Skip to main content
Dialogues in Clinical Neuroscience logoLink to Dialogues in Clinical Neuroscience
. 2012 Sep;14(3):281–292. doi: 10.31887/DCNS.2012.14.3/pchaste

Autism risk factors: genes, environment, and gene-environment interactions

Factores de riesgo del autisme: genes, ambiente e interacciones genes-ambiente

Facteurs de risque d'autisme: gènes, environnement et interactions gène-environnement

Pauline Chaste 1, Marion Leboyer 2,*
PMCID: PMC3513682  PMID: 23226953

Abstract

The aim of this review is to summarize the key findings from genetic and epidemiological research, which show that autism is a complex disorder resulting from the combination of genetic and environmental factors. Remarkable advances in the knowledge of genetic causes of autism have resulted from the great efforts made in the field of genetics. The identification of specific alleles contributing to the autism spectrum has supplied important pieces for the autism puzzle. However, many questions remain unanswered, and new questions are raised by recent results. Moreover, given the amount of evidence supporting a significant contribution of environmental factors to autism risk, it is now clear that the search for environmental factors should be reinforced. One aspect of this search that has been neglected so far is the study of interactions between genes and environmental factors.

Keywords: autism, risk, genetic, environment, interaction

Introduction

Autism was originally defined by Leo Kanner in 1943 as an innate inability to create normal, biologically determined, emotional contact with others. The primacy of the social deficit is widely recognized, and lack of social reciprocity is a central part of the diagnosis. Beyond that, there have been great changes in the past decade in the conceptualization of autism and related disorders, eventually reflected in the draft of the fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5, www.dsm5.org). Indeed, proposed revisions of the precedent edition of the manual (DSM IV-TR)1 include the combination of specific DSM-IV-TR diagnoses into a single broad autism spectrum disorder (ASD), and the identification of two domains of impairment (social communication and interaction, and restricted repetitive behavior) instead of three (social interaction, communication, and restricted repetitive and stereotyped patterns of behavior, interests and activities). These issues are discussed in detail by Volkmar et al in this issue of the journal.

Because of the high heritability estimates in autism, a major focus of research in autism has been on finding the underlying genetic causes, with less emphasis on potential environmental triggers or causes. Although remarkable advances in our knowledge of genetic causes have resulted from these great efforts made in the field of genetics, recent debates about increasing prevalence2 or heritability3 have highlighted the necessity to expand the research on environmental factors. In this review, we summarize the findings regarding genetic, epigenetic, and environmental risk factors identified in autism, and discuss the issue of gene x environment interactions (GxE).

Genetic risk factors

Genetic epidemiology

Heritability

The recurrence risk of pervasive developmental disorder in siblings of children with autism is 2% to 8%;4 and it rises to 12% to 20% if one takes into account the siblings showing impairment in one or two of the three domains impaired in autism respectively.5 Moreover, several twin studies have suggested that this aggregation within families is best explained by shared genes as opposed to shared environment.6-8 Interestingly, the variation of autistic traits in the general population has been shown to be highly heritable, at a similar level of genetic influence to autism itself, even though the results are heterogeneous (heritability 40% to 80%).9,10 These results have led to a huge effort in research to try to unravel the genetic factors underlying the disorder. However, two recent twin studies have provided intriguing results. One study showed that monozygotic twins had higher concordance rates than dizygotic twins for ASDs, attention deficit hyperactivity disorder (ADHD), developmental coordination disorder, and tic disorder with differences in cross-disorder effects between monozygotic and dizygotic twins, raising the question of the specificity of the underlying genetic factors.8 Another study recently challenged the high heritability model of autism, estimating the heritability of autism to be 55 %.3 This study generated considerable discussion, the main criticisms concerning the very large confidence interval of the odds ratio (9% to 81%) and the low participation rate. However, this study is the largest population-based twin study of autism that used contemporary standards for the diagnosis of autism.

The independent heritability of each of the domains of autistic symptomatology is still a matter of debate. While some argue that different autistic symptoms, to a considerable extent, have separate genetic influences,11,12 others argue that there is strong evidence in favor of the hypothesis that symptom domains represent correlated behavioral manifestations of a single underlying quantitative neurodevelopmental impairment.13

Transmission in simplex and multiplex families

According to two studies, the prevalence of de novo chromosomal rearrangements is higher in subjects from simplex families (one affected individual) compared with subjects from multiplex families,14,15 which is consistent with the high rate of notable de novo mutations identified in probands from simplex families.16 This is also consistent with the results of studies which have shown that familial aggregation of subclinical autistic traits may occur only in multiplex families, suggesting differential mechanisms of genetic transmission of autism in the population.17,18

Several biological pathways identified

Individuals with ASD vary in language ability, ranging from absent speech to fluent language, and in cognitive development, ranging from profound intellectual disability to above-average intellectual functioning. Individuals may also show associated medical comorbidities including epilepsy and minor physical anomalies, as well as psychiatric comorbidities, thus showing a wide clinical heterogeneity. The clinical heterogeneity of autism has long been a hindrance to understanding the pathophysiological mechanisms involved. However, although many questions remain and new questions are being raised, the last several years of investigation have brought important pieces to the autism puzzle. Indeed, the identification of specific alleles contributing to ASD has shed light on pathogenic mechanisms.

The only consensus regarding the mode of inheritance of autism is that it is not Mendelian, at least in a vast majority of cases. Several studies were initially in favor of a polygenic model.19-21 Therefore, the initial strategy to unravel genetic factors increasing autism risk was to build large cohorts for linkage and association studies. Given the lack of replication of the results, consortia gathering several cohorts were created to increase the power of the studies, but without clear results. With regard to nonparametric linkage, the largest study to date included 1181 multiplex families22 and did not identify highly significant evidence for linkage. Moreover, the three large studies using genome -wide association that have been published thus far each highlight a single, non-overlapping risk locus.23-25 These findings led some authors to predict that few, if any, common variants have a substantial impact on risk (odds ratio >1.2), but many common variants could have a more modest impact.26

Going back to an individual approach, already used in mental retardation, the search for rare mutations or chromosomal rearrangements was then used, allowing new hypotheses about the mechanisms involved in autism. While the existence of many genetic syndromes associated with autism first led to considering the existence of genetic heterogeneity mirroring the clinical variability, genetic studies in idiopathic autism confirmed the existence of different defects in common pathways. The results suggest that autism may be caused by a multitude of genetic alterations that ultimately affect only limited biological pathways of brain development and plasticity.

First, specific genetic syndromes like Rett syndrome or Fragile-X syndrome, or cytogenetic abnormalities, the most common being the 15qll-ql3 duplication of the maternal allele, associated with ASD affect synaptic plasticity Moreover, the first mutations identified in idiopathic autism involve synaptic genes like NLGN3 and NLGN4X,27,28 or SHANK329-31 Results were enriched by the development of whole-genome screening methodologies which have shown that genetic structural variation contributes significantly to autism. The detection of copy number variations (CNVs), with constantly increasing resolution, consistently confirmed the importance of the synaptic function in autism.22 Several subsequent studies showed CNV in the NLGN-NRXN-SHANK pathway, and other synaptic genes such as SynGAP and DLGAP215,32,33 (Table I).

Table I. Main copy number variation (CNV) studies.

Study Patients Controls Array Candidate region/ Genetic result region/ Other results
ref gene identified gene
112 350 cases (SSC) 337 NIMH controls CGH array FOXP1 De novo deletion Patients with autism without
DPP6, SCN4A, WNT3 mental retardation show only
and WNT9B modest increases in large CNV
burden compared with controls
39 852 quartets and 852 unaffected Illumina 1M 7q11.23 (Williams Recurrent de novo Only modest correlation with IQ
252 trios (SSC) siblings Beuren region), 1q21.1, deletions/duplications Rare inherited CNVs equally
16p13.2, CDH13 represented in patients and
unaffected siblings
Estimation of >234 distinct
genomic regions contributing to
large ASD-related de novo
structural variations
113 510 quartets and NimbleGen 7q11.23 (Williams Recurrent de novo
277 trios HD2 2.1 Beuren region), deletions/duplications
million 16p13.2, Rare homozygous
probe COMMD1, deletions
microarray CACNA2D4
33 996 cases (AGP) 1287 Illumina 1M SHANK2, SynGAP, De novo deletion N de novo CNV multiplex =
DLGAP2, PTCHD1 Maternal transmission simplex
(X chromosome) Common pathways/
intellectual disability
114 859 cases (ACC), 1409 llumina 550K PARK2, UBE3A Statistically significant
1336 cases (AGRE) RFWD2, and FBXO40 association
(ubiquitination), NLGN1 Inherited del/dup
CNTN4, 15q11, 22q11, absent in controls
NRXN1
115 859 cases (ACC), 1448 Illumina 550K CNTNAP2, NRXN1, Inherited deletion
912 families (AGRE) PCDH9, BZRAP1, absent in controls
MDGA2, RAI1, TSC2, Statistically significant
NLGN1 association
116 104 including Affymetrix 22q11 De novo deletion Role of inherited CNVs
88 consanguineous 500K et PCDH10, CNTN3 homozygous inherited
CGH array deletion
117 397 cases AGRE 372 CGH array 15q11, 22q11, 16p11 De novo deletion
15 427 (Can.) 500 Affymetrix 16p11 Statistically significant N de novo CNV
500K NLGN4 DLGAP2, Association simplex>multiplex
SHANK3, De novo deletion
22q11, 15q11-q13 16p11 De novo deletion
PTCHD1 Maternal transmission
(X chromosome)
NRXN1 Statistically significant
Association
43 1441 AGRE 1420 parents, Affymetrix 16p11 Statistically significant
2814 controls 5.0/Affymetrix Association
500K
40 712 AGRE 837 CGH array 16p11 Statistically significant
+ NIMH Association
14 264 families 99 families Agilent 85K FLI16237, SLC4A10, De novo deletion N CNV de novo greater in
AGRE A2BP1, FHIT De novo deletion/ probands from simplex families
15q11-q13,22q13.33, duplication
16p11.2
22 1496 families Unaffected Affymetrix NRXN1 De novo deletion in
AGP relatives two affected sisters
118 29 (Fr) - CGH array GRIA3 (Xq2S), Maternal transmission
15q11-q13 (X chromosome)
De novo duplication

The analysis of genes affected by rare CNVs has confirmed the crucial role of abnormalities in synapse formation and maintenance, but also identified other affected pathways, including cellular proliferation and motility, GTPase/Ras signaling, and neurogenesis.33-35 It is interesting to note that some de novo or inherited CNVs associated with ASD, which recur at the same locus among unrelated individuals, have so far resisted identification of specific ASD genes. One of the most frequent of these involves the 16p11 region. Moreover, as techniques are improving very fast, the first results of large-scale studies using whole-exome sequencing, ie, the mapping of every base of DNA across the exome, were recently released.36-38 These three studies report de novo mutations with a twofold to fourfold increase in de novo nonsense variants among affected subjects over that expected by chance. Interestingly, two of these studies report that spontaneous changes are correlated with paternal age.36-38 One of these studies strongly suggests the involvement of brain signaling as a new biological pathway.37

It is now clear that there is a huge genetic heterogeneity in ASD, involving both a locus heterogeneity and an allelic heterogeneity. The exome sequencing studies suggest that the recent results predicting up to 234 loci contributing to ASD risk39 are probably even an underestimation.37,38 Some important Web resources cataloguing genetic contributors in ASD include the SFARI Gene database (https://gene.sfari.org/autdb/), the AutDB database (http://www.mindspec.org/autdb.html), and the Autism Chromosome Rearrangement Database (http://projects.tcag.ca/autism/).

Remaining questions

Genotype/phenotype correlations

One of the most important remaining unsolved issues is the understanding of the relationships between genetic variation and phenotype, given the recent observations that identical mutations may be associated with highly divergent phenotype. Indeed, identical CNVs have been associated with autism and schizophrenia, notably 16p11 rearrangements.15,40-43 STIANK3 and NRXN1 genes were also suggested to be involved in schizophrenia,44-46 and genes implicated in autism and/or schizophrenia were significantly enriched in ADHD CNV genes in one study.47 A first hypothesis to explain this phenotypic heterogeneity is that a secondary insult is necessary during development to result in the phenotype, as in the “twohit model” proposed in developmental delay.48 Several studies recently supported the existence of such a combination of rare variants in some cases.39,49,50 Another hypothesis is the contribution of both rare and frequent variants. This would be consistent with the observations of broader subthreshold traits in siblings.51 Although, as we have already mentioned, association studies have not provided clear evidence of the contribution of common variants in autism, a recent analysis of genetic variations associated with ASD suggests that common and rare variants contribute to ASD by perturbation of common neuronal networks.35 The last hypothesis, which is not mutually exclusive with other hypotheses, is the contribution of environmental factors which modify the phenotype.

Sex ratio

Autism affects males four times more than females,52 and the cause for this difference is not well understood. Several theories have been proposed, among which the involvement of the sex chromosome in the etiology of ASD, and the role of hormonal influences in utero (for review see ref 53). However, none of these theories has been confirmed yet.

Intellectual disability

Intellectual disability (ID) is present in 65% to 75% ol individuals with a strict diagnosis of autistic disorder, and in 30% to 55% if all ASDs are considered.54,55 Two different models are proposed to explain this overlap. The first model proposes that intellectual disability and ASD share common genetic bases, common genes causing a continuum of developmental disorders that manifest in different ways depending on other genetic or environmental factors. This model is supported by the observation that all recurrent genetic defects reported in autism, including autism without mental retardation, have been causally implicated in intellectual disability,56 and that analysis of the genes affected by rare CNVs reveal that they are strongly functionally related to genes previously implicated in intellectual disability.33,34 Hie second model assumes that in patients with intellectual disability, the general cognitive disability unmasks the limitations in the individual's capacity for social reciprocity.57 This model is supported by evidence that indicates a continuous distribution of autistic traits in normal population9 and etiological similarity across ASD and autistic traits in the general population.58 However it is tempered by the results of a recent large-scale CNV study showing a strong effect of large rare genie de novo CNVs on the presence or absence of an ASD diagnosis, but did not support IQ as a useful predictor for probands carrying these risk variants.39

Environmental risk factors

Indirect evidence suggesting a contribution of environmental factors

Prevalence

Prevalence studies of autism spectrum disorders conducted in recent years have been the source of an important debate because of a steady and highly significant increase of estimates of the total prevalence of pervasive developmental disorders. Indeed, while the prevalence was estimated at 6 per 1000 in a population of school children in 2005,52 recent studies have gone so far as to estimate the prevalence to be one child in 38.59The last prevalence estimates in the United States, released by the Centers for Disease Control recently,60 reached 1 in 88 child in 2008, while their previous estimate was one in 110 in 2006. However, most of the studies are not comparable in method or in the populations studied. One hypothesis is that this increase is the result of enlargement of diagnostic criteria, and the growing importance of screening for ASDs. The results of an epidemiological study from England, based on a national sample from 2007, support this hypothesis. Indeed the authors found a rate of about 1% in adults across the entire age range, without a significant reduction in the older part of the sample, as one would expect if the prevalence had increased in recent years.61 However, another study suggested that diagnostic substitution, especially for the most severe cases, and better ascertainment, especially for children at the less severe end of the spectrum, explain only a part of the linear increase observed in the California registry.62

While the hypothesis of an increased incidence in relation to environmental factors could not be confirmed nor excluded definitely, studies using the same protocol several years apart are required.63 Nevertheless, it seems reasonable to think that there may be both a real increase in the number of cases and an increase in the detection of affected children, and one should not wait for the results of these studies to search for environmental factors increasing risk for autism.

Immune dysfunction

Several lines of evidence support the hypothesis of immune changes in autism. First, several studies have shown abnormalities in the peripheral immune system such as T-cell dysfunction, autoantibody production, increase in the number of activated B cells and NK cells, and increase in proinflammatory cytokines.64-66 Moreover, a landmark study provided evidence for microglial and astroglial activation in brain of patients with ASD.67 The most prominent microglial reaction was observed in the cerebellum and cerebral white matter. The authors also found, in the cerebrospinal fluid of other patients, an increase of proinflammatory and modulatory cytokines. Another study consistently reported microglial activation in the dorsolateral prefrontal cortex in brains of patients with ASD.68 This neuroglial response may result from either a primary disturbance of neuroglial function or unknown factors that disturb prenatal or postnatal CNS development.

Transcriptome

The first comprehensive gene-expression analysis of brains of patients with ASD recently reported differences in transcriptome organization between autistic and normal brain.69 The measure of messenger RNA levels, using Illumina microarrays, in three regions of post mortem brains from patients with autism and controls, showed 444 genes differentially expressed between the cerebral cortices of the autistic and control brains. Moreover, the authors identified two discrete modules of coexpressed genes associated with autism. While the first module, which is related to synaptic function and neuronal projection, was underexpressed in autism cases, the second module, which was enriched for immune genes and glial markers, was overexpressed. These results are consistent with the findings mentioned above, implicating synaptic dysfunction as well as immune dysregulation in autism. Interestingly, the first module shows a highly significant enrichment for variants genetically associated with autism, further supporting the genetic basis of synaptic dysfunction in ASD. On the contrary, the authors did not find any evidence for a genetic etiology for the upregulation of the genes of the second module, suggesting that it is probably a nongenetic, adaptive, or environmental process.

Epigenetic dysregulation in autism

Epigenetic marks define chromatin state and regulate the expression of many genes without affecting primary DNA sequence. These include DNA methylation, and histone methylation and acetylation, and can be modified in response to either genetic mutations or environmental exposure. Several elements indicate the existence of epigenetic dysregulation in autism. First, several syndromes associated with autism are caused by mutations in genes involved in epigenetic regulation. For example, there are abnormalities of transcriptional regulation in Rett syndrome, caused by a mutation of methyl-CpGbinding protein 2 (MeCP2). Indeed, MeCP2 binds to methylated DNA and represses the transcription of target genes.70 Second, several chromosomal regions subject to parental imprinting (transcriptional regulation of either the maternal allele or the paternal allele inducing monoallelic expression) were associated with autism. Notably, microduplications or microdeletions of the region 15qllql3, which is subject to parental imprinting, have been repeatedly reported in subjects with autism.71,72 Moreover, in Turner's syndrome, women who have monosomy of the X chromosome (X0), often have autistic traits which are correlated with the parental origin of the X chromosome they received.73 Regarding common variants, several studies have reported an association of ASD with single-nucleotide polymorphisms in a gene which is directly involved in methylation.74,75 Last, direct changes in DNA methylation profile in lymphoblastoid cells of autistic patients were reported recently76 showing a decreased expression of retinoic acid-related orphan receptor alpha gene (RORA) and B-cell lymphoma 2 (BCL-2).

Although most of the epigenetic modifications described above are underpinned by genetic mechanisms, the evidence of the contribution of epigenetic dysregulation in autism raises the issue of the role of epigenetic modifications by environmental factors. An example is assisted conception. Indeed, while it was shown that in vitro fertilization and ovulation induction can result in abnormal methylation and dysregulation of imprinted genes,77 epidemiologic studies on the use of assisted reproductive technology and the risk of autism found conflicting results.78

Direct evidence for the contribution of environmental factors

There has been much discussion about the initial suggestion that MMR (measles mumps, rubella) vaccine.79 However there is now a scientific consensus that the evidence favors rejection of a causal relationship between thimerosal-containing vaccines and autism, based on multiple epidemiologic studies which did not support a link between thimerosal-containing vaccines and ASD (see the review by Parker in ref 80). However, other environmental factors are likely to contribute to a significant proportion of ASD risk.

Prenatal and perinatal factors

A recent meta-analysis of prenatal factors, limited to pregnancy-related factors, identified few significant risk factors.81 The main factors are maternal gestational diabetes, maternal bleeding during pregnancy, and maternal medication. The latter issue will be further discussed later. Moreover, increased risk was also found in this meta-analysis for first-born children compared with children born third or later, and, in Nordic countries, for offspring of mothers born abroad. Exposure to intrauterine infections was associated with a significant increase in risk for autism in the analysis limited to the four studies that controlled for multiple covariates or used sibling controls. Hie association between maternal infection and autism risk is further supported by the results with rodent models of the maternal infection. In these animal models, gestational viral infection is mimicked by systemic administration of Poly I:C, a synthetic doublestranded RNA, which elicits an innate immune response. It seems that gestational viral infections trigger a maternal immune response, which can perturb fetal brain development, at least in part through interleukin-6.82

In another meta-analysis focusing on the perinatal and neonatal period,83 the same authors identified several potential risk factors, the main being fetal presentation, umbilieal-cord complications, fetal distress, birth injury or trauma, multiple birth, maternal hemorrhage, summer birth, low birth weight, small for gestational age, low 5minute Apgar score, meconium aspiration, neonatal anemia, ABO or Rh incompatibility, and hyperbilirubinemia. Feeding difficulties and congenital malformation that are also mentioned should rather be considered as symptoms of an underlying cause of autism. The identification of summer birth as a risk factor is consistent with the results of a recent study showing that maternal infection in the first trimester increases autism risk.84

Overall, preterm birth was not associated with the risk of autism. However, a recent study based on rigorous diagnostic assessment using validated instruments suggested an association between preterm birth and risk for ASD consistent with the results of most previous prospective studies.85 This study estimated the prevalence to be 5% in adolescents who had a birth weight <2000 g in the US, which is significantly greater than the last national prevalence estimates. Most of these subjects were born preterm (96.7%); however, 32.3% were small for gestational age and the authors did not use multivariate analyses to simultaneously control for birth weight and gestational age. This methodological issue concerns most of the studies on perinatal and neonatal risk factors, which makes it difficult to interpret the results of these studies, since many of the events studied are likely to occur at the same time. Another limitation to the interpretation of the results is that some studies have suggested that increased rates of birth and pregnancy complications are likely secondary to familial factors associated with autism.86

Socioeconomic status

Although one study did not find any association between risk of autism and socioeconomic status87 including maternal education level, the latter may significantly influence the age of first single words.88 Moreover, as already mentioned, autism risk was found to be significantly increased for the offspring of mothers born abroad in a meta-analysis.81 This risk was further specified in a very recent study89 showing that children of migrant parents are at an increased risk of autism with intellectual disability, especially when parents migrated to Sweden from regions with a low human development index, and a decreased risk of high-functioning autism. The risk for low-functioning autism peaked when migration occurred around the time of pregnancy. Different mechanisms can be proposed to explain these results, such as the high level of maternal stress or low immunity regarding common infections.

Drugs and toxic exposure

As previously mentioned, exposure to medication during pregnancy was found to increase autism risk in the most recent meta-analyses.81 Prenatal exposure to valproate is a recognized risk factor for ASD, especially in the first trimester of pregnancy. Children exposed in utero to valproate have 8-fold increased risk to have ASD.90 Interestingly, a downregulation of NLGN3 was observed in hippocampal subregions and the somatosensory cortex of mice prenatally exposed to valproate.91 Moreover, one ol the major concerns regarding medication exposure during pregnancy concerns the use of antidepressants, since selective serotonin reuptake inhibitor medication during pregnancy increased from 1.5% in 1996 to 6.4% in 2004 and 6.2% in 2005.92 It was suggested that antidepressant exposure during pregnancy modestly increases the risk of ASD, especially in the first semester.93 Lastly, exposure in utero to an organophosphate insecticide, chlorpyrifos, was found to increase ASD risk and it was suggested that synthetic chemicals should be far more explored.94

Gene-environment interaction

Genetic heterogeneity can be one explanation for the absence of replication of association studies in autism. However, these results could also be interpreted within the framework of a GxE interaction model.95 If, for example, an association has been found in a sample with subjects frequently exposed to a particular environmental risk but not in those infrequently exposed, and exposure was not ascertained, the source of nonreplication will remain elusive.

The existence of interactions between genetic background and environmental factors in autism was first suggested for perinatal complications. Indeed, in an epidemiological study on autism that included a comparison group of siblings,96 unaffected siblings had fewer prenatal and perinatal complications than their affected siblings, but more than control subjects. This suggested that individuals with autism may react differently to the same environmental stimuli and may have less tolerance to the prenatal experience compared with their siblings. Moreover, studies of animal models have suggested that genetic defects in synaptic function may alter sensitivity to the environment. Indeed a study has shown that neuroligin-deficient mutants of C. elegans nematodes are hypersensitive to oxidative stress.97 Another study reported that the hippocampal slices from MecP2- deficient mice are more susceptible to hypoxia.98 Conversely, it was shown in an animal models that the most significant pathology of the extremely premature brain is the disruption of synaptic development.99 It was thus hypothesized that synaptic gene defects could interact with environmental factor to increase autism risk. Another hypothesis is the interaction between genetic variations melatonin pathway genes and oxidative stress. Indeed, low plasma melatonin concentration is a frequent trait in ASD patients,100,101caused by a primary deficit in acetylserotonin-melhyl-transferase (ASMT) activity. It was suggested that genetic variations contribute to the enzymatic deficit.100 Several studies have suggested an antioxidant effect of melatonin in vitro,102,103 and it was shown that the administration of melatonin reduces oxidative stress in newborn infants exposed to infection or fetal distress,104 and promotes oligodendroglial maturation in the newborn rat with abnormal white matter related to fetal hypoxia.105 Thus it could have a neuroprotective effect in the newborn exposed to fetal distress. Interestingly Gardener et al81 noted that several of the perinatal and neonatal risk factors they identified may be associated with an increased risk of hypoxia. We can thus hypothesize that a deficit of melatonin could be taken into account in the consequences of perinatal distress.

Beyond these observations, available evidence for the contribution of GxE to autism risk comes from animal models. In a first study,106 mice haploinsufficient for the TSC2 gene demonstrated a lack of normal social approach behavior only when exposed to maternal immune activation. The authors propose that disinhibited TSC/mTOR signaling downstream of mediators of gestational immune activation effects amplifies their impact on the mutant mice fetal brain; or that the immune activation may be more pronounced in mutants because of the role of TSC/mTOR signaling in the regulation of the adaptive immune response. Moreover, exploring further the possible interaction between tuberous sclerosis and maternal immune activation in a cohort of individuals with tuberous sclerosis, the authors found an association of late gestation with peak seasonal flu activity specifically in individuals affected by ASD. These results suggest that late gestation is the main period of vulnerability of neurodevelopment to flu infection, which is in contradiction with results, discussed earlier, suggesting that summer birth and maternal infection during the first trimester are risk factors for ASD. However, we can reasonably hypothesize that the period of main vulnerability to infection during gestation may vary according to genetic factors, and that there is a specific period of vulnerability of neurodevelopment during late gestation in tuberous sclerosis. In another animal model,107 prenatal maternal immune activation and expression of a mutant DISC1 protein interacted to produce an altered pattern of sociability. This neurobehavioral profile was absent in untreated mice expressing the mutant.

Although these results are very encouraging, family and population-based association studies in autism have not been extended for GxE interaction yet. One of the main problems with this kind of study is that power to detect GxE interactions is even lower than power to detect genetic or environmental main effects, and the enthusiasm for GxE research in other psychiatric disorders has recently been tempered by the absence of replication of many positive results.108 Nevertheless, these studies are needed since they might help us to understand the inconsistency in results found in classical association studies and provide useful hints with regard to prevendon. Two large-scale prospective epidemiological studies aiming at exploring environmental factors and GxE interaction were recently launched. The National Children's study will follow 100 000 children in the US from conception to age 21.109 Biological samples are collected from each mother and child. The Autism Birth Cohort will follow 100 000 children from conception to age 7.110 Biological samples are collected from children and their parents. Interestingly, an encouraging result came from an association study in attention deficit with hyperactivity disorder (ADHD), which found GxE effects on ASD symptoms in children with ADHD. Multiple regression analyses for GxE effects showed that 5-HTTLPR S/S genotype interacted with maternal smoking during pregnancy, increasing problems in social interaction, and also interacted with low birth weight, increasing rigid behavior.111 Last, given the new understanding of the genetic architecture of autism, further study of the interaction of rare variants associated with ASD and environmental factors in populations carrying identical mutations would be useful but are difficult to perform due to the small number of carriers.

Conclusion

Contrary to the frequent assertion that we know only little of the risk of autism, major advances have been made in the past decade in this domain. In particular, recent advances in genetics have allowed a new conceptualization of molecular and cellular mechanisms of the pathology. At the same time new questions are raised, including the role of common variants and the relationship between genotype and phenotype. The contribution of environmental factors through additive or multiplicative effect needs to be further explored. New funding will need to be dedicated to this domain of research, which has been sparsely funded until very recently.

Contributor Information

Pauline Chaste, INSERM U 955, IMRB, Psychiatry Genetics, Creteil, France; FondaMental Foundation, Créteil, France.

Marion Leboyer, INSERM U 955, IMRB, Psychiatry Genetics, Creteil, France; FondaMental Foundation, Créteil, France ; APHP, Henri Mondor-Albert Chenevier Hospitals, Department of Psychiatry, Creteil, France; Paris-Est University, Faculty of Medicine, IFR10, Creteil, France.

REFERENCES

  • 1.American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington, DC: American Psychiatric Association; 1994 [Google Scholar]
  • 2.Weintraub K. The prevalence puzzle: autism counts. Nature. 2011;479:22–24. doi: 10.1038/479022a. [DOI] [PubMed] [Google Scholar]
  • 3.Hallmayer J., Cleveland S., Torres A., et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011;68:1095–1102. doi: 10.1001/archgenpsychiatry.2011.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Muhle R., Trentacoste SV., Rapin I. The genetics of autism. Pediatrics. 2004;113:e472–e486. doi: 10.1542/peds.113.5.e472. [DOI] [PubMed] [Google Scholar]
  • 5.Bolton P., Macdonald H., Pickles A., et al. A case-control family history study of autism. J Child Psychol Psychiatry. 1994;35:877–900. doi: 10.1111/j.1469-7610.1994.tb02300.x. [DOI] [PubMed] [Google Scholar]
  • 6.Bailey A., Le Couteur A., Gottesman I., et al. Autism as a strongly genetic disorder: evidence from a British twin study. Psychol Med. 1995;25:63–77. doi: 10.1017/s0033291700028099. [DOI] [PubMed] [Google Scholar]
  • 7.Folstein S., Rutter M. Infantile autism: a genetic study of 21 twin pairs. J Child Psychol Psychiatry. 1977;18:297–321. doi: 10.1111/j.1469-7610.1977.tb00443.x. [DOI] [PubMed] [Google Scholar]
  • 8.Lichtenstein P., Carlstrom E., Rastam M., Gillberg C., Anckarsater H. The genetics of autism spectrum disorders and related neuropsychiatric disorders in childhood. Am J Psychiatry. 2010;167:1357–1363. doi: 10.1176/appi.ajp.2010.10020223. [DOI] [PubMed] [Google Scholar]
  • 9.Constantino JN., Todd RD. Autistic traits in the general population: a twin study. Arch Gen Psychiatry. 2003;60:524–530. doi: 10.1001/archpsyc.60.5.524. [DOI] [PubMed] [Google Scholar]
  • 10.Ronald A., Happe F., Price TS., Baron-Cohen S., Plomin R. Phenotypic and genetic overlap between autistic traits at the extremes of the general population. J Am Acad Child Adolesc Psychiatry. 2006;45:1206–1214. doi: 10.1097/01.chi.0000230165.54117.41. [DOI] [PubMed] [Google Scholar]
  • 11.Ronald A., Larsson H., Anckarsater H., Lichtenstein P. A twin study of autism symptoms in Sweden. Mol Psychiatry. 2011;16:1039–1047. doi: 10.1038/mp.2010.82. [DOI] [PubMed] [Google Scholar]
  • 12.Happe F., Ronald A., Plomin R. Time to give up on a single explanation for autism. Nat Neurosci. 2006;9:1218–1220. doi: 10.1038/nn1770. [DOI] [PubMed] [Google Scholar]
  • 13.Constantino JN. The quantitative nature of autistic social impairment. Pediatr Res. 2011;69(5 Pt 2):55R–62R. doi: 10.1203/PDR.0b013e318212ec6e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sebat J., Lakshmi B., Malhotra D., et al. Strong association of de novo copy number mutations with autism. Science. 2007;316:445–449. doi: 10.1126/science.1138659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Marshall CR., Noor A., Vincent JB., et al. Structural variation of chromosomes in autism spectrum disorder. Am J Hum Genet. 2008;82:477–488. doi: 10.1016/j.ajhg.2007.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.O'Roak BJ., Deriziotis P., Lee C., et al. Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet. 2011;43:585–589. doi: 10.1038/ng.835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Constantino JN., Zhang Y., Frazier T., Abbacchi AM., Law P. Sibling recurrence and the genetic epidemiology of autism. Am J Psychiatry. 2010;167:1349–1356. doi: 10.1176/appi.ajp.2010.09101470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Virkud YV., Todd RD., Abbacchi AM., Zhang Y., Constantino JN. Familial aggregation of quantitative autistic traits in multiplex versus simplex autism. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:328–334. doi: 10.1002/ajmg.b.30810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jorde LB., Hasstedt SJ., Ritvo ER., et al. Complex segregation analysis of autism. Am J Hum Genet. 1991;49:932–938. [PMC free article] [PubMed] [Google Scholar]
  • 20.Pickles A., Bolton P., Macdonald H., et al. Latent-class analysis of recurrence risks for complex phenotypes with selection and measurement error: a twin and family history study of autism. Am J Hum Genet. 1995;57:717–726. [PMC free article] [PubMed] [Google Scholar]
  • 21.Risch N., Spiker D., Lotspeich L., et al. A genomic screen of autism: evidence for a multilocus etiology. Am J Hum Genet. 1999;65:493–507. doi: 10.1086/302497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Szatmari P., Paterson AD., Zwaigenbaum L., et al. Mapping autism risk loci using genetic linkage and chromosomal rearrangements. Nat Genet. 2007;39:319–328. doi: 10.1038/ng1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wang K., Zhang H., Ma D., et al. Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature. 2009;459:528–533. doi: 10.1038/nature07999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Weiss LA., Arking DE., Daly MJ., Chakravarti A. A genome-wide linkage and association scan reveals novel loci for autism. Nature. 2009;461:802–808. doi: 10.1038/nature08490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Anney R., Klei L., Pinto D., et al. A genome-wide scan for common alleles affecting risk for autism. Hum Mol Genet. 2010;19:4072–4082. doi: 10.1093/hmg/ddq307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Devlin B., Melhem N., Roeder K. Do common variants play a role in risk for autism? Evidence and theoretical musings. Brain Res. 2010;1380:78–84. doi: 10.1016/j.brainres.2010.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jamain S., Quach H., Betancur C., et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat Genet. 2003;34:27–29. doi: 10.1038/ng1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Laumonnier F., Bonnet-Brilhault F., Gomot M., et al. X-linked mental retardation and autism are associated with a mutation in the NLGN4 gene, a member of the neuroligin family. Am J Hum Genet. 2004;74:552–557. doi: 10.1086/382137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Durand CM., Betancur C., Boeckers TM., et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–27. doi: 10.1038/ng1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gauthier J., Spiegelman D., Piton A., et al. Novel de novo SHANK3 mutation in autistic patients. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:421–424. doi: 10.1002/ajmg.b.30822. [DOI] [PubMed] [Google Scholar]
  • 31.Moessner R., Marshall CR., Sutcliffe JS., et al. Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet. 2007;81:1289–1297. doi: 10.1086/522590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Berkel S., Marshall CR., Weiss B., et al. Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat Genet. 2010;42:489–491. doi: 10.1038/ng.589. [DOI] [PubMed] [Google Scholar]
  • 33.Pinto D., Pagnamenta AT., Klei L., et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature. 2010;466:368–372. doi: 10.1038/nature09146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gilman SR., Iossifov I., Levy D., Ronemus M., Wigler M., Vitkup D. Rare de novo variants associated with autism implicate a large functional network of genes involved in formation and function of synapses. Neuron. 2011;70:898–907. doi: 10.1016/j.neuron.2011.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ben-David E., Shifman S. Networks of neuronal genes affected by common and rare variants in autism spectrum disorders. PLoS Genet. 2012;8:e1002556. doi: 10.1371/journal.pgen.1002556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Neale BM., Kou Y., Liu L., et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature. 2012;485:242–245. doi: 10.1038/nature11011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.O'Roak BJ., Deriziotis P., Lee C., et al. Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet. 2012;44:471. doi: 10.1038/ng.835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sanders SJ., Murtha MT., Gupta AR., et al. De novo mutations revealed by whole-exome sequencing are strongly associated with autism. Nature. In press. doi: 10.1038/nature10945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sanders SJ., Ercan-Sencicek AG., Hus V., et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron. 2011;70:863–685. doi: 10.1016/j.neuron.2011.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kumar RA., KaraMohamed S., Sudi J., et al. Recurrent 16p11.2 microdeletions in autism. Hum Mol Genet. 2008;17:628–638. doi: 10.1093/hmg/ddm376. [DOI] [PubMed] [Google Scholar]
  • 41.McCarthy SE., Makarov V., Kirov G., et al Microduplications of 16p11.2 are associated with schizophrenia. Nat Genet. 2009;41:1223–1227. doi: 10.1038/ng.474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Vassos E., Collier DA., Holden S., et al. Penetrance for copy number variants associated with schizophrenia. Hum Mol Genet. 2010;19:3477–3481. doi: 10.1093/hmg/ddq259. [DOI] [PubMed] [Google Scholar]
  • 43.Weiss LA., Shen Y., Korn JM., et al. Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med. 2008;358:667–675. doi: 10.1056/NEJMoa075974. [DOI] [PubMed] [Google Scholar]
  • 44.Gauthier J., Champagne N., Lafreniere RG., et al. De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia. Proc Natl Acad Sci U S A. 2010;107:7863–7868. doi: 10.1073/pnas.0906232107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vrijenhoek T., Buizer-Voskamp JE., van der Stelt I., et al. Recurrent CNVs disrupt three candidate genes in schizophrenia patients. Am J Hum Genet. 2008;83:504–510. doi: 10.1016/j.ajhg.2008.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Need AC., Ge D., Weale ME., et al. A genome-wide investigation of SNPs and CNVs in schizophrenia. PLoS Genet. 2009;5:e1000373. doi: 10.1371/journal.pgen.1000373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Elia J., Gai X., Xie HM., et al. Rare structural variants found in attentiondeficit hyperactivity disorder are preferentially associated with neurodevelopmental genes. Mol Psychiatry. 2010;15:637–646. doi: 10.1038/mp.2009.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Girirajan S., Rosenfeld JA., Cooper GM., et al. A recurrent 16p12.1 microdeletion supports a two-hit model for severe developmental delay. Nat Genet. 2010;42:203–209. doi: 10.1038/ng.534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Leblond CS., Heinrich J., Delorme R., et al. Genetic and functional analyses of SHANK2 mutations suggest a multiple hit model of autism spectrum disorders. PLoS Genet. 2012;8:e1002521. doi: 10.1371/journal.pgen.1002521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vorstman JA., van Daalen E., Jalali GR., et al. A double hit implicates DIAPH3 as an autism risk gene. Mol Psychiatry. 2011;16:442–451. doi: 10.1038/mp.2010.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Constantino JN., Lajonchere C., Lutz M., et al. Autistic social impairment in the siblings of children with pervasive developmental disorders. Am J Psychiatry. 2006;163:294–296. doi: 10.1176/appi.ajp.163.2.294. [DOI] [PubMed] [Google Scholar]
  • 52.Fombonne E. Epidemiology of autistic disorder and other pervasive developmental disorders. J Clin Psychiatry. 2005;66 (suppl 10):3–8. [PubMed] [Google Scholar]
  • 53.Baron-Cohen S., Lombardo MV., Auyeung B., Ashwin E., Chakrabarti B., Knickmeyer R. Why are autism spectrum conditions more prevalent in males? PLoS Biol. 2011;9:e1001081. doi: 10.1371/journal.pbio.1001081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Baird G., Simonoff E., Pickles A., et al. Prevalence of disorders of the autism spectrum in a population cohort of children in South Thames: the Special Needs and Autism Project (SNAP). Lancet. 2006;368:210–215. doi: 10.1016/S0140-6736(06)69041-7. [DOI] [PubMed] [Google Scholar]
  • 55.Chakrabarti S., Fombonne E. Pervasive developmental disorders in preschool children: confirmation of high prevalence. Am J Psychiatry. 2005;162:1133–1141. doi: 10.1176/appi.ajp.162.6.1133. [DOI] [PubMed] [Google Scholar]
  • 56.Betancur C. Etiological heterogeneity in autism spectrum disorders: more than 100 genetic and genomic disorders and still counting. Brain Res. 2011;1380:42–77. doi: 10.1016/j.brainres.2010.11.078. [DOI] [PubMed] [Google Scholar]
  • 57.Skuse DH. Rethinking the nature of genetic vulnerability to autistic spectrum disorders. Trends Genet. 2007;23:387–395. doi: 10.1016/j.tig.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • 58.Lundstrom S., Chang Z., Rastam M., et al. Autism spectrum disorders and autistic like traits: similar etiology in the extreme end and the normal variation. Arch Gen Psychiatry. 2012;69:46–52. doi: 10.1001/archgenpsychiatry.2011.144. [DOI] [PubMed] [Google Scholar]
  • 59.Kim YS., Leventhal BL., Koh YJ., et al. Prevalence of autism spectrum disorders in a total population sample. Am J Psychiatry. 2011;168:904–912. doi: 10.1176/appi.ajp.2011.10101532. [DOI] [PubMed] [Google Scholar]
  • 60.Investigators. AaDDMNSYP. Prevalence of autism spectrum disorders autism and developmental disabilities monitoring network, 14 sites. United States, 2008. MMWR Surveill Summ. 2012;61:1–19. [PubMed] [Google Scholar]
  • 61.Brugha TS., McManus S., Bankart J., et al. Epidemiology of autism spectrum disorders in adults in the community in England. Arch Gen Psychiatry. 2011;68:459–465. doi: 10.1001/archgenpsychiatry.2011.38. [DOI] [PubMed] [Google Scholar]
  • 62.King M., Bearman P. Diagnostic change and the increased prevalence of autism. Int J Epidemiol. 2009;38:1224–1234. doi: 10.1093/ije/dyp261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fombonne E. Epidemiology of pervasive developmental disorders. Pediatr Res. 2009;65:591–598. doi: 10.1203/PDR.0b013e31819e7203. [DOI] [PubMed] [Google Scholar]
  • 64.Gupta S., Aggarwal S., Rashanravan B., Lee T. Th1 - and Th2-like cytokines in CD4+ and CD8+ T cells in autism. J Neuroimmunol. 1998;85:106–109. doi: 10.1016/s0165-5728(98)00021-6. [DOI] [PubMed] [Google Scholar]
  • 65.Singh VK., Warren R., Averett R., Ghaziuddin M. Circulating autoantibodies to neuronal and glial filament proteins in autism. Pediatr Neurol. 1997;17:88–90. doi: 10.1016/s0887-8994(97)00045-3. [DOI] [PubMed] [Google Scholar]
  • 66.Ashwood P., Corbett BA., Kantor A., Schulman H., Van de Water J., Amaral DG. In search of cellular immunophenotypes in the blood of children with autism. PLoS One. 2011;6:e19299. doi: 10.1371/journal.pone.0019299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Vargas DL., Nascimbene C., Krishnan C., Zimmerman AW., Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:67–81. doi: 10.1002/ana.20315. [DOI] [PubMed] [Google Scholar]
  • 68.Morgan JT., Chana G., Pardo CA., et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010;68:368–376. doi: 10.1016/j.biopsych.2010.05.024. [DOI] [PubMed] [Google Scholar]
  • 69.Voineagu I., Wang X., Johnston P., et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 2011;474:380–384. doi: 10.1038/nature10110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chahrour M., Jung SY., Shaw C., et al. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science. 2008;320:1224–1229. doi: 10.1126/science.1153252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Vorstman JA., Staal WG., van Daalen E., van Engeland H., Hochstenbach PF., Franke L. Identification of novel autism candidate regions through analysis of reported cytogenetic abnormalities associated with autism. Mol Psychiatry. 2006;11:18–28. doi: 10.1038/sj.mp.4001781. [DOI] [PubMed] [Google Scholar]
  • 72.Bremer A., Giacobini M., Nordenskjold M., et al. Screening for copy number alterations in loci associated with autism spectrum disorders by two-color multiplex ligation-dependent probe amplification. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:280–285. doi: 10.1002/ajmg.b.30954. [DOI] [PubMed] [Google Scholar]
  • 73.Skuse DH., James RS., Bishop DV., Coppin B., Dalton P., Aamodt-Leeper G., Bacarese-Hamilton M., Creswell C., McGurk R., Jacobs PA. Evidence from Turner's syndrome of an imprinted X-linked locus affecting cognitive function. Nature. 1997;387:705–708. doi: 10.1038/42706. [DOI] [PubMed] [Google Scholar]
  • 74.Mohammad NS., Jain JM., Chintakindi KP., Singh RP., Naik U., Akella RR. Aberrations in folate metabolic pathway and altered susceptibility to autism. Psychiatr Genet. 2009;19:171–176. doi: 10.1097/YPG.0b013e32832cebd2. [DOI] [PubMed] [Google Scholar]
  • 75.Liu X., Solehdin F., Cohen IL., et al. Population - and family-based studies associate the MTHFR gene with idiopathic autism in simplex families. J Autism Dev Disord. 2011;41:938–944. doi: 10.1007/s10803-010-1120-x. [DOI] [PubMed] [Google Scholar]
  • 76.Nguyen A., Rauch TA., Pfeifer GP., Hu VW. Global methylation profiling of lymphoblastoid cell lines reveals epigenetic contributions to autism spectrum disorders and a novel autism candidate gene, RORA, whose protein product is reduced in autistic brain. FASEB J. 2010;24:3036–3051. doi: 10.1096/fj.10-154484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fernandez-Gonzalez R., Moreira P., Bilbao A., et al. Long-term effect of in vitro culture of mouse embryos with serum on mRNA expression of imprinting genes, development, and behavior. Proc Natl Acad Sci U SA. 2004;101:5880–5885. doi: 10.1073/pnas.0308560101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hvidtjorn D., Schieve L., Schendel D., Jacobsson B., Svaerke C., Thorsen P. Cerebral palsy, autism spectrum disorders, and developmental delay in children born after assisted conception: a systematic review and meta-analysis. Arch Pediatr Adolesc Med. 2009;163:72–83. doi: 10.1001/archpediatrics.2008.507. [DOI] [PubMed] [Google Scholar]
  • 79.Wakefield AJ., Murch SH., Anthony A., et al. Ileal-lymphoid-nodular hyperplasia, non-specific colitis, and pervasive developmental disorder in children. Lancet. 1998;351:637–641. doi: 10.1016/s0140-6736(97)11096-0. [DOI] [PubMed] [Google Scholar]
  • 80.Parker SK., Schwartz B., Todd J., Pickering LK. Thimerosal-containing vaccines and autistic spectrum disorder: a critical review of published original data. Pediatrics. 2004;114:793–804. doi: 10.1542/peds.2004-0434. [DOI] [PubMed] [Google Scholar]
  • 81.Gardener H., Spiegelman D., Buka SL. Prenatal risk factors for autism: comprehensive meta-analysis. Br J Psychiatry. 2009;195:7–14. doi: 10.1192/bjp.bp.108.051672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Smith SE., Li J., Garbett K., Mirnics K., Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007;27:10695–10702. doi: 10.1523/JNEUROSCI.2178-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gardener H., Spiegelman D., Buka SL. Perinatal and neonatal risk factors for autism: a comprehensive meta-analysis. Pediatrics. 2011;128:344–355. doi: 10.1542/peds.2010-1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Atladottir HO., Thorsen P., Ostergaard L., et al. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J Autism Dev Disord. 2010;40:1423–1430. doi: 10.1007/s10803-010-1006-y. [DOI] [PubMed] [Google Scholar]
  • 85.Pinto-Martin JA., Levy SE., Feldman JF., Lorenz JM., Paneth N., Whitaker AH. Prevalence of autism spectrum disorder in adolescents born weighing <2000 grams. Pediatrics. 2011;128:883–891. doi: 10.1542/peds.2010-2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zwaigenbaum L., Szatmari P., Jones MB., et al. Pregnancy and birth complications in autism and liability to the broader autism phenotype. J Am Acad Child Adolesc Psychiatry. 2002;41:572–579. doi: 10.1097/00004583-200205000-00015. [DOI] [PubMed] [Google Scholar]
  • 87.Larsson HJ., Eaton WW., Madsen KM., et al. Risk factors for autism: perinatal factors, parental psychiatric history, and socioeconomic status. Am J Epidemiol. 2005;161:916–25; discussion 926-928. doi: 10.1093/aje/kwi123. [DOI] [PubMed] [Google Scholar]
  • 88.Grandgeorge M., Hausberger M., Tordjman S., Deleau M., Lazartigues A., Lemonnier E. Environmental factors influence language development in children with autism spectrum disorders. PLoS One. 2009;4:e4683. doi: 10.1371/journal.pone.0004683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Magnusson C., Rai D., Goodman A., et al. Migration and autism-spectrum disorder: population-based study. Br J Psychiatry. 2012;201:109–115. doi: 10.1192/bjp.bp.111.095125. [DOI] [PubMed] [Google Scholar]
  • 90.Rasalam AD., Hailey H., Williams JH., et al. Characteristics of fetal anticonvulsant syndrome associated autistic disorder. Dev Med Child Neurol. 2005;47:551–555. doi: 10.1017/s0012162205001076. [DOI] [PubMed] [Google Scholar]
  • 91.Kolozsi E., Mackenzie RN., Roullet FI., deCatanzaro D., Foster JA. Prenatal exposure to valproic acid leads to reduced expression of synaptic adhesion molecule neuroligin 3 in mice. Neuroscience. 2009;163:1201–1210. doi: 10.1016/j.neuroscience.2009.07.021. [DOI] [PubMed] [Google Scholar]
  • 92.Andrade SE., Raebel MA., Brown J., et al. Use of antidepressant medications during pregnancy: a multisite study. Am J Obstet Gynecol. 2008;198:194e1–e5. doi: 10.1016/j.ajog.2007.07.036. [DOI] [PubMed] [Google Scholar]
  • 93.Croen LA., Grether JK., Yoshida CK., Odouli R., Hendrick V. Antidepressant use during pregnancy and childhood autism spectrum disorders. Arch Gen Psychiatry. 2011;68:1104–1112. doi: 10.1001/archgenpsychiatry.2011.73. [DOI] [PubMed] [Google Scholar]
  • 94.Landrigan PJ. What causes autism? Exploring the environmental contribution. Curr Opin Pediatr. 2010;22:219–225. doi: 10.1097/MOP.0b013e328336eb9a. [DOI] [PubMed] [Google Scholar]
  • 95.D'Amelio M., Ricci I., Sacco R., et al. Paraoxonase gene variants are associated with autism in North America, but not in Italy: possible regional specificity in gene-environment interactions. Mol Psychiatry. 2005;10:1006–1016. doi: 10.1038/sj.mp.4001714. [DOI] [PubMed] [Google Scholar]
  • 96.Glasson EJ., Bower C., Petterson B., de Klerk N., Chaney G., Hallmayer JF. Perinatal factors and the development of autism: a population study. Arch Gen Psychiatry. 2004;61:618–627. doi: 10.1001/archpsyc.61.6.618. [DOI] [PubMed] [Google Scholar]
  • 97.Hunter JW., Mullen GP., McManus JR., Heatherly JM., Duke A., Rand JB. Neuroligin-deficient mutants of C. elegans have sensory processing deficits and are hypersensitive to oxidative stress and mercury toxicity. Dis Model Mech. 2010;3:366–376. doi: 10.1242/dmm.003442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Fischer M., Reuter J., Gerich FJ., et al. Enhanced hypoxia susceptibility in hippocampal slices from a mouse model of rett syndrome. J Neurophysiol. 2009;101:1016–1032. doi: 10.1152/jn.91124.2008. [DOI] [PubMed] [Google Scholar]
  • 99.Curristin SM., Cao A., Stewart WB., et al. Disrupted synaptic development in the hypoxic newborn brain. Proc Natl Acad Sci U S A. 2002;99:1 5729–1 5734. doi: 10.1073/pnas.232568799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Melke J., Goubran Botros H., Chaste P., et al. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13:90–98. doi: 10.1038/sj.mp.4002016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Tordjman S., Anderson GM., Pichard N., Charbuy H., Touitou Y. Nocturnal excretion of 6-sulphatoxymelatonin in children and adolescents with autistic disorder. Biol Psychiatry. 2005;57:134–138. doi: 10.1016/j.biopsych.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 102.Simonneaux V., Ribelayga C. Generation of the melatonin endocrine message in mammals: a review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev. 2003;55:325–395. doi: 10.1124/pr.55.2.2. [DOI] [PubMed] [Google Scholar]
  • 103.Jockers R., Maurice P., Boutin JA., Delagrange P. Melatonin receptors, heterodimerization, signal transduction and binding sites: what's new? Br J Pharmacol. 2008;154:1182–1195. doi: 10.1038/bjp.2008.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Gitto E., Pellegrino S., Gitto P., Barberi I., Reiter RJ. Oxidative stress of the newborn in the pre - and postnatal period and the clinical utility of melatonin. J Pineal Res. 2009;46:128–139. doi: 10.1111/j.1600-079X.2008.00649.x. [DOI] [PubMed] [Google Scholar]
  • 105.Olivier P., Fontaine RH., Loron G., et al. Melatonin promotes oligodendroglial maturation of injured white matter in neonatal rats. PLoS One. 2009;4:e7128. doi: 10.1371/journal.pone.0007128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ehninger D., Sano Y., de Vries PJ., et al. Gestational immune activation and Tsc2 haploinsufficiency cooperate to disrupt fetal survival and may perturb social behavior in adult mice. Mol Psychiatry. 2012;17:62–70. doi: 10.1038/mp.2010.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Abazyan B., Nomura J., Kannan G., et al. Prenatal interaction of mutant DISC1 and immune activation produces adult psychopathology. Biol Psychiatry. 2010;68:1172–1181. doi: 10.1016/j.biopsych.2010.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Duncan LE., Keller MC. A critical review of the first 10 years of candidate gene-by-environment interaction research in psychiatry. Am J Psychiatry. 2011;168:1041–1049. doi: 10.1176/appi.ajp.2011.11020191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Landrigan PJ., Trasande L., Thorpe LE., et al. The National Children's Study: a 21-year prospective study of 100,000 American children. Pediatrics. 2006;118:2173–2186. doi: 10.1542/peds.2006-0360. [DOI] [PubMed] [Google Scholar]
  • 110.Stoltenberg C., Schjolberg S., Bresnahan M., et al. The Autism Birth Cohort: a paradigm for gene-environment-timing research. Mol Psychiatry. 2010;15:676–680. doi: 10.1038/mp.2009.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nijmeijer JS., Hartman CA., Rommelse NN., et al. Perinatal risk factors interacting with catechol O-methyltransferase and the serotonin transporter gene predict ASD symptoms in children with ADHD. J Child Psychol Psychiatry. 2010;51:1242–1250. doi: 10.1111/j.1469-7610.2010.02277.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Girirajan S., Brkanac Z., Coe BP., et al. Relative burden of large CNVs on a range of neurodevelopmental phenotypes. PLoS Genet. 2011;7:e1002334. doi: 10.1371/journal.pgen.1002334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Levy D., Ronemus M., Yamrom B., et al. Rare de novo and transmitted copy-number variation in autistic spectrum disorders. Neuron. 70:886–897. doi: 10.1016/j.neuron.2011.05.015. [DOI] [PubMed] [Google Scholar]
  • 114.Glessner JT., Wang K., Cai G., et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature. 2009;459:569–573. doi: 10.1038/nature07953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bucan M., Abrahams BS., Wang K., et al. Genome-wide analyses of exonic copy number variants in a family-based study point to novel autism susceptibility genes. PLoS Genet. 2009;5:e1000536. doi: 10.1371/journal.pgen.1000536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Morrow EM., Yoo SY., Flavell SW., et al. Identifying autism loci and genes by tracing recent shared ancestry. Science. 2008;321:218–223. doi: 10.1126/science.1157657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Christian SL., Brune CW., Sudi J., et al. Novel submicroscopic chromosomal abnormalities detected in autism spectrum disorder. Biol Psychiatry. 2008;63:1111–1117. doi: 10.1016/j.biopsych.2008.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Jacquemont ML., Sanlaville D., Redon R., et al. Array-based comparative genomic hybridisation identifies high frequency of cryptic chromosomal rearrangements in patients with syndrom ic autism spectrum disorders. J Med Genet. 2006;43:843–849. doi: 10.1136/jmg.2006.043166. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Dialogues in Clinical Neuroscience are provided here courtesy of Taylor & Francis

RESOURCES