The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Influential PublicationFull Access

Innate Immune Dysfunction and Neuroinflammation in Autism Spectrum Disorder (ASD)

Abstract

Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation.

Appeared originally in Brain Behav Immun 2023; 108:245–254

Introduction

Typically diagnosed in early childhood, autism spectrum disorder (ASD) is a complex heterogeneous developmental disorder involving the early onset of behavioral abnormalities, social impairments, and communication deficits. ASD can present at various levels of severity from mild to severe and has a male preponderance, with as many as four males diagnosed per female; however, this may be changing due to our shifting understanding of the differences between male and female presentation of ASD. The prevalence of ASD has risen substantially over the past several decades to 1 in 44 children (Maenner, 2021). While the etiology of ASD remains unknown, several risk factors are associated with the development of ASD, including genetic and environmental risk factors.

Twin studies support a strong role of genetics, with differences in concordance rates among monozygotic versus dizygotic twins pointing to a heritable component of ASD. However, concordance rates from twin studies often vary, and shared environmental factors may be contributing to susceptibility (Hallmayer et al., 2011; Castelbaum et al., 2020; Pugsley et al., 2021). Many genes identified as contributing to ASD in large-scale genetic studies are common variants that carry low risk individually; however, additively, these variants increase the risk of ASD (Gaugler, 2014). Several high-confidence ASD variants converge onto inflammatory pathways, including the MTOR and PTEN genes. Rare inherited or de novo variants have also been identified and carry a substantially higher risk; however, these can only account for a small percentage of ASD cases (Iossifov, 2014) as do monogenic disorders such as Fragile X syndrome (FXS). Therefore, no single gene or common set of genes has been identified in the majority of ASD cases, and recent studies have identified environmental exposures implicated in ASD risk. These exposures may be exacerbated by gene-by-environment interactions and epigenetic mechanisms (Pugsley, 2021; Tordjman, 2014).

Several environmental risk factors have been clinically identified, with the majority occurring during gestation. Maternal autoimmunity confers a significantly increased risk for neurodevelopmental disorders (NDD) in offspring, including ASD (Chen et al., 2016). Maternal obesity, diabetes, and immune-mediated diseases such as asthma, also substantially increase the risk of NDD, as do exposure to toxicants, pesticides, and air pollution. These disorders and exposures share a commonality of immune activation and increased inflammation (Han, 2021). Notably, inflammation during gestation has been identified as a significant risk factor for ASD and other neurological disorders, although a recent population study in Sweden only identified an association, not a causal link, of maternal infection with ASD (Atladottir, 2010; Brown, 2014; Brynge, 2022). In support of these findings, animal models of maternal immune activation (MIA) with various immune initiators have revealed ASD-relevant behaviors and have provided evidence of innate immune activation in MIA offspring, suggesting that early exposure to maternal inflammation may be inappropriately priming the fetal immune response and may lead to future dysfunction of this arm of the immune system (Meyer, 2014; Patterson, 2011; Careaga et al., 2017).

The network of cellular and chemical components that make up the immune system confers protection from invaders through an intricate system designed to identify “self” from “other.” Sentinel cells of the innate immune system include neutrophils, monocytes, and their tissue counterparts—macrophages. They are activated when broadly recognizing pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) recognize conserved pathogen-associated molecular patterns (PAMPs) (Mogensen, 2009). Activation through these receptors results in a cascade of inflammatory responses that lead to removal of the invader and activation of the adaptive arm of the immune system, which later confers specific and prolonged protection to future invasion by the same pathogen. Associated with activation of the innate arm of the immune system, inflammatory cytokines such as interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-6 are acute phase response cytokines produced and released during the early stages of a response to a pathogen. Elevations in the innate cytokines may indicate chronic inflammation or innate immune dysfunction.

Innate Immune Findings in ASD

Inflammatory Cytokines

Cytokines are key chemical mediators of the immune response produced by cells when activated by exposure to an antigen or immunogen. They are fast-acting cell-signaling molecules that require appropriate regulation to maintain a balance between pro- and anti-inflammatory responses. This balance is especially critical during neurodevelopment as cytokines and their receptors are also expressed by neurons, and they are intricately involved in the proper development of the central nervous system (CNS). For example, these chemical messengers play essential roles in regulating synapse formation, maturation and elimination, cell renewal processes, and survival and apoptosis of cells of the CNS. Disrupted cytokine and chemokine expression during critical neurodevelopmental periods show the substantial influence of these signaling molecules on cells within the CNS, including radial glial cells and neural progenitors, neurons, microglia, astrocytes, and oligodendrocytes (reviewed in (Deverman and Patterson, 2009; Zengeler and Lukens, 2021).

Increases in these inflammatory cytokines have been identified repeatedly in ASD, providing evidence of peripheral innate activation and dysregulation compared to typically developing children (Table 1). Early studies in ASD cytokine research identified increases in inflammatory cytokines in whole blood cultures and TLR4-stimulated peripheral blood mononuclear cells (PBMC) (Croonenberghs et al., 2002; Jyonouchi et al., 2001). Activation of TLR4 on innate immune cells, including monocytes, initiates a proinflammatory cascade and production of cytokines including TNF-α, IL-1β, and IL-6. In adults with severe autism, serum IL-1β and IL-6 were significantly higher than healthy controls (Emanuele et al., 2010). In a large cohort of ASD children ages 2–5, significant elevations of IL-1β, IL-6, and IL-12p40 were seen compared to typically developing children. Moreover, these elevated inflammatory cytokines were associated with worsening behaviors (Ashwood et al., 2011a). Similar increases in IL-1β and IL-12 (p70) were also seen in a cohort of high-functioning ASD males, along with increases in IL-1RA and T helper (TH) cell-associated cytokines IL-5, IL-13, and IL-17 (Suzuki, 2011). In a recent study, individuals with ASD had increased inflammatory cytokines in serum, including IL-1β, IL-6, IL-12, IL-23, and TNF-α, providing additional evidence of peripheral innate immune activation (Ricci, 2013). We recently characterized a cohort of ASD children based on PBMC responses to lipopolysaccharide (LPS) stimulation and found those with higher innate inflammatory responses or increased T cell activation had more behavioral impairments (Careaga, 2017). A subset (endophenotype) of children with ASD that exhibited increased innate immune response and severe behavioral and cognitive problems emerged, accounting for at least a third of the subjects tested. When considering the balance of pro- and anti-inflammatory mediators, the regulatory cytokine TGF-β is often decreased in ASD and associated with worsening behaviors (Ashwood et al., 2008; Okada, 2007; Hashim et al., 2013). This cytokine is critical for dampening the inflammatory responses of monocytes and macrophages during immune response resolution, and deficiency may contribute to prolonged or dysfunctional activation of these cells.

TABLE 1. Altered cytokines and chemokines in ASDa

Author/YearSubjects (M/F)MethodsInnate immune findings in ASD
Jyonouchi et al. 200171 ASD (56/15) 23 SIB (16/7) 17 TD (7/ 10) 1–16 yrsPBMC culture supernatant ELISA↑ TNF-α, IL-1β, and/or IL-6 after activation of TLR4 in some ASD, high variability among subjects
Croonenberghs 200213 ASD (13/0) 13 TD (13/0) 12–18 yrsSerum and whole blood culture supernatant ELISA↑ IFN-γ and IL-1RA in whole blood cultures suggest ↑ activation of peripheral monocytes
Okada 200719 ASD (19/0) 21 TD (21/0) 18–28 yrsSerum ELISA↓ TGF-β1
Ashwood et al. 200875 ASD (68/7) 36 TD (24/12) 32 DD (28/4) Median age 3–4 yrsPlasma ELISA↓ TGF-β1, associated with worse behavioral scores
Ashwood et al. 2011a97 ASD (84/13) 87 TD (71/16) 39 DD (28/11) 2–5 yrsPlasma multi-plex assay↑ IL-1β, IL-6, IL-8, IL-12p40, increased cytokines were associated with worse behavioral scores
Suzuki 201128 ASD (28/0) 28 TD (28/0) 7–15 yrsPlasma multi-plex assay↑ IL-1β, IL-12(p70), GRO-a (CXCL1) (Also saw ↑ IL-1RA, IL-5, IL-8, IL-13, IL-17)
Abdallah et al. 2012a359 ASD (291/68) 741 CTL (595/146)Dried neonate blood spot multi-plex assay↓ RANTES, no differences in chemokines after adjustment (measured MCP-1, MIP-1α and RANTES)
Manzardo et al. 201299 ASD (74/25) 40 SIB (28/12) 5–10 yrsPlasma multi-plex assay↓ cytokines/chemokines
Ricci et al. 201329 ASD (27/2) 29 TD (27/2) 2–21 yrsSerum ELISA↑ IL-1β, IL-6, IL-23, IL-12 and TNF-α
Hashim et al. 201350 ASD (27/23) 50 TD (30/20) 50 DD (28/22) 6–12 yrsSerum ELISA↓ TGF-β1 and IL-23. Levels of TGF-β1 and IL-23 negatively correlated with severity of autism
Zerbo et al. 201484 ASD (73/11) 159 GP (139/20) 49 DD (29/20)Dried neonate blood spot multi-plex assay↑ MCP-1, ↓ RANTES
Mostafa and Al Ayadhi 201562 ASD (48/14) 62 TD (47/15) 4–12 yrsSerum ELISA↑ ENA-78 (CXCL5)
Masi 201517 studiesSystematic review and meta-analysis↑ IL-1β, IL-6, IFN-γ, eotaxin, IL-8 and MCP-1. ↓ TGF-β1
Shen 201642 ASD (38/4) 35 TD (19/16) 3–6 yrsPlasma multi-plex assay↑ RANTES, MIP-1α, and MIP-1β, ↓ IP-10 and MIG Levels of MIP-1α, MIP-1β and IP-10 associated with impairments in social behaviors
Saghazadeh et al. 201938 studiesSystematic review and meta-analysis↑ IL-1β, IL-6, TNF-α and IFN-γ

aASD = autism spectrum disorder; SIB: non-ASD sibling; TD = typically developing; PBMC = peripheral blood mononuclear cells; TLR = Toll-like receptor; DD = developmental disability/delay; CTL – control; GP = general population control.

TABLE 1. Altered cytokines and chemokines in ASDa

Enlarge table

Chemokines are cytokines that mediate chemotaxis and recruitment of immune cells to sites of inflammation. Several chemokines associated with innate immune activation are elevated in ASD. Increases in monocyte chemoattractant protein (MCP)-1, regulated on activation, normal T cell expressed and secreted (RANTES), eotaxin, IL-8, and C-X-C motif chemokine 1 (CXCL1, previously known as Gro-α) have been identified in children with ASD (Ashwood et al., 2011a; Suzuki, 2011). Supporting these findings, Shen et al. recently found elevations of RANTES, macrophage inflammatory protein (MIP)-1α, and MIP-1β and decreases in interferon gamma-induced protein (IP)-10 (also known as CXCL10) and monokine induced by gamma interferon (MIG, also known as CXCL9) in children with ASD. Furthermore, MIP-1α, MIP-1β, and IP-10 concentrations were associated with impairments in social behaviors (Shen, 2016). Increases in CXCL5 have also been seen in ASD (Mostafa and Al-Ayadhi, 2015). Additionally, neonatal blood spots from ASD children were found to have increased MCP-1 (Zerbo et al., 2014). Elevated MCP-1 in amniotic fluid was also associated with an increased risk of having a child with ASD (Abdallah et al., 2012b). These data suggest that chemotaxis and migration of monocytes/macrophages may be impacted in autism after diagnosis, at birth, and potentially in the womb during pregnancy.

Although the majority of ASD studies have identified increases in inflammatory cytokines and chemokines, findings are not always consistent. Reduced cytokines and chemokines were found in neonatal blood spots from ASD females, with no differences in ASD compared to typically developing (TD) in adjusted regression model estimates (Abdallah et al., 2012a). In one study utilizing multi-plex analysis of 29 cytokines in plasma, the concentration of eight cytokines/chemokines was consistently lower in ASD. However, this study used siblings of ASD as controls Manzardo, 2012). ASD is a highly heritable disorder, and families may broadly display immune dysfunction across members; therefore, using siblings as controls can be problematic. Other contradictory findings can be due in part to differences in patient populations and mismatched controls, variations in sample type and processing, and different immunological assays and technologies used for research. Systematic reviews and meta-analyses can help identify consistencies and eliminate some of the noise produced by differences in research. One such meta-analysis looked across 17 studies and concluded that there are consistent elevations in IL-1β, IL-6, and IFN-γ found in individuals with ASD. The chemokines eotaxin, IL-8, and MCP-1 are also frequently elevated, and concentrations of the critical regulatory cytokine active- TGF-β1 are decreased in ASD (Masi, 2015). Results from another recent meta-analysis support these findings, with elevations in IL-1β, IL-6, TNF-α, and IFN-γ seen across 38 studies (Saghazadeh, 2019).

Innate Immune Cells

Cytokines seen repeatedly in ASD, such as IL-1β, IL-6, and TNF-α, are produced during activation of the innate arm of the immune system. This arm is responsible for the first line of defense against foreign antigens. When physical or chemical barriers of the host are breached, cells of the innate arm called monocytes act as sentinels, which are recruited to assaulted tissues by chemokines released at sites of infection. Here, these cells join their tissue-resident counterparts to become macrophages. They are critical during the early immune response for producing inflammatory cytokines to initiate an acute phase response in an attempt to control the infection directly (Dey et al., 2015). Monocyte and macrophage mediators produced as a result of nuclear factor kappa light-chain-enhancer of activated B cells (NF-κB) binding to cytokine gene promoters and inflammasome activation influence downstream adaptive responses and later, under optimal conditions, lead to the resolution of inflammation. Conversely, dysfunction in the activity of these cells can lead to excessive and chronic inflammation, inappropriate adaptive immune responses, autoimmunity, and other immune mediated conditions—many of which have been seen in ASD (Hughes et al., 2018). A hallmark of macrophages is their plasticity. These cells skew to a spectrum of phenotypes depending on microenvironment exposures. In vitro, they polarize to a classical inflammatory “M1” phenotype through stimulation with LPS plus IFN-γ or TNF-α. This polarization leads to production of the canonical proinflammatory cytokines IL-1β, IL-6, and TNF-α and helps to drive T helper cell (TH)-1 differentiation. Conversely, macrophages can be alternatively activated to an “M2” phenotype when exposed to IL-4 and IL-13 in vitro, leading to production of IL-10 and suppression of the “M1” phenotype (Italiani and Boraschi, 2014).

Immune research has revealed increased numbers of innate immune circulating cells, including monocytes, in children with ASD (Sweeten et al., 2003; Tural Hesapcioglu, 2019) (Fig. 1). In some studies, monocyte numbers do not differ; however, monocyte activation is dysregulated. For example, Enstrom et al. found altered cytokine production after activation of TLRs on isolated peripheral CD14+ monocytes from ASD children, with increases in the canonical innate inflammatory cytokines TNF-α, IL-1β, and IL- 6 following activation of TLR2. IL-1β was also increased after TLR4 activation with LPS, and concentrations of IL-1β positively correlated with ASD-associated behaviors, including impaired social interactions and non-verbal communication (Enstrom, 2010). Monocytes from ASD children also had increased expression of CD95, which may reflect increased activation (Ashwood et al., 2011b). Whole peripheral blood cultures from ASD children also showed evidence of increased activation of monocytes through elevated production of innate-associated cytokines (Croonenberghs et al., 2002). Isolated PBMC showed elevated gene expression of caspases, including caspase-1 (also known as IL-1 converting enzyme). Caspase-1 is a proteolytic cleavage enzyme with critical involvement in inflammasome activity, leading to activation of IL-1β (Siniscalco et al. 2012).

FIGURE 1.

FIGURE 1. Cellular and cytokine dysregulation of the innate immune system in both the periphery and brain of ASD individuals. Inflammatory cytokines produced mainly by cells of the innate immune system are elevated in the periphery of ASD individuals. Increased frequencies and altered activation of innate cells such as monocytes, macrophages, and dendritic cells are found, contributing to the establishment and perpetuation of inflammation in ASD. Notably, inflammatory cells and cytokines of the innate immune system can have consequences on brain homeostasis. Peripheral cells and cytokines can breach barriers in the brain and upregulate inflammatory gene expression. Inflammatory conditions in the brain may also alter tissue-resident immune cells, as altered microglial activation and cellular density have been documented in several regions of the ASD brain. CD80, cluster of differentiation 80; CD86, cluster of differentiation 86; GM-CSF, granulocyte macrophage colony-stimulating factor; HLA-DR, human leukocyte antigen-DR isotype; IFNγ, interferon-gamma; IL-1β, interleukin-1beta; IL-6, interleukine-6; IL-8, interleukin-8; IL-10, interleukine-10; IL-12p40, interleukin-12subunit40; MCP-1, monocyte chemoattractant protein-1; NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells; TGF-β, transforming growth factor-beta; TNFα, tumor necrosis factor-alphaa

a[Publisher's Note: A color version of the figure, as originally published, appears in the online version of this article at focus.psychiatryonline.org.]

More recently, Jyonouchi et al. grouped ASD based on monocyte production ratios of IL-1β and IL-10 after stimulation of several TLRs (high, moderate, and low ratios). The IL-1β/IL-10 ratio groups differed significantly in monocyte expression of miRNAs and mitochondrial respiration. When probed with β-glucan, monocytes from the high-ratio group responded with increased pro-inflammatory responses (IL-1β and TNF-α), while the low-ratio group responded with increased anti-inflammatory responses (Jyonouchi and Geng, 2019; Jyonouchi et al., 2017; Jyonouchi et al., 2019). Overexpression of the IL-17A receptor on ASD monocytes has recently been identified, along with increased expression of NF-κB and inducible nitric oxide synthase (iNOS), markers of in flammatory activity (Nadeem, 2018). These markers further increased expression after treatment with IL-17A, the ligand for the IL-17A receptor. IL-17 is associated with autoimmune disorders, including rheumatoid arthritis and multiple sclerosis, and IL-17A was recently identified to have substantial involvement in the MIA rodent model of ASD (Choi et al., 2016). The transcription factor NF-κB plays a significant role in mediating innate inflammatory immune responses (Liu, 2017). Evidence supports increased NF-κB transcriptional activity, as elevated NF κB mRNA and protein are observed in ASD peripheral blood and ASD lymphoblastic cell lines. However, some inconsistencies exist across studies, potentially due to differing methodologies (reviewed in (Liao and Li, 2020). We recently investigated gene expression in TLR activated monocytes and found increased gene expression of NFKB1, a subunit of NF-κB, in TLR-activated monocytes from autistic children. We also found dysregulated gene expression of translational machinery in monocytes from ASD children compared to typically developing controls, which may be contributing to increased inflammatory responses in a subset of these children as a failure to dampen a prolonged immune response through regulation of translation (Hughes, 2022a). We further identified increased IL-6 production in LPS-stimulated monocytes from ASD children. This increase was associated with restricted and repetitive behaviors, supporting the role of excess inflammation associated with innate immune responses in aberrant behavior. Differences in gene expression and cytokine production were only seen after cells were activated; there were no differences at baseline (Hughes, 2022a; Hughes, 2022b). Similarly, Yamauchi et al. found increased gene expression of TNF-α in monocyte-derived macrophages from ASD in dividuals; however, this was only after differentiation to M1- macrophages. Neither baseline monocytes nor differentiated M2- macrophages showed this elevation (Yamauchi et al., 2021).

Aberrant numbers or activation of other innate immune cells are also present in ASD, including significantly increased numbers of peripheral myeloid dendritic cells (mDCs) (Breece et al., 2013) (Fig. 1). Changes in the mDC population may be due to increased differentiation of monocytes into dendritic cells. Further work has shown that peripheral mDCs from ASD children express more surface CD80/CD86 co-stimulatory molecules than TD children (Saad et al., 2017). Differences in DC correlated with amygdala volume, gastrointestinal issues, and impaired behaviors (Breece et al., 2013). In further studies, ASD children with gastrointestinal issues had significantly increased monocyte, eosinophil, and neutrophil infiltration in duodenum, colon, and terminal ileum biopsies (Ashwood et al., 2003; Furlano, 2001). In addition, significantly increased numbers of natural killer (NK) cells are seen in ASD children compared to controls (Ashwood et al., 2011b). Transcriptional profiling of whole blood from children with ASD also revealed increased expression of genes belonging to the NK cell KEGG pathway (Gregg et al., 2008). Later examination confirmed abnormalities in gene expression associated with NK cells, including increased expression of inhibitory killer cell immunoglobulin receptors (KIR) and cytolytic genes. Furthermore, ASD children had increased CD56+ NK cell frequency and dysfunctional activity in isolated NK cells under both resting and stimulative conditions (Enstrom et al., 2009). Function of NK cells was increased under basal conditions, but there was a failure to respond to stimulus in the form of tumor cells.

Decreased soluble adhesion molecules were also seen in ASD plasma, including soluble platelet endothelial cell adhesion molecule-1 (sPE-CAM-1), which plays an important role in mediating innate immune cell adhesion to and migration across the endothelial wall to facilitate recruitment to inflamed tissues (Onore, 2012). In mouse models, decreased sPECAM-1 is associated with decreased vascular integrity and increased blood-brain barrier permeability, and deficient mice are more prone to autoimmunity (Woodfin et al., 2007). See Table 2 for a comprehensive list of aberrant cellular findings associated with innate peripheral immune dysfunction.

TABLE 2. Altered peripheral innate cell numbers and functiona

Author/YearSubjects (M/F)MethodsInnate immune findings in ASD
Sweeten et al. 200331 ASD (27/4) 24 TD (24/4) 2–12 yrsWhole blood flow cytometry, plasma ELISA↑ numbers of monocyte, ↑ plasma neopterin indicative of monocyte/ macrophage activation
Enstrom et al., 200917 ASD (14/3) 16 TD (13/3) 2–5 yrsCD56 + NK cells isolated from PBMC, co-cultured with MHC-devoid K562 cells to assess cytotoxicity via flow cytometry↑ CD56 + NK cell frequency and dysfunctional cytotoxic activity under both resting and stimulation conditions. Increased expression of inhibitory killer cell immunoglobulin receptors (KIR) and cytolytic genes (N for gene expression studies: ASD 35, TD 11)
Enstrom 201017 ASD (14/3) 16 TD (13/3) 2–5 yrsSupernatant from TLR-stimulated monocytes isolated from PBMC—multi-plex assay↑ IL-1β, IL-6, TNF-α after TLR2 activation, ↑ IL-1β after TLR4 activation, ↑ IL-1β, IL-6, GM-CSF and MCP-1 after TLR9 activation
Ashwood et al. 2011b35 HFA (29/6) 35 LFA (29/6) 35 TD (29/6) 4–6 yrsWhole blood immune cell quantification and phenotyping via flow cytometry40 % ↑ in NK cells, increased CCD14+CD95+ monocytes (increases also seen in activated B and T cells)
Siniscalco et al. 201215 ASD 10 TDPBMC RT-PCR, Western blot, immunocytochemistryOver-expression of caspases at gene and protein level in PBMC, suggesting innate activation
Onore 201249 (42/7) ASD 31 (20/11) TD Median age: 2.91, 3.13Plasma adhesion molecules assessed by ELISA↓ sPECAM-1 and sP-selectin sPECAM-levels negatively correlated with repetitive behavior
Jyonouchi et al. 201769 (52/16 [sic])ASD 27 (16/11) TD 2.8–27 yrsmicroRNA sequencing of monocytes isolated from PBMC, supernatant from TLR-stimulated monocytes—ELISA,When grouped based on monocyte responses to TLR stimulation, “high” IL-1β/ IL-10 ratio responders had increased expression of miRNAs associated with immune signaling pathways.
Nadeem et al. 201840 (32/8) ASD 35 (30/5) TD 3–11 yrsFlow cytometry on isolated CD14+ isolated monocytes, real-time PCR, Western blot, ELISA↑ IL-17A receptor, NF-κB and iNOS on ASD monocytes Activation of the IL-17A receptor increased iNOS expression through NF-κB, which was reversed with IL-17RA antibody treatment.
Tural Hesapcioglu et al. 201945 ASD (36/9) 43 TD (33/10) Mean age: 13.5, 11.9Complete blood count↑ monocytes, ↓ lymphocyte-to-monocyte ratio.
Jyonouchi and Geng 2019152 (130/22) ASD 41 (27/17) Non-ASD 1.9–29.6 yrsMonocytes isolated from PBMC and stimulated with TLR-agonists and β-glucan, supernatant measured via ELISA.↑ IL-1β, TNF-α by β-glucan stimulated monocytes in “high” IL-1β/IL-10 ratio producing monocytes from ASD (previously identified after stimulation with TLR-agonists)
Yamauchi et al. 202129 ASD (24/5) 30 TD (25/5) Mean age: 28.0, 27.2CD14+ monocytes isolated from PBMC and cultured to differentiate into M1/M2 mRNA analyzed for gene expression using qRT-PCR.↑ TNF-α and IL-1β mRNA expression in monocyte-derived M1 macrophages ↑ TNF-α M1/M2 ratio.
Hughes 2022a17 AD (13/4) 9 PDDNOS (7/2) 22 TD (18/4) 4–9 yrsRNA seq analysis of mRNA from CD14+ monocytes cultured with TLR-2/4 agonists↑ gene expression associated with inflammatory pathways in AD compared to PDDNOS and TD.AD and PDDNOS monocytes also lacked the signficant downregulation of genes involved in translation seen in TD monocytes 24 h after stimulation.
Hughes et al., 2022b25 ASD (19/6) 20 TD (16/4) 4–9 yrsCD14+ monocytes isolated from PBMC and cultured with TLR-2/4 agonists Multi-plex analysis of supernatant↑ IL-6 after TLR4 activation IL-6 concentrations associated with worse scores on the Repetitive Behavior Scale.

aASD = autism spectrum disorder; TD = typically developing; NK cells = Natural Killer cells; PBMC = peripheral blood mononuclear cells; MHC = major histocompatibility complex; TLR = Toll-like receptor; NF‐κB = nuclear factor kappa B; iNOS = inducible nitric oxide synthase; AD = autistic disorder; PDDNOS = pervasive development disorder not otherwise specified.

TABLE 2. Altered peripheral innate cell numbers and functiona

Enlarge table

Neuroinflammation and Innate Immune Cells in the Brain

Microglia are specialized macrophages that reside in the brain and CNS. They play a critical role not only in neurodevelopment but also in CNS homeostasis throughout life. During neurodevelopment, microglia are responsible for phagocytosing excess neuronal precursor cells to regulate neurogenesis by restraining cell production (Cunningham et al., 2013). They also play a role in supporting neuronal survival while also limiting axon outgrowth (Li and Barres, 2018). Production of brain-derived neurotrophic factor (BDNF) by microglia promotes survival and differentiation of neurons, regulation of synaptic formation and transmission, and synaptic plasticity critical for memory and learning (Miranda, 2019; Parkhurst et al., 2013). Similar to peripheral monocytes, they are responsible for surveilling their local environment and can become activated when exposed to a foreign antigen or under inflammatory conditions following infection or injury. Under these conditions, microglia release pro-inflammatory cytokines and can experience aberrant activation, including excessive proliferation. If occurring during developmentally-delicate periods, prolonged inflammatory activity of microglia can alter neurodevelopment (Li and Barres, 2018). It was originally proposed that these cells polarized in a similar fashion to peripheral macrophages. However, more recent evidence suggests that microglia are not fully committed to one phenotype or activation state (Ransohoff, 2016).

Investigations of postmortem brain tissue from ASD individuals have revealed evidence of neuroinflammation in the brain, including microglia activation (detailed in Table 3) (Fig. 1). One of the first studies to identify the presence of neuroinflammation examined tissues from 11 ASD subjects and found regional neuronal loss and evidence of neuro inflammation in the cerebral cortex, white matter, and cerebellum. Increased staining of GFAP and HLA-DR in multiple brain regions suggested marked activation of microglia and astroglia. Increased MCP-1 was also seen in cerebrospinal fluid (CSF) (Vargas et al., 2005). A later study found significantly elevated cytokines associated with innate activation, including TNF-α, IL-6, and GM-CSF, and increased IFN-γ and IL-8 in postmortem tissue from the frontal cortex of 8 ASD subjects (ASD subject). Microglia were then characterized using immunostaining to identify differences in activation phenotypes; ASD tissues showed evidence of increased microglial activation, including amoeboid presentation. Additionally, density and somal volume of microglia were increased in ASD. These researchers also found no evidence of acute inflammation, suggesting microglia have long-standing activation (Morgan et al., 2010). Furthermore, the same group later found increased spatial proximity of microglia to neurons in the dorsolateral prefrontal cortex of ASD brain, with microglia processes frequently encircling the somal bodies of neurons in ASD samples (Morgan et al., 2012). Increased density of microglia has also been identified in two separate regions of cortex in the ASD brain compared to controls (Tetreault et al., 2012). NF-κB is increased in the postmortem ASD brain, with localization to glial and neuronal cells (Young et al., 2011), and an enzyme complex that activates NF κB is also elevated in the ASD brain (Malik et al., 2011). More recently, it was shown that cells of the adaptive arm of the immune system are increased in postmortem ASD brain and seen accumulating near blood vessels, further supporting evidence of neuroinflammation in ASD (DiStasio et al., 2019).

TABLE 3. Innate immune findings in postmortem ASD Braina

Author/YearSubjects (M/F)MethodsMain findings
Vargas et al., 2005Post-mortem brain: 15* ASD (12/3) 12* Non-ASD CTL (9/3) 5–50 yrs CSF: 6 ASD (4/2) 9 Non-ASD CTL (3/6) 2–45 yrs *n differed depending on fresh frozen vs fixed samplesICC staining on fixed brain samples from MFG and ACG from cortex, and GCL and white matter from cerebellum. Cytokine protein array and ELISA on homogenized samples from fresh frozen MFG, ACG, and cerebellum. Cytokine protein array on CSF.Loss of neurons in the Purkinje and GCL. ↑ staining of GFAP and HLA-DR in various regions including near Purkinje cells and GCL in cerebellum, indicating activation of astroglia and microglia. ↑ perivascular macrophages and monocytes in 4 of 10 ASD brains. Complement membrane attack complexes found in Purkinje cells, with accumulation of suspected microglia nearby.↑ IL-6 (31.4 fold) in the ACG, other cytokines/chemokines increased depending on region. Multiple cytokines/chemokines ↑ including MCP-1 in CSF
Li et al. 20098 ASD (5/3) 8 TD (5/3) 4–37 yrsMultiplex analysis of cerebral cortex homogenized extracts↑ TNFα, IL-6, IL-8, GM-CSF ↑ IFN-γ; ↑ IFN-γ:IL-10 ratio
Morgan et al., 201013 ASD (13/0) 9 TD (9/0) 3–44 yrsIHC staining of formalin-fixed dorsolateral prefrontal cortexEvidence of Iba-1+ microglia activation including amoeboid morphology, ↑ microglia somal volume in white matter (trend in gray matter), and ↑ microglia density in gray matter. Lack of Iba-1 colocalization with IL-1R1 suggests long-standing rather than acute inflammation.
Wei et al. 20116 ASD (4/2) 6 TD (4/2) 4–14 yrsIHC staining of formalin-fixed cerebellum CGC derived from C57BL/6J neonatal pups, cells were transfected with IL-6 GFP Adenovirus or GFP Adenovirus for control↑ IL-6 in cerebellum of ASD brain IL-6 stimulated formation of excitatory neurons and altered adhesion and migration of cerebellar granule cells in mice
Morgan et al. 201213 ASD (13/0) 9 TD (9/0) 3–44 yrsIHC staining and hematoxylin/eosin counter-staning of formalin-fixed dorsolateral prefrontal cortex tissue slices↑ spatial proximity of Iba-1+ microglia to neurons ↑ contact of microglia with neurons, including microglial processes encircling neuronal somal bodies
Tetreault et al. 201211 ASD (9/2) 12 TD (11/1) 2–23 yrsICC staining of formaldehyde-fixed fronto-insular (FI) and visual cortex (VC)↑ Iba-1+ microglia density in both regions of cortex examined
Young et al. 20119 ASD** (5/4) 9 TD (5/4) 5–79 yrs **Rett syndrome n=1IHC and WB of formalin-fixed orbitofrontal cortex or tissue microarray, depending on source↑ NF-κB in astrocytes and microglia ↑ CD11b+ and CD11c+ microglia with signficantly increased p65 nuclear translocation in ASD cells compared to TD.
Malik et al. 20117 ASD (5/2) 7 TD (4/3) 4–14 yrsWB and ELISA on fresh frozen cerebellum and frontal cortex homogenates. IHC on formalin-fixed cerebellar cortex.↑ IKKα expression in ASD cerebellum. IKKα phosphorylates the inhibitory subunit IκBα of NF-κB. No increases in protein expression or phosphorylation of IκBα and NF-κB p65 were seen in ASD brain.

aASD = autism spectrum disorder; CTL = control; CSF = cerebrospinal fluid; ICC = immunocytochemistry; MFG = middle frontal gyrus; ACG = anterior cingulate gyrus; GCL = granular cell layer; TD = typically developing; CGC = cerebellar granular cells; GFP = green fluorescent protein; IHC = immunohistochemistry.

TABLE 3. Innate immune findings in postmortem ASD Braina

Enlarge table

While the postmortem evaluation of ASD brain tissue has consistently shown neuroinflammation and microglial activation, ASD studies using positron emission tomography (PET) to study microglia ‐ in “real-time”- have provided mixed results. To date, PET evaluating microglia activation in ASD has relied on the radiolabeling of ligands that bind to the mitochondrial translocator protein (TSPO), a protein expressed on the outer mitochondrial membrane by activated microglia (Cosenza Nashat et al., 2009). The first study to evaluate microglial activity in this fashion used the first generation [11C]-PK11195 radiotracer to measure TSPO expression in 20 high-functioning male ASD and typically developing control subjects (Suzuki et al., 2013). Using the binding potential as the output PET parameter, [11C]-PC11195 binding was higher in the cerebellum, frontal, temporal parietal, limbic and subcortical regions. Higher radiotracer binding was interpreted as higher TSPO expression and activated microglia phenotypes in high functioning ASD subjects. However, recent studies do not corroborate increased TSPO expression. In one study evaluating the binding potential of the second generation radiotracer [11C]-PBR28 in 15 ASD males, TSPO expression was seen to be lower in several brain regions (Zürcher et al., 2021). Additionally, TSPO expression did not differ in low or high-functioning ASD. In another study using the total distribution volume as the primary PET output parameter and the second generation [18F]-FEPPA radiotracer, TSPO expression was also lower in both males and females with ASD (Simpson et al., 2022). Mixed findings between studies may be due to several rea sons. The radiotracer used since the first-generation [11C]-PK11195 has a high non-specific binding affinity. The PET output parameter (binding potential vs total distribution volume) can influence findings, as previously documented in a meta-analysis of patients with psychiatric disorders (Marques et al., 2019). Additionally, the measurement of TSPO for microglial activation may also introduce variability. While TSPO is expressed by activated microglia, endothelial cells during homeostasis and astrocytes in a disease context can also express TSPO (Cosenza Nashat et al., 2009). Lastly, Suzuki and colleagues did not identify if their research subjects had TSPO gene polymorphisms, as polymorphisms can influence radiotracer binding. Additional PET studies using microglial-specific proteins, correctly identifying subjects with high and low TSPO binding affinity, and analyzing both the binding affinity and total volume distribution will aid in clarifying the abovementioned findings.

Transcriptomic analyses of various brain regions support findings of increased immune responses in the ASD brain, with enrichment in immune-specific genes seen in several studies (Table 4). Increased expression of immune system-related transcripts was first seen in the temporal cortex of postmortem ASD brains. Pathway analysis resulted in 31 BioCarta gene sets that differed from controls, of which 19 involved immune pathways. Several of these belonged to innate inflammatory pathways, including NFKB, IL1R1, and IL6 (Garbett, 2008). Voineagu et al. found dysregulated genes in the ASD brain, with decreased expression of neuronal genes involved in synaptic function and upregulated genes associated with inflammatory responses and microglial activation. Using weighted-gene co-expression network analysis (WGCNA), they further identified two network modules associated with ASD, with the first involving under-expressed neuronal genes and overlapping with several autism susceptibility genes. The second module associated with ASD was enriched in overexpressed immune genes, and later work by this same group supported these findings (Voineagu et al., 2011; Parikshak et al., 2016). Similar results were also seen in a later study across several neuropsychiatric disorders, including ASD, schizophrenia, and bipolar disorder (Gandal, 2018). One study looked for enrichment of the immune module in a larger ASD data set and specifically identified M2 activation of microglia and type I interferon responses in the ASD brain. The authors argued that the increased expression of genes associated with M2 activation could be an effort to mediate M1 inflammatory skewing that occurs during type I IFN responses (Gupta et al., 2014). Meta-analysis of previous ASD cortex RNA-Seq studies utilizing data sets from GEO (Gene Expression Omnibus) unearthed newly identified differentially expressed genes in the ASD brain, including TNF signaling and complement cascade pathways (Rahman et al., 2020). Methylation studies have also reported hypomethylation of genes associated with the immune response, leading to increased expression of inflammatory genes (Nardone et al., 2014). Taken together, these studies provide evidence that neuroinflammation may play a role in ASD. However, sample sizes for postmortem ASD studies are limited and more research is needed in this area to elucidate potential mechanisms of neuroinflammation that contribute to ASD and other NDD.

TABLE 4. Brain transcriptome immune findingsa

Author/YearSubjects (M/F)MethodsMain findings
Garbett 2008Frozen samples of superior temporal gyrus from 6 ASD and 6 TDGeneChip microarrays on RNA isolated from frozen samples of superior temporal gyrus152 differentially expressed genes in ASD brain (↑ 130, ↓ 22). 19 BioCarta pathways involving immune function were altered in ASD including NFKB, IL1R, TOLL, INFLAM and IL6. Authors suggest an expression pattern associated with autoimmune rather than acute or non-specific immune activation
Voineagu et al. 2011Frontal and temporal cortex, and cerebellum from 19 ASD and 17 TD. Further analysis (after filtering microarray data for quality) on 58 cortex samples from 29 ASD, 29 TD; and 21 cerebellum samples from 11 ASD, 10 TD.Illumina microarrays on frontal and temporal cortex, cerebellum.444 DEGs in cortex, but only 2 DEGs in cerebellum. Of the DEGs in the cortex, GO enrichment analysis showed that ASD had 209 downregulated genes associated with synaptic function, and 235 upregulated genes involved in immune/inflammatory responses
Gupta et al. 2014Cortex samples from 32 ASD and 40 TDRNA Seq for differential gene expressionEnrichment of M2 microglia markers and Type I interfon signaling in ASD cortex, downregulation of genes associates with neuronal transmission and GABAergic signaling.
Parikshak et al. 2016251 samples of frontal cortex, temporal cortex and cerebellum from 48 ASD and 49 TDRNA Seq for differential gene expression, long non-coding RNA (lcnRNA) expresison, and alternative splicing.Pathway analysis of ASD DEGs in cortex showed decreased expression of genes associated with neuronal pathways, and upregulated genes enriched for microglia and astrocytes. Two brain-enriched lncRNAs, LINC00693 and LINC00689, were upregulated in ASD, while alternative splicing events for neuronal exons were downregulated.
Gandal 2018Data from N=1,695 samples that included ASD, SCZ and BDIntegration of gene expression and genotype data generated from 1695 frontal/temporal cortex samples from ASD, SCZ and BDWGCNA identified four neural-immune modules significantly upregulated depending on disorder: NF-κB signaling upregulated across all three disorders, astrocyte and IFN-response modules upregulated in ASD and SCZ, and microglia module upregulated in ASD but downregulated in SCZ and BD.
Rahman et al. 2020Data from 15 ASD and 15 TD cortex samplesMeta analysis of previous RNA Seq data235 new DEGs not detected in previous studies. GO enrichment analysis identified upregulation of genes involved in inflammatory immune response pathways, including TNF signaling.

aASD = autism spectrum disorder; TD = typically developing; DEGs = differentially expressed genes; GO = Gene Ontology; SCZ = schizophrenia; BD = bipolar disorder; MDD = depression; AAD = alcoholism; WGCNA = weighted gene correlation network analysis.

TABLE 4. Brain transcriptome immune findingsa

Enlarge table

Preclinical Evidence of Innate Immune Dysfunction

In support of clinical findings, preclinical studies relevant to ASD have also identified dysregulation of innate inflammatory responses and activation of macrophages and microglia. For example, in the poly(I:C)-induced MIA model, MIA drove increased inflammatory cytokine production by bone marrow-derived macrophages obtained from offspring (Onore, 2014). In an LPS-based MIA study, MIA offspring exhibited increased peripheral IL-1β and IL-6 throughout growth. Peripheral inflammation increased further after LPS re-exposure at 8 weeks, along with substantial increases in inflammatory cytokines, chemokines, and cell adhesion molecules in the brain (Hsueh et al., 2018). Other studies have revealed that MIA offspring exhibit altered microglia morphology and increased motility of microglia processes (Ikezu et al., 2021; Pratt et al., 2013; Ozaki et al., 2020). In an asthma model of MIA, offspring with increased gene expression and methylation patterns in multiple autism susceptibility genes exhibited increased microglial activation (Vogel Ciernia et al., 2018). A recent study using a two-hit model of MIA plus prenatal hypoxia found infiltrating monocytes and neuroinflammation in the brain. When monocytes were blocked from entering, neuroinflammation was ameliorated (Chen et al., 2020). Supporting the hypothesis that inflammation plays a significant role in aberrant behavior, in the MIA model, increased innate inflammatory cytokines in plasma at adulthood were associated with significantly impaired social behavior compared to those that did not exhibit this inflammatory response (Mueller et al., 2021). Other animal models have also documented associations between innate inflammatory cytokines and impaired behavior. In a non-human primate MIA model, resting and stimulated PBMCs from MIA offspring produced more innate inflammatory cytokines at one and four years post-birth, which was also associated with more stereotyped behaviors (Rose et al., 2017).

Hsaio et al. identified gastrointestinal dysfunction in poly(I:C) offspring, with increased inflammatory IL-6 in the colon coinciding with decreases in tight junction proteins. These offspring also had shifts in gut microbiota and alterations in microbial metabolites produced by the microbiota that were associated with worse behaviors. These behaviors were corrected with the addition of the commensal Bacteroides fragilis, suggesting that MIA-induced changes in the gut microbiota may be contributing to behaviors (Hsiao et al., 2013). The composition of maternal microbiota was also found to be a critical component for pathological outcomes in MIA—if dams lacked a certain commensal bacteria responsible for driving IL-17 responses, the offspring did not exhibit the typical MIA behavioral manifestations (Choi et al., 2016). Altered myeloid cell numbers were also observed in this model. Gut dysfunction is prevalent in ASD, and the microbiota composition are often altered (reviewed in (Hughes et al., 2018). Altered microbiota may be contributing to immune dysfunction, as these commensal microbes produce metabolites including short-chain fatty acids that are critical for gut-immune homeostasis (Parada Venegas et al., 2019), and have been shown to influence the behavior of monocytes and macrophages in culture (Wenzel et al., 2020; Ji et al., 2016).

Conclusion

There is growing evidence that innate immune dysfunction plays a role in ASD. Immune abnormalities involving excess inflammation and activation of innate immune cells are seen consistently across ASD studies. Aberrant inflammatory activation is also reflected in studies of ASD brain, suggesting a role for neuroinflammation. Maternal inflammation is risk factor for ASD, with maternal immune system activation during gestation resulting in ASD-like behavior and altered innate immune function in offspring in animal models. It is well understood that components of immune system, including microglia and the cytokines they produce, play an essential role in neurodevelopment. However, the underlying cause(s) of aberrant neurodevelopment and immune dysfunction in ASD remains elusive. The seemingly chronic immune dysregulation in ASD is likely influenced by mechanisms that govern long-term cellular phenotype and behavior. For instance, epigenetic modifications involved in trained immunity may be skewing myeloid cells towards an activated state, or alterations in the development of hematopoietic stem cells into myeloid cells may be skewed. Future studies will need to take into consideration the heterogeneity of ASD to fully elucidate etiology and the role of the immune system. Some studies have been successful in identifying subgroups of ASD children based on particular innate immune signatures. Further research and the use of unbiased technological approaches will aid in our understanding of how the innate immune system can be used to characterize ASD heterogeneity, ultimately improving potential personalized therapeutic strategies and the quality of life for autistic people and their families.

Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors); The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors).
Corresponding author at: Department of Microbiology and Immunology, M.I.N.D. Institute, 2805, 50th Street, Sacramento, CA 95817, USA. E-mail address: (P. Ashwood).

Acknowledgments: The material is based upon work supported by the National Institute of Child Health and Disease (R01HD090214, HD090214-04S1, and HD090214-04S2), the National Institute of Mental Health (R21MH116383, R01MH118209), Autism Research Institute, Brain Foundation, Jonty Foundation and the UC Davis MIND Institute.

Declaration of Competing Interest: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Data availability: No data was used for the research described in the article.

This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

References

Abdallah, M.W., et al., 2012a. Neonatal levels of cytokines and risk of autism spectrum disorders: an exploratory register-based historic birth cohort study utilizing the Danish Newborn Screening Biobank. J Neuroimmunol 252 (1–2), 75–82. CrossrefGoogle Scholar

Abdallah, M.W., et al., 2012b. Amniotic fluid chemokines and autism spectrum disorders: an exploratory study utilizing a Danish Historic Birth Cohort. Brain Behav Immun 26 (1), 170–176. CrossrefGoogle Scholar

Ashwood, P., et al., 2003. Intestinal Lymphocyte Populations in Children with Regressive Autism: Evidence for Extensive Mucosal Immunopathology. Journal of Clinical Immunology 23 (6), 504–517. CrossrefGoogle Scholar

Ashwood, P., et al., 2008. Decreased transforming growth factor beta1 in autism: a potential link between immune dysregulation and impairment in clinical behavioral outcomes. J Neuroimmunol 204 (1–2), 149–153. CrossrefGoogle Scholar

Ashwood, P., et al., 2011a. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun 25 (1), 40–45. CrossrefGoogle Scholar

Ashwood, P., et al., 2011b. In search of cellular immunophenotypes in the blood of children with autism. PLoS One 6 (5), e19299. CrossrefGoogle Scholar

Atladottir, H.O, et al., 2010. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J Autism Dev Disord 40 (12), 1423–1430. CrossrefGoogle Scholar

Breece, E., et al., 2013. Myeloid dendritic cells frequencies are increased in children with autism spectrum disorder and associated with amygdala volume and repetitive behaviors. Brain, Behavior, and Immunity 31, 69–75. CrossrefGoogle Scholar

Brown, A.S., et al., 2014. Elevated maternal C-reactive protein and autism in a national birth cohort. Molecular Psychiatry 19 (2), 259–264. CrossrefGoogle Scholar

Brynge, M., et al., 2022. Maternal infection during pregnancy and likelihood of autism and intellectual disability in children in Sweden: a negative control and sibling comparison cohort study. The Lancet Psychiatry 9 (10), 782–791. CrossrefGoogle Scholar

Careaga, M., et al., 2017. Immune Endophenotypes in Children With Autism Spectrum Disorder. Biol Psychiatry 81 (5), 434–441. CrossrefGoogle Scholar

Careaga, M., Murai, T., Bauman, M.D., 2017. Maternal Immune Activation and Autism Spectrum Disorder: From Rodents to Nonhuman and Human Primates. Biol Psychiatry 81 (5), 391–401. CrossrefGoogle Scholar

Castelbaum, L., et al., 2020. On the Nature of Monozygotic Twin Concordance and Discordance for Autistic Trait Severity: A Quantitative Analysis. Behavior Genetics 50 (4), 263–272. CrossrefGoogle Scholar

Chen, S‐W., et al., 2016. Maternal autoimmune diseases and the risk of autism spectrum disorders in offspring: A systematic review and meta-analysis. Behavioural Brain Research 296, 61–69. CrossrefGoogle Scholar

Chen, H‐R., et al., 2020. Monocytic Infiltrates Contribute to Autistic-like Behaviors in a Two-Hit Model of Neurodevelopmental Defects. The Journal of Neuroscience 40 (49), 9386. CrossrefGoogle Scholar

Choi, G.B., et al., 2016. The maternal interleukin-17a pathway in mice promotes autism‐like phenotypes in offspring. Science 351 (6276), 933–939. CrossrefGoogle Scholar

Cosenza-Nashat, M., et al., 2009. Expression of the translocator protein of 18 kDa by microglia, macrophages and astrocytes based on immunohistochemical localization in abnormal human brain. Neuropathology and Applied Neurobiology 35 (3), 306–328. CrossrefGoogle Scholar

Croonenberghs, J., et al., 2002. Activation of the Inflammatory Response System in Autism. Neuropsychobiology 45 (1), 1–6. CrossrefGoogle Scholar

Cunningham, C.L., Martinez-Cerdeno, V., Noctor, S.C., 2013. Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J Neurosci 33 (10), 4216–4233. CrossrefGoogle Scholar

Deverman, B.E., Patterson, P.H., 2009. Cytokines and CNS development. Neuron 64 (1), 61–78. CrossrefGoogle Scholar

Dey, A., Allen, J., Hankey-Giblin, P.A., 2015. Ontogeny and Polarization of Macrophages in Inflammation: Blood Monocytes Versus Tissue Macrophages. Frontiers in Immunology 5, 683. CrossrefGoogle Scholar

DiStasio, M.M., et al., 2019. T lymphocytes and cytotoxic astrocyte blebs correlate across autism brains. Ann Neurol 86 (6), 885–898. CrossrefGoogle Scholar

Emanuele, E., et al., 2010. Low-grade endotoxemia in patients with severe autism. Neurosci Lett 471 (3), 162–165. CrossrefGoogle Scholar

Enstrom, A.M., et al., 2009. Altered gene expression and function of peripheral blood natural killer cells in children with autism. Brain Behav Immun 23 (1), 124–133. CrossrefGoogle Scholar

Enstrom, A.M., et al., 2010. Differential monocyte responses to TLR ligands in children with autism spectrum disorders. Brain Behav Immun 24 (1), 64–71. CrossrefGoogle Scholar

Furlano, R.I., et al., 2001. Colonic CD8 and γδ T-cell infiltration with epithelial damage in children with autism. The Journal of Pediatrics 138 (3), 366–372. CrossrefGoogle Scholar

Gandal, M.J., et al., 2018. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science (New York, N.Y.) 362 (6420), 8127. CrossrefGoogle Scholar

Garbett, K., et al., 2008. Immune transcriptome alterations in the temporal cortex of subjects with autism. Neurobiol Dis 30 (3), 303–311. CrossrefGoogle Scholar

Gaugler, T., et al., 2014. Most genetic risk for autism resides with common variation. Nat Genet 46 (8), 881–885. CrossrefGoogle Scholar

Gregg, J.P., et al., 2008. Gene expression changes in children with autism. Genomics 91 (1), 22–29. CrossrefGoogle Scholar

Gupta, S., et al., 2014. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat Commun 5, 5748. CrossrefGoogle Scholar

Hallmayer, J., et al., 2011. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry 68 (11), 1095–1102. CrossrefGoogle Scholar

Han, V.X., et al., 2021. Maternal acute and chronic inflammation in pregnancy is associated with common neurodevelopmental disorders: a systematic review. Translational Psychiatry 11 (1), 71. CrossrefGoogle Scholar

Hashim, H., et al., 2013. Association between plasma levels of transforming growth factor-β1, IL-23 and IL-17 and the severity of autism in Egyptian children. Research in Autism Spectrum Disorders 7 (1), 199–204. CrossrefGoogle Scholar

Hsiao, E.Y., et al., 2013. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155 (7), 1451–1463. CrossrefGoogle Scholar

Hsueh, P.T., et al., 2018. Immune imbalance of global gene expression, and cytokine, chemokine and selectin levels in the brains of offspring with social deficits via maternal immune activation. Genes Brain Behav 17 (7), e12479. CrossrefGoogle Scholar

Hughes, H.K., et al., 2018. Immune Dysfunction and Autoimmunity as Pathological Mechanisms in Autism Spectrum Disorders. Front Cell Neurosci 12, 405. CrossrefGoogle Scholar

Hughes, H.K., et al., 2022a. Dysregulated gene expression associated with inflammatory and translation pathways in activated monocytes from children with autism spectrum disorder. Translational Psychiatry 12 (1), 39. CrossrefGoogle Scholar

Hughes, H.K., et al., 2022b. Increased Monocyte Production of IL-6 after Toll-like Receptor Activation in Children with Autism Spectrum Disorder (ASD) Is Associated with Repetitive and Restricted Behaviors. Brain Sciences 12 (2), 220. CrossrefGoogle Scholar

Hughes, H.K., Rose, D., Ashwood, P., 2018. The Gut Microbiota and Dysbiosis in Autism Spectrum Disorders. Curr Neurol Neurosci Rep 18 (11), 81. CrossrefGoogle Scholar

Ikezu, S., et al., 2021. Inhibition of colony stimulating factor 1 receptor corrects maternal inflammation-induced microglial and synaptic dysfunction and behavioral abnormalities. Molecular Psychiatry 26 (6), 1808–1831. CrossrefGoogle Scholar

Iossifov, I., et al., 2014. The contribution of de novo coding mutations to autism spectrum disorder. Nature 515 (7526), 216–221. CrossrefGoogle Scholar

Italiani, P., Boraschi, D., 2014. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Frontiers in Immunology 5, 514. Google Scholar

Ji, J., et al., 2016. Microbial metabolite butyrate facilitates M2 macrophage polarization and function. Sci Rep 6, 24838. CrossrefGoogle Scholar

Jyonouchi, H., et al., 2017. MicroRNA expression changes in association with changes in interleukin-1ss/interleukin10 ratios produced by monocytes in autism spectrum disorders: their association with neuropsychiatric symptoms and comorbid conditions (observational study). J Neuroinflammation 14 (1), 229. CrossrefGoogle Scholar

Jyonouchi, H., et al., 2019. Variations in Mitochondrial Respiration Differ in IL-1ß/IL-10 Ratio Based Subgroups in Autism Spectrum Disorders. Frontiers in psychiatry 10, 71. CrossrefGoogle Scholar

Jyonouchi, H., Geng, L., 2019. Associations between Monocyte and T Cell Cytokine Profiles in Autism Spectrum Disorders: Effects of Dysregulated Innate Immune Responses on Adaptive Responses to Recall Antigens in a Subset of ASD Children. International journal of molecular sciences 20 (19), 4731. CrossrefGoogle Scholar

Jyonouchi, H., Sun, S., Le, H., 2001. Proinflammatory and regulatory cytokine production associated with innate and adaptive immune responses in children with autism spectrum disorders and developmental regression. Journal of Neuroimmunology 120 (1–2), 170–179. CrossrefGoogle Scholar

Li, X., et al., 2009. Elevated immune response in the brain of autistic patients. J Neuroimmunol 207 (1–2), 111–116. CrossrefGoogle Scholar

Li, Q., Barres, B.A., 2018. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol 18 (4), 225–242. CrossrefGoogle Scholar

Liao, X., Li, Y., 2020. Nuclear Factor Kappa B in Autism Spectrum Disorder: A Systematic Review. Pharmacol Res 159, 104918. CrossrefGoogle Scholar

Liu, T., et al., 2017. NF-κB signaling in inflammation. Signal transduction and targeted therapy 2, 17023. CrossrefGoogle Scholar

Maenner, M.J., et al., 2021. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2018. MMWR Surveill Summ 70 (11), 1–16. CrossrefGoogle Scholar

Malik, M., et al., 2011. NF-κB signaling in the brain of autistic subjects. Mediators of inflammation 2011, 785265. CrossrefGoogle Scholar

Manzardo, A.M., et al., 2012. Plasma cytokine levels in children with autistic disorder and unrelated siblings. Int J Dev Neurosci 30 (2), 121–127. CrossrefGoogle Scholar

Marques, T.R., et al., 2019. Neuroinflammation in schizophrenia: meta-analysis of in vivo microglial imaging studies. Psychological Medicine 49 (13), 2186–2196. CrossrefGoogle Scholar

Masi, A., et al., 2015. Cytokine aberrations in autism spectrum disorder: a systematic review and meta-analysis. Mol Psychiatry 20 (4), 440–446. CrossrefGoogle Scholar

Meyer, U., 2014. Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biol Psychiatry 75 (4), 307–315. CrossrefGoogle Scholar

Miranda, M., et al., 2019. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Frontiers in Cellular Neuroscience 13. CrossrefGoogle Scholar

Mogensen, T.H., 2009. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev 22. CrossrefGoogle Scholar

Morgan, J.T., et al., 2010. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry 68 (4), 368–376. CrossrefGoogle Scholar

Morgan, J.T., et al., 2012. Abnormal microglial–neuronal spatial organization in the dorsolateral prefrontal cortex in autism. Brain Research 1456, 72–81. CrossrefGoogle Scholar

Mostafa, G.A., Al-Ayadhi, L.Y., 2015. The possible link between elevated serum levels of epithelial cell-derived neutrophil-activating peptide-78 (ENA-78/CXCL5) and autoimmunity in autistic children. Behav Brain Funct 11, 11. CrossrefGoogle Scholar

Mueller, F.S., et al., 2021. Behavioral, neuroanatomical, and molecular correlates of resilience and susceptibility to maternal immune activation. Mol Psychiatry 26 (2), 396–410. CrossrefGoogle Scholar

Nadeem, A., et al., 2018. Activation of IL-17 receptor leads to increased oxidative inflammation in peripheral monocytes of autistic children. Brain, Behavior, and Immunity 67, 335–344. CrossrefGoogle Scholar

Nardone, S., et al., 2014. DNA methylation analysis of the autistic brain reveals multiple dysregulated biological pathways. Translational Psychiatry 4 (9), e433. CrossrefGoogle Scholar

Okada, K., et al., 2007. Decreased serum levels of transforming growth factor-beta1 in patients with autism. Prog Neuropsychopharmacol Biol Psychiatry 31 (1), 187–190. CrossrefGoogle Scholar

Onore, C.E., et al., 2012. Levels of soluble platelet endothelial cell adhesion molecule-1 and P-selectin are decreased in children with autism spectrum disorder. Biol Psychiatry 72 (12), 1020–1025. CrossrefGoogle Scholar

Onore, C.E., et al., 2014. Maternal immune activation leads to activated inflammatory macrophages in offspring. Brain Behav Immun 38, 220–226. CrossrefGoogle Scholar

Ozaki, K., et al., 2020. Maternal immune activation induces sustained changes in fetal microglia motility. Scientific Reports 10 (1), 21378. CrossrefGoogle Scholar

Parada Venegas, D., et al., 2019. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Frontiers in immunology 10, 277. CrossrefGoogle Scholar

Parikshak, N.N., et al., 2016. Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism. Nature 540 (7633), 423–427. CrossrefGoogle Scholar

Parkhurst, C.N., et al., 2013. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155 (7), 1596–1609. CrossrefGoogle Scholar

Patterson, P.H., 2011. Maternal infection and immune involvement in autism. Trends Mol Med 17 (7), 389–394. CrossrefGoogle Scholar

Pratt, L., et al., 2013. Maternal inflammation promotes fetal microglial activation and increased cholinergic expression in the fetal basal forebrain: role of interleukin-6. Pediatr Res 74 (4), 393–401. CrossrefGoogle Scholar

Pugsley, K., et al., 2021. Environmental exposures associated with elevated risk for autism spectrum disorder may augment the burden of deleterious de novo mutations among probands. Molecular Psychiatry. Google Scholar

Rahman, M.R., et al., 2020. Comprehensive Analysis of RNA-Seq Gene Expression Profiling of Brain Transcriptomes Reveals Novel Genes, Regulators, and Pathways in Autism Spectrum Disorder. Brain Sci 10 (10). CrossrefGoogle Scholar

Ransohoff, R.M., 2016. A polarizing question: do M1 and M2 microglia exist? Nature Neuroscience 19 (8), 987–991. CrossrefGoogle Scholar

Ricci, S., et al., 2013. Altered cytokine and BDNF levels in autism spectrum disorder. Neurotox Res 24 (4), 491–501. CrossrefGoogle Scholar

Rose, D.R., et al., 2017. Long-term altered immune responses following fetal priming in a non-human primate model of maternal immune activation. Brain Behav Immun 63, 60–70. CrossrefGoogle Scholar

Saad, K., et al., 2017. Frequency of Dendritic Cells and Their Expression of Costimulatory Molecules in Children with Autism Spectrum Disorders. Journal of Autism and Developmental Disorders 47 (9), 2671–2678. CrossrefGoogle Scholar

Saghazadeh, A., et al., 2019. A meta-analysis of pro-inflammatory cytokines in autism spectrum disorders: Effects of age, gender, and latitude. J Psychiatr Res 115, 90–102. CrossrefGoogle Scholar

Shen, Y., et al., 2016. Altered plasma levels of chemokines in autism and their association with social behaviors. Psychiatry Research 244, 300–305. CrossrefGoogle Scholar

Simpson, D., et al., 2022. In vivo imaging translocator protein (TSPO) in autism spectrum disorder. Neuropsychopharmacology 47 (7), 1421–1427. CrossrefGoogle Scholar

Siniscalco, D., et al., 2012. The expression of caspases is enhanced in peripheral blood mononuclear cells of autism spectrum disorder patients. J Autism Dev Disord 42 (7), 1403–1410. CrossrefGoogle Scholar

Suzuki, K., et al., 2011. Plasma cytokine profiles in subjects with high-functioning autism spectrum disorders. PLoS One 6 (5), e20470. CrossrefGoogle Scholar

Suzuki, K., et al., 2013. Microglial Activation in Young Adults With Autism Spectrum Disorder. JAMA Psychiatry 70 (1), 49–58. CrossrefGoogle Scholar

Sweeten, T.L., Posey, D.J., McDougle, C.J., 2003. High Blood Monocyte Counts and Neopterin Levels in Children With Autistic Disorder. American Journal of Psychiatry 160 (9), 1691–1693. CrossrefGoogle Scholar

Tetreault, N.A., et al., 2012. Microglia in the Cerebral Cortex in Autism. Journal of Autism and Developmental Disorders 42 (12), 2569–2584. CrossrefGoogle Scholar

Tordjman, S., et al., 2014. Gene × Environment interactions in autism spectrum disorders: role of epigenetic mechanisms. Front Psychiatry 5, 53. CrossrefGoogle Scholar

Tural Hesapcioglu, S., et al., 2019. High monocyte level and low lymphocyte to monocyte ratio in autism spectrum disorders. International Journal of Developmental Disabilities 65 (2), 73–81. CrossrefGoogle Scholar

Vargas, D.L., et al., 2005. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol 57 (1), 67–81. CrossrefGoogle Scholar

Vogel Ciernia, A., et al., 2018. Microglia from offspring of dams with allergic asthma exhibit epigenomic alterations in genes dysregulated in autism. Glia 66 (3), 505–521. CrossrefGoogle Scholar

Voineagu, I., et al., 2011. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 474 (7351), 380–384. CrossrefGoogle Scholar

Wei, H., et al., 2011. IL-6 is increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and synaptic formation. Journal of Neuroinflammation 8 (1), 52. CrossrefGoogle Scholar

Wenzel, T.J., et al., 2020. Short-chain fatty acids (SCFAs) alone or in combination regulate select immune functions of microglia-like cells. Molecular and Cellular Neuroscience 105, 103493. CrossrefGoogle Scholar

Woodfin, A., Voisin, M.-B., Nourshargh, S., 2007. PECAM-1: A Multi-Functional Molecule in Inflammation and Vascular Biology. Arteriosclerosis, Thrombosis, and Vascular Biology 27 (12), 2514–2523. CrossrefGoogle Scholar

Yamauchi, T., et al., 2021. Tumor necrosis factor-α expression aberration of M1/M2 macrophages in adult high-functioning autism spectrum disorder. Autism Res 14 (11), 2330–2341. CrossrefGoogle Scholar

Young, A.M.H., et al., 2011. Aberrant NF-kappaB expression in autism spectrum condition: a mechanism for neuroinflammation. Frontiers in psychiatry 2, 27. CrossrefGoogle Scholar

Zengeler, K.E., Lukens, J.R., 2021. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nature Reviews Immunology 21 (7), 454–468. CrossrefGoogle Scholar

Zerbo, O., et al., 2014. Neonatal cytokines and chemokines and risk of Autism Spectrum Disorder: the Early Markers for Autism (EMA) study: a case-control study. Journal of Neuroinflammation 11, 113. CrossrefGoogle Scholar

Zürcher, N.R., et al., 2021. [11C]PBR28 MR–PET imaging reveals lower regional brain expression of translocator protein (TSPO) in young adult males with autism spectrum disorder. Molecular Psychiatry 26 (5), 1659–1669. CrossrefGoogle Scholar