Next Article in Journal
Plant Responses to Heat Stress: Physiology, Transcription, Noncoding RNAs, and Epigenetics
Next Article in Special Issue
The Role of Gut Dysbiosis in Acute-on-Chronic Liver Failure
Previous Article in Journal
Therapeutic Potential of the Molecular Chaperone and Matrix Metalloproteinase Inhibitor Clusterin for Dry Eye
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17

by
Yuanpeng Zheng
1,†,
Tessa A. Verhoeff
1,†,
Paula Perez Pardo
1,
Johan Garssen
1,2 and
Aletta D. Kraneveld
1,*
1
Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands
2
Global Centre of Excellence Immunology, Danone Nutricia Research B.V., 3584CT Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(1), 118; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010118
Submission received: 9 November 2020 / Revised: 20 December 2020 / Accepted: 21 December 2020 / Published: 24 December 2020
(This article belongs to the Special Issue Molecular Mechanisms in the Microbiome–Brain–Gut Axis)

Abstract

:
Autism Spectrum Disorder (ASD) is a spectrum of disorders that are characterized by problems in social interaction and repetitive behavior. The disease is thought to develop from changes in brain development at an early age, although the exact mechanisms are not known yet. In addition, a significant number of people with ASD develop problems in the intestinal tract. A Disintegrin And Metalloproteases (ADAMs) include a group of enzymes that are able to cleave membrane-bound proteins. ADAM10 and ADAM17 are two members of this family that are able to cleave protein substrates involved in ASD pathogenesis, such as specific proteins important for synapse formation, axon signaling and neuroinflammation. All these pathological mechanisms are involved in ASD. Besides the brain, ADAM10 and ADAM17 are also highly expressed in the intestines. ADAM10 and ADAM17 have implications in pathways that regulate gut permeability, homeostasis and inflammation. These metalloproteases might be involved in microbiota-gut–brain axis interactions in ASD through the regulation of immune and inflammatory responses in the intestinal tract. In this review, the potential roles of ADAM10 and ADAM17 in the pathology of ASD and as targets for new therapies will be discussed, with a focus on the gut–brain axis.

1. Introduction

Autism Spectrum Disorder (ASD) is a spectrum of neurodevelopmental disorders that are generally diagnosed early in life and might persist across the whole lifespan. It is mainly characterized by a deficiency in social interactions and communication and the presence of specific stereotyped behaviors [1]. The prevalence of ASD is approximately 1.5% in developed countries, as determined in 2012 by the World Health Organization [2], and predominantly affects males [3]. Globally, the incidence of ASD has shown a 35-fold increase (1 in 59 children in the USA and 1 in 89 in the EU are affected) compared to the 60s and 70s (Centers for Disease Control and Prevention Data & Statistics on Autism Spectrum Disorder, 2019 and Autism Spectrum Disorders in the European Union, 2018). A diagnosis is performed using a behavioral assessment due to the absence of reliable biomarkers.
Although the ASD pathogenesis remains elusive, it is thought that it results from early altered brain development and neural reorganization [4,5]. Some clinical aspects are found in large groups of people diagnosed with ASD, such as altered neuronal connectivity, increased synaptic density [6], neuroinflammation [7], microbiota dysbiosis [8,9,10], dysregulated immune responses [11,12,13,14,15] and gastrointestinal abnormalities [16,17,18]. In addition, the most replicated neuroanatomical finding in infants and the early childhoods of people diagnosed with ASD is an enlarged brain volume, called macrocephaly or megalocephaly [19,20]. Genetically, it is estimated that 400–1000 genes are involved in ASD susceptibility, and it is thought that there are more that have and will be discovered in the near future [21]. The estimated heritability of ASD was 83% in a recent population-based cohort of children born in Sweden [22], and Bailey et al. reported a similar result [23]. Among these genetic predispositions, the membrane-bound synaptic genes for the Amyloid Precursor Protein (APP), Neural glial-related Cell Adhesion Molecule (NrCAM), Neuroligins (NLGNs), Neurexins (NRXNs) and Protocadherins (PCDHs) were widely identified as candidate genes of ASD [24,25,26,27,28], as their deficiency in mice led to ASD-like behaviors, such as deficits in spatial memory and learning, increased repetitive and stereotype grooming behaviors and compromised social interactions [29,30,31]. Interestingly, the amount of these ASD-related proteins in the membrane is controlled by the metalloproteases ADAM10 and ADAM17 through proteolytically cleaving these transmembrane proteins by which ADAM10 and ADAM17 might be involved in ASD pathogenesis [32,33,34,35,36].
A Disintegrin And Metalloproteases (ADAMs) are a subgroup of the metzincin family of metalloproteases, which also consists of Matrix Metalloproteases (MMPs), ADAMs with thrombospondin motifs (ADAMTSs) and Snake Venom Metalloproteases (SVMPs) [37,38]. ADAMs are ubiquitously expressed and are able to regulate sperm–egg interactions, cell proliferation, differentiation, migration and cell fate determination [39]. ADAM10 and ADAM17 are the two most investigated members of the ADAM family. Interestingly, both ADAM10 and ADAM17 are highly expressed in the brain, as well as the intestines. In the Central Nervous System (CNS), ADAM10 and ADAM17 are able to regulate axon guidance and synaptic functions through controlling the cleavage of synaptic proteins, such as APP, NrCAM, NLGNs, NRXNs and PCDHs. Importantly, ADAM10 plays a vital role in synaptic pruning by cleaving the chemokine fractalkine (CX3CL1) that binds to its receptor CX3C chemokine receptor 1 (CX3CR1) to induce microglia-mediated synapse elimination [40]. ADAM17 also regulates neuroinflammation, attributing to its capacity of converting membrane-bound Tumor necrosis factor-α (TNF-α) into a soluble form that recognizes the TNF-α receptors I and II and, consequently, triggers inflammatory responses. In the intestinal tract, ADAM17 can regulate intestinal inflammation, intestinal barrier permeability and inflammatory responses by cleaving several cytokines, such as TNF-α and lymphotoxins [41,42,43,44]. Moreover, ADAM10 can control the intestinal permeability by cleaving the transmembrane proteins Notch [45,46,47] and E-cadherin [48]. However, there are only a few reports available that elucidate the roles and functions of ADAM10 and ADAM17 in the gut-to-brain pathology of ASD.
Taken together, ADAM10 and ADAM17 regulate synaptic functions, neuroinflammation and brain development, as well as intestinal barrier functions, inflammation and immunity, which are involved in the pathogenesis of ASD. In this review, we aim to elaborate on the potential role of ADAM10 and ADAM17 in the pathogenesis of ASD with a major focus on the gut–brain axis.

2. Structure of Metalloproteases

The metzincin family of metalloproteases has four family members: ADAMs, MMPs, ADAMTSs and SVMPs [37,38]. They are called metzincins for the conserved Met residue at the active site and the use of a zinc ion in the enzymatic reaction. Collectively, the metalloproteases are able to degrade all extracellular structures. These family members have some corresponding protein domains (Figure 1). All members start with a signal peptide at their N-terminal that allows them to be located at the secretory pathway. Immediately following the signal peptide is a pro-domain. This ensures enzyme latency until cleaved by pro-protein convertases [49]. After the pro-domain, all members of the metzincin family contain a metalloprotease domain, which holds its catalytic activity. After this, some major structure differences are found between members. For ADAMs, SVMPs and ADAMTSs, the metalloprotease domain is followed by a Disintegrin domain and then a cysteine-rich domain. For the ADAMTS, the cysteine-rich domain follows the Thrombospondin region. The ADAMs contain an epidermal growth factor (EGF)-like domain, followed by a transmembrane and cytoplasmic domain. SVMP and ADAMTS are soluble proteins. Membrane-type MMP and MMP contain a Hemapexin domain after their metalloprotease domain, necessary for substrate selectivity and for binding with Tissue Inhibitors of Metalloproteinases (TIMPs), the main MMP inhibitor [37], and the membrane-type matrix metalloproteinase (MT-MMP) contains a transmembrane region with a cytoplasmic tail [37,50,51].
In this review, the focus will be on two types of ADAMs: ADAM10 and ADAM17. The MMPs, MT-MMPs, ADAMTSs and SVMPs are beyond the scope of this review. Little is known about the possible role of MT-MMPs, ADAMTSs and SVMPs in ASD, however, it should be noted that we do not rule out that MMPs, such as MMP9, might also play a role in the pathogenesis of ASD [52,53].

ADAMs

ADAMs are a family of type I transmembrane proteins characterized mainly by their ability to cleave membrane-bound proteins at their extracellular domain. The cleavage generates a soluble protein fraction in the extracellular space, a process called “ectodomain shedding”. This will influence the signaling pathways of other cells by decreasing the amount of membrane-bound receptors or by increasing the amount of soluble ligands [50]. ADAMs are proteins of approximately 750 amino acids that contain several structurally conserved domains, which determine its biological function. The metalloprotease domain can contain a catalytic site with a zinc-binding motif, which is mediated by three histamine residues (HEXGHXXGXXHD) [54]. Around 24 ADAMs have been identified in humans, of which only 12 contain the metalloprotease domain with the active zinc-binding site [55,56]. The biological function of these proteolytically active ADAMs (ADAM8, 9, 10, 12, 15, 17, 19, 20, 21, 28, 30 and 33) is determined by their substrates and includes sperm–egg interactions, cell migration, axon guidance, inflammation and cell fate determination [39]. In addition, ADAMs have been implicated in different pathologies, such as cancer [57], inflammation [58] and Alzheimer’s Disease (AD) [59,60].

3. ADAM10 in the Central and Enteric Nervous Systems

The most extensively studied member of the ADAMs family is ADAM10. There are more than a hundred substrates cleaved by ADAM10 in the CNS [35], and its expression, maturation and substrates selectivity are regulated by the TspanC8 subfamily of tetraspanin, consisting of Tspan5, 10, 14, 15, 17 and 33 [61,62,63], as different Tspan-ADAM10 complexes might adopt different conformations and spaces to their substrates [62,64,65]. ADAM10 is ubiquitously expressed in the brain [66], where it is located at the synapse and in synaptic vesicles and functions as a sheddase of other synaptic proteins [67], which makes ADAM10 able to control CNS processes, such as development, synaptogenesis and axon targeting. In addition, ADAM10 is expressed in the intestinal tract [68]. In Section 5, the role of ADAM10 in the intestines is discussed. Table 1 lists the major ADAM10 substrates in the CNS that will be discussed below.

3.1. Amyloid-β Precursor Protein (APP)

APP is a transmembrane protein involved in cell adhesion and neurite pruning [69,70]. It consists of an extracellular N-terminus domain, a transmembrane region and a C-terminus, intracellular domain [71]. APP can be proteolytically cleaved by a group of secretases: α-, β- and γ-secretases (Figure 2). The β-site APP cleaving enzyme 1 (BACE1) and γ-secretases induce the amyloidogenic pathway, where APP is cleaved extracellularly to create a soluble fraction, sAPPβ, the main component of AD plaques, Amyloid β-peptide (Aβ) and Amyloid Precursor Intracellular Domain (AICD) [72]. α-Secretases activate the nonamyloidogenic pathway by creating the soluble fractions sAPPα, P3 and AICD. ADAM10 is the main α-secretase of APP in the nervous system [32]. ADAM10 cleaves APP in the Aβ domain, which inhibits the formation of the pathological plaques that cause AD and, consequently, creates sAPPα instead. Therefore, the nonamyloidogenic pathway of APP is thought to serve a neuroprotective function at this point [72]. Interestingly, studies show that while ADAM10 is the constitutive secretase, ADAM17 is the stimulatory secretase of APP [32].
Recent studies have started to investigate the roles of ADAM10 and ADAM17 in neurodevelopmental diseases. Of all ADAM10 substrates, APP is the most described in the context of ASD. It has been shown that there is an increase of sAPPα levels in the plasma of children diagnosed with severe ASD aged between 5–17 years [28,73]. In the fragile X mental retardation 1 knockout (Fmr1 KO) mouse model of Fragile X Syndrome, significantly increased levels of both sAPPα and ADAM10 are found at postnatal day 21 [74] but not in adulthood. These findings are in-line with the concept that ASD is a result of early altered brain development, as the prenatal and perinatal period is most critical for synaptogenesis. In parallel with these main findings, Westmark et al. found that genetically decreasing APP and Aβ levels are able to ameliorate the autistic phenotype in adult Fmr1 KO mice [75]. Additionally, Lahiri et al. hypothesized that increased sAPPα levels can activate neuroprotective pathways and microglia, which result in neuronal overgrowth and neuroinflammation, leading to an increased brain volume that is also observed in ASD [19,20,76].
Importantly, the N-terminal of APP (N-APP) is a ligand of Death receptor 6 (DR6), which is highly expressed in oligodendrocytes and neurons. N-APP is a cleavage product of sAPPβ by a still undetermined mechanism, and the specific cleavage site is unknown. The binding of N-APP to DR6 triggers neuronal death via caspase pathways in vitro and in vivo, and consequently, a role for N-APP/DR6 in neurodegeneration has been proposed [70]. Furthermore, DR6 negatively regulates oligodendrocyte survival, maturation and myelination, which is related to microglia activation, phagocytosis and neuroinflammation [77,78,79]. Colombo et al. demonstrated that the DR6 of Schwann cells (SCs) negatively regulates the myelination of the Peripheral Nervous System (PNS) and that DR6 KO mice showed precocious myelination in the PNS [80]. SCs underlie the sheath of most of peripheral nerves and regulate the myelination of the nervus vagus in the PNS [81,82,83]. Vagal stimulation is recognizably involved in ASD development. Sgritta et al. demonstrated that Lactobacillus reuteri (L. reuteri) rescued social behaviors in ASD mice (SH3 and multiple ankyrin repeated domains 3B KO mice) but not in vagotomized mice. These findings indicate that L. reuteri might ameliorate ASD-like behavior in the ventral tegmental area of ASD mice in a vagus nerve-dependent manner [84,85]. In addition, Jin et al. proposed transcutaneous vagus nerve stimulation is a promising treatment for ASD, but the exact mechanism is not clear [86]. Recently, DR6 cleavage was decreased by 50% in ADAM10-deficient murine neurons, and consequently, it is a substrate of ADAM10 [80]. However, the potential role of ADAM10-mediated cleavages of APP and its receptor DR6 in the pathology and treatment of ASD is barely investigated.
Overall, ADAM10 and, possibly, ADAM17 are able to regulate APP shedding and create sAPPα fractions at the expense of sAPPβ. As elevated sAPPα levels are found in the plasma of children with severe ASD behavior, it is probable that ADAM10/17-mediated APP shedding contributes to the development of disturbed brain development in ASD. More studies are needed to elucidate the specific mechanisms of APP shedding in ASD.

3.2. Neuroligins (NLGNs) and Neurexins (NRXNs)

NLGNs are synaptogenic adhesion proteins located at the post-synapse that trans-synaptically binds to the presynaptic partner NRXNs to form a NRXN/NLGN complex, necessary for efficient neurotransmission. These two proteins recruit key synaptic proteins, such as scaffolding proteins and neurotransmitter receptors, after the initial contact of an axon with its target cell. Therefore, they are essential for synaptic formation, maturation and differentiation [87,88,89]. There are five types of NLGNs (NLGN1, NLGN2, NLGN3, NLGN4 and NLGN4Y) and three NRXNs (NRXN1, NRXN2 and NRXN3) in the human genome. They are involved in ASD pathogenesis. Among the NLGNs, NLGN3 is the strongest candidate, followed by NLGN1, and of the NRXNs, NRXN1 is the strongest candidate [90]. Loss-of-function variants of NRXN1 have been found in ASD patients [24,26,27]. Interestingly, variants of the other two types, NRXN2 and NRXN3, are much rarer. Furthermore, NLGN-1 KO mice display deficits in spatial memory and learning and an increased repetitive, stereotypical grooming behavior, which is accompanied by a reduced ratio of NMDA to the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA) at the corticostriatal synapses [29]. NLGN-1 is shed by ADAM10 for 83% and NLGN-3 for 62%, as described in ADAM10-deficient neurons (Table 1) [35]. As the other types of NLGNs are not cleaved by ADAM10, we will not discuss them further. NLGN-1 is found exclusively on glutamatergic neurons, whereas NLGN-3 is found on both glutamatergic and GABAergic synapses [91,92]. The proteolytic cleavage by ADAM10 of membrane-bound NLGN-1 increases by either N-methyl-D-aspartate (NMDA) receptor activation or by binding to the secreted form of NRXNs [92]. Interestingly, the secreted form of NRXN2 and NRXN3 may also be generated by ADAM10 or ADAM17 [33,34]. A recent study discovered that NRXN1 is primarily cleaved by ADAM10 in hippocampal neurons [93].
Only limited reports are available on the role of intestinal NLGN-1, NLGN-3 and NRXN. NLGN-3 is expressed in the enteric nervous system as well, and gastrointestinal dysfunction is found in people and mice with a R451C missense mutation in this NLGN-3 that have an ASD phenotype [94]. Very recently, it was shown that ASD-associated NLGN-3 mutations, as well as NLGN-3 KO mice, have more cecal Nitric Oxide (NO)-producing neurons and more activated enteric macrophages [95]. These phenomena might explain the presence of intestinal symptoms in these NLGN-3-deficienct mice [94,96] and ASD patients [97,98], such as a disturbed intestinal transit and intestinal inflammation. Additionally, NLGN-1 and NRXN are shown to be important for the development of the enteric nervous system in rats [99]. It is rather speculative to link the enhanced intestinal ADAM10/17 expression to the loss of NLGN-1, NLGN-3 or NRXNs and ASD-related intestinal dysfunction.
Taken together, ADAM10 cleaves NLGN-1, NLGN-3 and, possibly, NRXNs. Following this, it can be hypothesized that increased ADAM10 levels or activity will lead to an increased cleavage of NLGN-1, NLGN-3 and NRXNs, therefore terminating their function at the synapse and disrupting the neurotransmissions. Disturbances in neurotransmissions are also described in people diagnosed with ASD. It has been shown that a decreased glutamate concentration in the striatum correlates with the severity of social ASD symptoms, suggesting that glutamate/γ-aminobutyric acid (GABA) abnormalities in the corticostriatal circuitry may contribute to ASD development [100]. Moreover, it was shown that NLGN-1 shedding has a negative effect on NRXN1 stability [101], which also seems to be primarily cleaved by ADAM10 [93]. In conclusion, ADAM10 is a major regulator of synaptic functions of NLGN/NRXN complexes, and the loss of NLGN-1 and NLGN-3 in mouse models induces autistic-like phenotypes [29,102,103], which highlights a potential connection between ASD and ADAM10-mediated cleavages of NLGN/NRXN complexes.

3.3. Protocadherins (PCDHs)

Cadherin is a family of calcium-dependent cell adhesion proteins consisting of three family members: classic cadherins, desmosomal cadherins and PCDHs [104]. PCDHs are mainly expressed in the developing CNS [105]. One member of this family that is an ADAM10 substrate is PCDH9 [35]. The conditional deletion of ADAM10 in primary neuron cultures resulted in a 71% reduced shedding of PCDH9 [35], which has been proposed to play a role in synaptogenesis [106]. It remains unclear what the effect of ADAM10 shedding is on PCDH9 functions. In PCDH9-deficient mice, long-term social and object recognition deficits were determined [30] but without any changes in perception, sociability or fear memory. Furthermore, the PCDH9 KO mice showed impairments in sensorimotor development and structural changes in layers of sensory cortices [30]. The behavioral dysfunctions in PCDH9 KO mice are also presented in ASD. Moreover, Copy Number Variations (CNV), including deletion, duplication, translocation and inversion, of the PCDH9 gene have been found in patients with ASD [24]. Additionally, another member of PCDHs, PCDH8, has also been identified as a substrate of ADAM10 [35] and regarded as a candidate ASD gene in Caucasian females [107]. Furthermore, Breuillard et al. demonstrated, although with a limited sample size, that PCDH19 genetic defects frequently emerged in female ASD children with epilepsy and intellectual disability [108]. Obviously, PCDHs play an important role in ASD pathogenesis, as it seems that ADAM10 is the main sheddase of PCDH8 and PCDH9 [108]. Indeed, the role of ADAM in the loss of function of PCDHs is not yet investigated in ASD. To elucidate this connection further, more research will be necessary.

3.4. Neural Glial-Related Cell Adhesion Molecules (NrCAM)

NrCAM is part of the L1 family of cell adhesion molecules of immunoglobulin superfamily (IgCAMs), and a cell adhesion molecule [109]. NrCAM is involved in brain development, where it controls dendritic spine densities, axon guidance and targeting and neurite outgrowth [110,111,112]. NrCAM was shown to be an ADAM10 substrate, where the deletion of ADAM10 in primary neurons reduces NrCAM shedding by 66% [35]. In addition, Brummer et al. recently showed that ADAM17 deletion in primary neurons did not alter NrCAM proteolysis and that ADAM10 controls NrCAM cell surface expression levels and NrCAM-dependent neurite outgrowth in vitro [36]. Furthermore, mice with a conditional ADAM10 knockout in neurons showed increased cell surface expression levels of NrCAM [35,36] and a reduced number of dendritic spines [113], whereas NrCAM knockout mice show increased dendritic spine densities. As NrCAM is a member of the Sema3F complex that mediates spine retraction [114], increased NrCAM surface levels would be expected to decrease spine density. A potential role for soluble NrCAM in axon signaling becomes apparent in NrCAM-deficient [115] and ADAM10-deficient mice [35]. Both mouse models present with axonal targeting deficits within their olfactory bulbs with axons overshooting their marks. This would suggest a functional role for the soluble fraction of NrCAM generated by ADAM10 cleavage, although more research is needed to obtain more information about this role. Currently, no studies report on the possible peripheral/intestinal role of NrCAM in relation to ASD.
In NrCAM-deficient male mice, the loss of NrCAM leads to ASD-related behavioral alterations in sociability, acquisition of a spatial task and reversal learning [31]. Marui et al. identified that seven single-nucleotide polymorphisms (SNPs) within the NrCAM gene are associated with ASD in Japanese children [25]. Bonora et al. also detected in 48 unrelated individuals with ASD several polymorphisms in the promoter and untranslated region of NrCAM and suggested that a reduction in the expression of this gene might be involved in ASD susceptibility [116]. In contrast, Hutcheson et al. showed that there is no association between ASD susceptibility and the NrCAM gene in the subsets of chromosome 7-linked families [117]. In order to further explore the relation between ASD and NrCAM, and the possible role of the ADAM10-induced cleavage of NrCAM, more studies are needed.

3.5. Fractakine (CX3CL1)

CX3CL1 is the only member of the δ subfamily of chemokines that is constitutively and abundantly expressed in the brain—specifically, in glial cells and neurons [118]. CX3CL1 recognizes its receptor CX3CR1, which is exclusively expressed in the microglia [119,120]. CX3CL1–CX3CR1 signaling is necessary for the immune response, neuroinflammation, synaptic pruning and brain development through maintaining the phagocytic function of the microglia [40,119]. Moreover, CX3CL1 was identified as a substrate of ADAM10 and cleaved by ADAM10 to generate a soluble CX3CL1 that acts as a ligand of CX3CR1 in the brain [35,121,122]. Currently, there is little known about the link between the ADAM10-mediated cleavage of CX3CL1, microglial phagocytosis and ASD. However, it has been shown that CX3CR1 KO mice exhibit a deficiency in microglia engulfment and, consequently, show an increased density of dendritic spines and immature synapses, as well as a synaptic pruning deficiency [119]. Furthermore, there is a defect in synaptic elimination in both CX3CL1 KO mice and CX3CR1 KO mice, which was phenocopied after ADAM10 inhibition in wild-type mice [40]. Insufficient synaptic elimination is a cause of hyperconnectivity in the brain, which is related to the macrocephaly found in ASD patients. Additionally, Rogers et al. demonstrated that CX3CR1 KO mice show impairments in synaptic plasticity and cognitive function [123], which are symptoms of neurodevelopmental diseases, including ASD.
There are limited reports available that discuss the contribution of ADAM10-mediated cleavage of CX3CL1 to the intestinal disturbances found in ASD. Interestingly, the CX3CL1–CX3CR1 levels are critical for the sex differences in high-fat food-induced obesity. For instance, females are much more resistant to diet-induced obesity due to the higher expression levels of CX3CL1 than in males. Strengthening this point, female CX3CR1 KO mice phenocopied “male-like” microglial activation and increased their susceptibility to diet-induced obesity. Conversely, increasing the CX3CL1 levels in the male mice brain converted them to a “female-like” metabolic phenotype with a decrease of microglial activation and weight gain [124]. These sex differences may be involved with gender differences in ASD. However, more research is necessary.
Taken together, the important role that CX3CL1–CX3CR1 signaling plays in the phagocytic function of microglia and, consequently, synaptic pruning emphasizes the potential of this ADAM10 substrate to contribute to ASD pathology. Importantly, more research will be needed to further explore alterations in CX3CL1 expression in people diagnosed with ASD and its contribution to the disease.

4. ADAM17 in the Central Nervous System

ADAM17 is one of most extensively studied member of the ADAMs family and is ubiquitously expressed in all tissues and cell types. Similar to the regulation of ADAM10 by TspanC8, the selectivity or specificity of substrates in ADAM17 is regulated by the iRhoms subfamily, which is one of the rhomboid superfamilies of intramembrane proteases and consists of iRhom 1 and iRhom 2 [125,126]. In addition, iRhoms also regulate ADAM17 enzymatic maturation from its exit from the endoplasmic reticulum to the membrane [125,127]. ADAM17 was first discovered as the enzyme responsible for the proteolytic cleavage of TNF-α, and therefore, ADAM17 was originally called the TNF-α Converting Enzyme [128]. Currently, it is known that ADAM17 is responsible for the shedding of over 90 substrates. Some substrates of ADAM17, such as TNF-α, Tumor necrosis factor-α receptor, Interleukin-6 receptor (IL6-R) and Triggering receptor expressed in myeloid cells-2 (TREM2), are involved in the onset of immune responses and neuroinflammation [129]. The influence of neuroinflammation in ASD has been recently reviewed [7]. Additionally, it was shown that ADAM17 expression levels increased with age in juvenile people diagnosed with ASD [130], although this was reported in relation to the elevation of sAPPα and not with respect to neuroinflammation, inflammation and immune responses. We will discuss the proteolytic cleavages of these substrates, focusing on neuroinflammation and immunity in the CNS and their role in ASD below. More information involving ADAM17 in the intestinal tract will be provided in Section 5.

4.1. Tumor Necrosis Factor-α (TNF-α)

ADAM17 is the main protease of TNFα, a proinflammatory cytokine that can elicit its proinflammatory potential only after being proteolytically released from the cell surface [128,131]. Although deletion of the ADAM17 gene abolishes TNF-α shedding by 90% in ADAM17−/− macrophages and neutrophils, it seems that there are other proteases, amongst others MMP7 and proteinase 3, that are also able of proteolytically cleaving TNF-α [131]. In addition, ADAM10 was identified as a major sheddase when ADAM17 is deficient in fibroblasts, indicating that there is a compensation between ADAM10 and ADAM17 [132]. These reports demonstrated that these two metalloproteases are important in TNF-α cleavage. It was shown that increased levels of TNF-α have been found in the cerebellum and hippocampus in a murine model for ASD induced by Valproic Acid (VPA) [133] and in the brain cortex of people diagnosed with ASD [134].
Together, there is an increase in proinflammatory cytokines in the brains of people diagnosed with ASD, and it seems to be that decreasing the levels of these specific cytokines has a beneficial effect on the disturbed sociability. As ADAM17 has been shown to be the main protease that controls TNF-α shedding from the cell membrane, an interesting connection between ADAM17-mediated TNF-α cleavage, neuroinflammation and ASD remains to be elucidated.

4.2. Interleukin-6 Receptor (IL6-R)

IL6-R is a known substrate of ADAM17. The cytokine IL-6 has both pro- and anti-inflammatory properties, which is determined by the receptor signaling type [129,135]. Signaling via the membrane-bound IL-6R is called classic signaling. This type of signaling can only occur on cell types that express IL-6R on their surface and is thus limited to hepatocytes and several leukocyte subsets and results in an anti-inflammatory response [136,137]. Signaling via soluble forms of the IL-6R, which is called trans-signaling, can occur on all cell types, because the IL-6/soluble IL-6R (sIL-6R) complex can directly bind to and activate the ubiquitously expressed glycoprotein 130 (gp130). The trans-signaling accounts mainly for the proinflammatory properties of IL-6 [137,138]. Interestingly, research in transgenic mice showed that the proteolytic cleavage of IL-6R to create a soluble form is carried out by both ADAM17 and ADAM10 [139] and that approximately 85% of sIL6-R is a result of a proteolytic cleavage in vivo [140]. Increased levels of IL-6 in the brain have been found in a murine model for ASD [133] and in the brain cortex of ASD patients [134]. Moreover, it has been shown that overexpressing IL-6 in the mouse brain mediates neuronal circuitry imbalances and induces ASD-like behavior [141]. Furthermore, one study showed that blocking the trans-signaling pathway of IL-6 led to improved social behavior in a murine ASD model by continuously infusing the IL-6 trans-signaling blocker sgp130Fc protein [142]. As ADAM17 and ADAM10 are capable of proteolytically cleaving IL6-R to create the soluble IL6/sIL6-R complex, it will be of interest to investigate whether reducing the levels of these metalloproteases and, consequently, the level of sIL6-R will lead to reduced ASD-like behavior [142].

4.3. Triggering Receptor Expressed in Myeloid Cells-2 (TREM2)

ADAM17 is the main protease shedding TREM2 under steady-state conditions [143]. Whether ADAM10 has a similar effect on TREM2 is still under debate [143]. TREM2 is a type I transmembrane protein and exclusively expressed by the microglia [144,145]. TREM2 deficiency in mice decreases the number of microglia and activated microglia in the hippocampus and increases the synaptic and spine density [145]. This may be involved with the increased expression of ligands of TREM2 induced by apoptotic neurons [146]. Furthermore, it is demonstrated that TREM2 is essential for initiating microglia-dependent synaptic pruning during early brain development [145]. Synaptic pruning is essential to remove synapses and keep normal brain connectivity during brain development. The shedding of TREM2 of microglial cells by ADAM17 might result in reduced synaptic pruning and associated neuronal overgrowth. The shedding of TREM2 results in a soluble fraction, sTREM2. Recently, Zhong et al. demonstrated that sTREM2 is able to activate the microglia and increases neuroinflammatory responses both in vitro and in vivo [147]; however, by which mechanisms remains to be elucidated.
Reports on the connection between TREM2 and ASD are currently scarce. Trem2-deficient mice display increased synaptic density, enhanced excitatory neurotransmission and reduced social and repetitive behaviors [145]. Additionally, in the post-mortem brain tissue of ASD patients, reduced TREM2 levels were found in the age group of 5–23 [145]. Furthermore, TREM2 protein levels of brain tissue were inversely correlated to the Autism Diagnostic Interview-Revised score [145]. However, the levels of sTREM2 were not determined in this study. In general, reduced TREM2 could result in an altered control of microglial pruning, and consequently, this would lead to increased synaptic density. In ASD patients, synaptic density is increased on apical dendrites of pyramidal neurons from cortical layer 2 in the frontal, temporal and parietal lobes and layer 5 only in the temporal lobe [6]. Therefore, the relationship between the lower TREM2 levels and increased synaptic density in ASD patients seems plausible.
Taken together, it is difficult to determine the contribution of ADAM17 or ADAM10-mediated shedding of TREM2 in people diagnosed with ASD. The decreased levels of TREM2 in juvenile ASD could be related to either lower baseline TREM2 protein levels or to increased shedding of the protein by proteases. This can be determined by measuring the sTREM2 levels in the same post-mortem tissue.

5. ADAM10 and ADAM17 in the Gut–Immune–Brain Axis

The contribution of the intestines to ASD pathogenesis remains a field of active research. Forty-six point eight percent of people diagnosed with ASD present with at least one intestinal symptom [148], such as constipation [149] and diarrhea [16]. Furthermore, a meta-analysis revealed that children with ASD show four times more intestinal symptoms than controls [150]. The gut–brain axis refers to the bidirectional interaction between these organs [151]. Alterations in this pathway could lead to the increased permeability of both the intestinal and brain barriers. Research emphasizing the role of the intestines show that, in the post-mortem duodenal tissue of people diagnosed with ASD, elevated levels of pore-forming proteins and decreased levels of barrier-forming proteins were found in the tight junction of the intestinal epithelium [53]. These findings suggest a “leaky gut”, which could lead to more circulating bacterial metabolites in the blood of people diagnosed with ASD and activation of the immune system associated with an enhanced cytokine response [152,153,154]. Entering the brain by circulating cytokines and bacterial metabolites via blood is regulated by the Blood–Brain Barrier (BBB). The BBB allows the selective entrance of compounds to the brain through the expression of receptors and transporters, which are necessary for maintaining brain homeostasis [155,156]. Disruption of the BBB—for example, by chronic systemic inflammation—will increase the permeability and allow cytokines and bacterial metabolites to enter the brain directly. In turn, this could result in neuroinflammation and neuronal dysfunction [156,157]. There are limited studies investigating the effect of ADAM10 and ADAM17 on the immune system, the intestinal homeostasis, intestinal microbiota, in intestinal inflammation and epithelial and endothelial (BBB) barrier functions. These studies are discussed below.

5.1. ADAM10 and ADAM17 and Blood–Brain Barrier Permeability

There is little known regarding the effects of ADAM10 or ADAM17 on the BBB permeability. Schulz et al. demonstrated ADAM10 increases the endothelial permeability by specifically cleaving Vascular Endothelial Cadherin (VE-cadherin) in human umbilical vein endothelial cells [158]. The low-density lipoprotein receptor-related protein 1 (LRP1) prevents the endocytic transport of Aβ [159,160]. LRP1 is located at the abluminal surface of the brain endothelium, by which Aβ is then released into the systematic circulation. ADAM10 KO and ADAM10 inhibition facilitate the clearance of Aβ in the brain through decreasing the proteolytic cleavage of LRP1 by ADAM10 in mice and in human brain microvessel endothelial cells [161]. It seems plausible that ADAM10 plays an important role in changing the BBB permeability through the proteolytic cleavage of junction and transporter proteins in the BBB.

5.2. ADAM10 in the Intestinal Tract

ADAM10 is widely expressed in intestinal epithelial cells [68] and involved in modulating the intestinal permeability by targeting its substrates Notch and E-cadherin [48]. Research in conditional ADAM10-deficient mice points out that, when ADAM10 is deleted in intestinal cells, there is an early lethality caused by altered intestinal morphology and changes in cell differentiation [45]. Furthermore, it has been shown that the intestinal morphology changes were due to the loss of Notch receptor signaling caused by the shedding of ADAM10 [45]. The Notch receptor is a recognized substrate of ADAM10 that is ubiquitously expressed in all epithelial cell types. The Notch receptor determines the intestinal stem cell fate and controls intestinal homeostasis [162]. An increase of cleaved Notch-1 decreases the transepithelial electrical resistance, indicative for a reduced intestinal barrier function, and tight junction protein Claudin-5 expression in Caco-2 cells. In addition, the levels of cleaved Notch-1 are increased in the colonic epithelium of patients with Crohn’s disease [47]. E-cadherin is one of the most important junction molecules involved in maintenance of the intestinal epithelial integrity. It was demonstrated that E-cadherin is specifically cleaved by ADAM10 in mouse embryonical fibroblasts and by the absence of soluble E-cadherin in ADAM10-deficient mice [48]. Taken together, these findings suggest that the ADAM10-mediated shedding of Notch receptor and E-cadherin downregulates epithelial cell migration and adhesion and influences intestinal barrier function.
Finally, there might be a role for ADAM10 (possibly, also, ADAM17) in cleaving APP in relation to weight gain, as children diagnosed with ASD have a higher risk for becoming overweight [163]. More recently, it has been demonstrated that APP mediates diet-dependent weight gain, probably through enhanced TNF-α and IL-6 secretion by macrophages, and the potentiation of cholesterol uptake by colonic epithelial cells [164,165,166]. Moreover, high-fat diet-induced APP production in white adipose tissue leads to mitochondrial dysfunction [167]. However, the role of ADAM10 in cleaving APP in relation to weight gain has not been studied. Taken together, it can be hypothesized that, possibly, ADAM10-induced dysregulated APP can be involved in the development of obesity in ASD.
In conclusion, ADAM10 has an important role in maintaining intestinal homeostasis. However, there are no studies conducted about the role of enhanced ADAM10 on intestinal functioning in ASD, and this will be crucial to deepening our understanding of this involvement.

5.3. ADAM17 in the Intestinal Tract

ADAM17 is ubiquitously expressed in all intestinal epithelial cells. In a murine model of decreased ADAM17 expression, where its activity is significantly reduced, normal intestinal epithelial cell proliferation is not compromised. However, there is a less effective response of the intestines to inflammation [168]. ADAM17 seems to be an essential component in regulating intestinal inflammation. The proinflammatory cytokines TNF-α and IL-6 [8] can disrupt the tight junction structure in the intestine and contribute to inflammation [169,170]. As ADAM17 cleaves TNF-α [171] and IL-6R [139], it seems that ADAM17 activity is tightly connected to the intestinal barrier integrity via a proinflammatory route. Recently, research pointed out that the polyphenol, resveratrol, is able to ameliorate social deficits in the VPA mouse model of ASD, probably attributing to its anti-inflammatory properties [172]. Resveratrol also reduced the proinflammatory cytokine levels, such as IL-6 and TNF-α in the BTBR T+tf/J mouse model of ASD, which indicates that the inhibition of inflammation may be promising to the treatment of ASD [173]. Additionally, with the observed increased levels of TNF-α, sIL6R/IL6 complexes [134,174] and ADAM17 [130] in ASD patients, it is tempting to assume that this metalloprotease could be crucial in the development of ASD by alteration of the gut–brain axis.

5.4. ADAMs and Intestinal Microbiota

Another important aspect of the gut–brain axis is the intestinal microbiome. Intestinal microbiotas consist of cohabitating microorganisms involved in regulating the host immunity and inflammation [175,176,177]. There is bidirectional communication between the intestinal microbiota and the brain [178,179,180]. Although there are limited reports on the contribution of ADAM10 or ADAM17 to intestinal microbiota dysbiosis, it is possible to connect several studies and hypothesize.
The composition of the intestinal microbiome is altered in ASD children compared to normal healthy individuals [8,9,84,151]. Their microbial composition contains a higher proportion of Gram-negative bacteria [181], which increases the expression of lipopolysaccharides (LPS) [182]. LPS can activate the innate immune system through the activation of Toll-Like Receptor 4 (TLR4) [183,184]. The activation of TLR4 stimulates the production of proinflammatory cytokines in the intestines and causes neuroinflammation in the brain by activation of the microglia [185,186]. Strikingly, the activation of TLR4 stimulates the ADAM17-dependent shedding of TNF-α [187]. Furthermore, proinflammatory cytokines and LPS treatments are both able to increase the active ADAM10 levels in vitro [188]. It can be suggested that the altered microbiome in people diagnosed with ASD might be able to activate ADAM17 and ADAM10 through increased LPS and proinflammatory cytokines production.
A higher incidence of Clostridium perfringens in fecal samples of ASD children has been described compared to healthy children [189,190]. In the intestines, several species of Clostridium perfringens generate potent toxins that are the causatives of fatal intestinal and CNS diseases in animals [191]. Delta-toxin is one of these that perturbs the intestinal epithelial barrier function in human intestinal epithelial Caco-2 cells through enhancing the ADAM10 activity in a dose- and time-dependent way, which is blocked in the presence of the ADAM10 inhibitor [192,193]. Therefore, the altered microbiome and related metabolites in ASD patients seem to be able to activate ADAM17 and ADAM10 by increasing the production of LPS and Delta-toxin. However, there is little known about the effects of other bacterial-generated metabolites on ADAM10 and ADAM17, such as p-cresol, its derivative p-cresyl sulfate (pCS) and 4-ethylphenylsulfate (4EPS). The levels of p-cresol and its conjugated derivative pCS are increased in urine and fecal samples in ASD children [97,194,195]. Urinary p-cresol has been suggested as a biomarker for ASD in small children because of its significant elevation [195]. A 4EPS treatment induced ASD-like behavior in mice [8]. Mishra et al. showed that there was no difference in the cecal bacterial microbiota composition and load between ADAM17 conditional KO mice and control mice, but the conditional knockout of ADAM17 decreased the peritoneal spread of bacteria following sepsis induction compared to control mice [196], which might be involved with reduced cleavages of TNF-α shedding and other proinflammatory cytokines by ADAM17.
Additionally, some substrates of ADAM17 or ADAM10 can regulate microbiota composition. Angiotensin-converting enzyme 2 (ACE2), a substrate of ADAM17 [197,198], plays an emerging role in the pathogenesis of cardiovascular and lung diseases through changing the composition of the intestinal microbiota, such as increasing the ratio of the Firmicutes to Bacteroidetes and decreasing the Bifidobacterium genus, which raises the potential relation of ADAM17-mediated ACE2 shedding and intestinal dysbiosis [199,200,201]. Meantime, it is demonstrated that an enteric infection coupled with chronic Notch receptor pathway inhibition in mice is associated with bacterial dysbiosis compared to control mice, indicated as a significant decrease in the Bacteroidetes phyla, with concomitant increases in the Firmicutes, Proteobacteria and Verrucomicrobia phyla [46]. It seems that the Notch receptor inhibition changed the microbiota composition in enteric-infected mice, which implies that ADAM10 overexpression may have a similar effect on microbiota composition through cleavage of the Notch receptor. Taken together, intestinal microbes can regulate ADAM10 or ADAM17 activity by producing bacterial metabolites, and, in turn, ADAM10 or ADAM17 can also change the intestinal microbiome composition. However, these connections remain unclear, and more research is needed.

5.5. ADAMs and the Immune System

The development and function of the immune system is highly dependent on the intestinal microbiota, as demonstrated by the limited immune activity in germ-free mice [202]. Intestinal bacterial fermentation produces a wide range of metabolites on the basis of tryptophan, tyrosine and phenylalanine from our daily diet, such as serotonin, short-chain fatty acids (SCFAs), indole-containing metabolites and p-cresol [203,204,205]. The role of SCFAs and other not-mentioned bacterial metabolites in ASD have been extensively reviewed [177]. These metabolites can regulate immune responses and inflammatory responses by recognizing their receptor on epithelial cells or entering into the systemic circulation or the brain. SCFAs promote the number, function and differentiation of colonic T-reg cells in mice [203,206]. In addition, SCFA—specifically, butyrate—being fuel for epithelial cells, promote intestinal barrier integrity [207,208]. In the brain, SCFAs also increase the microglia maturation and functions in mice [204]. However, the exact mechanisms how these metabolites affect host immune and brain functions remains to be investigated. In recent years, alterations in the gut–brain axis have been presented as possible pathological causes of ASD, and targeting the intestinal microbes has been recognized as a promising treatment for ASD [8,16,178,209]. For example, maternal immune activation (MIA) induced by polyinosinic:polycytidylic acid (polyI:C) injection led to intestinal dysbiosis in the male offspring associated with defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors. The oral administration of Bacteroides fragilis restored these ASD-like symptoms [8]. The roles of ADAM10 and ADAM17 in the effects of the bacterial metabolites in the immune system in ASD has not been researched well; however, it is possible to speculate about its potential involvement.
Transforming growth factor β1 (TGF-β1) is one member of the TGFβ family and, generally, regulates T lymphocytes and antigen-presenting cells as an immunosuppressor [210]. Some studies showed that there is a significant decrease of the TGF-β1 level in the plasma or serum of ASD children [211,212]. Moreover, one study showed that TGF-β1 might be considered as a biomarker of ASD severity. Increasing TGF-β1 levels in the plasma of ASD children consequently improved the behavioral rating score [213]. TGF-β1 also is essential to the microglial development, phenotypes and functions in vitro and in vivo, which is connected with ASD pathogenesis [214,215,216]. Besides, TGF-β1 plays a vital role in modulating social interactions and repetitive behaviors in mice hippocampus. It was demonstrated that adult hippocampal TGF-β1 overexpression increases social interactions and decreases self-grooming and depression-related behaviors, and early hippocampal TGF-β1 overexpression reversely decreases those behaviors [217]. Kawasaki et al. illustrated that TGF-β1 signaling is dependent on ADAM 17 activity, and thus, modifying ADAM17 genetic variants enhances TGF-β1 signaling activity through cleaving less type 1 TGF-beta receptor (TGF-βR1) in mice and humans [218]. It is demonstrated that TGF-βR1 is a substrate of ADAM17, and its cleavage by ADAM17 downregulates TGF-β1 signaling through the decreasing cell surface TGF-βR1 [218,219]. In addition, Vasorin is a type 1 transmembrane protein, and it is cleaved by ADAM17 to generate soluble Vasorin that binds to TGF-β1 as a suppressor [220,221]. To our knowledge, there is little known about the ADAM17-mediated TGF-β1 signaling in ASD pathogenesis.
T-helper 17 lymphocytes (TH17) cells and their effector cytokines interleukin 17 (IL-17) are necessary for immune responses against extracellular bacteria and fungi, and their dysregulation is thought to underlie numerous inflammatory and autoimmune diseases, such as inflammatory bowel disease and multiple sclerosis [222]. Choi et al. demonstrated that maternal immune activation induced ASD-like behavioral phenotypes in the offspring and compromised their cortical brain development in an IL-17a-dependent manner. IL17a KO or IL-17a blockage with an antibody rescued the ASD-like phenotypes [223]. The proinflammatory cytokine IL-6 inhibits TH17 cell differentiation from the CD4+ T-cell subset as an upstream regulator of IL-17a [224,225]. Therefore, IL-6 is also necessary for maternal immune activation-induced ASD-like phenotypes in the offspring [226]. It is demonstrated that ADAM17 regulates IL-6 signaling through controlling the IL-6 receptor (IL-6R) in vitro and in vivo [227]. Horiuchi et al. reported that conditional ADAM17 KO mice have increased serum levels of IL-17 compared with control littermates, indicating that decreased ADAM17 activity is associated with a downregulation of IL-17 secretion in vivo [187], but the role of the IL-17 receptor in this process was not investigated, and the definite mechanism remains to be elucidated. It is possible that the increased IL-17 level in ADAM17 KO mice is a result from the increased cleavage of the IL-17 receptor by ADAM17 [228], but it is barely studied. These findings make it promising to investigate the potential roles of ADAM10 and ADAM17 in the pathogenesis of ASD from an immunological perspective.

6. Metalloproteases ADAM10 and ADAM17 as Therapeutic Targets for Autism Spectrum Disorders

The metalloproteinases ADAM10 and ADAM17 are or might be involved in different aspects of ASD pathogenesis. Strikingly, as can be concluded from Table 2, it seems that the enhanced expression and activity of ADAM10 or ADAM17 might contribute to several aspects of ASD. Therefore, the reduction or inhibition of these targets could be interesting as therapeutic strategies for ASD. Although not much attention has been given in research to targeting metalloproteases, some possible therapeutic options will be discussed below.

6.1. TIMPs

The natural inhibitors of the metalloproteases are the Tissue Inhibitors of Metalloproteases (TIMPs). There are four members of the TIMP family [229,230]. In general, the TIMPs can inhibit all MMPs, but the strength of MMP inhibition differs between the TIMPs. Interestingly, TIMPs inhibit ADAMs with higher specificity. For example, ADAM10 is specifically inhibited by TIMP-1 and TIMP-3 [231]. Moreover, TIMP-3 also inhibits ADAM17 [232]. The main limitation of metalloprotease inhibitors is their lack of selectivity. Therefore, an inhibitor can affect other enzymes as well, which could lead to undesirable side effects. For instance, increasing the TIMP-3 levels can be an interesting therapeutic target, as this can possibly decrease both the ADAM10 and ADAM17 activity in ASD patients; however, TIMP-3 also has an inhibitory effect on most MMPs [233,234]. Thus, the search for selective inhibitors is of critical importance in order to be used as a therapeutic drug.

6.2. ADAM Inhibitors

In search for molecules with a great selectivity for ADAM10 and ADAM17, the GI254023X compound has been identified as a potent and selective inhibitor of ADAM10, with 100-fold higher selectivity than ADAM17 [235,236]. In addition, Mahasenan et al. recently synthesized and tested the compound (1R,3S,4S)-3-(hydroxycarbamoyl)-4-(4-phenylpiperidine-1-carbonyl) cyclohexyl pyrrolidine-1-carboxylate, which showed a high potency of inhibiting ADAM10 [237]. This compound can also cross the BBB [237]. Furthermore, Hirata et al. showed that the inhibitor KP-457 has over 50 times higher selectivity for ADAM17 than ADAM10 or any other MMPs [238].
TspanC8 members regulate ADAM10 maturation and substrate selectivity. Six TspanC8 members can form six different Tspan–ADAM10 complexes, which preferentially cleave different substrates as six scissors [36,62,65,125]. Therefore, the development of inhibitors targeting these complexes is beneficial compared to the side effects of broad ADAM inhibitors. Taken together, these studies showed that effective inhibitors are available and might be beneficial for ASD treatment.

6.3. Probiotics, Bacterial Metabolites and Prebiotics?

As discussed above, intestinal dysbiosis has been frequently described in children suffering from ASD. Pro-, prebiotic and even microbiota transfer therapy (MTT) interventions have been proposed as promising treatments for ASD children [16,239,240,241,242]. Of interest are the bacterial metabolites 4EPS, as well bacterial toxin LPS, that induce ASD-like behavior in mice by unknown mechanisms [8,243]. Furthermore, a bacterial LPS-induced increase of ADAM10 expression is important for proinflammatory immune cell responses [244]. Other important bacterial metabolites are SCFAs. The increase of enteric SCFAs levels in ASD mice [245], as well as in ASD children, are demonstrated [246,247]. The precise mechanism of action of SCFAs in relation to ASD-like behaviors is not known, but the effects on the mitochondrial function or epigenetic alterations in the brain may be involved [248]. Given the important roles of ADAM10 and ADAM17 in the gut functions, immunity and brain, it will be interesting to study whether and how intestinal microbiota-derived metabolites, such as 4EPS, LPS, p-cresol and SCFAs, affect ADAM10/17 activity in the intestinal tract and in the brain related to ASD. When, indeed, an important role of these bacterial metabolites on the ADAM10/17 activity is established, then targeting the intestinal microbiota with pre- and probiotics, as well as MTT, may be useful in ASD.

6.4. Targeting ADAM10 and ADAM17 in ASD: Some Considerations

The strategy of targeting ADAM 10 and/or ADAM17 as future treatments in ASD raises several issues. First, the processes in which ADAMs are involved are critical for cell, tissue and organ functioning; therefore, ADAM10 and ADAM17 inhibitors might have serious side effects. Both proteases contribute to developmental and regenerative processes; for example, the disruption of ADAM17 in mice leads to death, and studies in KO mice show that ADAM10 is vital for early development [125]. The specific targeting of ADAM10 or ADAM17 at the right time and in the right location might be the way to go. Secondly, it should be investigated at which location ADAM10 and ADAM17 should be targeted: in the intestines or the brain. For the latter, compounds that are able to pass the BBB are essential. Indirect targeting through manipulation of the intestinal microbiota with pre-, pro or postbiotics might be a safer way to inhibit the enhanced ADAM10 and ADAM 17 activity in ASD.

7. Outlook and Conclusions

ASD is a highly heterogeneous disorder that includes multiple affected genes, altered synaptic density, neuroinflammation, low-grade systemic immune activation and an intestinal phenotype, including a “leaky gut”. Therefore, it is challenging to pinpoint what the exact underlying cause of this neurodevelopmental disease is. The ASD-associated enhanced expression and/or activity of the metalloproteases ADAM10 and ADAM17 provide an overarching hypothesis that affects many different aspects that seem to be involved, at least in part, in ASD pathology. ADAM10 is responsible for the proteolytic cleavage of several key proteins involved in synapse formation, axon signaling and cell adhesion and for regulating the intestinal permeability. Furthermore, ADAM17 has a pivotal role in the shedding of proteins that regulate the onset of (neuro)inflammation and immune responses. Additionally, the effects of ADAM10 and ADAM17 on the intestinal microbiota composition and the effects of bacterial metabolites on ADAM10 and ADAM17 expression and activity remain to be investigated. Taken together, these two metalloproteases seem responsible for activating key pathways that seem to be altered in ASD pathogenesis. Figure 3 provides an overview of the pathways where ADAM10 and ADAM17 are possibly involved in the pathogenesis of ASD. In conclusion, the enhanced expression and/or activity of ADAM10 or ADAM17 could possibly be involved in the induction and maintenance of ASD-like phenotypes in the brain, as well as systemically and in the intestinal tract. Consequently, this hypothesis suggests that reducing the levels or activity of ADAM10 or ADAM17 could be a potential therapeutic target in ASD patients.
In order to provide more evidence to support this hypothesis, it is necessary to further determine if there is any altered ADAM10 and ADAM17 expression and/or activity in ASD-associated mouse models or in ASD patients. Moreover, more studies need to be conducted to investigate the role and molecular mechanisms of ADAM10 and ADAM17, which will shed light on the molecular pathogenesis and possible targets for the treatment of ASD. Additionally, it will be interesting to screen metalloprotease-specific inhibitors and then test the specific inhibitors of ADAM10 and ADAM17 in ASD animal models.

Author Contributions

Writing—original draft preparation: Y.Z. and T.A.V. and writing—review and editing: Y.Z., T.A.V., P.P.P., J.G. and A.D.K. All authors have read and agreed to the published version of the manuscript.

Funding

This review was written in the context of The GEMMA project (grant number 00002771-13001252) and partly funded by the China Scholarship Council (CSC) (grant number 201706210077).

Conflicts of Interest

Y.Z., T.A.V., P.P.P. and A.D.K. declare no conflict of interest. J.G. works at Danone Nutricia Research B.V., Utrecht, The Netherlands.

References

  1. Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism spectrum disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef]
  2. Lyall, K.; Croen, L.; Daniels, J.; Fallin, M.D.; Ladd-Acosta, C.; Lee, B.K.; Park, B.Y.; Snyder, N.W.; Schendel, D.; Volk, H.; et al. The Changing Epidemiology of Autism Spectrum Disorders. Annu. Rev. Public Health 2017, 38, 81–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Maenner, M.J.; Shaw, K.A.; Baio, J.; Washington, A.; Patrick, M.; DiRienzo, M.; Christensen, D.L.; Wiggins, L.D.; Pettygrove, S.; Andrews, J.G.; et al. Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2016. Surveill. Summ. 2020, 69, 1–12. [Google Scholar] [CrossRef] [PubMed]
  4. Bauman, M.L.; Kemper, T.L. Neuroanatomic observations of the brain in autism: A review and future directions. Int. J. Dev. Neurosci. 2005, 23, 183–187. [Google Scholar] [CrossRef]
  5. O’Reilly, C.; Lewis, J.D.; Elsabbagh, M. Is functional brain connectivity atypical in autism? A systematic review of EEG and MEG studies. PLoS ONE 2017, 12, e0175870. [Google Scholar] [CrossRef]
  6. Hutsler, J.J.; Zhang, H. Increased dendritic spine densities on cortical projection neurons in autism spectrum disorders. Brain Res. 2010, 1309, 83–94. [Google Scholar] [CrossRef]
  7. Matta, S.M.; Hill-Yardin, E.L.; Crack, P.J. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav. Immun. 2019, 79, 75–90. [Google Scholar] [CrossRef]
  8. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
  9. Sharon, G.; Cruz, N.J.; Kang, D.W.; Gandal, M.J.; Wang, B.; Kim, Y.M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; Lane, C.J.; et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019, 177, 1600–1618. [Google Scholar] [CrossRef] [Green Version]
  10. Roussin, L.; Prince, N.; Perez-Pardo, P.; Kraneveld, A.D.; Rabot, S.; Naudon, L. Role of the Gut Microbiota in the Pathophysiology of Autism Spectrum Disorder: Clinical and Preclinical Evidence. Microorganisms 2020, 8, 1369. [Google Scholar] [CrossRef]
  11. DiStasio, M.M.; Nagakura, I.; Nadler, M.J.; Anderson, M.P. T lymphocytes and cytotoxic astrocyte blebs correlate across autism brains. Ann. Neurol. 2019, 86, 885–898. [Google Scholar] [CrossRef] [PubMed]
  12. Rossignol, D.A.; Frye, R.E. A review of research trends in physiological abnormalities in autism spectrum disorders: Immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Mol. Psychiatry 2012, 17, 389–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Meltzer, A.; Van de Water, J. The Role of the Immune System in Autism Spectrum Disorder. Neuropsychopharmacology 2017, 42, 284–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Masi, A.; Glozier, N.; Dale, R.; Guastella, A.J. The Immune System, Cytokines, and Biomarkers in Autism Spectrum Disorder. Neurosci. Bull. 2017, 33, 194–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ormstad, H.; Bryn, V.; Saugstad, O.D.; Skjeldal, O.; Maes, M. Role of the Immune System in Autism Spectrum Disorders (ASD). CNS Neurol. Disord. Drug Targets 2018, 17, 489–495. [Google Scholar] [CrossRef]
  16. Kang, D.W.; Adams, J.B.; Coleman, D.M.; Pollard, E.L.; Maldonado, J.; McDonough-Means, S.; Caporaso, J.G.; Krajmalnik-Brown, R. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Sci. Rep. 2019, 9, 5821. [Google Scholar] [CrossRef]
  17. Ferguson, B.J.; Dovgan, K.; Takahashi, N.; Beversdorf, D.Q. The Relationship among Gastrointestinal Symptoms, Problem Behaviors, and Internalizing Symptoms in Children and Adolescents with Autism Spectrum Disorder. Front. Psychiatry 2019, 10, 194. [Google Scholar] [CrossRef] [Green Version]
  18. Ding, H.T.; Taur, Y.; Walkup, J.T. Gut Microbiota and Autism: Key Concepts and Findings. J. Autism Dev. Disord. 2017, 47, 480–489. [Google Scholar] [CrossRef]
  19. Hazlett, H.C.; Gu, H.; Munsell, B.C.; Kim, S.H.; Styner, M.; Wolff, J.J.; Elison, J.T.; Swanson, M.R.; Zhu, H.; Botteron, K.N.; et al. Early brain development in infants at high risk for autism spectrum disorder. Nature 2017, 542, 348–351. [Google Scholar] [CrossRef]
  20. Ecker, C.; Bookheimer, S.Y.; Murphy, D.G. Neuroimaging in autism spectrum disorder: Brain structure and function across the lifespan. Lancet Neurol. 2015, 14, 1121–1134. [Google Scholar] [CrossRef] [Green Version]
  21. Geschwind, D.H.; State, M.W. Gene hunting in autism spectrum disorder: On the path to precision medicine. Lancet Neurol. 2015, 14, 1109–1120. [Google Scholar] [CrossRef] [Green Version]
  22. Sandin, S.; Lichtenstein, P.; Kuja-Halkola, R.; Hultman, C.; Larsson, H.; Reichenberg, A. The Heritability of Autism Spectrum Disorder. JAMA 2017, 318, 1182–1184. [Google Scholar] [CrossRef] [PubMed]
  23. Bailey, A.; Le Couteur, A.; Gottesman, I.; Bolton, P.; Simonoff, E.; Yuzda, E.; Rutter, M. Autism as a strongly genetic disorder: Evidence from a British twin study. Psychol. Med. 1995, 25, 63–77. [Google Scholar] [CrossRef] [PubMed]
  24. Marshall, C.R.; Noor, A.; Vincent, J.B.; Lionel, A.C.; Feuk, L.; Skaug, J.; Shago, M.; Moessner, R.; Pinto, D.; Ren, Y.; et al. Structural variation of chromosomes in autism spectrum disorder. Am. J. Hum. Genet. 2008, 82, 477–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Marui, T.; Funatogawa, I.; Koishi, S.; Yamamoto, K.; Matsumoto, H.; Hashimoto, O.; Nanba, E.; Nishida, H.; Sugiyama, T.; Kasai, K.; et al. Association of the neuronal cell adhesion molecule (NRCAM) gene variants with autism. Int. J. Neuropsychopharmacol. 2009, 12, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Morrow, E.M.; Yoo, S.Y.; Flavell, S.W.; Kim, T.K.; Lin, Y.; Hill, R.S.; Mukaddes, N.M.; Balkhy, S.; Gascon, G.; Hashmi, A.; et al. Identifying autism loci and genes by tracing recent shared ancestry. Science 2008, 321, 218–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Glessner, J.T.; Wang, K.; Cai, G.; Korvatska, O.; Kim, C.E.; Wood, S.; Zhang, H.; Estes, A.; Brune, C.W.; Bradfield, J.P.; et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature 2009, 459, 569–573. [Google Scholar] [CrossRef]
  28. Ray, B.; Long, J.M.; Sokol, D.K.; Lahiri, D.K. Increased secreted amyloid precursor protein-α (sAPPα) in severe autism: Proposal of a specific, anabolic pathway and putative biomarker. PLoS ONE 2011, 6, e20405. [Google Scholar] [CrossRef] [Green Version]
  29. Blundell, J.; Blaiss, C.A.; Etherton, M.R.; Espinosa, F.; Tabuchi, K.; Walz, C.; Bolliger, M.F.; Südhof, T.C.; Powell, C.M. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J. Neurosci. 2010, 30, 2115–2129. [Google Scholar] [CrossRef] [Green Version]
  30. Bruining, H.; Matsui, A.; Oguro-Ando, A.; Kahn, R.S.; Van’t Spijker, H.M.; Akkermans, G.; Stiedl, O.; van Engeland, H.; Koopmans, B.; van Lith, H.A.; et al. Genetic Mapping in Mice Reveals the Involvement of Pcdh9 in Long-Term Social and Object Recognition and Sensorimotor Development. Biol. Psychiatry 2015, 78, 485–495. [Google Scholar] [CrossRef]
  31. Moy, S.S.; Nonneman, R.J.; Young, N.B.; Demyanenko, G.P.; Maness, P.F. Impaired sociability and cognitive function in Nrcam-null mice. Behav. Brain Res. 2009, 205, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kuhn, P.H.; Wang, H.; Dislich, B.; Colombo, A.; Zeitschel, U.; Ellwart, J.W.; Kremmer, E.; Rossner, S.; Lichtenthaler, S.F. ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons. EMBO J. 2010, 29, 3020–3032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bot, N.; Schweizer, C.; Ben Halima, S.; Fraering, P.C. Processing of the synaptic cell adhesion molecule neurexin-3beta by Alzheimer disease alpha- and gamma-secretases. J. Biol. Chem. 2011, 286, 2762–2773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Borcel, E.; Palczynska, M.; Krzisch, M.; Dimitrov, M.; Ulrich, G.; Toni, N.; Fraering, P.C. Shedding of neurexin 3β ectodomain by ADAM10 releases a soluble fragment that affects the development of newborn neurons. Sci. Rep. 2016, 6, 39310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kuhn, P.H.; Colombo, A.V.; Schusser, B.; Dreymueller, D.; Wetzel, S.; Schepers, U.; Herber, J.; Ludwig, A.; Kremmer, E.; Montag, D.; et al. Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife 2016, 5, e12748. [Google Scholar] [CrossRef] [PubMed]
  36. Brummer, T.; Muller, S.A.; Pan-Montojo, F.; Yoshida, F.; Fellgiebel, A.; Tomita, T.; Endres, K.; Lichtenthaler, S.F. NrCAM is a marker for substrate-selective activation of ADAM10 in Alzheimer’s disease. EMBO Mol. Med. 2019, 11, e9695. [Google Scholar] [CrossRef] [PubMed]
  37. Rivera, S.; Khrestchatisky, M.; Kaczmarek, L.; Rosenberg, G.A.; Jaworski, D.M. Metzincin proteases and their inhibitors: Foes or friends in nervous system physiology? J. Neurosci. 2010, 30, 15337–15357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Huovila, A.P.; Turner, A.J.; Pelto-Huikko, M.; Kärkkäinen, I.; Ortiz, R.M. Shedding light on ADAM metalloproteinases. Trends Biochem. Sci. 2005, 30, 413–422. [Google Scholar] [CrossRef]
  39. Edwards, D.R.; Handsley, M.M.; Pennington, C.J. The ADAM metalloproteinases. Mol. Asp. Med. 2008, 29, 258–289. [Google Scholar] [CrossRef]
  40. Gunner, G.; Cheadle, L.; Johnson, K.M.; Ayata, P.; Badimon, A.; Mondo, E.; Nagy, M.A.; Liu, L.; Bemiller, S.M.; Kim, K.W.; et al. Sensory lesioning induces microglial synapse elimination via ADAM10 and fractalkine signaling. Nat. Neurosci. 2019, 22, 1075–1088. [Google Scholar] [CrossRef]
  41. Moss, M.L.; Minond, D. Recent Advances in ADAM17 Research: A Promising Target for Cancer and Inflammation. Mediat. Inflamm. 2017, 2017, 9673537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Young, J.; Yu, X.; Wolslegel, K.; Nguyen, A.; Kung, C.; Chiang, E.; Kolumam, G.; Wei, N.; Wong, W.L.; DeForge, L.; et al. Lymphotoxin-alphabeta heterotrimers are cleaved by metalloproteinases and contribute to synovitis in rheumatoid arthritis. Cytokine 2010, 51, 78–86. [Google Scholar] [CrossRef] [PubMed]
  43. Freour, T.; Jarry, A.; Bach-Ngohou, K.; Dejoie, T.; Bou-Hanna, C.; Denis, M.G.; Mosnier, J.F.; Laboisse, C.L.; Masson, D. TACE inhibition amplifies TNF-alpha-mediated colonic epithelial barrier disruption. Int. J. Mol. Med. 2009, 23, 41–48. [Google Scholar] [PubMed]
  44. Geesala, R.; Schanz, W.; Biggs, M.; Dixit, G.; Skurski, J.; Gurung, P.; Meyerholz, D.K.; Elliott, D.; Issuree, P.D.; Maretzky, T. Loss of RHBDF2 results in an early-onset spontaneous murine colitis. J. Leukoc. Biol. 2019, 105, 767–781. [Google Scholar] [CrossRef]
  45. Tsai, Y.H.; VanDussen, K.L.; Sawey, E.T.; Wade, A.W.; Kasper, C.; Rakshit, S.; Bhatt, R.G.; Stoeck, A.; Maillard, I.; Crawford, H.C.; et al. ADAM10 regulates Notch function in intestinal stem cells of mice. Gastroenterology 2014, 147, 822–834. [Google Scholar] [CrossRef] [Green Version]
  46. Ahmed, I.; Roy, B.C.; Raach, R.T.; Owens, S.M.; Xia, L.; Anant, S.; Sampath, V.; Umar, S. Enteric infection coupled with chronic Notch pathway inhibition alters colonic mucus composition leading to dysbiosis, barrier disruption and colitis. PLoS ONE 2018, 13, e0206701. [Google Scholar] [CrossRef]
  47. Dahan, S.; Rabinowitz, K.M.; Martin, A.P.; Berin, M.C.; Unkeless, J.C.; Mayer, L. Notch-1 signaling regulates intestinal epithelial barrier function, through interaction with CD4+ T cells, in mice and humans. Gastroenterology 2011, 140, 550–559. [Google Scholar] [CrossRef] [Green Version]
  48. Maretzky, T.; Reiss, K.; Ludwig, A.; Buchholz, J.; Scholz, F.; Proksch, E.; de Strooper, B.; Hartmann, D.; Saftig, P. ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. Proc. Natl. Acad. Sci. USA 2005, 102, 9182–9187. [Google Scholar] [CrossRef] [Green Version]
  49. Lum, L.; Reid, M.S.; Blobel, C.P. Intracellular maturation of the mouse metalloprotease disintegrin MDC15. J. Biol. Chem. 1998, 273, 26236–26247. [Google Scholar] [CrossRef] [Green Version]
  50. Reiss, K.; Saftig, P. The “a disintegrin and metalloprotease” (ADAM) family of sheddases: Physiological and cellular functions. Semin. Cell Dev. Biol. 2009, 20, 126–137. [Google Scholar] [CrossRef]
  51. Fanjul-Fernández, M.; Folgueras, A.R.; Cabrera, S.; López-Otín, C. Matrix metalloproteinases: Evolution, gene regulation and functional analysis in mouse models. Biochim. Biophys. Acta 2010, 1803, 3–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Abdallah, M.W.; Pearce, B.D.; Larsen, N.; Greaves-Lord, K.; Nørgaard-Pedersen, B.; Hougaard, D.M.; Mortensen, E.L.; Grove, J. Amniotic fluid MMP-9 and neurotrophins in autism spectrum disorders: An exploratory study. Autism Res. 2012, 5, 428–433. [Google Scholar] [CrossRef] [PubMed]
  53. Fiorentino, M.; Sapone, A.; Senger, S.; Camhi, S.S.; Kadzielski, S.M.; Buie, T.M.; Kelly, D.L.; Cascella, N.; Fasano, A. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol. Autism 2016, 7, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Stöcker, W.; Grams, F.; Baumann, U.; Reinemer, P.; Gomis-Rüth, F.X.; McKay, D.B.; Bode, W. The metzincins--topological and sequential relations between the astacins, adamalysins, serralysins, and matrixins (collagenases) define a superfamily of zinc-peptidases. Protein Sci. Publ. Protein Soc. 1995, 4, 823–840. [Google Scholar] [CrossRef] [Green Version]
  55. Puente, X.S.; López-Otín, C. A genomic analysis of rat proteases and protease inhibitors. Genome Res. 2004, 14, 609–622. [Google Scholar] [CrossRef] [Green Version]
  56. Bode, W.; Gomis-Rüth, F.X.; Stöckler, W. Astacins, serralysins, snake venom and matrix metalloproteinases exhibit identical zinc-binding environments (HEXXHXXGXXH and Met-turn) and topologies and should be grouped into a common family, the ‘metzincins’. FEBS Lett. 1993, 331, 134–140. [Google Scholar] [CrossRef] [Green Version]
  57. Mochizuki, S.; Okada, Y. ADAMs in cancer cell proliferation and progression. Cancer Sci. 2007, 98, 621–628. [Google Scholar] [CrossRef]
  58. Düsterhöft, S.; Lokau, J.; Garbers, C. The metalloprotease ADAM17 in inflammation and cancer. Pathol. Res. Pract. 2019, 215, 152410. [Google Scholar] [CrossRef]
  59. Qian, M.; Shen, X.; Wang, H. The Distinct Role of ADAM17 in APP Proteolysis and Microglial Activation Related to Alzheimer’s Disease. Cell. Mol. Neurobiol. 2016, 36, 471–482. [Google Scholar] [CrossRef]
  60. Marcello, E.; Borroni, B.; Pelucchi, S.; Gardoni, F.; Di Luca, M. ADAM10 as a therapeutic target for brain diseases: From developmental disorders to Alzheimer’s disease. Expert Opin. Ther. Targets 2017, 21, 1017–1026. [Google Scholar] [CrossRef]
  61. Noy, P.J.; Yang, J.; Reyat, J.S.; Matthews, A.L.; Charlton, A.E.; Furmston, J.; Rogers, D.A.; Rainger, G.E.; Tomlinson, M.G. TspanC8 Tetraspanins and A Disintegrin and Metalloprotease 10 (ADAM10) Interact via Their Extracellular Regions: EVIDENCE FOR DISTINCT BINDING MECHANISMS FOR DIFFERENT TspanC8 PROTEINS. J. Biol. Chem. 2016, 291, 3145–3157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Matthews, A.L.; Szyroka, J.; Collier, R.; Noy, P.J.; Tomlinson, M.G. Scissor sisters: Regulation of ADAM10 by the TspanC8 tetraspanins. Biochem. Soc. Trans. 2017, 45, 719–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Haining, E.J.; Yang, J.; Bailey, R.L.; Khan, K.; Collier, R.; Tsai, S.; Watson, S.P.; Frampton, J.; Garcia, P.; Tomlinson, M.G. The TspanC8 subgroup of tetraspanins interacts with A disintegrin and metalloprotease 10 (ADAM10) and regulates its maturation and cell surface expression. J. Biol. Chem. 2012, 287, 39753–39765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Seipold, L.; Altmeppen, H.; Koudelka, T.; Tholey, A.; Kasparek, P.; Sedlacek, R.; Schweizer, M.; Bar, J.; Mikhaylova, M.; Glatzel, M.; et al. In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15. Cell. Mol. Life Sci. 2018, 75, 3251–3267. [Google Scholar] [CrossRef] [PubMed]
  65. Jouannet, S.; Saint-Pol, J.; Fernandez, L.; Nguyen, V.; Charrin, S.; Boucheix, C.; Brou, C.; Milhiet, P.E.; Rubinstein, E. TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, Notch activation and ADAM10 membrane compartmentalization. Cell. Mol. Life Sci. 2016, 73, 1895–1915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sharma, K.; Schmitt, S.; Bergner, C.G.; Tyanova, S.; Kannaiyan, N.; Manrique-Hoyos, N.; Kongi, K.; Cantuti, L.; Hanisch, U.K.; Philips, M.A.; et al. Cell type- and brain region-resolved mouse brain proteome. Nat. Neurosci. 2015, 18, 1819–1831. [Google Scholar] [CrossRef]
  67. Lundgren, J.L.; Ahmed, S.; Schedin-Weiss, S.; Gouras, G.K.; Winblad, B.; Tjernberg, L.O.; Frykman, S. ADAM10 and BACE1 are localized to synaptic vesicles. J. Neurochem. 2015, 135, 606–615. [Google Scholar] [CrossRef] [Green Version]
  68. Dempsey, P.J. Role of ADAM10 in intestinal crypt homeostasis and tumorigenesis. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2228–2239. [Google Scholar] [CrossRef]
  69. Thinakaran, G.; Koo, E.H. Amyloid precursor protein trafficking, processing, and function. J. Biol. Chem. 2008, 283, 29615–29619. [Google Scholar] [CrossRef] [Green Version]
  70. Nikolaev, A.; McLaughlin, T.; O’Leary, D.D.; Tessier-Lavigne, M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 2009, 457, 981–989. [Google Scholar] [CrossRef] [Green Version]
  71. Kang, J.; Lemaire, H.G.; Unterbeck, A.; Salbaum, J.M.; Masters, C.L.; Grzeschik, K.H.; Multhaup, G.; Beyreuther, K.; Müller-Hill, B. The precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface receptor. Nature 1987, 325, 733–736. [Google Scholar] [CrossRef] [PubMed]
  72. Haass, C.; Selkoe, D.J. Cellular processing of beta-amyloid precursor protein and the genesis of amyloid beta-peptide. Cell 1993, 75, 1039–1042. [Google Scholar] [CrossRef]
  73. Sokol, D.K.; Chen, D.; Farlow, M.R.; Dunn, D.W.; Maloney, B.; Zimmer, J.A.; Lahiri, D.K. High levels of Alzheimer beta-amyloid precursor protein (APP) in children with severely autistic behavior and aggression. J. Child Neurol. 2006, 21, 444–449. [Google Scholar] [CrossRef] [PubMed]
  74. Pasciuto, E.; Ahmed, T.; Wahle, T.; Gardoni, F.; D’Andrea, L.; Pacini, L.; Jacquemont, S.; Tassone, F.; Balschun, D.; Dotti, C.G.; et al. Dysregulated ADAM10-Mediated Processing of APP during a Critical Time Window Leads to Synaptic Deficits in Fragile X Syndrome. Neuron 2015, 87, 382–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Westmark, C.J.; Westmark, P.R.; O’Riordan, K.J.; Ray, B.C.; Hervey, C.M.; Salamat, M.S.; Abozeid, S.H.; Stein, K.M.; Stodola, L.A.; Tranfaglia, M.; et al. Reversal of fragile X phenotypes by manipulation of AβPP/Aβ levels in Fmr1KO mice. PLoS ONE 2011, 6, e26549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lahiri, D.K.; Sokol, D.K.; Erickson, C.; Ray, B.; Ho, C.Y.; Maloney, B. Autism as early neurodevelopmental disorder: Evidence for an sAPPα-mediated anabolic pathway. Front. Cell. Neurosci. 2013, 7, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Mc Guire, C.; Beyaert, R.; van Loo, G. Death receptor signalling in central nervous system inflammation and demyelination. Trends Neurosci. 2011, 34, 619–628. [Google Scholar] [CrossRef]
  78. Mi, S.; Lee, X.; Hu, Y.; Ji, B.; Shao, Z.; Yang, W.; Huang, G.; Walus, L.; Rhodes, K.; Gong, B.J.; et al. Death receptor 6 negatively regulates oligodendrocyte survival, maturation and myelination. Nat. Med. 2011, 17, 816–821. [Google Scholar] [CrossRef]
  79. Popko, B. Downregulating DR6 to drive remyelination. Nat. Med. 2011, 17, 779–780. [Google Scholar] [CrossRef]
  80. Colombo, A.; Hsia, H.E.; Wang, M.; Kuhn, P.H.; Brill, M.S.; Canevazzi, P.; Feederle, R.; Taveggia, C.; Misgeld, T.; Lichtenthaler, S.F. Non-cell-autonomous function of DR6 in Schwann cell proliferation. EMBO J. 2018, 37, e97390. [Google Scholar] [CrossRef]
  81. Corfas, G.; Velardez, M.O.; Ko, C.P.; Ratner, N.; Peles, E. Mechanisms and roles of axon-Schwann cell interactions. J. Neurosci. 2004, 24, 9250–9260. [Google Scholar] [CrossRef] [PubMed]
  82. Espinosa-Medina, I.; Jevans, B.; Boismoreau, F.; Chettouh, Z.; Enomoto, H.; Muller, T.; Birchmeier, C.; Burns, A.J.; Brunet, J.F. Dual origin of enteric neurons in vagal Schwann cell precursors and the sympathetic neural crest. Proc. Natl. Acad. Sci. USA 2017, 114, 11980–11985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Vega, C.; Martiel, J.L.; Drouhault, D.; Burckhart, M.F.; Coles, J.A. Uptake of locally applied deoxyglucose, glucose and lactate by axons and Schwann cells of rat vagus nerve. J. Physiol. 2003, 546, 551–564. [Google Scholar] [CrossRef]
  84. Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. van Hoorn, A.; Carpenter, T.; Oak, K.; Laugharne, R.; Ring, H.; Shankar, R. Neuromodulation of autism spectrum disorders using vagal nerve stimulation. J. Clin. Neurosci. 2019, 63, 8–12. [Google Scholar] [CrossRef]
  86. Jin, Y.; Kong, J. Transcutaneous Vagus Nerve Stimulation: A Promising Method for Treatment of Autism Spectrum Disorders. Front. Neurosci. 2016, 10, 609. [Google Scholar] [CrossRef] [Green Version]
  87. Krueger, D.D.; Tuffy, L.P.; Papadopoulos, T.; Brose, N. The role of neurexins and neuroligins in the formation, maturation, and function of vertebrate synapses. Curr. Opin. Neurobiol. 2012, 22, 412–422. [Google Scholar] [CrossRef]
  88. Reissner, C.; Runkel, F.; Missler, M. Neurexins. Genome Biol. 2013, 14, 213. [Google Scholar] [CrossRef]
  89. Südhof, T.C. Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits. Cell 2017, 171, 745–769. [Google Scholar] [CrossRef] [Green Version]
  90. Guang, S.; Pang, N.; Deng, X.; Yang, L.; He, F.; Wu, L.; Chen, C.; Yin, F.; Peng, J. Synaptopathology Involved in Autism Spectrum Disorder. Front. Cell. Neurosci. 2018, 12, 470. [Google Scholar] [CrossRef] [Green Version]
  91. Budreck, E.C.; Scheiffele, P. Neuroligin-3 is a neuronal adhesion protein at GABAergic and glutamatergic synapses. Eur. J. Neurosci. 2007, 26, 1738–1748. [Google Scholar] [CrossRef] [PubMed]
  92. Suzuki, K.; Hayashi, Y.; Nakahara, S.; Kumazaki, H.; Prox, J.; Horiuchi, K.; Zeng, M.; Tanimura, S.; Nishiyama, Y.; Osawa, S.; et al. Activity-dependent proteolytic cleavage of neuroligin-1. Neuron 2012, 76, 410–422. [Google Scholar] [CrossRef] [Green Version]
  93. Trotter, J.H.; Hao, J.; Maxeiner, S.; Tsetsenis, T.; Liu, Z.; Zhuang, X.; Südhof, T.C. Synaptic neurexin-1 assembles into dynamically regulated active zone nanoclusters. J. Cell Biol. 2019, 218, 2677–2698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Hosie, S.; Ellis, M.; Swaminathan, M.; Ramalhosa, F.; Seger, G.O.; Balasuriya, G.K.; Gillberg, C.; Råstam, M.; Churilov, L.; McKeown, S.J.; et al. Gastrointestinal dysfunction in patients and mice expressing the autism-associated R451C mutation in neuroligin-3. Autism Res. 2019, 12, 1043–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Sharna, S.S.; Balasuriya, G.K.; Hosie, S.; Nithianantharajah, J.; Franks, A.E.; Hill-Yardin, E.L. Altered Caecal Neuroimmune Interactions in the Neuroligin-3(R451C) Mouse Model of Autism. Front. Cell. Neurosci. 2020, 14, 85. [Google Scholar] [CrossRef] [PubMed]
  96. Leembruggen, A.J.L.; Balasuriya, G.K.; Zhang, J.; Schokman, S.; Swiderski, K.; Bornstein, J.C.; Nithianantharajah, J.; Hill-Yardin, E.L. Colonic dilation and altered ex vivo gastrointestinal motility in the neuroligin-3 knockout mouse. Autism Res. 2020, 13, 691–701. [Google Scholar] [CrossRef] [Green Version]
  97. Gabriele, S.; Sacco, R.; Altieri, L.; Neri, C.; Urbani, A.; Bravaccio, C.; Riccio, M.P.; Iovene, M.R.; Bombace, F.; De Magistris, L.; et al. Slow intestinal transit contributes to elevate urinary p-cresol level in Italian autistic children. Autism Res. 2016, 9, 752–759. [Google Scholar] [CrossRef] [PubMed]
  98. Walker, S.J.; Langefeld, C.D.; Zimmerman, K.; Schwartz, M.Z.; Krigsman, A. A molecular biomarker for prediction of clinical outcome in children with ASD, constipation, and intestinal inflammation. Sci. Rep. 2019, 9, 5987. [Google Scholar] [CrossRef]
  99. Wang, D.; Pan, J.; Song, G.; Gao, N.; Zheng, Y.; Zhang, Q.; Li, A. Abundance and Significance of Neuroligin-1 and Neurexin II in the Enteric Nervous System of Embryonic Rats. BioMed Res. Int. 2017, 2017, 1209360. [Google Scholar] [CrossRef] [Green Version]
  100. Horder, J.; Petrinovic, M.M.; Mendez, M.A.; Bruns, A.; Takumi, T.; Spooren, W.; Barker, G.J.; Künnecke, B.; Murphy, D.G. Glutamate and GABA in autism spectrum disorder-a translational magnetic resonance spectroscopy study in man and rodent models. Transl. Psychiatry 2018, 8, 106. [Google Scholar] [CrossRef] [Green Version]
  101. Peixoto, R.T.; Kunz, P.A.; Kwon, H.; Mabb, A.M.; Sabatini, B.L.; Philpot, B.D.; Ehlers, M.D. Transsynaptic signaling by activity-dependent cleavage of neuroligin-1. Neuron 2012, 76, 396–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Radyushkin, K.; Hammerschmidt, K.; Boretius, S.; Varoqueaux, F.; El-Kordi, A.; Ronnenberg, A.; Winter, D.; Frahm, J.; Fischer, J.; Brose, N.; et al. Neuroligin-3-deficient mice: Model of a monogenic heritable form of autism with an olfactory deficit. Genes Brain Behav. 2009, 8, 416–425. [Google Scholar] [CrossRef]
  103. Ellegood, J.; Lerch, J.P.; Henkelman, R.M. Brain abnormalities in a Neuroligin3 R451C knockin mouse model associated with autism. Autism Res. 2011, 4, 368–376. [Google Scholar] [CrossRef] [PubMed]
  104. Halbleib, J.M.; Nelson, W.J. Cadherins in development: Cell adhesion, sorting, and tissue morphogenesis. Genes Dev. 2006, 20, 3199–3214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Sano, K.; Tanihara, H.; Heimark, R.L.; Obata, S.; Davidson, M.; St John, T.; Taketani, S.; Suzuki, S. Protocadherins: A large family of cadherin-related molecules in central nervous system. EMBO J. 1993, 12, 2249–2256. [Google Scholar] [CrossRef] [PubMed]
  106. Yasuda, S.; Tanaka, H.; Sugiura, H.; Okamura, K.; Sakaguchi, T.; Tran, U.; Takemiya, T.; Mizoguchi, A.; Yagita, Y.; Sakurai, T.; et al. Activity-induced protocadherin arcadlin regulates dendritic spine number by triggering N-cadherin endocytosis via TAO2beta and p38 MAP kinases. Neuron 2007, 56, 456–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Butler, M.G.; Rafi, S.K.; Hossain, W.; Stephan, D.A.; Manzardo, A.M. Whole exome sequencing in females with autism implicates novel and candidate genes. Int. J. Mol. Sci. 2015, 16, 1312–1335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Breuillard, D.; Leunen, D.; Chemaly, N.; Auclair, L.; Pinard, J.M.; Kaminska, A.; Desguerre, I.; Ouss, L.; Nabbout, R. Autism spectrum disorder phenotype and intellectual disability in females with epilepsy and PCDH-19 mutations. Epilepsy Behav. 2016, 60, 75–80. [Google Scholar] [CrossRef]
  109. Grumet, M.; Mauro, V.; Burgoon, M.P.; Edelman, G.M.; Cunningham, B.A. Structure of a new nervous system glycoprotein, Nr-CAM, and its relationship to subgroups of neural cell adhesion molecules. J. Cell Biol. 1991, 113, 1399–1412. [Google Scholar] [CrossRef]
  110. Zelina, P.; Avci, H.X.; Thelen, K.; Pollerberg, G.E. The cell adhesion molecule NrCAM is crucial for growth cone behaviour and pathfinding of retinal ganglion cell axons. Development 2005, 132, 3609–3618. [Google Scholar] [CrossRef] [Green Version]
  111. Demyanenko, G.P.; Mohan, V.; Zhang, X.; Brennaman, L.H.; Dharbal, K.E.; Tran, T.S.; Manis, P.B.; Maness, P.F. Neural cell adhesion molecule NrCAM regulates Semaphorin 3F-induced dendritic spine remodeling. J. Neurosci. 2014, 34, 11274–11287. [Google Scholar] [CrossRef] [Green Version]
  112. Sakurai, T.; Lustig, M.; Nativ, M.; Hemperly, J.J.; Schlessinger, J.; Peles, E.; Grumet, M. Induction of neurite outgrowth through contactin and Nr-CAM by extracellular regions of glial receptor tyrosine phosphatase beta. J. Cell Biol. 1997, 136, 907–918. [Google Scholar] [CrossRef]
  113. Prox, J.; Bernreuther, C.; Altmeppen, H.; Grendel, J.; Glatzel, M.; D’Hooge, R.; Stroobants, S.; Ahmed, T.; Balschun, D.; Willem, M.; et al. Postnatal disruption of the disintegrin/metalloproteinase ADAM10 in brain causes epileptic seizures, learning deficits, altered spine morphology, and defective synaptic functions. J. Neurosci. 2013, 33, 12915–12928. [Google Scholar] [CrossRef]
  114. Mohan, V.; Sullivan, C.S.; Guo, J.; Wade, S.D.; Majumder, S.; Agarwal, A.; Anton, E.S.; Temple, B.S.; Maness, P.F. Temporal Regulation of Dendritic Spines Through NrCAM-Semaphorin3F Receptor Signaling in Developing Cortical Pyramidal Neurons. Cereb. Cortex 2019, 29, 963–977. [Google Scholar] [CrossRef] [PubMed]
  115. Heyden, A.; Angenstein, F.; Sallaz, M.; Seidenbecher, C.; Montag, D. Abnormal axonal guidance and brain anatomy in mouse mutants for the cell recognition molecules close homolog of L1 and NgCAM-related cell adhesion molecule. Neuroscience 2008, 155, 221–233. [Google Scholar] [CrossRef] [PubMed]
  116. Bonora, E.; Lamb, J.A.; Barnby, G.; Sykes, N.; Moberly, T.; Beyer, K.S.; Klauck, S.M.; Poustka, F.; Bacchelli, E.; Blasi, F.; et al. Mutation screening and association analysis of six candidate genes for autism on chromosome 7q. Eur. J. Hum. Genet. 2005, 13, 198–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hutcheson, H.B.; Olson, L.M.; Bradford, Y.; Folstein, S.E.; Santangelo, S.L.; Sutcliffe, J.S.; Haines, J.L. Examination of NRCAM, LRRN3, KIAA0716, and LAMB1 as autism candidate genes. BMC Med. Genet. 2004, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  118. Rostene, W.; Kitabgi, P.; Parsadaniantz, S.M. Chemokines: A new class of neuromodulator? Nat. Rev. Neurosci. 2007, 8, 895–903. [Google Scholar] [CrossRef]
  119. Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [Green Version]
  120. Harrison, J.K.; Jiang, Y.; Chen, S.; Xia, Y.; Maciejewski, D.; McNamara, R.K.; Streit, W.J.; Salafranca, M.N.; Adhikari, S.; Thompson, D.A.; et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc. Natl. Acad. Sci. USA 1998, 95, 10896–10901. [Google Scholar] [CrossRef] [Green Version]
  121. O’Sullivan, S.A.; Gasparini, F.; Mir, A.K.; Dev, K.K. Fractalkine shedding is mediated by p38 and the ADAM10 protease under pro-inflammatory conditions in human astrocytes. J. Neuroinflamm. 2016, 13, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Hundhausen, C.; Misztela, D.; Berkhout, T.A.; Broadway, N.; Saftig, P.; Reiss, K.; Hartmann, D.; Fahrenholz, F.; Postina, R.; Matthews, V.; et al. The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion. Blood 2003, 102, 1186–1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rogers, J.T.; Morganti, J.M.; Bachstetter, A.D.; Hudson, C.E.; Peters, M.M.; Grimmig, B.A.; Weeber, E.J.; Bickford, P.C.; Gemma, C. CX3CR1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. J. Neurosci. 2011, 31, 16241–16250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Dorfman, M.D.; Krull, J.E.; Douglass, J.D.; Fasnacht, R.; Lara-Lince, F.; Meek, T.H.; Shi, X.; Damian, V.; Nguyen, H.T.; Matsen, M.E.; et al. Sex differences in microglial CX3CR1 signalling determine obesity susceptibility in mice. Nat. Commun. 2017, 8, 14556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Matthews, A.L.; Noy, P.J.; Reyat, J.S.; Tomlinson, M.G. Regulation of A disintegrin and metalloproteinase (ADAM) family sheddases ADAM10 and ADAM17: The emerging role of tetraspanins and rhomboids. Platelets 2017, 28, 333–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Maretzky, T.; McIlwain, D.R.; Issuree, P.D.; Li, X.; Malapeira, J.; Amin, S.; Lang, P.A.; Mak, T.W.; Blobel, C.P. iRhom2 controls the substrate selectivity of stimulated ADAM17-dependent ectodomain shedding. Proc. Natl. Acad. Sci. USA 2013, 110, 11433–11438. [Google Scholar] [CrossRef] [Green Version]
  127. Adrain, C.; Zettl, M.; Christova, Y.; Taylor, N.; Freeman, M. Tumor necrosis factor signaling requires iRhom2 to promote trafficking and activation of TACE. Science 2012, 335, 225–228. [Google Scholar] [CrossRef] [Green Version]
  128. Black, R.A.; Rauch, C.T.; Kozlosky, C.J.; Peschon, J.J.; Slack, J.L.; Wolfson, M.F.; Castner, B.J.; Stocking, K.L.; Reddy, P.; Srinivasan, S.; et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 1997, 385, 729–733. [Google Scholar] [CrossRef]
  129. Scheller, J.; Chalaris, A.; Garbers, C.; Rose-John, S. ADAM17: A molecular switch to control inflammation and tissue regeneration. Trends Immunol. 2011, 32, 380–387. [Google Scholar] [CrossRef]
  130. Ray, B.; Sokol, D.K.; Maloney, B.; Lahiri, D.K. Finding novel distinctions between the sAPPα-mediated anabolic biochemical pathways in Autism Spectrum Disorder and Fragile X Syndrome plasma and brain tissue. Sci. Rep. 2016, 6, 26052. [Google Scholar] [CrossRef]
  131. Bell, J.H.; Herrera, A.H.; Li, Y.; Walcheck, B. Role of ADAM17 in the ectodomain shedding of TNF-alpha and its receptors by neutrophils and macrophages. J. Leukoc. Biol. 2007, 82, 173–176. [Google Scholar] [CrossRef] [PubMed]
  132. Mezyk-Kopec, R.; Bzowska, M.; Stalinska, K.; Chelmicki, T.; Podkalicki, M.; Jucha, J.; Kowalczyk, K.; Mak, P.; Bereta, J. Identification of ADAM10 as a major TNF sheddase in ADAM17-deficient fibroblasts. Cytokine 2009, 46, 309–315. [Google Scholar] [CrossRef] [PubMed]
  133. Lucchina, L.; Depino, A.M. Altered peripheral and central inflammatory responses in a mouse model of autism. Autism Res. 2014, 7, 273–289. [Google Scholar] [CrossRef] [PubMed]
  134. Li, X.; Chauhan, A.; Sheikh, A.M.; Patil, S.; Chauhan, V.; Li, X.M.; Ji, L.; Brown, T.; Malik, M. Elevated immune response in the brain of autistic patients. J. Neuroimmunol. 2009, 207, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Wolf, J.; Rose-John, S.; Garbers, C. Interleukin-6 and its receptors: A highly regulated and dynamic system. Cytokine 2014, 70, 11–20. [Google Scholar] [CrossRef] [PubMed]
  137. Rose-John, S. IL-6 trans-signaling via the soluble IL-6 receptor: Importance for the pro-inflammatory activities of IL-6. Int. J. Biol. Sci. 2012, 8, 1237–1247. [Google Scholar] [CrossRef]
  138. Chalaris, A.; Garbers, C.; Rabe, B.; Rose-John, S.; Scheller, J. The soluble Interleukin 6 receptor: Generation and role in inflammation and cancer. Eur. J. Cell Biol. 2011, 90, 484–494. [Google Scholar] [CrossRef]
  139. Garbers, C.; Janner, N.; Chalaris, A.; Moss, M.L.; Floss, D.M.; Meyer, D.; Koch-Nolte, F.; Rose-John, S.; Scheller, J. Species specificity of ADAM10 and ADAM17 proteins in interleukin-6 (IL-6) trans-signaling and novel role of ADAM10 in inducible IL-6 receptor shedding. J. Biol. Chem. 2011, 286, 14804–14811. [Google Scholar] [CrossRef] [Green Version]
  140. Riethmueller, S.; Somasundaram, P.; Ehlers, J.C.; Hung, C.W.; Flynn, C.M.; Lokau, J.; Agthe, M.; Dusterhoft, S.; Zhu, Y.; Grotzinger, J.; et al. Proteolytic Origin of the Soluble Human IL-6R In Vivo and a Decisive Role of N-Glycosylation. PLoS Biol. 2017, 15, e2000080. [Google Scholar] [CrossRef]
  141. Wei, H.; Chadman, K.K.; McCloskey, D.P.; Sheikh, A.M.; Malik, M.; Brown, W.T.; Li, X. Brain IL-6 elevation causes neuronal circuitry imbalances and mediates autism-like behaviors. Biochim. Biophys. Acta 2012, 1822, 831–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Wei, H.; Ma, Y.; Liu, J.; Ding, C.; Jin, G.; Wang, Y.; Hu, F.; Yu, L. Inhibition of IL-6 trans-signaling in the brain increases sociability in the BTBR mouse model of autism. Biochim. Biophys. Acta 2016, 1862, 1918–1925. [Google Scholar] [CrossRef] [PubMed]
  143. Feuerbach, D.; Schindler, P.; Barske, C.; Joller, S.; Beng-Louka, E.; Worringer, K.A.; Kommineni, S.; Kaykas, A.; Ho, D.J.; Ye, C.; et al. ADAM17 is the main sheddase for the generation of human triggering receptor expressed in myeloid cells (hTREM2) ectodomain and cleaves TREM2 after Histidine 157. Neurosci. Lett. 2017, 660, 109–114. [Google Scholar] [CrossRef] [PubMed]
  144. Bouchon, A.; Hernández-Munain, C.; Cella, M.; Colonna, M. A DAP12-mediated pathway regulates expression of CC chemokine receptor 7 and maturation of human dendritic cells. J. Exp. Med. 2001, 194, 1111–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Filipello, F.; Morini, R.; Corradini, I.; Zerbi, V.; Canzi, A.; Michalski, B.; Erreni, M.; Markicevic, M.; Starvaggi-Cucuzza, C.; Otero, K.; et al. The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity. Immunity 2018, 48, 979–991. [Google Scholar] [CrossRef] [Green Version]
  146. Hsieh, C.L.; Koike, M.; Spusta, S.C.; Niemi, E.C.; Yenari, M.; Nakamura, M.C.; Seaman, W.E. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. J. Neurochem. 2009, 109, 1144–1156. [Google Scholar] [CrossRef] [Green Version]
  147. Zhong, L.; Chen, X.F.; Wang, T.; Wang, Z.; Liao, C.; Wang, Z.; Huang, R.; Wang, D.; Li, X.; Wu, L.; et al. Soluble TREM2 induces inflammatory responses and enhances microglial survival. J. Exp. Med. 2017, 214, 597–607. [Google Scholar] [CrossRef] [Green Version]
  148. Holingue, C.; Newill, C.; Lee, L.C.; Pasricha, P.J.; Daniele Fallin, M. Gastrointestinal symptoms in autism spectrum disorder: A review of the literature on ascertainment and prevalence. Autism Res. 2018, 11, 24–36. [Google Scholar] [CrossRef]
  149. Golubeva, A.V.; Joyce, S.A.; Moloney, G.; Burokas, A.; Sherwin, E.; Arboleya, S.; Flynn, I.; Khochanskiy, D.; Moya-Perez, A.; Peterson, V.; et al. Microbiota-related Changes in Bile Acid & Tryptophan Metabolism are Associated with Gastrointestinal Dysfunction in a Mouse Model of Autism. EBioMedicine 2017, 24, 166–178. [Google Scholar] [CrossRef] [Green Version]
  150. McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal symptoms in autism spectrum disorder: A meta-analysis. Pediatrics 2014, 133, 872–883. [Google Scholar] [CrossRef] [Green Version]
  151. Lasheras, I.; Seral, P.; Latorre, E.; Barroso, E.; Gracia-Garcia, P.; Santabarbara, J. Microbiota and gut-brain axis dysfunction in autism spectrum disorder: Evidence for functional gastrointestinal disorders. Asian J. Psychiatry 2020, 47, 101874. [Google Scholar] [CrossRef] [PubMed]
  152. Fowlie, G.; Cohen, N.; Ming, X. The Perturbance of Microbiome and Gut-Brain Axis in Autism Spectrum Disorders. Int. J. Mol. Sci. 2018, 19, 2251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. de Magistris, L.; Familiari, V.; Pascotto, A.; Sapone, A.; Frolli, A.; Iardino, P.; Carteni, M.; De Rosa, M.; Francavilla, R.; Riegler, G.; et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J. Pediatric Gastroenterol. Nutr. 2010, 51, 418–424. [Google Scholar] [CrossRef] [Green Version]
  154. Liu, Z.; Li, N.; Neu, J. Tight junctions, leaky intestines, and pediatric diseases. Acta Paediatr. 2005, 94, 386–393. [Google Scholar] [CrossRef] [PubMed]
  155. Banks, W.A. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain Behav. Immun. 2015, 44, 1–8. [Google Scholar] [CrossRef] [Green Version]
  156. Erickson, M.A.; Dohi, K.; Banks, W.A. Neuroinflammation: A common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation 2012, 19, 121–130. [Google Scholar] [CrossRef] [Green Version]
  157. Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.S.; Knapp, D.J.; Crews, F.T. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef] [Green Version]
  158. Schulz, B.; Pruessmeyer, J.; Maretzky, T.; Ludwig, A.; Blobel, C.P.; Saftig, P.; Reiss, K. ADAM10 regulates endothelial permeability and T-Cell transmigration by proteolysis of vascular endothelial cadherin. Circ. Res. 2008, 102, 1192–1201. [Google Scholar] [CrossRef] [Green Version]
  159. Candela, P.; Saint-Pol, J.; Kuntz, M.; Boucau, M.C.; Lamartiniere, Y.; Gosselet, F.; Fenart, L. In vitro discrimination of the role of LRP1 at the BBB cellular level: Focus on brain capillary endothelial cells and brain pericytes. Brain Res. 2015, 1594, 15–26. [Google Scholar] [CrossRef]
  160. Storck, S.E.; Meister, S.; Nahrath, J.; Meißner, J.N.; Schubert, N.; Di Spiezio, A.; Baches, S.; Vandenbroucke, R.E.; Bouter, Y.; Prikulis, I.; et al. Endothelial LRP1 transports amyloid-β(1-42) across the blood-brain barrier. J. Clin. Investig. 2016, 126, 123–136. [Google Scholar] [CrossRef] [Green Version]
  161. Shackleton, B.; Crawford, F.; Bachmeier, C. Inhibition of ADAM10 promotes the clearance of Aβ across the BBB by reducing LRP1 ectodomain shedding. Fluids Barriers CNS 2016, 13, 14. [Google Scholar] [CrossRef] [Green Version]
  162. Liang, S.J.; Li, X.G.; Wang, X.Q. Notch Signaling in Mammalian Intestinal Stem Cells: Determining Cell Fate and Maintaining Homeostasis. Curr. Stem Cell Res. Ther. 2019, 14, 583–590. [Google Scholar] [CrossRef] [PubMed]
  163. Dhaliwal, K.K.; Orsso, C.E.; Richard, C.; Haqq, A.M.; Zwaigenbaum, L. Risk Factors for Unhealthy Weight Gain and Obesity among Children with Autism Spectrum Disorder. Int. J. Mol. Sci. 2019, 20, 3285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Puig, K.L.; Brose, S.A.; Zhou, X.; Sens, M.A.; Combs, G.F.; Jensen, M.D.; Golovko, M.Y.; Combs, C.K. Amyloid precursor protein modulates macrophage phenotype and diet-dependent weight gain. Sci. Rep. 2017, 7, 43725. [Google Scholar] [CrossRef]
  165. Puig, K.L.; Manocha, G.D.; Combs, C.K. Amyloid precursor protein mediated changes in intestinal epithelial phenotype in vitro. PLoS ONE 2015, 10, e0119534. [Google Scholar] [CrossRef] [PubMed]
  166. Puig, K.L.; Swigost, A.J.; Zhou, X.; Sens, M.A.; Combs, C.K. Amyloid precursor protein expression modulates intestine immune phenotype. J. Neuroimmune Pharmacol. 2012, 7, 215–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. An, Y.A.; Crewe, C.; Asterholm, I.W.; Sun, K.; Chen, S.; Zhang, F.; Shao, M.; Funcke, J.B.; Zhang, Z.; Straub, L.; et al. Dysregulation of Amyloid Precursor Protein Impairs Adipose Tissue Mitochondrial Function and Promotes Obesity. Nat. Metab. 2019, 1, 1243–1257. [Google Scholar] [CrossRef] [PubMed]
  168. Chalaris, A.; Adam, N.; Sina, C.; Rosenstiel, P.; Lehmann-Koch, J.; Schirmacher, P.; Hartmann, D.; Cichy, J.; Gavrilova, O.; Schreiber, S.; et al. Critical role of the disintegrin metalloprotease ADAM17 for intestinal inflammation and regeneration in mice. J. Exp. Med. 2010, 207, 1617–1624. [Google Scholar] [CrossRef] [Green Version]
  169. Wang, F.; Graham, W.V.; Wang, Y.; Witkowski, E.D.; Schwarz, B.T.; Turner, J.R. Interferon-gamma and tumor necrosis factor-alpha synergize to induce intestinal epithelial barrier dysfunction by up-regulating myosin light chain kinase expression. Am. J. Pathol. 2005, 166, 409–419. [Google Scholar] [CrossRef]
  170. Moriez, R.; Salvador-Cartier, C.; Theodorou, V.; Fioramonti, J.; Eutamene, H.; Bueno, L. Myosin light chain kinase is involved in lipopolysaccharide-induced disruption of colonic epithelial barrier and bacterial translocation in rats. Am. J. Pathol. 2005, 167, 1071–1079. [Google Scholar] [CrossRef] [Green Version]
  171. Peschon, J.J.; Slack, J.L.; Reddy, P.; Stocking, K.L.; Sunnarborg, S.W.; Lee, D.C.; Russell, W.E.; Castner, B.J.; Johnson, R.S.; Fitzner, J.N.; et al. An essential role for ectodomain shedding in mammalian development. Science 1998, 282, 1281–1284. [Google Scholar] [CrossRef] [PubMed]
  172. Bambini-Junior, V.; Zanatta, G.; Della Flora Nunes, G.; Mueller de Melo, G.; Michels, M.; Fontes-Dutra, M.; Nogueira Freire, V.; Riesgo, R.; Gottfried, C. Resveratrol prevents social deficits in animal model of autism induced by valproic acid. Neurosci. Lett. 2014, 583, 176–181. [Google Scholar] [CrossRef]
  173. Ahmad, S.F.; Ansari, M.A.; Nadeem, A.; Bakheet, S.A.; Alzahrani, M.Z.; Alshammari, M.A.; Alanazi, W.A.; Alasmari, A.F.; Attia, S.M. Resveratrol attenuates pro-inflammatory cytokines and activation of JAK1-STAT3 in BTBR T(+) Itpr3(tf)/J autistic mice. Eur. J. Pharmacol. 2018, 829, 70–78. [Google Scholar] [CrossRef] [PubMed]
  174. Masi, A.; Quintana, D.S.; Glozier, N.; Lloyd, A.R.; Hickie, I.B.; Guastella, A.J. Cytokine aberrations in autism spectrum disorder: A systematic review and meta-analysis. Mol. Psychiatry 2015, 20, 440–446. [Google Scholar] [CrossRef] [PubMed]
  175. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef]
  178. Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef]
  179. Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef]
  180. Sampson, T.R.; Mazmanian, S.K. Control of brain development, function, and behavior by the microbiome. Cell Host Microbe 2015, 17, 565–576. [Google Scholar] [CrossRef] [Green Version]
  181. Coretti, L.; Paparo, L.; Riccio, M.P.; Amato, F.; Cuomo, M.; Natale, A.; Borrelli, L.; Corrado, G.; Comegna, M.; Buommino, E.; et al. Gut Microbiota Features in Young Children With Autism Spectrum Disorders. Front. Microbiol. 2018, 9, 3146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Emanuele, E.; Orsi, P.; Boso, M.; Broglia, D.; Brondino, N.; Barale, F.; di Nemi, S.U.; Politi, P. Low-grade endotoxemia in patients with severe autism. Neurosci. Lett. 2010, 471, 162–165. [Google Scholar] [CrossRef] [PubMed]
  183. Guijarro-Muñoz, I.; Compte, M.; Álvarez-Cienfuegos, A.; Álvarez-Vallina, L.; Sanz, L. Lipopolysaccharide activates Toll-like receptor 4 (TLR4)-mediated NF-κB signaling pathway and proinflammatory response in human pericytes. J. Biol. Chem. 2014, 289, 2457–2468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Park, B.S.; Lee, J.O. Recognition of lipopolysaccharide pattern by TLR4 complexes. Exp. Mol. Med. 2013, 45, e66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Fiebich, B.L.; Batista, C.R.A.; Saliba, S.W.; Yousif, N.M.; de Oliveira, A.C.P. Role of Microglia TLRs in Neurodegeneration. Front. Cell. Neurosci. 2018, 12, 329. [Google Scholar] [CrossRef] [Green Version]
  186. Yao, L.; Kan, E.M.; Lu, J.; Hao, A.; Dheen, S.T.; Kaur, C.; Ling, E.A. Toll-like receptor 4 mediates microglial activation and production of inflammatory mediators in neonatal rat brain following hypoxia: Role of TLR4 in hypoxic microglia. J. Neuroinflamm. 2013, 10, 23. [Google Scholar] [CrossRef] [Green Version]
  187. Horiuchi, K.; Kimura, T.; Miyamoto, T.; Takaishi, H.; Okada, Y.; Toyama, Y.; Blobel, C.P. Cutting edge: TNF-alpha-converting enzyme (TACE/ADAM17) inactivation in mouse myeloid cells prevents lethality from endotoxin shock. J. Immunol. 2007, 179, 2686–2689. [Google Scholar] [CrossRef] [Green Version]
  188. Maretzky, T.; Scholz, F.; Köten, B.; Proksch, E.; Saftig, P.; Reiss, K. ADAM10-mediated E-cadherin release is regulated by proinflammatory cytokines and modulates keratinocyte cohesion in eczematous dermatitis. J. Investig. Dermatol. 2008, 128, 1737–1746. [Google Scholar] [CrossRef] [Green Version]
  189. Alshammari, M.K.; AlKhulaifi, M.M.; Al Farraj, D.A.; Somily, A.M.; Albarrag, A.M. Incidence of Clostridium perfringens and its toxin genes in the gut of children with autism spectrum disorder. Anaerobe 2020, 61, 102114. [Google Scholar] [CrossRef]
  190. Gora, B.; Gofron, Z.; Grosiak, M.; Aptekorz, M.; Kazek, B.; Kocelak, P.; Radosz-Komoniewska, H.; Chudek, J.; Martirosian, G. Toxin profile of fecal Clostridium perfringens strains isolated from children with autism spectrum disorders. Anaerobe 2018, 51, 73–77. [Google Scholar] [CrossRef]
  191. Popoff, M.R.; Bouvet, P. Clostridial toxins. Future Microbiol. 2009, 4, 1021–1064. [Google Scholar] [CrossRef] [PubMed]
  192. Seike, S.; Takehara, M.; Kobayashi, K.; Nagahama, M. Clostridium perfringens Delta-Toxin Damages the Mouse Small Intestine. Toxins 2019, 11, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Seike, S.; Takehara, M.; Takagishi, T.; Miyamoto, K.; Kobayashi, K.; Nagahama, M. Delta-toxin from Clostridium perfringens perturbs intestinal epithelial barrier function in Caco-2 cell monolayers. Biochim. Biophys. Acta Biomembr. 2018, 1860, 428–433. [Google Scholar] [CrossRef] [PubMed]
  194. Persico, A.M.; Napolioni, V. Urinary p-cresol in autism spectrum disorder. Neurotoxicol. Teratol. 2013, 36, 82–90. [Google Scholar] [CrossRef]
  195. Altieri, L.; Neri, C.; Sacco, R.; Curatolo, P.; Benvenuto, A.; Muratori, F.; Santocchi, E.; Bravaccio, C.; Lenti, C.; Saccani, M.; et al. Urinary p-cresol is elevated in small children with severe autism spectrum disorder. Biomarkers 2011, 16, 252–260. [Google Scholar] [CrossRef]
  196. Mishra, H.K.; Johnson, T.J.; Seelig, D.M.; Walcheck, B. Targeting ADAM17 in leukocytes increases neutrophil recruitment and reduces bacterial spread during polymicrobial sepsis. J. Leukoc. Biol. 2016, 100, 999–1004. [Google Scholar] [CrossRef] [Green Version]
  197. Xu, J.; Sriramula, S.; Xia, H.; Moreno-Walton, L.; Culicchia, F.; Domenig, O.; Poglitsch, M.; Lazartigues, E. Clinical Relevance and Role of Neuronal AT(1) Receptors in ADAM17-Mediated ACE2 Shedding in Neurogenic Hypertension. Circ. Res. 2017, 121, 43–55. [Google Scholar] [CrossRef]
  198. Pedersen, K.B.; Chodavarapu, H.; Porretta, C.; Robinson, L.K.; Lazartigues, E. Dynamics of ADAM17-Mediated Shedding of ACE2 Applied to Pancreatic Islets of Male db/db Mice. Endocrinology 2015, 156, 4411–4425. [Google Scholar] [CrossRef] [Green Version]
  199. Cole-Jeffrey, C.T.; Liu, M.; Katovich, M.J.; Raizada, M.K.; Shenoy, V. ACE2 and Microbiota: Emerging Targets for Cardiopulmonary Disease Therapy. J. Cardiovasc. Pharmacol. 2015, 66, 540–550. [Google Scholar] [CrossRef] [Green Version]
  200. Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef]
  201. Santisteban, M.M.; Qi, Y.; Zubcevic, J.; Kim, S.; Yang, T.; Shenoy, V.; Cole-Jeffrey, C.T.; Lobaton, G.O.; Stewart, D.C.; Rubiano, A.; et al. Hypertension-Linked Pathophysiological Alterations in the Gut. Circ. Res. 2017, 120, 312–323. [Google Scholar] [CrossRef] [PubMed]
  202. Lerner, A.; Neidhöfer, S.; Matthias, T. The Gut Microbiome Feelings of the Brain: A Perspective for Non-Microbiologists. Microorganisms 2017, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  203. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  205. Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698–3703. [Google Scholar] [CrossRef] [Green Version]
  206. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  207. Peng, L.; Li, Z.R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef]
  208. Wang, H.B.; Wang, P.Y.; Wang, X.; Wan, Y.L.; Liu, Y.C. Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein Claudin-1 transcription. Dig. Dis. Sci. 2012, 57, 3126–3135. [Google Scholar] [CrossRef]
  209. Buffington, S.A.; Di Prisco, G.V.; Auchtung, T.A.; Ajami, N.J.; Petrosino, J.F.; Costa-Mattioli, M. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016, 165, 1762–1775. [Google Scholar] [CrossRef]
  210. Cerwenka, A.; Swain, S.L. TGF-beta1: Immunosuppressant and viability factor for T lymphocytes. Microbes Infect. 1999, 1, 1291–1296. [Google Scholar] [CrossRef]
  211. Ashwood, P.; Enstrom, A.; Krakowiak, P.; Hertz-Picciotto, I.; Hansen, R.L.; Croen, L.A.; Ozonoff, S.; Pessah, I.N.; Van de Water, J. Decreased transforming growth factor beta1 in autism: A potential link between immune dysregulation and impairment in clinical behavioral outcomes. J. Neuroimmunol. 2008, 204, 149–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Okada, K.; Hashimoto, K.; Iwata, Y.; Nakamura, K.; Tsujii, M.; Tsuchiya, K.J.; Sekine, Y.; Suda, S.; Suzuki, K.; Sugihara, G.; et al. Decreased serum levels of transforming growth factor-beta1 in patients with autism. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 31, 187–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Al-Ayadhi, L.; Alhowikan, A.M.; Halepoto, D.M. Impact of Auditory Integrative Training on Transforming Growth Factor-β1 and Its Effect on Behavioral and Social Emotions in Children with Autism Spectrum Disorder. Med. Princ. Pract. Int. J. Kuwait Univ. Health Sci. Cent. 2018, 27, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Butovsky, O.; Jedrychowski, M.P.; Moore, C.S.; Cialic, R.; Lanser, A.J.; Gabriely, G.; Koeglsperger, T.; Dake, B.; Wu, P.M.; Doykan, C.E.; et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 2014, 17, 131–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Taylor, R.A.; Chang, C.F.; Goods, B.A.; Hammond, M.D.; Mac Grory, B.; Ai, Y.; Steinschneider, A.F.; Renfroe, S.C.; Askenase, M.H.; McCullough, L.D.; et al. TGF-β1 modulates microglial phenotype and promotes recovery after intracerebral hemorrhage. J. Clin. Investig. 2017, 127, 280–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Chen, J.H.; Ke, K.F.; Lu, J.H.; Qiu, Y.H.; Peng, Y.P. Protection of TGF-β1 against neuroinflammation and neurodegeneration in Aβ1-42-induced Alzheimer’s disease model rats. PLoS ONE 2015, 10, e0116549. [Google Scholar] [CrossRef] [Green Version]
  217. Depino, A.M.; Lucchina, L.; Pitossi, F. Early and adult hippocampal TGF-β1 overexpression have opposite effects on behavior. Brain Behav. Immun. 2011, 25, 1582–1591. [Google Scholar] [CrossRef]
  218. Kawasaki, K.; Freimuth, J.; Meyer, D.S.; Lee, M.M.; Tochimoto-Okamoto, A.; Benzinou, M.; Clermont, F.F.; Wu, G.; Roy, R.; Letteboer, T.G.; et al. Genetic variants of Adam17 differentially regulate TGFβ signaling to modify vascular pathology in mice and humans. Proc. Natl. Acad. Sci. USA 2014, 111, 7723–7728. [Google Scholar] [CrossRef] [Green Version]
  219. Liu, C.; Xu, P.; Lamouille, S.; Xu, J.; Derynck, R. TACE-mediated ectodomain shedding of the type I TGF-beta receptor downregulates TGF-beta signaling. Mol. Cell 2009, 35, 26–36. [Google Scholar] [CrossRef] [Green Version]
  220. Malapeira, J.; Esselens, C.; Bech-Serra, J.J.; Canals, F.; Arribas, J. ADAM17 (TACE) regulates TGFβ signaling through the cleavage of vasorin. Oncogene 2011, 30, 1912–1922. [Google Scholar] [CrossRef] [Green Version]
  221. Ikeda, Y.; Imai, Y.; Kumagai, H.; Nosaka, T.; Morikawa, Y.; Hisaoka, T.; Manabe, I.; Maemura, K.; Nakaoka, T.; Imamura, T.; et al. Vasorin, a transforming growth factor beta-binding protein expressed in vascular smooth muscle cells, modulates the arterial response to injury in vivo. Proc. Natl. Acad. Sci. USA 2004, 101, 10732–10737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Wilke, C.M.; Bishop, K.; Fox, D.; Zou, W. Deciphering the role of Th17 cells in human disease. Trends Immunol. 2011, 32, 603–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Choi, G.B.; Yim, Y.S.; Wong, H.; Kim, S.; Kim, H.; Kim, S.V.; Hoeffer, C.A.; Littman, D.R.; Huh, J.R. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 2016, 351, 933–939. [Google Scholar] [CrossRef] [Green Version]
  224. Kuchroo, V.K.; Awasthi, A. Emerging new roles of Th17 cells. Eur. J. Immunol. 2012, 42, 2211–2214. [Google Scholar] [CrossRef] [PubMed]
  225. Bettelli, E.; Carrier, Y.; Gao, W.; Korn, T.; Strom, T.B.; Oukka, M.; Weiner, H.L.; Kuchroo, V.K. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, 441, 235–238. [Google Scholar] [CrossRef]
  226. Smith, S.E.; Li, J.; Garbett, K.; Mirnics, K.; Patterson, P.H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 2007, 27, 10695–10702. [Google Scholar] [CrossRef] [Green Version]
  227. Yan, I.; Schwarz, J.; Lücke, K.; Schumacher, N.; Schumacher, V.; Schmidt, S.; Rabe, B.; Saftig, P.; Donners, M.; Rose-John, S.; et al. ADAM17 controls IL-6 signaling by cleavage of the murine IL-6Rα from the cell surface of leukocytes during inflammatory responses. J. Leukoc. Biol. 2016, 99, 749–760. [Google Scholar] [CrossRef]
  228. Liu, S.; Fu, Y.; Mei, K.; Jiang, Y.; Sun, X.; Wang, Y.; Ren, F.; Jiang, C.; Meng, L.; Lu, S.; et al. A shedding soluble form of interleukin-17 receptor D exacerbates collagen-induced arthritis through facilitating TNF-α-dependent receptor clustering. Cell. Mol. Immunol. 2020. [Google Scholar] [CrossRef]
  229. Baker, A.H.; Edwards, D.R.; Murphy, G. Metalloproteinase inhibitors: Biological actions and therapeutic opportunities. J. Cell Sci. 2002, 115, 3719–3727. [Google Scholar] [CrossRef] [Green Version]
  230. Brew, K.; Dinakarpandian, D.; Nagase, H. Tissue inhibitors of metalloproteinases: Evolution, structure and function. Biochim. Biophys. Acta 2000, 1477, 267–283. [Google Scholar] [CrossRef]
  231. Amour, A.; Knight, C.G.; Webster, A.; Slocombe, P.M.; Stephens, P.E.; Knäuper, V.; Docherty, A.J.; Murphy, G. The in vitro activity of ADAM-10 is inhibited by TIMP-1 and TIMP-3. FEBS Lett. 2000, 473, 275–279. [Google Scholar] [CrossRef] [Green Version]
  232. Amour, A.; Slocombe, P.M.; Webster, A.; Butler, M.; Knight, C.G.; Smith, B.J.; Stephens, P.E.; Shelley, C.; Hutton, M.; Knäuper, V.; et al. TNF-alpha converting enzyme (TACE) is inhibited by TIMP-3. FEBS Lett. 1998, 435, 39–44. [Google Scholar] [CrossRef] [Green Version]
  233. Arpino, V.; Brock, M.; Gill, S.E. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol. J. Int. Soc. Matrix Biol. 2015, 44, 247–254. [Google Scholar] [CrossRef] [PubMed]
  234. Sawicki, G.; Menon, V.; Jugdutt, B.I. Improved balance between TIMP-3 and MMP-9 after regional myocardial ischemia-reperfusion during AT1 receptor blockade. J. Card. Fail. 2004, 10, 442–449. [Google Scholar] [CrossRef]
  235. Hoettecke, N.; Ludwig, A.; Foro, S.; Schmidt, B. Improved synthesis of ADAM10 inhibitor GI254023X. Neuro-Degener. Dis. 2010, 7, 232–238. [Google Scholar] [CrossRef]
  236. Ludwig, A.; Hundhausen, C.; Lambert, M.H.; Broadway, N.; Andrews, R.C.; Bickett, D.M.; Leesnitzer, M.A.; Becherer, J.D. Metalloproteinase inhibitors for the disintegrin-like metalloproteinases ADAM10 and ADAM17 that differentially block constitutive and phorbol ester-inducible shedding of cell surface molecules. Comb. Chem. High Throughput Screen. 2005, 8, 161–171. [Google Scholar] [CrossRef]
  237. Mahasenan, K.V.; Ding, D.; Gao, M.; Nguyen, T.T.; Suckow, M.A.; Schroeder, V.A.; Wolter, W.R.; Chang, M.; Mobashery, S. In Search of Selectivity in Inhibition of ADAM10. ACS Med. Chem. Lett. 2018, 9, 708–713. [Google Scholar] [CrossRef]
  238. Hirata, S.; Murata, T.; Suzuki, D.; Nakamura, S.; Jono-Ohnishi, R.; Hirose, H.; Sawaguchi, A.; Nishimura, S.; Sugimoto, N.; Eto, K. Selective Inhibition of ADAM17 Efficiently Mediates Glycoprotein Ibα Retention During Ex Vivo Generation of Human Induced Pluripotent Stem Cell-Derived Platelets. Stem Cells Transl. Med. 2017, 6, 720–730. [Google Scholar] [CrossRef]
  239. Abdellatif, B.; McVeigh, C.; Bendriss, G.; Chaari, A. The Promising Role of Probiotics in Managing the Altered Gut in Autism Spectrum Disorders. Int. J. Mol. Sci. 2020, 21, 4159. [Google Scholar] [CrossRef]
  240. Navarro, F.; Liu, Y.; Rhoads, J.M. Can probiotics benefit children with autism spectrum disorders? World J. Gastroenterol. 2016, 22, 10093–10102. [Google Scholar] [CrossRef]
  241. Liu, J.; Wan, G.B.; Huang, M.S.; Agyapong, G.; Zou, T.L.; Zhang, X.Y.; Liu, Y.W.; Song, Y.Q.; Tsai, Y.C.; Kong, X.J. Probiotic Therapy for Treating Behavioral and Gastrointestinal Symptoms in Autism Spectrum Disorder: A Systematic Review of Clinical Trials. Curr. Med. Sci. 2019, 39, 173–184. [Google Scholar] [CrossRef] [PubMed]
  242. Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejía, J.L.; Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome 2018, 6, 133. [Google Scholar] [CrossRef]
  243. Carlezon, W.A., Jr.; Kim, W.; Missig, G.; Finger, B.C.; Landino, S.M.; Alexander, A.J.; Mokler, E.L.; Robbins, J.O.; Li, Y.; Bolshakov, V.Y.; 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, 16928. [Google Scholar] [CrossRef] [PubMed]
  244. Zhang, W.; Lu, F.; Xie, Y.; Lin, Y.; Zhao, T.; Tao, S.; Lai, Z.; Wei, N.; Yang, R.; Shao, Y.; et al. miR-23b Negatively Regulates Sepsis-Induced Inflammatory Responses by Targeting ADAM10 in Human THP-1 Monocytes. Mediat. Inflamm. 2019, 2019, 5306541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. De Theije, C.G.; Wopereis, H.; Ramadan, M.; van Eijndthoven, T.; Lambert, J.; Knol, J.; Garssen, J.; Kraneveld, A.D.; Oozeer, R. Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav. Immun. 2014, 37, 197–206. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 2012, 57, 2096–2102. [Google Scholar] [CrossRef] [PubMed]
  247. Ristori, M.V.; Quagliariello, A.; Reddel, S.; Ianiro, G.; Vicari, S.; Gasbarrini, A.; Putignani, L. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients 2019, 11, 2812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F., 3rd; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
  249. Gauthier, J.; Bonnel, A.; St-Onge, J.; Karemera, L.; Laurent, S.; Mottron, L.; Fombonne, E.; Joober, R.; Rouleau, G.A. NLGN3/NLGN4 gene mutations are not responsible for autism in the Quebec population. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2005, 132, 74–75. [Google Scholar] [CrossRef]
  250. Talebizadeh, Z.; Bittel, D.C.; Veatch, O.J.; Butler, M.G.; Takahashi, T.N.; Miles, J.H. Do known mutations in neuroligin genes (NLGN3 and NLGN4) cause autism? J. Autism Dev. Disord. 2004, 34, 735–736. [Google Scholar] [CrossRef]
  251. Jamain, S.; Quach, H.; Betancur, C.; Råstam, M.; Colineaux, C.; Gillberg, I.C.; Soderstrom, H.; Giros, B.; Leboyer, M.; Gillberg, C.; et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat. Genet. 2003, 34, 27–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Földy, C.; Malenka, R.C.; Südhof, T.C. Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron 2013, 78, 498–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Protein structure of the members of the metzincin family of metalloproteases: A Disintegrin And Metalloprotease (ADAM), Snake Venom Metalloprotease (SVMP), A Disintegrin And Metalloprotease Thrombospondin motif (ADAMTS), membrane-type matrix metalloproteinases (MT-MMP) and MMP.
Figure 1. Protein structure of the members of the metzincin family of metalloproteases: A Disintegrin And Metalloprotease (ADAM), Snake Venom Metalloprotease (SVMP), A Disintegrin And Metalloprotease Thrombospondin motif (ADAMTS), membrane-type matrix metalloproteinases (MT-MMP) and MMP.
Ijms 22 00118 g001
Figure 2. Overview of cleavage products of the Amyloid Precursor Protein (APP) by different secretases. The cleavage of APP by the β-site APP cleaving enzyme 1 (BACE1) initiates the amyloidogenic pathway, where the soluble fraction of APP (sAPP)β and C-Terminal Fragment 99 (CTF99) are created. γ-secretases further process the CTF99 to create the neurotoxic Aβ protein and the Amyloid Precursor Intracellular Domain (AICD). Meantime, sAPPβ is also cleaved by additional secretases at an unknown site to generate an N-terminal of APP (N-APP). The cleavage of APP by either ADAM10 or ADAM17 initiates the nonamyloidogenic pathway, which creates sAPPα and C-Terminal Fragment 83 (CTF83). γ-secretases then cleave CTF83 to create P3 and AICD.
Figure 2. Overview of cleavage products of the Amyloid Precursor Protein (APP) by different secretases. The cleavage of APP by the β-site APP cleaving enzyme 1 (BACE1) initiates the amyloidogenic pathway, where the soluble fraction of APP (sAPP)β and C-Terminal Fragment 99 (CTF99) are created. γ-secretases further process the CTF99 to create the neurotoxic Aβ protein and the Amyloid Precursor Intracellular Domain (AICD). Meantime, sAPPβ is also cleaved by additional secretases at an unknown site to generate an N-terminal of APP (N-APP). The cleavage of APP by either ADAM10 or ADAM17 initiates the nonamyloidogenic pathway, which creates sAPPα and C-Terminal Fragment 83 (CTF83). γ-secretases then cleave CTF83 to create P3 and AICD.
Ijms 22 00118 g002
Figure 3. Overview of the proposed role of ADAM10 and ADAM17 in ASD pathology, with a focus on the gut–immune–brain axis. In the intestines, the increased activity or levels of ADAM10 and ADAM17 will lead to an increased intestinal permeability by cleaving more E-cadherin and Notch, increased intestinal inflammation by cleaving more IL-6R and TNF-α, disrupted intestinal transit by cleaving more NLGN3 and increased myelination of nervus vagus by cleaving more DR6 in the Enteric Nervous System; these contribute to the intestinal dysfunctions. In the brain, the elevated activity of ADAM10 and ADAM17 will result in increased neuronal growth, synaptic density, dendritic spines densities and larger brain volume by cleaving more synaptic molecules, such as NLGN, NRXN, NrCAM and APP. Furthermore, it will result in decreased synaptic elimination and microglial phagocytosis by cleaving more CX3CL1 and TREM2; increased neuroinflammation by cleaving more IL-6R, TNF-α and TREM2 and increased BBB permeability by cleaving more VE-cadherin and LRP1. These processes will lead to altered brain development and functions. Additionally, increased ADAM10 and ADAM17 activity will lead to immune activation by cleaving more IL-17R, IL-6R and TNF-α. All of these can participate ASD development and be involved in ASD pathogenesis.
Figure 3. Overview of the proposed role of ADAM10 and ADAM17 in ASD pathology, with a focus on the gut–immune–brain axis. In the intestines, the increased activity or levels of ADAM10 and ADAM17 will lead to an increased intestinal permeability by cleaving more E-cadherin and Notch, increased intestinal inflammation by cleaving more IL-6R and TNF-α, disrupted intestinal transit by cleaving more NLGN3 and increased myelination of nervus vagus by cleaving more DR6 in the Enteric Nervous System; these contribute to the intestinal dysfunctions. In the brain, the elevated activity of ADAM10 and ADAM17 will result in increased neuronal growth, synaptic density, dendritic spines densities and larger brain volume by cleaving more synaptic molecules, such as NLGN, NRXN, NrCAM and APP. Furthermore, it will result in decreased synaptic elimination and microglial phagocytosis by cleaving more CX3CL1 and TREM2; increased neuroinflammation by cleaving more IL-6R, TNF-α and TREM2 and increased BBB permeability by cleaving more VE-cadherin and LRP1. These processes will lead to altered brain development and functions. Additionally, increased ADAM10 and ADAM17 activity will lead to immune activation by cleaving more IL-17R, IL-6R and TNF-α. All of these can participate ASD development and be involved in ASD pathogenesis.
Ijms 22 00118 g003
Table 1. The reduction in shedding of the A Disintegrin And Metalloprotease (ADAM)10 substrates after the conditional deletion of ADAM10 in embryonic primary cortical neuron cultures [35]. Abbreviations: Fractalkine (Cx3cl1), Neuroligin-1 (NLGN-1), Protocadherin-9 (PCDH9), Neural glial-related Cell Adhesion Molecule (NrCAM), Neuroligin-3 (NLGN-3) and Amyloid-β Precursor Protein (APP).
Table 1. The reduction in shedding of the A Disintegrin And Metalloprotease (ADAM)10 substrates after the conditional deletion of ADAM10 in embryonic primary cortical neuron cultures [35]. Abbreviations: Fractalkine (Cx3cl1), Neuroligin-1 (NLGN-1), Protocadherin-9 (PCDH9), Neural glial-related Cell Adhesion Molecule (NrCAM), Neuroligin-3 (NLGN-3) and Amyloid-β Precursor Protein (APP).
ADAM10 SubstratesShedding Reduction in ADAM10–/– Neurons
Cx3cl191%
NLGN-183%
PCDH971%
NrCAM66%
NLGN-362%
APP20%
Table 2. The effect of ADAM10 and ADAM17 on different substrates and their involvement in Autism Spectrum Disorder (ASD) patients. ↓: downregulated; ↑: upregulated
Table 2. The effect of ADAM10 and ADAM17 on different substrates and their involvement in Autism Spectrum Disorder (ASD) patients. ↓: downregulated; ↑: upregulated
Protein NameGene SymbolADAM10 SheddingADAM17 SheddingASD Patients
Amyloid Precursor ProteinAPP↑sAPPα [32]↑sAPPα [32]↑ sAPPα [28]
Neuroligin-1NLGN-1↓ Synaptogenic activity [92] Common variants [249,250]
Neuroligin-3NLGN-3↓ Synaptogenic activity [92] R415C transition [251,252]
Common variants [250]
Neurexin-1NRXN-1↓ Synaptogenic activity [93] Loss-of-function variants [24,26,27]
Neural glial-related Cell Adhesion MoleculeNrCAM↑ Axon targeting activity [35] SNPs & Common variants [25]
Protocadherin9PCDH9No data available Copy Number Variants [24]
FractalkineCX3CL1↑ Synaptic pruning [35,121,122] No data available
Tumor Necrosis Factor-αTNF-α ↑ pro-inflammatory activity [131]↑ in blood and brain [134]
Interleukin-6 ReceptorIL-6R↑ pro-inflammatory pathways [139]↑ pro-inflammatory pathways [139]↑ IL-6 in blood and brain [134]
Triggering Receptor Expressed in Myeloid cells 2TREM2 ↓ TREM2 membrane receptor levels [143]↓ in post-mortem brain tissue age 5–23 [145]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zheng, Y.; Verhoeff, T.A.; Perez Pardo, P.; Garssen, J.; Kraneveld, A.D. The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17. Int. J. Mol. Sci. 2021, 22, 118. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010118

AMA Style

Zheng Y, Verhoeff TA, Perez Pardo P, Garssen J, Kraneveld AD. The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17. International Journal of Molecular Sciences. 2021; 22(1):118. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010118

Chicago/Turabian Style

Zheng, Yuanpeng, Tessa A. Verhoeff, Paula Perez Pardo, Johan Garssen, and Aletta D. Kraneveld. 2021. "The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17" International Journal of Molecular Sciences 22, no. 1: 118. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22010118

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

Article Metrics

Back to TopTop