMed] [Google Scholar]
  • 36.Lintas C, Sacco R, Garbett K, Mirnics K, Militerni R, Bravaccio C, Curatolo P, Manzi B, Schneider C, Melmed R, Elia M et al. : Involvement of the PRKCB1 gene in autistic disorder: Significant genetic association and reduced neocortical gene expression. Mol Psychiatry (2008):doi: 10.1038/mp.2008.21. [DOI] [PubMed] [Google Scholar]
  • 37.Philippi A, Roschmann E, Tores F, Lindenbaum P, Benajou A, Germain-Lederc L, Marcaillou C, Fontaine K, Vanpeene M, Roy S, Maillard S et al. : Haplotypes in the gene encoding protein kinase c-ß (PRKCB1) on chromosome 16 are associated with autism. Mol Psychiatry (2005) 10(10):950–960. [DOI] [PubMed] [Google Scholar]
  • 38.Yang MS, Cochrane L, Conroy J, Hawi Z, Fitzgerald M, Gallagher L, Gill M: Protein kinase C-ß 1 gene variants are not associated with autism in the Irish population. Psychiatric Genetics (2007) 17(1):39–41. [DOI] [PubMed] [Google Scholar]
  • 39.Yang MS, Conroy J, Hawi Z, Gallagher L, Gill M: PRKCB1 is not associated with autism in the Irish population. Am J Med Genetics Part B-Neuropsychiatric Genetics (2006) 141B(7):Abs 766. [Google Scholar]
  • 40.Gregg JP, Lit L, Baron CA, Hertz-Picciotto I, Walker WL, Davis RA, Croen LA, Ozonoff S, Hansen R, Pessah IN, Sharp FR: Gene expression changes in children with autism. Genomics (2008) 91(1):22–29. [DOI] [PubMed] [Google Scholar]
  • 41.Enstrom AM, Lit L, Onore CE, Gregg JP, Hansen RL, Pessah IN, Hertz-Picciotto I, Van de Water JA, Sharp FR, Ashwood P: Altered gene expression and function of peripheral blood natural killer cells in children with autism. Brain Behav Immun (2009) 23(1):124–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Warren RP, Foster A, Margaretten NC: Reduced natural killer cell activity in autism. J Am Acad Child Adolesc Psychiatry (1987) 26(3):333–335. [DOI] [PubMed] [Google Scholar]
  • 43.Vojdani A, Mumper E, Granpeesheh D, Mielke L, Traver D, Bock K, Hirani K, Neubrander J, Woeller KN, O’Hara N, Usman A et al. : Low natural killer cell cytotoxic activity in autism: The role of glutathione, IL-2 and IL-15. J Neuroimmunol (2008) 205(1–2):148–154. [DOI] [PubMed] [Google Scholar]
  • 44.Skaar DA, Shao Y, Haines JL, Stenger JE, Jaworski J, Martin ER, DeLong GR, Moore JH, McCauley JL, Sutcliffe JS, Ashley-Koch AE et al. : Analysis of the RELN gene as a genetic risk factor for autism. Molecular Psychiatry (2005) 10(6):563–571. [DOI] [PubMed] [Google Scholar]
  • 45.Butler MG, Dasouki MJ, Zhou XP, Talebizadeh Z, Brown M, Takahashi TN, Miles JH, Wang CH, Stratton R, Pilarski R, Eng C: Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet (2005) 42(4):318–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Persico AM, D’Agruma L, Maiorano N, Totaro A, Militerni R, Bravaccio C, Wassink TH, Schneider C, Melmed R, Trillo S, Montecchi F et al. : Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder. Mol Psychiatry (2001) 6(2):150–159. [DOI] [PubMed] [Google Scholar]
  • 47.Oliveira G, Ataide A, Marques C, Miguel TS, Coutinho AM, Mota-Vieira L, Goncalves E, Lopes NM, Rodrigues V, Carmona da Mota H, Vicente AM: Epidemiology of autism spectrum disorder in Portugal: Prevalence, clinical characterization, and medical conditions. Dev Med Child Neurol (2007) 49(10):726–733. [DOI] [PubMed] [Google Scholar]
  • 48.Li L, Lu J, Tay SS, Moochhala SM, He BP: The function of microglia, either neuroprotection or neurotoxicity, is determined by the equilibrium among factors released from activated microglia in vitro. Brain Res (2007) 1159:8–17. [DOI] [PubMed] [Google Scholar]
  • 49.Munch G, Gasic-Milenkovic J, Dukic-Stefanovic S, Kuhla B, Heinrich K, Riederer P, Huttunen HJ, Founds H, Sajithlal G: Microglial activation induces cell death, inhibits neurite outgrowth and causes neurite retraction of differentiated neuroblastoma cells. Exp Brain Res (2003) 150(1):1–8. [DOI] [PubMed] [Google Scholar]
  • 50.Stoll G, Jander S: The role of microglia and macrophages in the pathophysiology of the CNS. Prog Neurobiol (1999) 58(3):233–247. [DOI] [PubMed] [Google Scholar]
  • 51.Gehrmann J, Matsumoto Y, Kreutzberg GW: Microglia: Intrinsic immuneffector cell of the brain. Brain Res Brain Res Rev (1995) 20(3):269–287. [DOI] [PubMed] [Google Scholar]
  • 52.Gilmore JH, Jarskog LF, Vadlamudi S: Maternal poly I:C exposure during pregnancy regulates TNFa, BDNF, and NGF expression in neonatal brain and the maternal-fetal unit of the rat. J Neuroimmunol (2005) 159(1–2):106–112. [DOI] [PubMed] [Google Scholar]
  • 53.Shi L, Fatemi SH, Sidwell RW, Patterson PH: Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci (2003) 23(1):297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mehler MF, Kessler JA: Cytokines in brain development and function. Adv Protein Chem (1998) 52:223–251. [DOI] [PubMed] [Google Scholar]
  • 55.Gadient RA, Patterson PH: Leukemia inhibitory factor, interleukin and other cytokines using the GP130 transducing receptor: Roles in inflammation and injury. Stem Cells (1999) 17(3):127–137. [DOI] [PubMed] [Google Scholar]
  • 56.Munoz-Fernandez MA, Fresno M: The role of tumour necrosis factor, interleukin 6, interferon-γ and inducible nitric oxide synthase in the development and pathology of the nervous system. Prog Neurobiol (1998) 56(3):307–340. [DOI] [PubMed] [Google Scholar]
  • 57.Barker V, Middleton G, Davey F, Davies AM: TNFα contributes to the death of NGF-dependent neurons during development. Nat Neurosci (2001) 4(12):1194–1198. [DOI] [PubMed] [Google Scholar]
  • 58.Cacci E, Ajmone-Cat MA, Anelli T, Biagioni S, Minghetti L: In vitro neuronal and glial differentiation from embryonic or adult neural precursor cells are differently affected by chronic or acute activation of microglia. Glia (2008) 56(4):412–425. [DOI] [PubMed] [Google Scholar]
  • 59.Muller N, Ackenheil M: Psychoneuroimmunology and the cytokine action in the CNS: Implications for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry (1998) 22(1):1–33. [DOI] [PubMed] [Google Scholar]
  • 60.Yovel G, Sirota P, Mazeh D, Shakhar G, Rosenne E, Ben-Eliyahu S: Higher natural killer cell activity in schizophrenic patients: The impact of serum factors, medication, and smoking. Brain Behav Immun (2000) 14(3):153–169. [DOI] [PubMed] [Google Scholar]
  • 61.Ashwood P, Wills S, Van de Water J: The immune response in autism: A new frontier for autism research. J Leukoc Biol (2006) 80(1):1–15. [DOI] [PubMed] [Google Scholar]
  • 62.Cohly HH, Panja A: Immunological findings in autism. Int Rev Neurobiol (2005) 71:317–341. [DOI] [PubMed] [Google Scholar]
  • 63.Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li X-M, Ji L, Brown T, Malik M: Elevated immune response in the brain of autistic patients. J Neuroimmunol (2009) 207(1–2):111–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA: Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol (2005) 57(1):67–81.•• Reported significant neuroinflammation in post-mortem human brain specimens and CSF from individuals with autism. Increased levels of proinflammatory cytokines were observed in many brain regions, and significant innate immune activation was observed, particularly in microglia and astroglia. Similar observations were not reported in matched controls.
  • 65.Gupta S, Aggarwal S, Rashanravan B, Lee T: Th1- and Th2-like cytokines in CD4+ and CD8+ T cells in autism. J Neuroimmunol (1998) 85(1):106–109. [DOI] [PubMed] [Google Scholar]
  • 66.Molloy CA, Morrow AL, Meinzen-Derr J, Schleifer K, Dienger K, Manning-Courtney P, Altaye M, Wills-Karp M: Elevated cytokine levels in children with autism spectrum disorder. J Neuroimmunol (2006) 172(1–2):198–205. [DOI] [PubMed] [Google Scholar]
  • 67.Croonenberghs J, Bosmans E, Deboutte D, Kenis G, Maes M: Activation of the inflammatory response system in autism. Neuropsychobiology (2002) 45(1):1–6. [DOI] [PubMed] [Google Scholar]
  • 68.Jyonouchi H, Geng L, Ruby A, Zimmerman-Bier B: Dysregulated innate immune responses in young children with autism spectrum disorders: Their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology (2005) 51(2):77–85. [DOI] [PubMed] [Google Scholar]
  • 69.Sweeten TL, Posey DJ, Shankar S, McDougle CJ: High nitric oxide production in autistic disorder: A possible role for interferon-γ. Biol Psychiatry (2004) 55(4):434–437. [DOI] [PubMed] [Google Scholar]
  • 70.Ashwood P, Enstrom A, Krakowiak P, Hertz-Picciotto I, Hansen RL, Croen LA, Ozonoff S, Pessah IN, Van de Water J: Decreased transforming growth factor pi in autism: A potential link between immune dysregulation and impairment in clinical behavioral outcomes. J Neuroimmunol (2008) 204(1–2):149–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Okada K, Hashimoto K, Iwata Y, Nakamura K, Tsujii M, Tsuchiya KJ, Sekine Y, Suda S, Suzuki K, Sugihara G, Matsuzaki H et al. : Decreased serum levels of transforming growth factor-p1 in patients with autism. Prog Neuropsychopharmacol Biol Psychiatry (2007) 31(1):187–190. [DOI] [PubMed] [Google Scholar]
  • 72.Fombonne E: Are measles infections or measles immunizations linked to autism? J Autism Dev Disord (1999) 29(4):349–350. [DOI] [PubMed] [Google Scholar]
  • 73.Chess S: Autism in children with congenital rubella. J Autism Child Schizophr (1971) 1(1):33–47. [DOI] [PubMed] [Google Scholar]
  • 74.Gillberg C, Coleman M (Eds): The Biology of Autistic Syndromes: 3rd edition Cambridge University Press, Cambridge, UK: (2000). [Google Scholar]
  • 75.Niehus R, Lord C: Early medical history of children with autism spectrum disorders. J Dev Behav Pediatr (2006) 27(Suppl 2):S120–S127. [DOI] [PubMed] [Google Scholar]
  • 76.Rosen NJ, Yoshida CK, Croen LA: Infection in the first 2 years of life and autism spectrum disorders. Pediatrics (2007) 119(1): 61–69. [DOI] [PubMed] [Google Scholar]
  • 77.Todd RD, Hickok JM, Anderson GM, Cohen DJ: Antibrain antibodies in infantile autism. Biol Psychiatry (1988) 23(6):644–647. [DOI] [PubMed] [Google Scholar]
  • 78.Connolly AM, Chez MG, Pestronk A, Arnold ST, Mehta S, Deuel RK: Serum autoantibodies to brain in Landau-Kleffner variant, autism, and other neurologic disorders. J Pediatr (1999) 134(5):607–613. [DOI] [PubMed] [Google Scholar]
  • 79.Plioplys AV, Greaves A, Kazemi K, Silverman E: Immunoglobin reactivity in autism and Retts syndrome. Dev Brain Dysfunct (1994) 7(1):12–16. [DOI] [PubMed] [Google Scholar]
  • 80.Singh VK, Rivas WH: Prevalence of serum antibodies to caudate nucleus in autistic children. Neurosci Lett (2004) 355(1–2):53–56. [DOI] [PubMed] [Google Scholar]
  • 81.Singh VK, Lin SX, Newell E, Nelson C: Abnormal measles-mumps- rubella antibodies and CNS autoimmunity in children with autism. J Biomed Sci (2002) 9(4):359–364. [DOI] [PubMed] [Google Scholar]
  • 82.Singh VK, Lin SX, Yang VC: Serological association of measles virus and human herpesvirus-6 with brain autoantibodies in autism. Clin Immunol Immunopathol (1998) 89(1):105–108. [DOI] [PubMed] [Google Scholar]
  • 83.Vojdani A, Campbell AW, Anyanwu E, Kashanian A, Bock K, Vojdani E: Antibodies to neuron-specific antigens in children with autism: Possible cross-reaction with encephalitogenic proteins from milk, Chlamydia pneumoniae and Streptococcus group A. J Neuroimmunol (2002) 129(1–2):168–177. [DOI] [PubMed] [Google Scholar]
  • 84.Silva SC, Correia C, Fesel C, Barreto M, Coutinho AM, Marques C, Miguel TS, Ataide A, Bento C, Borges L, Oliveira G et al. : Autoantibody repertoires to brain tissue in autism nuclear families. J Neuroimmunol (2004) 152(1–2):176–182. [DOI] [PubMed] [Google Scholar]
  • 85.Libbey JE, Coon HH, Kirkman NJ, Sweeten TL, Miller JN, Stevenson EK, Lainhart JE, McMahon WM, Fujinami RS: Are there enhanced MBP autoantibodies in autism? J Autism Dev Disord 38(2):324–332. [DOI] [PubMed] [Google Scholar]
  • 86.Kirkman NJ, Libbey JE, Sweeten TL, Coon HH, Miller JN, Stevenson EK, Lainhart JE, McMahon WM, Fujinami RS: How relevant are GFAP autoantibodies in autism and Tourette syndrome? J Autism Dev Disord (2008) 38(2):333–341. [DOI] [PubMed] [Google Scholar]
  • 87.Comi AM, Zimmerman AW, Frye VH, Law PA, Peeden JN: Familial clustering of autoimmune disorders and evaluation of medical risk factors in autism. J Child Neurol (1999) 14(6):388–394. [DOI] [PubMed] [Google Scholar]
  • 88.Croen LA, Grether JK, Yoshida CK, Odouli R, Van de Water J: Maternal autoimmune diseases, asthma and allergies, and childhood autism spectrum disorders: A case-control study. Arch Pediatr Adolesc Med (2005) 159(2):151–157. [DOI] [PubMed] [Google Scholar]
  • 89.Molloy CA, Morrow AL, Meinzen-Derr J, Dawson G, Bernier R, Dunn M, Hyman SL, McMahon WM, Goudie-Nice J, Hepburn S, Minshew N et al. : Familial autoimmune thyroid disease as a risk factor for regression in children with autism spectrum disorder: A CPEA study. J Autism Dev Disord (2006) 36(3): 317–324. [DOI] [PubMed] [Google Scholar]
  • 90.Money J, Bobrow NA, Clarke FC: Autism and autoimmune disease: A family study. J Autism Child Schizophr (1971) 1(2):146–160. [DOI] [PubMed] [Google Scholar]
  • 91.Mouridsen SE, Rich B, Isager T, Nedergaard NJ: Autoimmune diseases in parents of children with infantile autism: A case-control study. Dev Med Child Neurol (2007) 49(6):429–432. [DOI] [PubMed] [Google Scholar]
  • 92.Sweeten TL, Bowyer SL, Posey DJ, Halberstadt GM, McDougle CJ: Increased prevalence of familial autoimmunity in probands with pervasive developmental disorders. Pediatrics (2003) 112(5):e420. [DOI] [PubMed] [Google Scholar]
  • 93.Dalton P, Deacon R, Blamire A, Pike M, McKinlay I, Stein J, Styles P, Vincent A: Maternal neuronal antibodies associated with autism and a language disorder. Ann Neurol (2003) 53(4):533–537. [DOI] [PubMed] [Google Scholar]
  • 94.Warren RP, Cole P, Odell JD, Pingree CB, Warren WL, White E, Yonk J, Singh VK: Detection of maternal antibodies in infantile autism. J Am Acad Child Adolesc Psychiatry (1990) 29(6):873–877. [DOI] [PubMed] [Google Scholar]
  • 95.Martin LA, Ashwood P, Braunschweig D, Cabanlit M, Van de Water J, Amaral DG: Stereotypies and hyperactivity in rhesus monkeys exposed to IgG from mothers of children with autism. Brain Behav Immun (2008) 22(6):806–816.•• The first study to demonstrate in a non-human primate model that anti-fetal brain antibodies from mothers of children with autism can result in behavioral changes in offspring that are consistent with some autism-like behaviors. Prenatal administration to rhesus monkeys of anti-fetal brain antibodies purified from mothers of children with autism resulted in social and behavioral changes, including whole body stereotypies.
  • 96.Stigler KA, McDougle CJ: Pharmacotherapy of irritability in pervasive developmental disorders. Child Adolesc Psychiatr Clin N Am (2008) 17(4):739–752. [DOI] [PubMed] [Google Scholar]
  • 97.Soorya L, Kiarashi J, Hollander E: Psychopharmacologic interventions for repetitive behaviors in autism spectrum disorders. Child Adolesc Psychiatr Clin N Am (2008) 17(4):753–771. [DOI] [PubMed] [Google Scholar]
  • 98.McDougle CJ, Stigler KA, Erickson CA, Posey DJ: Atypical antipsychotics in children and adolescents with autistic and other pervasive developmental disorders. J Clin Psychiatry (2008) 69(Suppl 4):15–20. [PubMed] [Google Scholar]
  • 99.Sugino H, Futamura T, Mitsumoto Y, Maeda K, Marunaka Y: Atypical antipsychotics suppress production of proinflammatory cytokines and up-regulate interleukin-10 in lipopolysaccharide-treated mice. Prog Neuropsychopharmacol Biol Psychiatry (2009) 32(2):303–307. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang XY, Zhou DF, Cao LY, Wu GY, Shen YC: Cortisol and cytokines in chronic and treatment-resistant patients with schizophrenia: Association with psychopathology and response to antipsychotics. Neuropsychopharmacology (2005) 30(8):1532–1538. [DOI] [PubMed] [Google Scholar]
  • 101.Zhang XY, Zhou DF, Cao LY, Zhang PY, Wu GY, Shen YC: Changes in serum interleukin-2, −6, and −8 levels before and during treatment with risperidone and haloperidol: Relationship to outcome in schizophrenia. J Clin Psychiatry (2004) 65(7):940–947. [DOI] [PubMed] [Google Scholar]
  • 102.Cazzullo CL, Sacchetti E, Galluzzo A, Panariello A, Adorni A, Pegoraro M, Bosis S, Colombo F, Trabattoni D, Zagliani A, Clerici M: Cytokine profiles in schizophrenic patients treated with risperidone: A 3-month follow-up study. Prog Neuropsychopharmacol Biol Psychiatry (2002) 26(1):33–39. [DOI] [PubMed] [Google Scholar]
  • 103.Bradstreet JJ, Smith S, Granpeesheh D, El-Dahr JM, Rossignol D: Spironolactone might be a desirable immunologic and hormonal intervention in autism spectrum disorders. Med Hypotheses (2007) 68(5):979–987. [DOI] [PubMed] [Google Scholar]
  • 104.Geier MR, Geier DA: The potential importance of steroids in the treatment of autistic spectrum disorders and other disorders involving mercury toxicity. Med Hypotheses (2005) 64(5):946–954. [DOI] [PubMed] [Google Scholar]
  • 105.Matarazzo EB: Treatment of late onset autism as a consequence of probable autoimmune processes related to chronic bacterial infection. World J Biol Psychiatry (2002) 3(3):162–166. [DOI] [PubMed] [Google Scholar]
  • 106.Levy SE, Hyman SL: Novel treatments for autistic spectrum disorders. Ment Retard Dev Disabil Res Rev (2005) 11(2):131–142. [DOI] [PubMed] [Google Scholar]
  • 107.DelGiudice-Asch G, Simon L, Schmeidler J, Cunningham-Rundles C, Hollander E: Brief report: A pilot open clinical trial of intravenous immunoglobulin in childhood autism. J Autism Dev Disord (1999) 29(2):157–160. [DOI] [PubMed] [Google Scholar]
  • 108.Plioplys AV: Intravenous immunoglobulin treatment of children with autism. J Child Neurol (1998) 13(2):79–82. [DOI] [PubMed] [Google Scholar]
  • 109.Schneider CK, Melmed RD, Barstow LE, Enriquez FJ, Ranger-Moore J, Ostrem JA: Oral human immunoglobulin for children with autism and gastrointestinal dysfunction: A prospective, open- label study. J Autism Dev Disord (2006) 36(8):1053–1064. [DOI] [PubMed] [Google Scholar]
  • 110.Levy SE, Hyman SL: Use of complementary and alternative treatments for children with autistic spectrum disorders is increasing. Pediatr Ann (2003) 32(10):685–691. [DOI] [PubMed] [Google Scholar]
  • 111.Hyman SL, Levy SE: Introduction: Novel therapies in developmental disabilities - hope, reason, and evidence. Ment Retard Dev Disabil Res Rev (2005) 11(2):107–109. [DOI] [PubMed] [Google Scholar]
  • 112.Hanson E, Kalish LA, Bunce E, Curtis C, McDaniel S, Ware J, Petry J: Use of complementary and alternative medicine among children diagnosed with autism spectrum disorder. J Autism Dev Disord (2007) 37(4):628–636. [DOI] [PubMed] [Google Scholar]
  • 113.Harrington JW, Rosen L, Garnecho A, Patrick PA: Parental perceptions and use of complementary and alternative medicine practices for children with autistic spectrum disorders in private practice. J Dev Behav Pediatr (2006) 27(2 Suppl):S156–S161. [DOI] [PubMed] [Google Scholar]
  • 114.Wong HH, Smith RG: Patterns of complementary and alternative medical therapy use in children diagnosed with autism spectrum disorders. J Autism Dev Disord (2006) 36(7):901–909. [DOI] [PubMed] [Google Scholar]
  • 115.Jyonouchi H, Sun S, Le H: Proinflammatory and regulatory cytokine production associated with innate and adaptive immune responses in children with autism spectrum disorders and developmental regression. J Neuroimmunol (2001) 120(1–2):170–179. [DOI] [PubMed] [Google Scholar]
  • 116.Jyonouchi H, Sun S, Itokazu N: Innate immunity associated with inflammatory responses and cytokine production against common dietary proteins in patients with autism spectrum disorder. Neuropsychobiology (2002) 46(2):76–84. [DOI] [PubMed] [Google Scholar]
  • 117.Mostafa GA, Kitchener N: Serum anti-nuclear antibodies as a marker of autoimmunity in Egyptian autistic children. Pediatr Neurol (2009) 40(2):107–112. [DOI] [PubMed] [Google Scholar]
  • 118.Singh VK, Rivas WH: Detection of antinuclear and antilaminin antibodies in autistic children who received thimerosal-containing vaccines. J Biomed Sci (2004) 11(5):607–610. [DOI] [PubMed] [Google Scholar]
  • 119.Evers M, Cunningham-Rundles C, Hollander E: Heat shock protein 90 antibodies in autism. Mol Psychiatry (2002) 7(Suppl 2):S26–S28. [DOI] [PubMed] [Google Scholar]
  • 120.Vojdani A, Bazargan M, Vojdani E, Samadi J, Nourian AA, Eghbalieh N, Cooper EL: Heat shock protein and gliadin peptide promote development of peptidase antibodies in children with autism and patients with autoimmune disease. Clin Diagn Lab Immunol (2004) 11(3):515–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Russo AF: Anti-metallothionein IgG and levels of metallothionein in autistic families. Swiss Med Wkly (2008) 138(5–6):70–77. [DOI] [PubMed] [Google Scholar]
  • 122.Singh VK, Hanson J: Assessment of metallothionein and antibodies to metallothionein in normal and autistic children having exposure to vaccine-derived thimerosal. Pediatr Allergy Immunol (2006) 17(4):291–296. [DOI] [PubMed] [Google Scholar]

RESOURCES