Next Article in Journal
Risk of Adverse Drug Events Following the Virtual Addition of COVID-19 Repurposed Drugs to Drug Regimens of Frail Older Adults with Polypharmacy
Previous Article in Journal
Key Aspects in Nutritional Management of COVID-19 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity

1
NHS Foundation Trust, Oxford University Hospitals, Oxford OX3 9DU, UK
2
Nuffield Department of Clinical Neurosciences and MRC, Weatherall Institute of Molecular Medicine, Oxford OX3 9DU, UK
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(8), 2590; https://doi.org/10.3390/jcm9082590
Submission received: 20 July 2020 / Revised: 5 August 2020 / Accepted: 6 August 2020 / Published: 10 August 2020
(This article belongs to the Section Clinical Neurology)

Abstract

:
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders with unknown heterogeneous aetiologies. Epidemiological studies have found an association between maternal infection and development of ASD in the offspring, and clinical findings reveal a state of immune dysregulation in the pre- and postnatal period of affected subjects. Maternal immune activation (MIA) has been proposed to mediate this association by altering fetal neurodevelopment and leading to autism. Although animal models have supported a causal link between MIA and development of ASD, their validity needs to be explored. Moreover, considering that only a small proportion of affected offspring develop autism, and that MIA has been implicated in related diseases such as schizophrenia, a key unsolved question is how disease specificity and phenotypic outcome are determined. Here, we have integrated preclinical and clinical evidence, including the use of animal models for establishing causality, to explore the role of maternal infections in ASD. A proposed priming/multi-hit model may offer insights into the clinical heterogeneity of ASD, its convergence with related disorders, and therapeutic strategies.

1. Introduction

Autism spectrum disorders (ASD) are neurodevelopmental disorders characterised by impairments in social interaction and communication along with restricted/repetitive behaviours [1]. Despite their epidemiological significance, with estimates revealing a rising prevalence of 1 in 68 children, the aetiology of ASD remains unknown. Twin studies have shown a variable but incomplete concordance between monozygotic twins, indicating both genetic and environmental contributions [2,3,4,5].
Several lines of evidence implicate the prenatal innate and adaptive immune environment in ASD. Meta-analyses and epidemiological studies have reported an association between maternal infection and incidence of ASD in the offspring [6,7,8], and certain in utero inflammatory markers have been associated with ASD [6]. Animal studies of maternal immune activation (MIA) have characterised a prominent pro-inflammatory phenotype with is associated with impairments in communication and stereotypic behaviours [9,10,11,12,13]. Furthermore, transcriptomic and cytokine profiling studies in individuals with ASD have also revealed a state of immune dysregulation and elevated pro-inflammatory cytokines [14,15,16,17].
However, clinical findings only show correlation and cannot establish a causal link between MIA or maternal antibodies and ASD. Supporting this link are animal models of MIA that serve to elucidate the mechanisms by which fetal development can be altered.

2. Search Strategy

The PubMed, Embase, and Web of Science databases were searched on 2 May 2020 using the search strategy: (((prenatal OR mother OR maternal) AND immun*) OR (influenza OR flu) OR (polyinosinic:polycytidylic acid OR polyI:C OR poly I:C OR poly(I:C)) OR (lipopolysaccharide OR LPS) OR (interleukin)) AND (autis* OR ASD OR neurodevelopment*). Reference lists of relevant articles were reviewed to locate additional studies that may have been missed during the search. Search results were restricted to English-language studies only.

3. Animal Models of Maternal Immune Activation (MIA)

The main paradigms used to model MIA consist of applying live influenza to pregnant rodents or mimicking infection with immunogenic molecules. Influenza models were pioneered by Fatemi et al. [18] to extend epidemiological findings associating maternal infection with autism and schizophrenia. Maternal influenza administration results in deficits in cortical and hippocampal cytoarchitecture and in social interaction related to ASD [18,19,20] (Table 1). Despite displaying the strongest construct validity, this model does not allow dissociating the effects of the pathogen from those of MIA.
Models based on the bacterial mimic lipopolysaccharide (LPS) or the viral mimic polyinosinic–polycytidylic acid (Poly(I:C)) overcome this limitation and afford precise control of the duration of the immune response. Both induction protocols elicit overlapping findings with maternal influenza injection (Table 1), suggesting that the maternal reaction is more critical than the pathogen per se. Malkova et al. found that poly(I:C) injection during pregnancy resulted in offspring exhibiting the core symptoms of ASD: decreases in sociability and communication and increases in repetitive/stereotypic behaviours [21]. These findings were reproduced by subsequent studies and extended to include other ASD-related symptoms, establishing construct and face validity [10,11,22]. The range of animal models of MIA continues to expand, with more recent models using exposures to Staphylococcal enterotoxin [23], toll-like receptor 7 agonists [24], and allergens [25].
Although these studies support a causal link between MIA and autism, LPS and poly(I:C) injection differ from a live influenza infection, which is acquired through a respiratory route and induces both innate and adaptive arms of immunity. Moreover, maternal infection might have confounding effects on postpartum behaviour, which can influence offspring phenotype [26]. Litter effects, arising from shared genetic factors and maternal environment, may also result in inflated effect sizes. These methodological issues are often neglected and could be overcome by postnatal cross-fostering designs and inclusion of offspring from each litter in investigations, respectively.
It is also unclear to what extent these behavioural findings are representative of ASD symptoms. Although ultrasonic vocalisations (USVs) have been used as a measure of communication deficits [27,28], their intentional communicative nature is unknown, considering the importance of olfactory communication in rodents. Similarly, decreased sniffing behaviour is typically explained in terms of sociability but could be equally ascribed to elevated anxiety or olfactory dysfunction, which should be independently investigated.
Another limitation is that MIA models are restricted by specificity and are unlikely to apply solely to ASD. Most studies have evaluated behaviours that are more relevant to schizophrenia than to autism, such as prepulse inhibition (PPI) and latent inhibition (LI). Behavioural testing is normally conducted during adulthood, which relates more closely to the onset of schizophrenia. Further specificity might be achieved by characterising disease-differentiating features (e.g., antipsychotic response in schizophrenia, stereotyped motor behaviours and macrocephaly in autism); such dissociation might be unfeasible since both diseases share overlapping genetic, neuropathological, and symptomatic traits and could lie across a neurodevelopmental spectrum [29,30].
Future studies could incorporate assays investigating the “theory of mind” deficit in ASD. Mice exhibit greater pain sensitivity after observing cagemates undergo painful stimuli, indicating that some elements of empathy may be testable in mice. Changes in the reward value of social interactions could also be assessed with operant chambers requiring lever presses to access partners. Histological and electrophysiological recordings in the first days of postnatal life are scarce and may inform how neurobiology relates to disease progression. Investigations could also expand to peripheral systems disrupted in ASD patients, such as emerging alterations in gut microbiota [31,32].
Another promising avenue are primate models, due to their complex social structure and their neuroanatomical homology. Patterson and colleagues found that maternal poly(I:C) injection in rhesus macaques led to abnormal social approach, affiliative vocalisations, and repetitive behaviours, validating the MIA primate model [33] (Table 2). Future investigations could analyse ASD deficits that are difficult to explore in rodents, such as joint attention and multisensory temporal integration. Similarly, neuroimaging may be employed to probe the validity of MIA at the level of neural circuitry.

4. MIA and Disease Specificity

Epidemiological studies have shown that MIA is a risk factor shared with related disorders including schizophrenia and epilepsy [34]. Although the incidence of infection during pregnancy is estimated at 64%, only around 1% of children develop ASD [8]. This raises a key unsolved question in the MIA field regarding phenotypic outcome: in the presence of prenatal immune challenge, what determines whether the offspring develop ASD, a related disease, or none?
Two hypotheses have been put forward to explain the relationship between MIA and ASD [34,35,36]: (1) MIA constitutes a disease-specific risk factor that depends on inherent characteristics of the immune challenge, or (2) MIA is a general vulnerability factor that predisposes to specific diseases depending on individual genetic, epigenetic, and environmental influences. Below, these two possibilities are discussed.

5. Disease Specificity Arising from Immune Challenge

The idea that the properties of the immune challenge (nature, intensity, and timing) can bias progression towards a specific disease is compelling, as different immunogens administered at critical developmental windows could lead to pathogenic cascades manifesting as distinct phenotypes.
Epidemiological evidence indicates that the nature of the immune challenge is less important than the maternal response, with ASD being associated with both bacterial and viral infections [7,8]. Interestingly, a Danish birth cohort study found an association between ASD and maternal viral infection in the first trimester and bacterial infection in the second [8], suggesting that the nature of the pathogen may be important. Major limitations were the exclusion of infectious episodes not requiring hospitalisation and the lack of control for multiple comparisons. In a larger study, Lee et al. observed that the association between maternal infection and ASD was independent of both infectious agent and trimester [7].
The relative importance of the maternal response over the pathogen is also highlighted by animal models, which exhibit similar deficits regardless of the immunogen (Table 1). Nevertheless, there might be subtle differences in the downstream effects of influenza, LPS, and poly(I:C), as they lead to opposite effects in dopaminergic systems [26] and white matter volume [20,37]. These could be related to their respective downstream signalling cascades: LPS acts via Toll-like receptor 4 (TLR4) pathways, including the myeloid differentiation primary response 88 (Myd88) and the TIR-domain-containing adapter-inducing interferon-β (TRIF) dependent pathways. This leads to strong induction of tumour necrosis factor alpha (TNFα). On the other hand, poly(I:C) acts via TLR3 TRIF-dependent pathways mainly inducing type I interferons [38]. Both TNFα and type I and II interferons are increased in the maternal plasma and the blood of individuals with ASD [39,40] and are involved in microglial dysfunction and neurogenesis modulation [14,41,42].
The intensity of the immunogen could also be relevant since different doses may trigger immune responses of varying severity, with disease only occurring when a specific MIA threshold is surpassed. Indeed, rodent models using higher doses of immunogen typically induce more striking behavioural deficits and cytokine alterations in the fetal brain (Table 1; Table 3), and several studies point to a dose-dependent effect of poly(I:C) on behavioural impairments. However, a direct relation between dose and disease development may be over-simplistic, as it ignores maternal variability to immune challenge arising from different gene-environment influences. Studies have revealed that different maternal responses can determine whether offspring develop abnormal behaviour [43].
Timing of prenatal exposure is crucial as immune activation at different critical windows could impact disease-specific neurodevelopmental processes. Although several epidemiological studies support an association between prenatal stressors during mid- and late gestational periods and ASD, others have found associations with early gestation or no association [2,3,7,8]. These discrepancies could be attributed to the lack of substratification in clinical designs, since patient subpopulations may be especially vulnerable to infection due to genetic or environmental factors, compared to others with different aetiologies. In the future, substratification according to infection vulnerability may be conducted to test this hypothesis.
The poly(I:C) model is useful to study timing since it induces a controlled immune reaction lasting 24–48 h [32,44]. Poly(I:C) administration at embryonic day (E) 9 or 12.5 (equivalent to middle-to-late first trimester in humans) typically results in schizophrenia-like behaviour in offspring mice26 (Table 1). By contrast, poly(I:C) at E16 (middle second trimester) increases seizure susceptibility while impairing sociability, reminiscent of autism symptoms [45]. Neuroanatomically, mice exposed to early MIA display ventriculomegaly characteristic of schizophrenia, and mice exposed late exhibit decreased cerebrospinal fluid (CSF) volume, perhaps relating to autism. Thus, MIA effects can be dissociated according to the early or late timing of the challenge [46], which may bias development along a spectrum towards schizophrenia or autism-like features, respectively. However, more precise control of the immune reaction is required. This could be achieved with models of reversible interleukin-6 (IL-6) overexpression with tetracycline-controlled transcriptional activation systems, where tetracyclines reversibly switch transcription on or off with spatiotemporal specificity.
It would be informative to interrogate to what extent the neural systems affected at specific stages account for symptom clusters. For example, the hippocampus and corpus callosum develop around E16 in mice. However, although the Black and Tan Brachyury (BTBR) mouse exhibiting agenesis of the corpus callosum is a widely used model of autism [21], the hippocampus is not typically involved in ASD. Collectively, many findings seem to indicate that the characteristics of the immune challenge can result in partly dissociable neuroanatomical and behavioural phenotypes, accounting to a certain extent for disease specificity. Future studies will need to discern whether different immunogens, doses, and administration windows truly result in distinct outcomes within a single study. It will be crucial to use a wider range of measures for ASD and schizophrenia, to avoid falling into a confirmation bias in which results are predetermined by assays capturing specific disease phenotypes. Despite the emergence of neuroinformatic approaches [47], extrapolation of developmental timing from rodents to humans remains spurious, and primate models could help bridge the gap due to their comparably similar development.
Table 1. Rodent models of maternal immune activation (MIA).
Table 1. Rodent models of maternal immune activation (MIA).
ReferenceTreatment and Species Timing of ChallengeBehavioural FindingsNeuropathological Findings
Fatemi et al. (1999) [18]Influenza (IN)
Mouse
E9ND↓ Reelin expression in cortical layer I and hippocampus
↓ Ventricular size ↑ Brain size
Defective corticogenesis, pyramidal cell atrophy
Shi et al. (2003) [19]Influenza (IN)
Mouse
E9.5↓ PPI ↓ Exploratory behaviour
↓ Social interaction
ND
Shi et al. (2009) [27]Influenza (IN)
Poly(I:C) (20 mg/kg, IP)
Mouse
E9.5 (influenza)
E12.5 (Poly(I:C))
ND↓ Purkinje cells in lobule VII of cerebellum
Delayed migration of granule cells
Kirsten et al. (2012) [48]LPS (100 μg/kg, IP)
Rat
E9.5↓ Play behaviour ↓ Social interaction ↓ USVs
↓ Exploratory behaviour ↑ Repetitive behaviour
No change in anxiety-like behaviour
ND
Xuan et al. (2014) [49]LPS (75 μg/kg, IP)
Poly(I:C) (20 mg/kg, IP)
Mouse
E11.5, 12 (LPS)
E12.5 (Poly(I:C))
↓ Locomotor activity * ↓ Social approach *
↑ Repetitive behaviour *
ND
Sharova et al. (2014) [50]LPS (45 μg/kg, IP)
Mouse
E11.5 ND↓ Neuron number in forebrain area
↑ Neuron number in nasal and olfactory bulb
Altered gonadotropin-releasing hormone neuron migration
Foley et al. (2015) [51]LPS (50 μg/kg, SC)
Rat
E15, 16↑ Acoustic startle *
No change in PPI *
ND
Wischhof et al. (2015) [52]LPS (100 μg/kg, IP)
Rat
E15, 16↓ PPI * ↓ Recognition memory↓ Myelination in cortex and limbic regions
↓ PARV-expressing interneurons in medial prefrontal cortex, entorhinal cortex, and hippocampus
Batinić et al. (2016) [53]LPS (100 μg/kg, IP)
Rat
E15, 16↓ Locomotion * ↓ Response to amphetamines *
↓ Spatial learning and memory *
ND
Fernández de Cossío et al. (2017) [28]LPS (100 μg/kg, IP)
Mouse
E15↓ USV duration ↓ Social behaviour
↑ Stereotypical behaviour
↑ Repetitive behaviour *
Increased spine density in granule cells *
No effect on pyramidal cells
Kirsten et al. (2017) [48]LPS (100 μg/kg, IP)
Rats
E9.5↑ Repetitive behaviour Reduced dopaminergic activity in the hypothalamus
Wu et al. (2018) [54]LPS (75 μg/kg, IP)
Mouse
E14.5↑ Anxiety-like behaviour ↓ Social interaction
↑ Depression-like behaviour
Aberrant cytoarchitecture and lamination in neocortex
↓ Intermediate progenitor cells and astrocytes in neocortex
Meyer et al. (2006, 2008) [26,55]
Li et al. (2009) [46]
Poly(I:C) (5 mg/kg, IV)
Mouse
E9, 17↓ Exploratory behaviour * ↓ Reversal learning
↓ PPI * ↑ Repetitive behaviour *
↑ Locomotor activity
↓ Hippocampal reelin expression ↓ Ventricular volume * ↑ Dopamine-relater marker expression
Altered glutamate-related marker expression
Abazyan et al. (2010) [56]
Lipina et al. (2013) [57]
Poly(I:C) (5 mg/kg, IP)
Mouse
E9↓ PPI ↓ LI ↓ Exploratory behaviour
Hyperactivity
ND
Hsiao et al. (2012) [44]Poly(I:C) (20 mg/kg, IP)
IL-6 (5 μg, IP)
Mouse
E12.5↓ PPI ↓ LI ↓ Exploratory behaviourND
Malkova et al. (2012) [21]Poly(I:C) (5 mg/kg, IP)
Mouse
E10.5, 12.5, 14.5↓ USVs ↓ Social interaction ↓ Female-induced scent marking ↑ Repetitive behaviourND
Ehninger et al. (2012) [58]Poly(I:C) (20 mg/kg, IP)
Mouse
E10.5, 12.5, 14.5No change in social approach, exploratory behaviour, or olfactory functionND
Hsiao et al. (2013) [32] Schwartzer et al. (2013) [25] Wu et al. (2015) [29]Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↓ PPI ↓ USVs ↓ LI ↓ Social approach
↑ Repetitive behaviour ↑ Anxiety-like behaviour
↑ Depressive-like behaviour
↓ Neuronal proliferation, maturation and survival in dentate gyrus
↓ VEGFA-VEGFR2 hippocampal expression
Impaired hippocampal long-term potentiation
Giovanoli et al. (2013) [59]Poly(I:C) (1 mg/kg, IP)
Mouse
E9↓ LI
No change in PPI or anxiety-like behaviour
↑ Reelin expression
No change in microglia activation or GABAergic interneurons in CA regions
Missault et al. (2014) [43]Poly(I:C) (4 mg/kg, IP, IV)
Rat
E15↓ PPI ↑ Depressive-like behaviour
No change in locomotor activity
No change in microglial number
Giovanoli et al. (2016) [60]Poly(I:C) (5 mg/kg, IV)
Mouse
E9NDPresynaptic hippocampal deficits
No change in microglia or astrocyte density
Pendyala et al. (2017) [12]Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↓ USVs ↑ Repetitive behaviour
No change in motor coordination
Proinflammatory state in cerebellum
Reduction in cerebellar synaptic organising proteins
Li et al. (2018) [61]Poly(I:C) (10 mg/kg, IV)
Rat
E9↑ Locomotion ↓ PPI ↓ LIIncreased prefrontal cortex and hippocampus activity
Increased activation of microglia
Murray et al. (2019) [9]Poly(I:C) (10 mg/kg, IP)
Rat
E15NDIncreased activation of microglia in hippocampus *
Lins et al. (2019) [62]Poly(I:C) (20 mg/kg, IP)
Rat
E15↓ Social interaction *
No change in startle or PPI
ND
Amodeo et al. (2019) [10]Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↓ Social behaviour ↓Reversal learningDysregulation of potassium ion channel activity in frontal cortex
Carlezon et al. (2019) [22]Poly(I:C) (20 mg/kg, IP)
LPS (10 mg/kg, SC, postnatal day 9)
Mouse
E12.5↓ USVs ↓ Social behaviour *
↑ Anxiety-like behaviour *
Pro-inflammatory state in prefrontal cortex, amygdala, hippocampus and thalamus *
Haida et al. (2019) [11]Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↓ Social behaviour * ↓Motor coordination *Reduced number of Purkinje cells in cerebellum *
Reduced number of neurons in motor cortex*
Samuelsson et al. (2006) [63]IL-6 (9 μg/kg, IP)
Rat
E10, 12, 16 or E16, 18, 20↓ Spatial learning↓ Neuron number in CA regions ↑ Astrogliosis
↑ Apoptosis ↑ GFAP expression
Smith et al. (2007) [64]IL-6 (5 μg, IP)
Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↓ PPI ↓ LI ↓ Exploratory behaviour
↓ Social interaction
ND
IN, intranasal; E, embryonic day; ND, not determined; PPI, prepulse inhibition; LPS, lipopolysaccharide; IP, intraperitoneal; IV, intravenous; USV, ultrasonic vocalisations; LI, latent inhibition; poly(I:C), polyinosinic:polycytidylic acid; IL-6, interleukin-6; CA, cornu Ammonis. * Effects were sex-, age-, or genotype-dependent.
Table 2. Primate models of MIA.
Table 2. Primate models of MIA.
ReferenceTreatment and Species Timing of ChallengeBehavioural Findings Neuropathological Findings
Short et al. (2010) [20]Influenza (IN)
Rhesus macaque
Early 3rd trimesterAltered mother-infant interaction↓ Total and cortical grey matter
Willette et al. (2011) [37]LPS (2 or 4 ng/kg, IV)
Rhesus macaque
Early 3rd trimester↓ PPI
↑ Emotionality
No change in social interaction
↓ Medial temporal lobe grey matter
↑ Total white matter volume
Bauman et al. (2014) [33]Modified poly(I:C) (0.25 mg/kg, IV)
Rhesus macaque
Late 1st and 2nd trimester↓ Affiliative vocalisations
↓ Social approach
↑ Motor stereotypy
Abnormal social behaviour
Abnormal attachment to mother
ND
Machado et al. (2015) [65]Modified poly(I:C) (0.25 mg/kg, IV)
Rhesus macaque
Late 1st trimester↓ Social attention
↓ Visual fixation
ND
Weir et al. (2015) [66]Modified poly(I:C) (0.25, 0.5, or 1 mg/kg, IV)
Rhesus macaque
Late 1st trimesterND↓ Apical dendrite size in prefrontal cortex
↑ Number of oblique dendrites
Rose et al. (2017) [67]Modified poly(I:C) (0.25 mg/kg, IV)Late 1st or 2nd trimester↑ Stereotyped behavioursND
Bauman et al. (2019) [68]Modified poly(I:C) (0.25 mg/kg, IV)Late 1st or 2nd trimesterND↑ Striatal dopamine in late adolescence
IN, intranasal; LPS, lipopolysaccharide; IV, intravenous; PPI, prepulse inhibition; ND, not determined; poly(I:C), polyinosinic:polycytidylic acid.
Table 3. MIA-induced cytokine increases in the fetal brain of animal models.
Table 3. MIA-induced cytokine increases in the fetal brain of animal models.
ReferenceTreatment and Species Timing of ChallengeImmune Findings
Meyer et al. (2006, 2008) [26,55]Poly(I:C) (5 mg/kg, IV)
Mouse
E9, 17↑ TNFα, IL-1α, IL-6, IL-10
Abazyan et al. (2010) [56]Poly(I:C) (5 mg/kg, IP)
Mouse
E9↑ IL-1α, IL-4, IL-5
Arrode-Brusés et al. (2012) [69]Poly(I:C) (20 mg/kg, IP)
Mouse
E16↑ TNFα, IL-1α, IL-7, IL-13, MCP-1, MIP-1α
Lipina et al. (2013) [57]Poly(I:C) (2.5 or 5 mg/kg, IV)
Mouse
E9↑ IL-6
Wu et al. (2015) [29]Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↑ IL-6
Giovanoli et al. (2016) [60]Poly(I:C) (5 mg/kg, IV)
Mouse
E9↑ IL-1
Pendyala et al. (2017) [12] Poly(I:C) (20 mg/kg, IP)
Mouse
E12.5↑ IL-2, IL-3, IL-6, TNFRI, TNF-α, FasL
Rose et al. (2017) [67]Modified poly(I:C) (0.25 mg/kg, IV)
Rhesus macaque
1st or 2nd trimester↑ IL-1α, IL-4, IL-13
Poly(I:C), polyinosinic:polycytidylic acid; IV, intravenous; E, embryonic day; IL, interleukin; TNFα, tumour necrosis factor alpha; MCP-1, monocyte chemoattract protein-1; MIP-1α, macrophage inflammatory protein-1-alpha; TNFRI, tumour necrosis factor 1; FasL, Fas-ligand.

6. Disease Specificity Arising from Genetic and Environmental Factors

Another possibility is that MIA only reveals its neuropathological impact in subjects with genetic predisposition. Several cohorts have highlighted that ASD may have a significant genetic component, with heritability estimates ranging from 50% using a Cox model [70] to 83–97% using liability-threshold models [2,3]. A meta-analysis of twin studies calculated the heritability estimate at 64–91% [4].
Transcriptomic analyses have found that ASD is correlated with gene expression modules corresponding to immune genes and M2-activated microglia [17], suggesting that genetic factors may modulate the fetal response to MIA. Mazina et al. found that ASD individuals with copy number variations and history of maternal infection showed more severe impairments in communication and repetitive behaviours, implying an interaction between genetic susceptibility and prenatal immune challenge [71]. However, this does not necessarily indicate interaction as their effects could be independent and additive rather than synergistic. This could be investigated by exploring whether maternal infection results in ASD solely in subjects with a family history of neurodevelopmental disorders. A meta-analysis of common genetic variants in ASD failed to accurately identify variants that contribute to the aetiology of ASD, suggesting that either common variants do not play a major role in the genetic contribution or that larger population studies may be needed to achieve sufficient statistical power [51].
Multiple multi-trait models have investigated gene-MIA interactions. Ehninger et al. found that MIA resulted in disrupted social behaviour in mice offspring only in the presence of ASD-associated tuberous sclerosis complex 2 (TSC2) mutations [58]. Similarly, expression of mutant human disrupted-in-schizophrenia 1 (mhDisc1) or disrupted in schizophrenia 1 (Disc1) L100P mutations, linked to autism and schizophrenia, revealed synergistic effects with low-dose MIA on social and cognitive deficits, reduced amygdala volume, and elevated IL-6 levels in mice [32]. These findings support the notion that gene-MIA interactions could account for incomplete penetrance in ASD.
However, these genetic models are confounded by the effects of sex and background strain, which can affect the expression of autism-like phenotypes [25,72]. Although most studies only test male offspring, comparison of sex-specific effects may provide insights into the 4:1 male-to-female ratio in ASD. Broader issues concern the validity of using single gene manipulations, since the genetic architecture of ASD in humans is complex and heterogeneous [73], and most genetic risk in ASD arises from common variation, with rare variants contributing only around 3% [74].
Technologies such as the CRISPR/Cas (clustered regularly interspaced short palindromic repeats) system allow targeting multiple genes and could be used to investigate the additive effects of gene variants and their interaction with MIA. However, using transgenic models of common variants of small effect could compromise construct validity. Two categories of genes that could be further examined in combination with MIA are susceptibility factors that do not recapitulate all core symptoms of autism when manipulated alone ((FMRP translational regulator 1 (FMR1), methyl CpG binding protein 2 (MECP2)) and immune-related genes associated with ASD (human leukocyte antigen (HLA), phosphatase and tensin homolog (PTEN)).
On the other hand, the possibility that MIA may require exposure to an additional environmental insult to trigger full-blown pathology remains underexplored. Animal models combining MIA with postnatal stress have found synergistic effects on PPI deficits and microglia activation, supporting this “two-hit” model [59]. Nonetheless, considering the early onset of ASD, secondary environmental insults during pre- or perinatal life may be more relevant, and the examination of MIA convergence with such insults may help develop preventive strategies.
In terms of genetic versus environmental contribution, one can propose a framework in which either extreme genetic predisposition or environmental insults can cause ASD, with a proportion of ASD cases resulting from gene-environment interactions in the middle of this spectrum (Figure 1).
The hypotheses of disease specificity arising from MIA properties or gene-environment interactions are not mutually exclusive. It is possible to integrate these factors into a single priming/multi-hit model, in which MIA serves as a developmental primer that increases vulnerability towards disease-specific pathways depending on its nature, intensity, and timing. Another layer of phenotypic specificity is provided in the form of genetic, epigenetic, and environmental factors that are required for the progression from vulnerability to full-blown disease (Figure 2). Two meta-analysis of genome-wide association studies have identified novel loci that are shared between ASD and schizophrenia, raising the possibility of a shared pathway for disruption of neurodevelopment in these conditions [75,76].

7. Mechanistic Insights

How does MIA result in autism-like behaviours in the offspring? One hypothesis is that an MIA-induced elevation in proinflammatory cytokines alters fetal neurodevelopment [77]. IL-6 has been identified as a key mediator, since maternal injection of poly(I:C) and IL-6 lead to the same behavioural deficits, which are prevented by co-injection of an anti-IL-6 antibody or IL-6 knock-out (KO) [63,64].
Cytokines could exert their effects at three levels: the maternal circulation, the maternal-fetal interface, and the fetal brain (Figure 3). Injection of poly(I:C) into pregnant mice leads to increases in IL-6 in the placenta, derived from uterine natural killer cells and decidual macrophages, and activate the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in the spongiotrophoblast layer expressing glycoprotein 130 (gp130) and IL-6 receptors [32,44]. Changes in STAT transcription factors increase expression of acute phase genes (suppressor of cytokine signalling 3 (SOCS3), tissue inhibitor of metalloproteinases 1 (TIMP1)) and disrupt the growth hormone (GH)–insulin-like growth factor (IGF)-I axis [32,44]. This constitutes an indirect mechanism by which MIA could alter fetal development through disrupting trophoblast function, nutrient partitioning, and maternal-fetal tolerance.
A more direct mechanism could involve IL-6 crossing the placenta to enter the fetal circulation. However, the ability of cytokines to cross the placenta is disputed. While some studies have shown that radiolabelled IL-6 crosses the rat placenta and that IL-6 is transported across the human placenta ex vivo [78], others have found negligible transport [79]. These discrepancies could be ascribed to differences in measurement timing, as it is unknown how placental permeability to cytokines changes across gestation. Despite being hemochorial, rodent placentas show significant differences in interdigitation with human placentas [80]. Further insights into transplacental transport could be acquired by measuring cytokine concentrations in maternal and fetal compartments of the placenta in women undergoing caesarean section, an approach used to measure glucose transfer [81].
Elevated cytokine levels are found in the fetal brain (Table 3), possibly arising from local expression or from cytokines traversing the blood–brain barrier (BBB) via circumventricular organs or active transport [63]. However, the timing of BBB development and cytokine transporter expression in humans remains to be investigated. It will be equally important to delimit the site of action of IL-6, and this could be accomplished by investigating the effects of MIA in tissue-specific Cre-mediated IL-6R KO models.
Another unsolved question is how an acute immune challenge results in permanent behavioural deficits. MIA could lead to chronic neuronal injury underpinning later pathophysiology [55], or MIA-induced epigenetic alterations could affect the expression of inflammatory mediators in postnatal life [44], resulting in chronic elevations in proinflammatory cytokines responsible for behavioural deficits. This intriguing possibility may be tested through combining in vivo microdialysis with the inhibition of inflammatory mediators in specific brain regions.
Finally, the evidence that maternal antibodies to membrane proteins such as contactin-associated protein-like 2 (CASPR2) and (N-methyl-d-aspartate receptor) NMDAR can influence the behaviour of mice in maternal-to-fetal transfer models [82], suggests that MIAs may, by stimulating a maternal immune response, lead to potentially harmful autoantibodies. Alternatively, the infections and the associated cytokine responses could provide an inflammatory milieu that increases the transfer of immunoglobulin G (IgG) antibodies or their pathogenic effects. There are many possibilities that remain unexplored.

8. Conclusions

The role of MIA in the development of ASD and related disorders is highlighted by animal models linking prenatal infection and ASD-relevant deficits and clinical studies providing correlative evidence. However, key questions about the validity, mechanism, and the source of disease specificity in this hypothesis remain unsolved. A priming/multi-hit model, in which MIA acts as a primer requiring a substantial genetic contribution and further environmental factors to trigger the clinical manifestation of ASD, could be used in future studies to develop increasingly valid multi-trait animal models. Applying this framework to answer the MIA disease specificity question may offer insight into the clinical heterogeneity of ASD and its convergence with related diseases such as schizophrenia, with the ultimate goal of guiding diagnostic classification and therapeutic strategies in neurodevelopmental psychiatric disorders.

Funding

This review received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism spectrum disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef]
  2. Taylor, M.J.; Rosenqvist, M.A.; Larsson, H.; Gillberg, C.; D’Onofrio, B.M.; Lichtenstein, P.; Lundstrom, S. Etiology of Autism Spectrum Disorders and Autistic Traits Over Time. JAMA Psychiatry 2020. [Google Scholar] [CrossRef] [PubMed]
  3. Sandin, S.; Lichtenstein, P.; Kuja-Halkola, R.; Hultman, C.; Larsson, H.; Reichenberg, A. The Heritability of Autism Spectrum Disorder. JAMA 2017, 318, 1182–1184. [Google Scholar] [CrossRef] [PubMed]
  4. Tick, B.; Bolton, P.; Happé, F.; Rutter, M.; Rijsdijk, F. Heritability of autism spectrum disorders: A meta-analysis of twin studies. J. Child. Psychol. Psychiatry 2016, 57, 585–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hallmayer, J.; Cleveland, S.; Torres, A.; Phillips, J.; Cohen, B.; Torigoe, T.; Miller, J.; Fedele, A.; Collins, J.; Smith, K.; et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch. Gen. Psychiatry 2011, 68, 1095–1102. [Google Scholar] [CrossRef] [PubMed]
  6. Jiang, H.-Y.; Xu, L.-L.; Shao, L.; Xia, R.-M.; Yu, Z.-H.; Ling, Z.-X.; Yang, F.; Deng, M.; Ruan, B. Maternal infection during pregnancy and risk of autism spectrum disorders: A systematic review and meta-analysis. Brain Behav. Immun. 2016, 58, 165–172. [Google Scholar] [CrossRef] [PubMed]
  7. Lee, B.K.; Magnusson, C.; Gardner, R.M.; Blomstrom, A.; Newschaffer, C.J.; Burstyn, I.; Karlsson, H.; Dalman, C. Maternal hospitalization with infection during pregnancy and risk of autism spectrum disorders. Brain Behav. Immun. 2015, 44, 100–105. [Google Scholar] [CrossRef] [Green Version]
  8. Atladóttir, H.O.; Thorsen, P.; Schendel, D.E.; Østergaard, L.; Lemcke, S.; Partner, E.T. Association of hospitalization for infection in childhood with diagnosis of autism spectrum disorders: A Danish cohort study. Arch. Pediatr. Adolesc. Med. 2010, 164, 470–477. [Google Scholar] [CrossRef] [Green Version]
  9. Murray, K.N.; Edye, M.E.; Manca, M.; Vernon, A.C.; Oladipo, J.M.; Fasolina, V.; Harte, M.K.; Mason, V.; Grayson, B.; McHugh, P.C. Evolution of a maternal immune activation (mIA) model in rats: Early developmental effects. Brain Behav. Immun. 2019, 75, 48–59. [Google Scholar] [CrossRef] [Green Version]
  10. Amodeo, D.A.; Lai, C.-Y.; Hassan, O.; Mukamel, E.A.; Behrens, M.M.; Powell, S.B. Maternal immune activation impairs cognitive flexibility and alters transcription in frontal cortex. Neurobiol. Dis. 2019, 125, 211–218. [Google Scholar] [CrossRef] [Green Version]
  11. Haida, O.; Al Sagheer, T.; Balbous, A.; Francheteau, M.; Matas, E.; Soria, F.; Fernagut, P.O.; Jaber, M. Sex-dependent behavioral deficits and neuropathology in a maternal immune activation model of autism. Transl. Psychiatry 2019, 9, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pendyala, G.; Chou, S.; Jung, Y.; Coiro, P.; Spartz, E.; Padmashri, R.; Li, M.; Dunaevsky, A. Maternal Immune Activation Causes Behavioral Impairments and Altered Cerebellar Cytokine and Synaptic Protein Expression. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2017, 42, 1435–1446. [Google Scholar] [CrossRef] [Green Version]
  13. Kirsten, T.B.; Bernardi, M.M. Prenatal lipopolysaccharide induces hypothalamic dopaminergic hypoactivity and autistic-like behaviors: Repetitive self-grooming and stereotypies. Behav. Brain Res. 2017, 331, 25–29. [Google Scholar] [CrossRef] [PubMed]
  14. Gandal, M.J.; Zhang, P.; Hadjimichael, E.; Walker, R.L.; Chen, C.; Liu, S.; Won, H.; van Bakel, H.; Varghese, M.; Wang, Y.; et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 2018, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Eftekharian, M.M.; Ghafouri-Fard, S.; Noroozi, R.; Omrani, M.D.; Arsang-jang, S.; Ganji, M.; Gharzi, V.; Noorozi, H.; Komaki, A.; Mazdeh, M.; et al. Cytokine profile in autistic patients. Cytokine 2018, 108, 120–126. [Google Scholar] [CrossRef]
  16. Tylee, D.S.; Hess, J.L.; Quinn, T.P.; Barve, R.; Huang, H.; Zhang-James, Y.; Chang, J.; Stamova, B.S.; Sharp, F.R.; Hertz-Picciotto, I. Blood transcriptomic comparison of individuals with and without autism spectrum disorder: A combined-samples mega-analysis. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2017, 174, 181–201. [Google Scholar] [CrossRef] [Green Version]
  17. Gupta, S.; Ellis, S.E.; Ashar, F.N.; Moes, A.; Bader, J.S.; Zhan, J.; West, A.B.; Arking, D.E. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat. Commun. 2014, 5, 5748. [Google Scholar] [CrossRef]
  18. Fatemi, S.H.; Emamian, E.S.; Kist, D.; Sidwell, R.W.; Nakajima, K.; Akhter, P.; Shier, A.; Sheikh, S.; Bailey, K. Defective corticogenesis and reduction in Reelin immunoreactivity in cortex and hippocampus of prenatally infected neonatal mice. Mol. Psychiatry 1999, 4, 145–154. [Google Scholar] [CrossRef] [Green Version]
  19. Shi, L.; Fatemi, S.H.; Sidwell, R.W.; Patterson, P.H. Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J. Neurosci. Off. J. Soc. Neurosci. 2003, 23, 297–302. [Google Scholar] [CrossRef]
  20. Short, S.J.; Lubach, G.R.; Karasin, A.I.; Olsen, K.W.; Styner, M.; Knickmeyer, R.C.; Gilmore, J.H.; Coe, C.L. Maternal influenza infection during pregnancy impacts postnatal brain development in the rhesus monkey. Biol. Psychiatry 2010, 67, 965–973. [Google Scholar] [CrossRef] [Green Version]
  21. Malkova, N.V.; Yu, C.Z.; Hsiao, E.Y.; Moore, M.J.; Patterson, P.H. Maternal immune activation yields offspring displaying mouse versions of the three core symptoms of autism. Brain Behav. Immun. 2012, 26, 607–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Carlezon, W.A.; Kim, W.; Missig, G.; Finger, B.C.; Landino, S.M.; Alexander, A.J.; Mokler, E.L.; Robbins, J.O.; Li, Y.; Bolshakov, V.Y. Maternal and early postnatal immune activation produce sex-specific effects on autism-like behaviors and neuroimmune function in mice. Sci. Rep. 2019, 9, 16928. [Google Scholar] [CrossRef] [PubMed]
  23. Glass, R.; Norton, S.; Fox, N.; Kusnecov, A.W. Maternal immune activation with staphylococcal enterotoxin A produces unique behavioral changes in C57BL/6 mouse offspring. Brain Behav. Immun. 2019, 75, 12–25. [Google Scholar] [CrossRef]
  24. Missig, G.; Robbins, J.O.; Mokler, E.L.; Mccullough, K.; Bilbo, S.D.; McDougle, C.J.; Carlezon, W.A. Sex-dependent neurobiological features of prenatal immune activation via TLR7. Mol. Psychiatry 2019. [Google Scholar] [CrossRef] [PubMed]
  25. Schwartzer, J.J.; Careaga, M.; Chang, C.; Onore, C.E.; Ashwood, P. Allergic fetal priming leads to developmental, behavioral and neurobiological changes in mice. Transl. Psychiatry 2015, 5, e543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Meyer, U.; Nyffeler, M.; Schwendener, S.; Knuesel, I.; Yee, B.K.; Feldon, J. Relative prenatal and postnatal maternal contributions to schizophrenia-related neurochemical dysfunction after in utero immune challenge. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2008, 33, 441–456. [Google Scholar] [CrossRef]
  27. Shi, L.; Smith, S.E.P.; Malkova, N.; Tse, D.; Su, Y.; Patterson, P.H. Activation of the maternal immune system alters cerebellar development in the offspring. Brain Behav. Immun. 2009, 23, 116–123. [Google Scholar] [CrossRef] [Green Version]
  28. Fernández de Cossío, L.; Guzmán, A.; van der Veldt, S.; Luheshi, G.N. Prenatal infection leads to ASD-like behavior and altered synaptic pruning in the mouse offspring. Brain Behav. Immun. 2017, 63, 88–98. [Google Scholar] [CrossRef]
  29. Wu, W.-L.; Adams, C.E.; Stevens, K.E.; Chow, K.-H.; Freedman, R.; Patterson, P.H. The interaction between maternal immune activation and alpha 7 nicotinic acetylcholine receptor in regulating behaviors in the offspring. Brain Behav. Immun. 2015, 46, 192–202. [Google Scholar] [CrossRef] [Green Version]
  30. DiLalla, L.F.; McCrary, M.; Diaz, E. A review of endophenotypes in schizophrenia and autism: The next phase for understanding genetic etiologies. Am. J. Med. Genet. C Semin. Med. Genet. 2017, 175, 354–361. [Google Scholar] [CrossRef]
  31. Bjorklund, G.; Pivina, L.; Dadar, M.; Meguid, N.A.; Semenova, Y.; Anwar, M.; Chirumbolo, S. Gastrointestinal Alterations in Autism Spectrum Disorder: What Do We Know? Neurosci. Biobehav. Rev. 2020. [Google Scholar] [CrossRef] [PubMed]
  32. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bauman, M.D.; Iosif, A.-M.; Smith, S.E.P.; Bregere, C.; Amaral, D.G.; Patterson, P.H. Activation of the maternal immune system during pregnancy alters behavioral development of rhesus monkey offspring. Biol. Psychiatry 2014, 75, 332–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gumusoglu, S.B.; Stevens, H.E. Maternal Inflammation and Neurodevelopmental Programming: A Review of Preclinical Outcomes and Implications for Translational Psychiatry. Biol. Psychiatry 2019, 85, 107–121. [Google Scholar] [CrossRef] [PubMed]
  35. Brown, A.S.; Meyer, U. Maternal Immune Activation and Neuropsychiatric Illness: A Translational Research Perspective. Am. J. Psychiatry 2018, 175, 1073–1083. [Google Scholar] [CrossRef] [Green Version]
  36. Estes, M.L.; McAllister, A.K. Maternal immune activation: Implications for neuropsychiatric disorders. Science 2016, 353, 772–777. [Google Scholar] [CrossRef] [Green Version]
  37. Willette, A.A.; Lubach, G.R.; Knickmeyer, R.C.; Short, S.J.; Styner, M.; Gilmore, J.H.; Coe, C.L. Brain enlargement and increased behavioral and cytokine reactivity in infant monkeys following acute prenatal endotoxemia. Behav. Brain Res. 2011, 219, 108–115. [Google Scholar] [CrossRef] [Green Version]
  38. Oshiumi, H.; Matsumoto, M.; Funami, K.; Akazawa, T.; Seya, T. TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon-beta induction. Nat. Immunol. 2003, 4, 161–167. [Google Scholar] [CrossRef]
  39. Goines, P.E.; Croen, L.A.; Braunschweig, D.; Yoshida, C.K.; Grether, J.; Hansen, R.; Kharrazi, M.; Ashwood, P.; Van de Water, J. Increased midgestational IFN-γ, IL-4 and IL-5 in women bearing a child with autism: A case-control study. Mol. Autism 2011, 2, 13. [Google Scholar] [CrossRef] [Green Version]
  40. Xie, J.; Huang, L.; Li, X.; Li, H.; Zhou, Y.; Zhu, H.; Pan, T.; Kendrick, K.M.; Xu, W. Immunological cytokine profiling identifies TNF-α as a key molecule dysregulated in autistic children. Oncotarget 2017, 8, 82390–82398. [Google Scholar] [CrossRef] [Green Version]
  41. Ben-Yehuda, H.; Matcovitch-Natan, O.; Kertser, A.; Spinrad, A.; Prinz, M.; Amit, I.; Schwartz, M. Maternal Type-I interferon signaling adversely affects the microglia and the behavior of the offspring accompanied by increased sensitivity to stress. Mol. Psychiatry 2020, 25, 1050–1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Dulken, B.W.; Buckley, M.T.; Navarro Negredo, P.; Saligrama, N.; Cayrol, R.; Leeman, D.S.; George, B.M.; Boutet, S.C.; Hebestreit, K.; Pluvinage, J.V.; et al. Single-cell analysis reveals T cell infiltration in old neurogenic niches. Nature 2019, 571, 205–210. [Google Scholar] [CrossRef] [PubMed]
  43. Missault, S.; Van den Eynde, K.; Vanden Berghe, W.; Fransen, E.; Weeren, A.; Timmermans, J.P.; Kuman-Singh, S.; Dedeurwaedere, S. The risk for behavioural deficits is determined by the maternal immune response to prenatal immune challenge in a neurodevelopmental model. Brain Behav. Immun. 2014, 42, 138–146. [Google Scholar] [CrossRef] [PubMed]
  44. Hsiao, E.Y.; McBride, S.W.; Chow, J.; Mazmanian, S.K.; Patterson, P.H. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl. Acad. Sci. USA 2012, 109, 12776–12781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pineda, E.; Shin, D.; You, S.J.; Auvin, S.; Sankar, R.; Mazarati, A. Maternal immune activation promotes hippocampal kindling epileptogenesis in mice. Ann. Neurol. 2013, 74, 11–19. [Google Scholar] [CrossRef] [PubMed]
  46. Li, Q.; Cheung, C.; Wei, R.; Hui, E.S.; Feldon, J.; Meyer, U.; Chung, S.; Chue, S.E.; Sham, P.C.; Wu, E.X.; et al. Prenatal immune challenge is an environmental risk factor for brain and behavior change relevant to schizophrenia: Evidence from MRI in a mouse model. PLoS ONE 2009, 4, e6354. [Google Scholar] [CrossRef] [Green Version]
  47. Clancy, B.; Darlington, R.B.; Finlay, B.L. Translating developmental time across mammalian species. Neuroscience 2001, 105, 7–17. [Google Scholar] [CrossRef] [Green Version]
  48. Kirsten, T.B.; Chaves-Kirsten, G.P.; Chaible, L.M.; Silva, A.C.; Martins, D.O.; Britto, L.R.G.; Dagli, M.L.Z.; Torrao, A.S.; Palermo-Neto, J.; Bernardi, M.M. Hypoactivity of the central dopaminergic system and autistic-like behavior induced by a single early prenatal exposure to lipopolysaccharide. J. Neurosci. Res. 2012, 90, 1903–1912. [Google Scholar] [CrossRef]
  49. Xuan, I.C.Y.; Hampson, D.R. Gender-dependent effects of maternal immune activation on the behavior of mouse offspring. PLoS ONE 2014, 9, e104433. [Google Scholar] [CrossRef]
  50. Sharova, V.S.; Izvolskaia, M.S.; Zakharova, L.A. Lipopolysaccharide-induced maternal inflammation affects the gonadotropin-releasing hormone neuron development in fetal mice. Neuroimmunomodulation 2015, 22, 222–232. [Google Scholar] [CrossRef]
  51. Warrier, V.; Chee, V.; Smith, P.; Chakrabarti, B.; Baron-Cohen, S. A comprehensive meta-analysis of common genetic variants in autism spectrum conditions. Mol. Autism 2015, 6, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Wischhof, L.; Irrsack, E.; Osorio, C.; Koch, M. Prenatal LPS-exposure—A neurodevelopmental rat model of schizophrenia—Differentially affects cognitive functions, myelination and parvalbumin expression in male and female offspring. Prog. Neuropsychopharmacol. Biol. Psychiatry 2015, 57, 17–30. [Google Scholar] [CrossRef] [PubMed]
  53. Batinić, B.; Santrač, A.; Divović, B.; Timic, T.; Stankovic, T.; Obradovic, A.L.; Joksimovic, S.; Savic, M.M. Lipopolysaccharide exposure during late embryogenesis results in diminished locomotor activity and amphetamine response in females and spatial cognition impairment in males in adult, but not adolescent rat offspring. Behav. Brain Res. 2016, 299, 72–80. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, Y.; Qi, F.; Song, D.; He, Z.; Zuo, Z.; Yang, Y.; Liu, Q.; Hu, S.; Wang, X.; Zheng, X.; et al. Prenatal influenza vaccination rescues impairments of social behavior and lamination in a mouse model of autism. J. Neuroinflamm. 2018, 15, 228. [Google Scholar] [CrossRef] [PubMed]
  55. Meyer, U.; Nyffeler, M.; Engler, A.; Urwyler, A.; Schedlowski, M.; Knuesel, I.; Yee, B.K.; Feldon, J. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J. Neurosci. Off. J. Soc. Neurosci. 2006, 26, 4752–4762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Abazyan, B.; Nomura, J.; Kannan, G.; Ishizuka, K.; Tamashiro, K.L.; Nucifora, F.; Pogorelov, V.; Ladenheim, B.; Yang, C.; Krasnova, I.N.; et al. Prenatal interaction of mutant DISC1 and immune activation produces adult psychopathology. Biol. Psychiatry 2010, 68, 1172–1181. [Google Scholar] [CrossRef] [Green Version]
  57. Lipina, T.V.; Zai, C.; Hlousek, D.; Order, J.; Wong, A.H.C. Maternal immune activation during gestation interacts with Disc1 point mutation to exacerbate schizophrenia-related behaviors in mice. J. Neurosci. Off. J. Soc. Neurosci. 2013, 33, 7654–7666. [Google Scholar] [CrossRef] [Green Version]
  58. Ehninger, D.; Sano, Y.; de Vries, P.J.; Dies, K.; Franz, D.; Geschwind, D.H.; Kaur, M.; Lee, Y.-S.; Li, W.; Lowe, J.K.; 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. [Google Scholar] [CrossRef]
  59. Giovanoli, S.; Engler, H.; Engler, A.; Richetto, J.; Voget, M.; Willi, R.; Winter, C.; Riva, M.A.; Brobech, P.; Schedlowski, M. Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice. Science 2013, 339, 1095–1099. [Google Scholar] [CrossRef] [Green Version]
  60. Giovanoli, S.; Weber-Stadlbauer, U.; Schedlowski, M.; Meyer, U.; Engler, H. Prenatal immune activation causes hippocampal synaptic deficits in the absence of overt microglia anomalies. Brain Behav. Immun. 2016, 55, 25–38. [Google Scholar] [CrossRef] [Green Version]
  61. Li, X.; Tian, X.; Lv, L.; Hei, G.; Huang, X.; Fan, X.; Zhang, J.; Zhang, J.; Pang, L.; Song, X. Microglia activation in the offspring of prenatal Poly I: C exposed rats: A PET imaging and immunohistochemistry study. Gen. Psychiatry 2018, 31, e000006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Lins, B.R.; Marks, W.N.; Zabder, N.K.; Greba, Q.; Howland, J.G. Maternal Immune Activation during Pregnancy Alters the Behavior Profile of Female Offspring of Sprague Dawley Rats. eNeuro 2019, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Samuelsson, A.-M.; Jennische, E.; Hansson, H.-A.; Holmäng, A. Prenatal exposure to interleukin-6 results in inflammatory neurodegeneration in hippocampus with NMDA/GABA(A) dysregulation and impaired spatial learning. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006, 290, R1345–R1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Smith, S.E.P.; Li, J.; Garbett, K.; Mirnics, K.; Patterson, P.H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. Off. J. Soc. Neurosci. 2007, 27, 10695–10702. [Google Scholar] [CrossRef] [Green Version]
  65. Machado, C.J.; Whitaker, A.M.; Smith, S.E.P.; Patterson, P.H.; Bauman, M.D. Maternal immune activation in nonhuman primates alters social attention in juvenile offspring. Biol. Psychiatry 2015, 77, 823–832. [Google Scholar] [CrossRef] [Green Version]
  66. Weir, R.K.; Forghany, R.; Smith, S.E.P.; Patterson, P.H.; McAllister, A.K.; Schumann, C.M.; Bauman, M.D. Preliminary evidence of neuropathology in nonhuman primates prenatally exposed to maternal immune activation. Brain Behav. Immun. 2015, 48, 139–146. [Google Scholar] [CrossRef] [Green Version]
  67. Rose, D.R.; Careaga, M.; Van de Water, J.; McAllister, K.; Bauman, M.D.; Ashwood, P. Long-term altered immune responses following fetal priming in a non-human primate model of maternal immune activation. Brain Behav. Immun. 2017, 63, 60–70. [Google Scholar] [CrossRef]
  68. Bauman, M.D.; Lesh, T.A.; Rowland, D.J.; Schumann, C.M.; Smucny, J.; Kukis, D.L.; Cherry, S.R.; MacAllister, A.K.; Carter, C.S. Preliminary evidence of increased striatal dopamine in a nonhuman primate model of maternal immune activation. Transl. Psychiatry 2019, 9, 135. [Google Scholar] [CrossRef] [Green Version]
  69. Arrode-Brusés, G.; Brusés, J.L. Maternal immune activation by poly I:C induces expression of cytokines IL-1β and IL-13, chemokine MCP-1 and colony stimulating factor VEGF in fetal mouse brain. J. Neuroinflamm. 2012, 9, 83. [Google Scholar] [CrossRef] [Green Version]
  70. Sandin, S.; Lichtenstein, P.; Kuja-Halkola, R.; Larsson, H.; Hultman, C.M.; Reichenberg, A. The familial risk of autism. JAMA 2014, 311, 1770–1777. [Google Scholar] [CrossRef]
  71. Mazina, V.; Gerdts, J.; Trinh, S.; Ankenman, K.; Ward, T.; Dennis, M.Y.; Girirajan, S.; Eichler, E.E.; Berniner, R. Epigenetics of autism-related impairment: Copy number variation and maternal infection. J. Dev. Behav. Pediatr. JDBP 2015, 36, 61–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Foley, K.A.; MacFabe, D.F.; Kavaliers, M.; Ossenkopp, K.-P. Sexually dimorphic effects of prenatal exposure to lipopolysaccharide, and prenatal and postnatal exposure to propionic acid, on acoustic startle response and prepulse inhibition in adolescent rats: Relevance to autism spectrum disorders. Behav. Brain Res. 2015, 278, 244–256. [Google Scholar] [CrossRef] [PubMed]
  73. Yuen, R.K.C.; Thiruvahindrapuram, B.; Merico, D.; Walker, S.; Tammimies, K.; Hoang, N.; Crysler, C.; Nalpathamkalam, T.; Pellechia, G.; Liu, Y.; et al. Whole-genome sequencing of quartet families with autism spectrum disorder. Nat. Med. 2015, 21, 185–191. [Google Scholar] [CrossRef] [PubMed]
  74. Gaugler, T.; Klei, L.; Sanders, S.J.; Bodea, C.A.; Goldberg, A.P.; Lee, A.B.; Mahajan, M.; Manna, D.; Pawitan, Y.; Reichert, J.; et al. Most genetic risk for autism resides with common variation. Nat. Genet. 2014, 46, 881–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Autism Spectrum Disorders Working Group of the Psychiatric Genomics Consortium. Meta-analysis of GWAS of over 16,000 individuals with autism spectrum disorder highlights a novel locus at 10q24.32 and a significant overlap with schizophrenia. Mol. Autism 2017, 8. [Google Scholar] [CrossRef]
  76. Grove, J.; Ripke, S.; Als, T.D.; Mattheisen, M.; Walters, R.K.; Won, H.; Pallasen, J.; Agerbo, E.; Andreassen, O.A.; Anney, R. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 2019, 51, 431–444. [Google Scholar] [CrossRef] [Green Version]
  77. Patterson, P.H. Maternal infection and immune involvement in autism. Trends Mol. Med. 2011, 17, 389–394. [Google Scholar] [CrossRef] [Green Version]
  78. Zaretsky, M.V.; Alexander, J.M.; Byrd, W.; Bawdon, R.E. Transfer of inflammatory cytokines across the placenta. Obstet. Gynecol. 2004, 103, 546–550. [Google Scholar] [CrossRef]
  79. Aaltonen, R.; Heikkinen, T.; Hakala, K.; Laine, K.; Alanen, A. Transfer of proinflammatory cytokines across term placenta. Obstet. Gynecol. 2005, 106, 802–807. [Google Scholar] [CrossRef]
  80. PrabhuDas, M.; Bonney, E.; Caron, K.; Dey, S.; Erlebacher, A.; Fazleabas, A.; Fisher, S.; Golos, T.; Matzuk, M.; McCune, J.M.; et al. Immune mechanisms at the maternal-fetal interface: Perspectives and challenges. Nat. Immunol. 2015, 16, 328–334. [Google Scholar] [CrossRef]
  81. Holme, A.M.; Roland, M.C.P.; Lorentzen, B.; Michelsen, T.M.; Henriksen, T. Placental glucose transfer: A human in vivo study. PLoS ONE 2015, 10, e0117084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Coutinho, E.; Menassa, D.A.; Jacobson, L.; West, S.J.; Domingos, J.; Moloney, T.C.; Lang, B.; Harrison, B.J.; Bennett, D.L.H.; Bannerman, D.; et al. Persistent microglial activation and synaptic loss with behavioral abnormalities in mouse offspring exposed to CASPR2-antibodies in utero. Acta Neuropathol. 2017, 134, 567–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. A spectrum of genetic versus environmental contribution in the aetiology of autism spectrum disorder (ASD).
Figure 1. A spectrum of genetic versus environmental contribution in the aetiology of autism spectrum disorder (ASD).
Jcm 09 02590 g001
Figure 2. A priming/multi-hit model for MIA and disease specificity.
Figure 2. A priming/multi-hit model for MIA and disease specificity.
Jcm 09 02590 g002
Figure 3. Sites of action and mechanisms of MIA.
Figure 3. Sites of action and mechanisms of MIA.
Jcm 09 02590 g003

Share and Cite

MDPI and ACS Style

Ji-Xu, A.; Vincent, A. Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. J. Clin. Med. 2020, 9, 2590. https://doi.org/10.3390/jcm9082590

AMA Style

Ji-Xu A, Vincent A. Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. Journal of Clinical Medicine. 2020; 9(8):2590. https://doi.org/10.3390/jcm9082590

Chicago/Turabian Style

Ji-Xu, Antonio, and Angela Vincent. 2020. "Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity" Journal of Clinical Medicine 9, no. 8: 2590. https://doi.org/10.3390/jcm9082590

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop