Review

Etiologies underlying sex bias in autism spectrum disorder: a narrative review of preclinical rodent models

Taeyoung Lee1https://orcid.org/0009-0005-2317-3451, Eunha Kim1,2,*https://orcid.org/0000-0001-7041-1727
Author Information & Copyright
1BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
2Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
*Corresponding author: Eunha Kim, BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea, E-mail: Eunha_Kim@korea.ac.kr

© Copyright 2024 Ewha Womans University College of Medicine and Ewha Medical Research Institute. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Mar 11, 2024; Revised: Apr 22, 2024; Accepted: Apr 24, 2024

Published Online: Apr 30, 2024

Abstract

Neurodevelopmental disorders, which emerge early in development, include a range of neurological phenotypes and exhibit marked differences in prevalence between sexes. A male predominance is particularly pronounced in autism spectrum disorder (ASD). Although the precise cause of ASD is still unknown, certain genetic variations and environmental influences have been implicated as risk factors. Preclinical ASD models have been instrumental in shedding light on the mechanisms behind the sexual dimorphism observed in this disorder. In this review, we explore the potential processes contributing to sex bias by examining both intrinsic differences in neuronal mechanisms and the influence of external factors. We organize these mechanisms into six categories: 1) sexually dimorphic phenotypes in mice with mutations in ASD-associated genes related to synaptic dysfunction; 2) sex-specific microglial activity, which may disrupt neural circuit development by excessively pruning synapses during critical periods; 3) sex steroid hormones, such as testosterone and allopregnanolone, that differentially influence brain structure and function; 4) escape from X chromosome inactivation of the O-linked-N-acetylglucosamine transferase gene in the placenta; 5) sexually dimorphic activation of the integrated stress response pathway following maternal immune activation; and 6) immunological responses that are differentially regulated by sex. Understanding these mechanisms is essential for deciphering the underlying causes of ASD and may offer insights into other disorders with notable sex disparities.

Keywords: Autism spectrum disorder; Genetic variation; Pregnancy; Risk factors; Sex characteristics

Introduction

Background

Neurodevelopmental disorders (NDDs) are a heterogenous group of conditions that manifest during the developmental period and are characterized by impairments in various aspects of neurological functioning [1]. As defined by the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, NDDs include autism spectrum disorder (ASD), intellectual disability/developmental delay, attention-deficit/hyperactivity disorder, motor and tic disorders, and specific language disorders. These conditions can lead to a range of developmental deficits, from specific challenges in learning or executive function management to more extensive impairments in social skills or intellectual abilities [2]. This review is primarily focused on ASD, a NDD marked by deficits in social interaction and engagement in repetitive and stereotyped behaviors [2].

The prevalence, age of onset, pathophysiology, and symptomatology of many NDDs vary substantially by sex. A pronounced male bias is evident in ASD, with a male-to-female prevalence of approximately four to one [3,4]. The potential underdiagnosis of females with ASD has raised concerns, suggesting the need for distinct diagnostic criteria in their assessment [5,6]. Even apart from variations in diagnostic practices, a sex bias persists in the prevalence of ASD, with a male-to-female ratio ranging from at least 2:1 to 3:1. This highlights the critical need to explore the biological basis of sexual dimorphism, which may be key to understanding the processes underlying ASD pathogenesis [7].

Here, we explore the potential mechanisms contributing to the sex-biased prevalence disparity in ASD using various preclinical models (Table 1). While preclinical rodent models of ASD cannot fully replicate the spectrum of human ASD phenotypes [8], potentially due to differences in brain structures and developmental trajectories [9], they remain invaluable for gaining mechanistic insights into the pathogenesis of ASD and for exploring potential therapeutic strategies [10,11].

Table 1. Potential contributing mechanisms underlying the sex-biased prevalence of ASD as demonstrated in preclinical rodent models
Potential contributing mechanisms Preclinical models showing sexually dimorphic ASD-like phenotypes Suggested mechanism(s) References
Synaptic dysfunction Shank3 KO Reduced levels of mGluR5 in male mice [27]
Chd8+/N2373K Sexually dimorphic changes in neuronal activity, synaptic transmission, and transcriptomic profiles [40]
Fmr1 KO Sexually dimorphic upregulation of ASD risk genes
(male↑: Ctnnb1a and Grin1a, female↑: Homer1a, Ptgs2a, Drd1a, Pik3cab, and Csnk1g1b)
[44]
Microglial abnormalities Cntnap2 KO Activated morphology and phagocytosis of synaptic structures in male microglia [58]
DEP/MS Hyper-ramified phenotype in male microglia [63]
Hormones VPA-induced ASD mouse model Lower levels of TH expression in the AVPV of male mice [70]
Placenta-specific Akr1c14 KO Male mouse-specific abnormalities in cerebellar white matter [75]
Escape from X chromosome inactivation Prenatal stress model Placental OGT expression levels are twice as high for female fetuses as for male fetuses; this results in sexually distinct gene expression in trophoblasts through epigenetic modulation by histone methylation [79,80]
Integrated stress response pathway MIA (Poly[I:C]) Hyperactivation of the ISR pathway in male MIA offspring, resulting in reduced nascent protein synthesis in the brain [85]
Immune pathways Prenatal GBS infection Heightened levels of pro-inflammatory cytokines and chemokines such as IL-1β and CINC-1/CXCL1 in male fetuses [98]
MIA (LPS) Male MIA offspring exhibit heightened cortical hypoxia, reduced mitosis of radial glial cells, disrupted E/I balance within the brain, severe placental necrosis, elevated inflammation, and reduced placental growth [99]
MIA (two-hit model) The anti-inflammatory cytokines IL-10 and TGF-β1 are decreased in male offspring but increased in female mice [100]

upregulated,

a high-confidence risk genes for ASD,

b suggestive risk genes for ASD.

ASD, autism spectrum disorder; Shank3, SH3 and multiple ankyrin repeat domains 3; KO, knockout; mGluR5, metabotropic glutamate receptor 5; Chd8, chromodomain helicase DNA-binding protein 8; Fmr1, fragile X mental retardation 1; Ctnnb1, catenin beta 1; Grin1, glutamate ionotropic receptor NMDA type subunit 1; Homer1, homer scaffold protein 1; Ptgs2, prostaglandin-endoperoxide synthase 2; Drd1, dopamine receptor D1; Pik3ca, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; Csnk1g1, casein kinase 1 gamma 1; Cntnap2, contactin-associated protein 2; DEP/MS, diesel exhaust particles and maternal stress; VPA, valproic acid; TH, tyrosine hydroxylase; AVPV, anteroventral periventricular nucleus; Akr1c14, aldo-keto reductase family 1 member C4; OGT, O-linked-N-acetylglucosamine transferase; MIA, maternal immune activation; poly(I:C), polyinosinic:polycytidylic acid; ISR, integrated stress response; GBS, Group B Streptococcus; IL-1β, interleukin 1 beta; CINC-1/CXCL1, cytokine-induced neutrophil chemoattractant-1; LPS, lipopolysaccharide; E/I, excitation/inhibition; IL-10, interleukin 10; TGF-β1, transforming growth factor beta 1.

Download Excel Table
Objectives

This review aims to elucidate the potential contributing mechanisms underlying the sex-biased prevalence of ASD as demonstrated in preclinical rodent models. These include synaptic dysfunction, microglial abnormalities, the influence of sex hormones, escape from X chromosome inactivation, the integrated stress response (ISR) pathway, and immune pathways.

Methods

Ethics statement

The present study is based on a review of the literature; consequently, neither approval from an institutional review board nor the acquisition of informed consent was necessary.

Study design

This study is a narrative review.

Literature search: information sources and search strategies

We searched the PubMed database for articles published from 1990 up to April 2024. Only articles published in English were included.

Results

This review encompasses a total of 104 articles. A list of articles pertaining to each topic is available in the references.

Synaptic dysfunction

Previous studies have reported that those with ASD exhibit different brain connectivity patterns compared to typically developing individuals [12]. Patterns of widespread cortical underconnectivity, local overconnectivity, or a combination of these suggest that disrupted brain connectivity may represent a potential neural signature of ASD [13]. Brain connectivity is largely determined by the characteristics of neurons and synapses, with synapses being highly specialized, asymmetric cell-to-cell junctions that constitute the fundamental units of brain communication [14].

According to the Simons Foundation Autism Research Initiative (SFARI) gene database, hundreds of genes have been identified as being associated with ASD [1518]. Among these, genes such as those of the SH3 and multiple ankyrin repeat domains (SHANK) family, fragile X mental retardation 1 (FMR1), and chromodomain helicase DNA-binding protein 8 (CHD8) are linked to common cellular pathways that converge at synapses [1921]. This convergence suggests that synaptic dysfunction may contribute to the development of ASD, potentially leading to functional and cognitive impairments [14].

SHANK, also known as ProSAP, is a family of postsynaptic proteins found at glutamatergic synapses and includes three major isoforms: SHANK1, SHANK2, and SHANK3. These proteins act as master scaffolding proteins at excitatory synapses [22,23]. They interact with over 30 synaptic proteins across multiple domains and are critical for synaptic formation, glutamate receptor trafficking, and neuronal signaling [24]. Genetic screenings have identified mutations, rare variants, or disruptions of the SHANK3 gene in patients with ASD [22]. Mice with a genetic disruption of Shank3 display compulsive/repetitive behaviors and social interaction deficits, which reflect clinical features of ASD [25]. Studies using Shank3 knockout (KO) mouse models have reported sexually dimorphic phenotypes [26,27]. Matas et al. found that male Shank3 KO mice with a mutation in the C-terminal regions (exons 21–22) [28] exhibit more pronounced gait deficits than their female siblings [27]. Further research into cerebellar glutamate levels and postsynaptic receptors showed that metabotropic glutamate receptor 5 levels were reduced only in male Shank3 KO mice, suggesting a potential cause for the varied behavioral outcomes [27].

CHD8, a chromatin remodeling factor, is essential for regulating the transcription of a wide variety of genes [29,30], including approximately 1,000 ASD risk genes identified in the SFARI gene database [30]. Mice with homozygous deletions of Chd8 die early in embryonic development [31], while those with heterozygous mutations or gene knockdown display a range of ASD-like phenotypes [3236]. These include impaired social interaction, repetitive behaviors, and cognitive impairments, resembling characteristics of individuals with CHD8 mutations [20,30,3739]. Jung et al. found that a heterozygous mutation in Chd8, specifically the substitution of asparagine with lysine at position 2373—the first mutation identified as an ASD risk factor in human CHD8—results in sexually dimorphic effects that range from transcriptional to behavioral changes in mice [40]. Male Chd8+/N2373K mice exhibited various abnormal behaviors at the pup, juvenile, and adult stages, such as increased ultrasonic vocalizations when seeking their mother, heightened attachment upon reunion with their mother, and increased self-grooming when isolated. In contrast, their female counterparts did not exhibit these behaviors. This behavioral disparity is thought to be associated with sexual dimorphism in neuronal activity, synaptic transmission, and transcriptomic profiles.

The FMR1 gene encodes the fragile X mental retardation protein (FMRP), which acts as a messenger RNA-binding translational suppressor. It also modulates activity-dependent calcium signaling during critical developmental periods [41]. In mice, FMRP is most abundantly expressed in the hippocampus and cerebral cortex, with peak levels occurring between 2 to 4 weeks postnatally—a crucial time frame for synaptic development and maturation [42]. A deficiency in FMRP results in abnormal synaptic plasticity and structural remodeling [43]. Notably, male Fmr1 KO mice exhibit more severe anxiety, deficiencies in social preference, and repetitive behaviors than their female counterparts [44]. Differential gene expression analysis of the hippocampus in wild-type (WT) versus Fmr1 KO mice revealed that in male Fmr1 KO mice, Ctnnb1 and Grin1—genes considered high-confidence risk factors for ASD—are highly upregulated. In contrast, female Fmr1 KO mice exhibited upregulation of genes such as Homer1, Ptgs2, and Drd1, which are strong ASD risk gene candidates, as well as Pik3ca and Csnk1g1, which provide suggestive evidence of risk for ASD. These findings suggest that the loss of FMRP leads to sexually dimorphic phenotypes, potentially due to different patterns of gene expression regulation resulting from the absence of FMR1.

The collective evidence from these reports suggests that synaptic dysfunction and disrupted connectivity could be responsible for sex-specific functional and cognitive impairments observed in ASD [45].

Microglial abnormalities

The balance between excitation and inhibition (E/I) in neural circuits is critical for maintaining brain homeostasis [46]. Disruption of this E/I balance has been implicated as a potential cause of behavioral phenotypes associated with ASD [47]. Microglia, the phagocytic cells that reside in the brain from the developmental period, engulf the synaptic materials, thus pruning synapses and supporting synaptic maturation. [48]. When microglial function is compromised, improper synaptic pruning can disrupt the E/I balance and potentially contribute to the pathogenesis of ASD [49]. Notably, microglia exhibit sexually dimorphic transcriptional and translational profiles [50]. Furthermore, the morphology and number of microglia in the developing rat brain differ between male and female rats [51]. During the early postnatal period, male rats have significantly higher numbers of microglia compared to female rats. These sex-based differences in microglial numbers appear to be functionally related to sex-specific behaviors [52].

The contactin-associated protein 2 (CNTNAP2) gene encodes the CASPR2 protein, which is a neurexin-related synaptic cell adhesion molecule. A study utilizing high-density single nucleotide polymorphisms identified CNTNAP2 as a strong candidate gene implicated in the etiology of ASD [53]. Subsequent loss-of-function studies in Cntnap2 KO mice demonstrated that the absence of Cntnap2 leads to a decrease in dendritic spine density [54], disruptions in synaptic function [55], imbalances in E/I signaling, and impaired neural oscillations [56]. These Cntnap2 KO mice also display core ASD-like behavioral phenotypes, including impairments in sociability and repetitive behaviors [57]. Dawson et al. found that male Cntnap2 KO mice exhibited pronounced social deficits, whereas their female counterparts did not. Further investigation into the anterior cingulate cortex—a region critical for social behavior regulation through its connections with other intracortical and subcortical areas—revealed a more activated morphology and increased phagocytosis of synaptic structures in male KO mice compared to WT mice, a distinction not observed in female KO versus WT mice [58].

In addition to genetic models, differences in microglial morphology and function have been observed in preclinical models that incorporate environmental risk factors. High levels of air pollution, particularly during development [59,60], and maternal stress (MS) during gestation [61,62] have been linked to an increased risk of ASD. Smith et al. investigated the combined effects of these two risk factors and found that prenatal exposure to air pollution—specifically diesel exhaust particles (DEP)—along with MS in mice led to sociability deficits exclusively in male offspring [63]. These behavioral impairments were paralleled by alterations in microglial morphology and gene expression, with DEP/MS exposure resulting in a hyper-ramified microglial phenotype in male but not female animals.

The collective evidence from these reports suggests that sexually dimorphic microglial activity could play a role in the etiology of ASD. This activity may disrupt the development of neural circuits responsible for social behavior by excessively pruning synapses during a critical period of development [49].

Hormones

Sex steroid hormones are known to contribute to sex differences in neural activity and behaviors in mammals through their interactions with specific nuclear hormone receptors [64]. Testosterone plays a crucial role during prenatal development in shaping sex differences, influencing brain structure, neurotransmitter and receptor levels, neurogenesis, immune responses, neuropeptide signaling, and cellular processes such as apoptosis, migration, and differentiation [65]. Clinical reports have correlated high levels of testosterone with autistic behavior [66,67], and this association is supported by Erdogan et al., who showed that prenatal testosterone exposure led to ASD-like behaviors in the offspring of Wistar rats [68]. Both male and female rats exposed to testosterone exhibited reduced interaction times with a stranger rat during the three-chamber sociability and social novelty test, indicating a decrease in social interaction and a phenotype with characteristics resembling ASD. In line with these findings, studies on the valproic acid (VPA)-induced ASD mouse model, which is based on a medication known to increase the risk of ASD in humans [69], have shown that elevated plasma testosterone levels resulting from VPA treatment led to significantly lower levels of tyrosine hydroxylase (TH) expression in the anteroventral periventricular nucleus of male mice. In contrast, TH levels in female mice were unaffected [70].

Allopregnanolone (ALLO), a 3ɑ, 5ɑ progesterone metabolite [71], is a key GABAergic neurosteroid [72,73]. Reduced ALLO levels are correlated with a greater severity of restricted and repetitive behaviors [74]. Penn and colleagues have shown that ALLO plays a vital role as a placental hormone in shaping the fetal brain, leading to sexually dimorphic behavioral outcomes [75]. Specifically, a deficiency of placental ALLO in mice resulted in male-specific abnormalities in cerebellar white matter and core ASD symptoms, such as diminished social preference and increased repetitive behaviors. Notably, this study observed sex-linked dysregulation of myelin proteins in the cerebellar vermis of preterm infants, which aligns with human data.

These results highlight the influence of hormones in molding the early brain environment, potentially leading to sexually dimorphic behavioral outcomes.

Escape from X chromosome inactivation

In a mouse model of early prenatal stress, male offspring exposed to MS during gestation exhibited certain NDD phenotypes [76,77]. The placenta plays a critical role during pregnancy, acting as a mediator in response to disturbances within the intrauterine environment [78]. MS leads to sexually dimorphic changes in the placental expression of O-linked-N-acetylglucosamine transferase (OGT), an X-linked gene essential for the regulation of proteins involved in chromatin remodeling [79]. Notably, OGT escapes X chromosome inactivation in the placenta, resulting in placental levels that are approximately twice as high in female animals than in male animals. Crucially, this finding also translates to humans: levels of both OGT and its biochemical marker, O-GlcNAcylation, have been found to be considerably lower for male fetuses and are further reduced by prenatal stress [79]. Nugent et al. demonstrated that OGT levels establish a sex-specific gene expression pattern in trophoblasts through regulation of a canonical histone repressive mark, H3K27me3 [80]. Higher placental levels of H3K27me3 for female offspring provided a protective effect against the altered hypothalamic programming associated with prenatal stress exposure. Consequently, lower levels of OGT may predispose male offspring to a higher risk of ASD. Future studies should explore the molecular mechanisms underlying this increased male susceptibility.

Integrated stress response pathway

Maternal immune activation (MIA) during pregnancy is linked to a heightened risk of ASD in offspring [81]. This phenomenon has been extensively investigated using a rodent MIA model. In this model, pregnant mice received intraperitoneal injections of polyinosinic:polycytidylic acid (poly[I:C]), a synthetic analog of double-stranded RNA that simulates viral infection. The offspring exhibited significant neurodevelopmental impairments, including diminished social interaction and increased repetitive behaviors [8284]. Kalish et al. found that MIA exerts a sexually dimorphic effect in utero, leading to different behavioral outcomes. Male offspring exhibited MIA-induced behavioral abnormalities, whereas female offspring did not [85]. Notably, when gene expression was examined at the single-cell level, changes in the fetal cortex were observed to be sexually dimorphic. In male fetuses, these changes were predominantly characterized by reduced gene expression related to protein translation, followed by an overactive ISR pathway. In eukaryotic cells, the ISR signaling pathway regulates protein synthesis in response to various stresses, both physiological and pathological, to restore cellular homeostasis [86]. Dysregulation of protein synthesis has been implicated in ASD-related traits and other neurological disorders [8789]. Male-specific activation of the ISR is dependent on the maternal induction of interleukin (IL)-17A following MIA, which has been shown to be necessary for the development of MIA-induced ASD-like behaviors in mouse offspring [83]. The genetic and pharmacological inhibition of ISR pathway hyperactivation was sufficient to protect male offspring from MIA-induced behavioral abnormalities. This study offers valuable insights into potential preventative strategies for ASD-like phenotypes that may result from prenatal immune activation.

Immune pathways

Preterm delivery is associated with a higher likelihood of ASD in children compared to those born at full term [9093]. Chorioamnionitis, caused by Group B Streptococcus (GBS; Streptococcus agalactiae), is one of the most common maternal infections and accounts for 40% to 70% of preterm births [9496]. This condition typically involves an inflammatory intrauterine environment, even in the absence of bacterial translocation from mother to fetus [97]. Allard et al. demonstrated that prenatal infection with live GBS in rats resulted in social impairments in male but not in female offspring [98]. A prominent inflammatory state was noted in male animals, with higher levels of the pro-inflammatory cytokine IL-1β and the cytokine-induced neutrophil chemoattractant-1 (CINC-1/CXCL1), compared to female rats. These findings suggest that sex-specific inflammatory profiles may contribute to the observed sexually dimorphic behavioral outcomes [98].

Consistent with this notion, in a model of MIA induced by lipopolysaccharide (LPS), a toll-like receptor 4 agonist, Braun et al. investigated sex-specific pro-inflammatory responses in both the placenta and fetus, as well as their effects on behavioral outcomes. Male offspring of mothers exposed to LPS exhibited behavioral abnormalities in social interaction and learning, as well as increased repetitive behavior, whereas female offspring were unaffected [99]. Male MIA offspring showed increased cortical hypoxia, decreased mitosis of radial glial cells, and disrupted E/I balance in the brain. Additionally, severe placental necrosis, heightened inflammation, and reduced placental growth were specifically observed in male mice affected by MIA, suggesting that unique sex-specific placental characteristics may make male offspring more susceptible to intrauterine disturbances.

Carlezon Jr et al. demonstrated sex-specific behavioral effects and immune responses in the brain using a combined rodent “two-hit” immune activation model. This model involved treatment with poly (I:C) to induce MIA and the administration of LPS to produce postnatal immune activation [100]. Exposure to early-life immune activation (EIA) was shown to lead to reduced social interaction and increased repetitive behaviors in male animals, while female rodents displayed no significant changes. Molecular studies indicated that EIA resulted in pronounced sex-specific alterations in the expression of inflammation-related genes in the brain. Both male and female rodents exposed to EIA exhibited elevated levels of pro-inflammatory factors in the brain, such as tumor necrosis factor alpha, inducible nitric oxide synthase, IL-6, and IL-1β. Conversely, the expression of anti-inflammatory factors like IL-10 and transforming growth factor beta 1 was reduced in male mice but elevated in female animals [100].

The collective findings of these studies suggest that sexually dimorphic inflammatory responses could potentially contribute to the sex-specific effects of MS on the neurobehavioral outcomes of offspring.

Discussion

Implication and suggestion

In this review, we have comprehensively examined the potential mechanisms by which genetic variants and environmental factors contribute to sex differences, as demonstrated by preclinical models. Our analysis encompassed both intrinsic differences in the brain, such as synaptic connectivity and microglial activity, and the potential influence of extrinsic factors, including sex hormones and the placenta. These elements may either increase male susceptibility or bolster female resilience. Notably, beyond the intrinsic factors of the brain, the hormonal profile, epigenetic landscape, and immune pathways associated with the placenta have been implicated in contributing to sexually dimorphic outcomes in mouse models exhibiting ASD-like behaviors. This indicates that a deeper understanding of the placenta as a temporary but dynamic interface during prenatal development could provide valuable insights into the sex biases observed in NDDs.

Although preclinical mouse models of ASD have important limitations, such as their inability to engage neural circuitry comparable to that observed in humans or to recapitulate all human ASD phenotypes [8], they remain valuable tools for gaining mechanistic insights into ASD pathogenesis and for developing potential therapeutic approaches [10,11]. Further investigation is imperative to unravel the mechanistic basis of sexual dimorphism in ASD, as well as in other NDDs. For example, utilizing large-scale transcriptomics from postmortem brain studies [101], generating a single-cell atlas [102], and conducting multi-omic profiling of somatic mutations [103] in brains exhibiting ASD could help uncover sex-specific changes in genes or pathways during early neurodevelopment. Given that ASD is a developmental disorder, it is crucial to conduct further longitudinal investigations to understand how risk factors evolve across the developmental trajectory. For instance, a longitudinal cohort study of children with developmental disabilities suggested that de novo protein-truncating variants were correlated with clinical characteristics [104].

Conclusion

Overall, understanding sex-specific mechanisms is pivotal for comprehending the fundamental causes of ASD and may illuminate the pathologies of other diseases characterized by prominent sex biases.

Authors' contributions

Project administration: not applicable

Conceptualization: Lee T, Kim E

Methodology & data curation: not applicable

Funding acquisition: Kim E

Writing – original draft: Lee T

Writing – review & editing: Lee T, Kim E

Conflict of interest

Eunha Kim serves as a consultant for Interon Laboratories.

Funding

Eunha Kim received support through a Korea University grant (K2225821).

Data availability

Not applicable.

Acknowledgments

We would like to express our gratitude to all members of the Kim laboratory for their insightful comments. We also give special thanks to Hyun Je for providing technical assistance in structuring the manuscript.

Supplementary materials

Not applicable.

References

1.

Morris-Rosendahl DJ, Crocq MA. Neurodevelopmental disorders—the history and future of a diagnostic concept. Dialogues Clin Neurosci. 2020; 22(1):65-72

2.

American Psychiatric Association. Diagnostic and statistical manual of mental disorders (DSM-5®). 5th ed Washington: American Psychiatric Association. 2013

3.

Fombonne E. Epidemiology of pervasive developmental disorders. Pediatr Res. 2009; 65(6):591-598

4.

Werling DM, Geschwind DH. Sex differences in autism spectrum disorders. Curr Opin Neurol. 2013; 26(2):146-153

5.

Loomes R, Hull L, Mandy WPL. What is the male-to-female ratio in autism spectrum disorder? A systematic review and meta-analysis. J Am Acad Child Adolesc Psychiatry. 2017; 56(6):466-474

6.

Hull L, Petrides KV, Mandy W. The female autism phenotype and camouflaging: a narrative review. Rev J Autism Dev Disord. 2020; 7(4):306-317

7.

Zwaigenbaum L, Bryson SE, Szatmari P, Brian J, Smith IM, Roberts W, et al. Sex differences in children with autism spectrum disorder identified within a high-risk infant cohort. J Autism Dev Disord. 2012; 42(12):2585-2596

8.

Silverman JL, Thurm A, Ethridge SB, Soller MM, Petkova SP, Abel T, et al. Reconsidering animal models used to study autism spectrum disorder: current state and optimizing future. Genes Brain Behav. 2022; 21(5)e12803

9.

Pensado-López A, Veiga-Rúa S, Carracedo Á, Allegue C, Sánchez L. Experimental models to study autism spectrum disorders: hiPSCs, rodents and zebrafish. Genes. 2020; 11(11):1376

10.

Kazdoba TM, Leach PT, Yang M, Silverman JL, Solomon M, Crawley JN. Translational mouse models of autism: advancing toward pharmacological therapeutics. Curr Top Behav Neurosci. 2016; 28:1-52

11.

Jabarin R, Netser S, Wagner S. Beyond the three-chamber test: toward a multimodal and objective assessment of social behavior in rodents. Mol Autism. 2022; 13(1):41

12.

Mohammad-Rezazadeh I, Frohlich J, Loo SK, Jeste SS. Brain connectivity in autism spectrum disorder. Curr Opin Neurol. 2016; 29(2):137-147

13.

Maximo JO, Cadena EJ, Kana RK. The implications of brain connectivity in the neuropsychology of autism. Neuropsychol Rev. 2014; 24(1):16-31

14.

Guang S, Pang N, Deng X, Yang L, He F, Wu L, et al. Synaptopathology involved in autism spectrum disorder. Front Cell Neurosci. 2018; 12:470

15.

Banerjee-Basu S, Packer A. SFARI Gene: an evolving database for the autism research community. Dis Model Mech. 2010; 3(3-4):133-135

16.

Abrahams BS, Arking DE, Campbell DB, Mefford HC, Morrow EM, Weiss LA, et al. SFARI Gene 2.0: a community-driven knowledgebase for the autism spectrum disorders (ASDs). Mol Autism. 2013; 4(1):36

17.

Iossifov I, O'Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature. 2014; 515(7526):216-221

18.

Choi L, An JY. Genetic architecture of autism spectrum disorder: lessons from large-scale genomic studies. Neurosci Biobehav Rev. 2021; 128:244-257

19.

Wang T, Guo H, Xiong B, Stessman HAF, Wu H, Coe BP, et al. De novo genic mutations among a Chinese autism spectrum disorder cohort. Nat Commun. 2016; 7:13316

20.

Stessman HAF, Xiong B, Coe BP, Wang T, Hoekzema K, Fenckova M, et al. Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases. Nat Genet. 2017; 49(4):515-526

21.

Ellingford RA, Panasiuk MJ, de Meritens ER, Shaunak R, Naybour L, Browne L, et al. Cell-type-specific synaptic imbalance and disrupted homeostatic plasticity in cortical circuits of ASD-associated Chd8 haploinsufficient mice. Mol Psychiatry. 2021; 26(7):3614-3624

22.

Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007; 39(1):25-27

23.

Betancur C, Buxbaum JD. SHANK3 haploinsufficiency: a "common" but underdiagnosed highly penetrant monogenic cause of autism spectrum disorders. Mol Autism. 2013; 4(1):17

24.

Monteiro P, Feng G. SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat Rev Neurosci. 2017; 18(3):147-157

25.

Peca J, Feliciano C, Ting JT, Wang W, Wells MF, Venkatraman TN, et al. Shank3 mutant mice display autistic-like behaviours and striatal dysfunction. Nature. 2011; 472(7344):437-442

26.

Maloney SE, Sarafinovska S, Weichselbaum C, McCullough KB, Swift RG, Liu Y, et al. A comprehensive assay of social motivation reveals sex-specific roles of autism-associated genes and oxytocin. Cell Rep Methods. 2023; 3(6):100504

27.

Matas E, Maisterrena A, Thabault M, Balado E, Francheteau M, Balbous A, et al. Major motor and gait deficits with sexual dimorphism in a Shank3 mutant mouse model. Mol Autism. 2021; 12(1):2

28.

Moretto E, Murru L, Martano G, Sassone J, Passafaro M. Glutamatergic synapses in neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2018; 84(Pt B):328-342

29.

Bourgeron T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat Rev Neurosci. 2015; 16(9):551-563

30.

Barnard RA, Pomaville MB, O'Roak BJ. Mutations and modeling of the chromatin remodeler CHD8 define an emerging autism etiology. Front Neurosci. 2015; 9:477

31.

Nishiyama M, Nakayama K, Tsunematsu R, Tsukiyama T, Kikuchi A, Nakayama KI. Early embryonic death in mice lacking the β-catenin-binding protein Duplin. Mol Cell Biol. 2004; 24(19):8386-8394

32.

Katayama Y, Nishiyama M, Shoji H, Ohkawa Y, Kawamura A, Sato T, et al. CHD8 haploinsufficiency results in autistic-like phenotypes in mice. Nature. 2016; 537(7622):675-679

33.

Durak O, Gao F, Kaeser-Woo YJ, Rueda R, Martorell AJ, Nott A, et al. Chd8 mediates cortical neurogenesis via transcriptional regulation of cell cycle and Wnt signaling. Nat Neurosci. 2016; 19(11):1477-1488

34.

Gompers AL, Su-Feher L, Ellegood J, Copping NA, Asrafuzzaman Riyadh M, Stradleigh TW, et al. Germline Chd8 haploinsufficiency alters brain development in mouse. Nat Neurosci. 2017; 20(8):1062-1073

35.

Platt RJ, Zhou Y, Slaymaker IM, Shetty AS, Weisbach NR, Kim JA, et al. Chd8 mutation leads to autistic-like behaviors and impaired striatal circuits. Cell Rep. 2017; 19(2):335-350

36.

Suetterlin P, Hurley S, Mohan C, Riegman KLH, Pagani M, Caruso A, et al. Altered neocortical gene expression, brain overgrowth and functional over-connectivity in Chd8 haploinsufficient mice. Cereb Cortex. 2018; 28(6):2192-2206

37.

Bernier R, Golzio C, Xiong B, Stessman HA, Coe BP, Penn O, et al. Disruptive CHD8 mutations define a subtype of autism early in development. Cell. 2014; 158(2):263-276

38.

O'Roak BJ, Vives L, Fu W, Egertson JD, Stanaway IB, Phelps IG, et al. Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science. 2012; 338(6114):1619-1622

39.

Merner N, Forgeot d'Arc B, Bell SC, Maussion G, Peng H, Gauthier J, et al. A de novo frameshift mutation in chromodomain helicase DNA-binding domain 8 (CHD8): a case report and literature review. Am J Med Genet A. 2016; 170(5):1225-1235

40.

Jung H, Park H, Choi Y, Kang H, Lee E, Kweon H, et al. Sexually dimorphic behavior, neuronal activity, and gene expression in Chd8-mutant mice. Nat Neurosci. 2018; 21(9):1218-1228

41.

Davis JK, Broadie K. Multifarious functions of the fragile X mental retardation protein. Trends Genet. 2017; 33(10):703-714

42.

Pacey LKK, Xuan ICY, Guan S, Sussman D, Mark Henkelman R, Chen Y, et al. Delayed myelination in a mouse model of fragile X syndrome. Hum Mol Genet. 2013; 22(19):3920-3930

43.

Qin M, Entezam A, Usdin K, Huang T, Liu ZH, Hoffman GE, et al. A mouse model of the fragile X premutation: effects on behavior, dendrite morphology, and regional rates of cerebral protein synthesis. Neurobiol Dis. 2011; 42(1):85-98

44.

Wang Z, Qiao D, Chen H, Zhang S, Zhang B, Zhang J, et al. Effects of Fmr1 gene mutations on sex differences in autism-like behavior and dendritic spine development in mice and transcriptomic studies. Neuroscience. 2023; 534:16-28

45.

Luo J, Norris RH, Gordon SL, Nithianantharajah J. Neurodevelopmental synaptopathies: insights from behaviour in rodent models of synapse gene mutations. Prog Neuropsychopharmacol Biol Psychiatry. 2018; 84(Pt B):424-439

46.

Tatti R, Haley MS, Swanson OK, Tselha T, Maffei A. Neurophysiology and regulation of the balance between excitation and inhibition in neocortical circuits. Biol Psychiatry. 2017; 81(10):821-831

47.

Hollestein V, Poelmans G, Forde NJ, Beckmann CF, Ecker C, Mann C, et al. Excitatory/inhibitory imbalance in autism: the role of glutamate and GABA gene-sets in symptoms and cortical brain structure. Transl Psychiatry. 2023; 13(1):18

48.

Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011; 333(6048):1456-1458

49.

Koyama R, Ikegaya Y. Microglia in the pathogenesis of autism spectrum disorders. Neurosci Res. 2015; 100:1-5

50.

Guneykaya D, Ivanov A, Hernandez DP, Haage V, Wojtas B, Meyer N, et al. Transcriptional and translational differences of microglia from male and female brains. Cell Rep. 2018; 24(10):2773-2783.E6

51.

Schwarz JM, Sholar PW, Bilbo SD. Sex differences in microglial colonization of the developing rat brain. J Neurochem. 2012; 120(6):948-963

52.

McCarthy MM, Nugent BM, Lenz KM. Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat Rev Neurosci. 2017; 18(8):471-484

53.

Alarcón M, Abrahams BS, Stone JL, Duvall JA, Perederiy JV, Bomar JM, et al. Linkage, association, and gene-expression analyses identify CNTNAP2 as an autism-susceptibility gene. Am J Hum Genet. 2008; 82(1):150-159

54.

Varea O, Martin-de-Saavedra MD, Kopeikina KJ, Schürmann B, Fleming HJ, Fawcett-Patel JM, et al. Synaptic abnormalities and cytoplasmic glutamate receptor aggregates in contactin associated protein-like 2/Caspr2 knockout neurons. Proc Natl Acad Sci USA. 2015; 112(19):6176-6181

55.

Anderson GR, Galfin T, Xu W, Aoto J, Malenka RC, Südhof TC. Candidate autism gene screen identifies critical role for cell-adhesion molecule CASPR2 in dendritic arborization and spine development. Proc Natl Acad Sci USA. 2012; 109(44):18120-18125

56.

Lazaro MT, Taxidis J, Shuman T, Bachmutsky I, Ikrar T, Santos R, et al. Reduced prefrontal synaptic connectivity and disturbed oscillatory population dynamics in the CNTNAP2 model of autism. Cell Rep. 2019; 27(9):2567-2578.E6

57.

Peñagarikano O, Geschwind DH. What does CNTNAP2 reveal about autism spectrum disorder?. Trends Mol Med. 2012; 18(3):156-163

58.

Dawson MS, Gordon-Fleet K, Yan L, Tardos V, He H, Mui K, et al. Sexual dimorphism in the social behaviour of Cntnap2-null mice correlates with disrupted synaptic connectivity and increased microglial activity in the anterior cingulate cortex. Commun Biol. 2023; 6(1):846

59.

Rahman MM, Shu YH, Chow T, Lurmann FW, Yu X, Martinez MP, et al. Prenatal exposure to air pollution and autism spectrum disorder: sensitive windows of exposure and sex differences. Environ Health Perspect. 2022; 130(1):017008-1-017008-9

60.

Volk HE, Lurmann F, Penfold B, Hertz-Picciotto I, McConnell R. Traffic-related air pollution, particulate matter, and autism. JAMA Psychiatry. 2013; 70(1):71-77

61.

Roberts AL, Koenen KC, Lyall K, Ascherio A, Weisskopf MG. Women's posttraumatic stress symptoms and autism spectrum disorder in their children. Res Autism Spectr Disord. 2014; 8(6):608-616

62.

Kinney DK, Munir KM, Crowley DJ, Miller AM. Prenatal stress and risk for autism. Neurosci Biobehav Rev. 2008; 32(8):1519-1532

63.

Smith CJ, Rendina DN, Kingsbury MA, Malacon KE, Nguyen DM, Tran JJ, et al. Microbial modulation via cross-fostering prevents the effects of pervasive environmental stressors on microglia and social behavior, but not the dopamine system. Mol Psychiatry. 2023; 28(6):2549-2562

64.

Gegenhuber B, Wu MV, Bronstein R, Tollkuhn J. Gene regulation by gonadal hormone receptors underlies brain sex differences. Nature. 2022; 606(7912):153-159

65.

Ferri SL, Abel T, Brodkin ES. Sex differences in autism spectrum disorder: a review. Curr Psychiatry Rep. 2018; 20(2):9

66.

Baron-Cohen S, Auyeung B, Nørgaard-Pedersen B, Hougaard DM, Abdallah MW, Melgaard L, et al. Elevated fetal steroidogenic activity in autism. Mol Psychiatry. 2015; 20(3):369-376

67.

Majewska MD, Hill M, Urbanowicz E, Rok-Bujko P, Bieńkowski P, Namyslowska I, et al. Marked elevation of adrenal steroids, especially androgens, in saliva of prepubertal autistic children. Eur Child Adolesc Psychiatry. 2014; 23(6):485-498

68.

Erdogan MA, Bozkurt MF, Erbas O. Effects of prenatal testosterone exposure on the development of autism-like behaviours in offspring of Wistar rats. Int J Dev Neurosci. 2022; 83(2):201-215

69.

Christensen J, Grønborg TK, Sørensen MJ, Schendel D, Parner ET, Pedersen LH, et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013; 309(16):1696-1703

70.

Grgurevic N. Testing the extreme male hypothesis in the valproate mouse model; sex-specific effects on plasma testosterone levels and tyrosine hydroxylase expression in the anteroventral periventricular nucleus, but not on parental behavior. Front Behav Neurosci. 2023; 17:1107226

71.

Diviccaro S, Cioffi L, Falvo E, Giatti S, Melcangi RC. Allopregnanolone: an overview on its synthesis and effects. J Neuroendocrinol. 2021; 34(2)e12996

72.

Lambert JJ, Belelli D, Hill-Venning C, Peters JA. Neurosteroids and GABAA receptor function. Trends Pharmacol Sci. 1995; 16(9):295-303

73.

Pinna G, Uzunova V, Matsumoto K, Puia G, Mienville JM, Costa E, et al. Brain allopregnanolone regulates the potency of the GABAA receptor agonist muscimol. Neuropharmacology. 2000; 39(3):440-448

74.

Chew L, Sun KL, Sun W, Wang Z, Rajadas J, Flores RE, et al. Association of serum allopregnanolone with restricted and repetitive behaviors in adult males with autism. Psychoneuroendocrinology. 2021; 123:105039

75.

Vacher CM, Lacaille H, O'Reilly JJ, Salzbank J, Bakalar D, Sebaoui S, et al. Placental endocrine function shapes cerebellar development and social behavior. Nat Neurosci. 2021; 24(10):1392-1401

76.

Mueller BR, Bale TL. Early prenatal stress impact on coping strategies and learning performance is sex dependent. Physiol Behav. 2007; 91(1):55-65

77.

Mueller BR, Bale TL. Sex-specific programming of offspring emotionality after stress early in pregnancy. J Neurosci. 2008; 28(36):9055-9065

78.

Marsit CJ, Maccani MA, Padbury JF, Lester BM. Placental 11-beta hydroxysteroid dehydrogenase methylation is associated with newborn growth and a measure of neurobehavioral outcome. PLoS ONE. 2012; 7(3)e33794

79.

Howerton CL, Morgan CP, Fischer DB, Bale TL. O-GlcNAc transferase (OGT) as a placental biomarker of maternal stress and reprogramming of CNS gene transcription in development. Proc Natl Acad Sci USA. 2013; 110(13):5169-5174

80.

Nugent BM, O'Donnell CM, Neill Epperson C, Bale TL. Placental H3K27me3 establishes female resilience to prenatal insults. Nat Commun. 2018; 9(1):2555

81.

Estes ML, Kimberley McAllister A. Maternal immune activation: implications for neuropsychiatric disorders. Science. 2016; 353(6301):772-777

82.

Smith SEP, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci. 2007; 27(40):10695-10702

83.

Choi GB, Yim YS, Wong H, Kim S, Kim H, Kim SV, et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science. 2016; 351(6276):933-939

84.

Shin Yim Y, Park A, Berrios J, Lafourcade M, Pascual LM, Soares N, et al. Reversing behavioural abnormalities in mice exposed to maternal inflammation. Nature. 2017; 549(7673):482-487

85.

Kalish BT, Kim E, Finander B, Duffy EE, Kim H, Gilman CK, et al. Maternal immune activation in mice disrupts proteostasis in the fetal brain. Nat Neurosci. 2021; 24(2):204-213

86.

Pakos‐Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, Gorman AM. The integrated stress response. EMBO Rep. 2016; 17(10):1374-1395

87.

Kelleher RJ, Bear MF. The autistic neuron: troubled translation?. Cell. 2008; 135(3):401-406

88.

Torossian A, Saré RM, Loutaev I, Smith CB. Increased rates of cerebral protein synthesis in Shank3 knockout mice: implications for a link between synaptic protein deficit and dysregulated protein synthesis in autism spectrum disorder/intellectual disability. Neurobiol Dis. 2021; 148:105213

89.

Wood H. Integrated stress response mediates cognitive decline in Down syndrome. Nat Rev Neurol. 2020; 16(1):3

90.

Dudova I, Kasparova M, Markova D, Zemankova J, Beranova S, Urbanek T, et al. Screening for autism in preterm children with extremely low and very low birth weight. Neuropsychiatr Dis Treat. 2014; 10:277-282

91.

Guy A, Seaton SE, Boyle EM, Draper ES, Field DJ, Manktelow BN, et al. Infants born late/moderately preterm are at increased risk for a positive autism screen at 2 years of age. J Pediatr. 2015; 166(2):269-275.E3

92.

Kuzniewicz MW, Wi S, Qian Y, Walsh EM, Armstrong MA, Croen LA. Prevalence and neonatal factors associated with autism spectrum disorders in preterm infants. J Pediatr. 2014; 164(1):20-25

93.

Limperopoulos C, Bassan H, Sullivan NR, Soul JS, Robertson RL, Moore M, et al. Positive screening for autism in ex-preterm infants: prevalence and risk factors. Pediatrics. 2008; 121(4):758-765

94.

Jain VG, Willis KA, Jobe A, Ambalavanan N. Chorioamnionitis and neonatal outcomes. Pediatr Res. 2022; 91(2):289-296

95.

Horvath B, Lakatos F, Tóth C, Bödecs T, Bódis J. Silent chorioamnionitis and associated pregnancy outcomes: a review of clinical data gathered over a 16-year period. J Perinat Med. 2014; 42(4):441-447

96.

Larsen JW, Sever JL. Group B Streptococcus and pregnancy: a review. Am J Obstet Gynecol. 2008; 198(4):440-450

97.

Nasef N, Shabaan AE, Schurr P, Iaboni D, Choudhury J, Church P, et al. Effect of clinical and histological chorioamnionitis on the outcome of preterm infants. Am J Perinatol. 2013; 30(01):059-068

98.

Allard MJ, Bergeron JD, Baharnoori M, Srivastava LK, Fortier LC, Poyart C, et al. A sexually dichotomous, autistic-like phenotype is induced by group B Streptococcus maternofetal immune activation. Autism Res. 2016; 10(2):233-245

99.

Braun AE, Carpentier PA, Babineau BA, Narayan AR, Kielhold ML, Moon HM, et al. "Females are not just 'Protected' males": sex-specific vulnerabilities in placenta and brain after prenatal immune disruption. eNeuro. 2019; 6(6):ENEURO.0358-19.2019

100.

Carlezon WA, Kim W, Missig G, Finger BC, Landino SM, Alexander AJ, et al. Maternal and early postnatal immune activation produce sex-specific effects on autism-like behaviors and neuroimmune function in mice. Sci Rep. 2019; 9(1):16928

101.

Werling DM, Pochareddy S, Choi J, An JY, Sheppard B, Peng M, et al. Whole-genome and RNA sequencing reveal variation and transcriptomic coordination in the developing human prefrontal cortex. Cell Rep. 2020; 31(1):107489

102.

Kim S, Lee J, Koh IG, Ji J, Kim HJ, Kim E, et al. An integrative single-cell atlas to explore the cellular and temporal specificity of neurological disorder genes during human brain development [Internet]. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory. c2024[cited 2024 Apr 11]Available from: https://www.biorxiv.org/content/10.1101/2024.04.09.588220v1

103.

Chung C, Yang X, Bae T, Vong KI, Mittal S, Donkels C, et al. Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development. Nat Genet. 2023; 55(2):209-220

104.

Lee T, Lee H, Kim S, Park KJ, An JY, Kim HW. Brief report: risk variants could inform early neurodevelopmental outcome in children with developmental disabilities. J Autism Dev Disord. 2022 Sep 7; [Epub]