Next Article in Journal
High Mobility Group Box 1 (HMGB1): Potential Target in Sepsis-Associated Encephalopathy
Next Article in Special Issue
Glucocorticoid Hormones as Modulators of the Kynurenine Pathway in Chronic Pain Conditions
Previous Article in Journal
Quadra-Stable Dynamics of p53 and PTEN in the DNA Damage Response
Previous Article in Special Issue
Differential Levels of Tryptophan–Kynurenine Pathway Metabolites in the Hippocampus, Anterior Temporal Lobe, and Neocortex in an Animal Model of Temporal Lobe Epilepsy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder

1
Department of Hygiene and Public Health, Kansai Medical University, Hirakata 573-1010, Japan
2
Department of Architecture and Architectual Systems Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
3
Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Suita 565-0871, Japan
4
Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
5
Department of Biology, Kanazawa Medical University, Kanazawa 920-0293, Japan
6
Center for Baby Science, Doshisha University, Kyotanabe 619-0225, Japan
7
Healthcare and Medical Data Multi-Level Integration Platform Group, RIKEN Medical Sciences Innovation Hub Program, Yokohama 230-0045, Japan
*
Author to whom correspondence should be addressed.
Deceased 6 September 2019.
Cells 2023, 12(7), 1087; https://doi.org/10.3390/cells12071087
Submission received: 7 February 2023 / Revised: 27 March 2023 / Accepted: 31 March 2023 / Published: 4 April 2023
(This article belongs to the Special Issue Kynurenine Pathway in Health and Disease)

Abstract

:
Several studies show that genetic and environmental factors contribute to the onset and progression of neurodevelopmental disorders. Maternal immune activation (MIA) during gestation is considered one of the major environmental factors driving this process. The kynurenine pathway (KP) is a major route of the essential amino acid L-tryptophan (Trp) catabolism in mammalian cells. Activation of the KP following neuro-inflammation can generate various endogenous neuroactive metabolites that may impact brain functions and behaviors. Additionally, neurotoxic metabolites and excitotoxicity cause long-term changes in the trophic support, glutamatergic system, and synaptic function following KP activation. Therefore, investigating the role of KP metabolites during neurodevelopment will likely promote further understanding of additional pathophysiology of neurodevelopmental disorders, including autism spectrum disorder (ASD). In this review, we describe the changes in KP metabolism in the brain during pregnancy and represent how maternal inflammation and genetic factors influence the KP during development. We overview the patients with ASD clinical data and animal models designed to verify the role of perinatal KP elevation in long-lasting biochemical, neuropathological, and behavioral deficits later in life. Our review will help shed light on new therapeutic strategies and interventions targeting the KP for neurodevelopmental disorders.

1. Introduction

Maternal inflammation with elevated kynurenine (Kyn) metabolites has been implicated in developing various central nervous system (CNS) disorders. Kyn metabolites are produced as part of the tryptophan (Trp) catabolism pathway, and increased levels of these metabolites have been found in response to inflammation. Maternal inflammation during pregnancy can increase in pro-inflammatory cytokines, which can stimulate the production of Kyn metabolites.
Studies have shown that elevated inflammation and Kyn metabolites during pregnancy are associated with an increased risk of several CNS disorders, including autism spectrum disorder (ASD) [1,2,3], schizophrenia (SCZ) [4,5], and depression [3,6,7]. In ASD patients, for example, alterations in the Kyn pathway (KP) have been observed in both the peripheral and central nervous systems [1,2,8]. It has been shown that inflammation during pregnancy may contribute to the dysregulation of the KP and increase the risk of ASD. Similarly, elevated levels of Kyn metabolites, especially kynurenic acid (Kyna), have been observed in individuals with SCZ [9,10,11]. It has been shown that inflammation and oxidative stress may contribute to the dysregulation of the KP in this disorder. In depression, altered levels of Kyn metabolites have also been reported, with higher levels of Kyn and lower levels of its downstream metabolite, Kyna, observed in individuals with depression [12,13]. The relationship between maternal inflammation with elevated Kyn metabolites and CNS disorders is complex and poorly understood. However, studies show that dysregulation of the KP because of inflammation may contribute to the development of various CNS disorders.
ASD is a complex developmental disorder associated with impaired social interactions/affective function, communication abnormalities, stereotypic behavior, a repetitive repertoire of interests and activities [14], and secondary causes of cognitive impairment and depression. In 2018, the Autism and Developmental Disabilities Monitoring (ADDM) Network reported that one in 44 (23.0 per 1000) children aged 8 years were estimated to have ASD, and the ASD prevalence has kept increasing in the last decade [15,16]. Additionally, several genetic factors of ASD have been identified, and twin studies estimate its heritability to be 64–91% [17,18]. However, maternal immune activation (MIA) following viral infection is associated with an increased ASD prevalence and is one of the most impacting environmental risk factors [19,20,21].
Recent studies have demonstrated that the neuroimmune response and the abnormality of KP metabolites are linked to the pathophysiology of ASD. Some evidence demonstrates that children with ASD show increased levels of serum inflammatory cytokines [8,22]. As the KP of Trp degradation is activated in neuroinflammatory states, previous studies also demonstrate inflammation-induced KP activation in children with ASD [1,8]. Activation of the KP can generate various endogenous neuroactive KP metabolites, such as Kyn and Kyna. In particular, increasing evidence shows that these metabolites play important and unique roles in brain development [23]. We summarize the possible relationship between the KP, as well as its neuroactive metabolites, and inflammation and also the risk of abnormal brain development and behavioral changes to provide insights into the pathological mechanisms of ASD.

2. Interleukin (IL)-17a as a Potential Mediator of ASD

2.1. Pro-Inflammatory Cytokines and ASD

An altered and abnormal immune system has been widely observed in the periphery of patients with ASD and in experimental animal models. Several studies have found chronic neuroinflammation to be associated with ASD, indicated by an increased number of activated microglia and astrocytes, and the production of cytokines and chemokines in the brain [8,24,25,26,27,28]. Additionally, ASD has been associated with chronic inflammatory and allergic conditions, such as maternal infection and autoimmune diseases in the first trimester of pregnancy [19,29,30,31]. In the brain of the patient with ASD, IL-6 elevation was a repeated finding [24,27,32], and consistent with these findings, Smith et al. showed that maternal IL-6 was critical for mediating the behavioral and transcriptional changes in the offspring using IL-6 blocking antibody and IL-6 gene-deficient mice [33]. Moreover, Rudolph et al. indicated that functional connectivity within and between multiple neonatal brain networks could be modeled to estimate the maternal IL-6 concentration during pregnancy [34]. However, IL-6 is a multifaceted signal capable of triggering the expression of other cytokines and immune regulatory genes that may additionally impact the developing brain or are responsible for precipitating ASD. T helper (Th)17 cells have been proposed to play important roles in immune responses against extracellular bacteria and fungi. Their dysregulation is related to various autoimmune diseases, such as allergies, asthma, rheumatoid arthritis, and inflammatory bowel diseases (IBDs) [35,36]. The differentiation of Th17 cells from naïve Th cells is initiated by stimulation with professional antigen-presenting cells (APCs) and particular cytokines, including IL-6, IL-21, and transforming growth factor-beta (TGF-β) [36,37,38,39]. Maturated Th17 cells produce the signature cytokines IL-17a, IL-17f, and IL-21, essential for mucosal host defense against extracellular bacteria and fungi, and recruit neutrophils by inducing chemokines under inflammation [36]. Recently, cytokines produced by Th17 cells have been shown to have a role in ASD. IL-17a is the predominant Th17 cytokine, and elevated levels of IL-17a have been found in the blood and correlated with the severity of behavioral symptoms in individuals with ASD [8,28,40,41]. A MIA model mouse injected intraperitoneally with synthetic double-stranded RNA [polyinosinic:polycytidylic acid; poly(I:C)], a mimic of viral infection, showed increased levels of IL-17a in the maternal blood and postnatal fetal brain [42,43]. Offspring from poly(I:C)-treated dam showed ASD-like behaviors, such as abnormal communication and social interest, and increased stereotypic and anxiety-like behaviors [44]. Inhibited IL-17a signaling by antibody blockade of the cytokine in poly(I:C)-treated pregnant mice can prevent ASD-like phonotypes in the offspring [42].
Additionally, mice directly injected with IL-17a into the fetal lateral ventricles on embryonic day (E) 14.5 showed similar ASD-like behaviors and cortical disorganization as the offspring of poly(I:C)-treated dam [42], and activated cortical microglia, which excessively phagocytosed neuronal progenitor cells in the ventricular zone [45]. Furthermore, chronic gestational IL-17a causes ASD-like phenotypes early and persistently in male offspring and leads to 320 differentially expressed genes related to “neuron-neuron synaptic transmission” and “cell cycles” [46]. IL-17a also blocks the proliferation of neuronal stem cells, resulting in a significantly reduced number of astrocytes and oligodendrocyte precursor cells [47]. Kim et al. showed that ASD phenotypes following MIA in offspring necessitate maternal intestinal bacteria, including segmented filamentous bacteria (SFB), to promote Th17 cell differentiation and produce IL-17a [48]. Pretreatment of antibiotic vancomycin in poly(I:C)-injected dam canceled the development of all behavioral abnormalities in MIA-offspring. Generally, in all the findings, dysregulation of maternal Th17 differentiation or the Th17/IL-17a pathway may play a critical role in the pathophysiology of MIA-induced ASD.

2.2. Proinflammatory Cytokines Regulate KP Enzymes

The correlation between proinflammatory cytokines and KP enzymes is well-known. Indoleamine 2,3-dioxygenase1 (IDO1) is an enzyme that is the first rate-limiting step of Trp degradation and leads to the production of a series of downstream metabolites in KP. During pregnancy, IDO1 is mainly expressed in the trophoblast [49] and placenta [50] to protect the fetus from the attack of the maternal immune system by inhibition of T-lymphocyte responses through Trp consumption and Trp catabolism defects [51,52]. In maternal circulation, the ratio of Kyn and Trp (Kyn/Trp ratio) is also significantly increased in a normal pregnancy at late gestation stages, compared to non-pregnant women [53]. CD4-positive Th cells may release various cytokines after stimulation, such as classified Th1-, Th2-, and Th17-type cytokines. The predominant Th2 immunity, which overrules the Th1 immunity at the placental implantation site, protects the fetus by balancing the Th1 immunity and accommodates fetal and placental development [54]. IL-6 is a major Th2-type cytokine, which can induce the upregulation of IDO1 in the chorionic villi and decidua of women in early pregnancy, revealing that Th-2type cytokines can induce maternal immunotolerance via activated IDO1 [55,56]. In the normal physiological state of very early gestation (around 8 weeks of gestation), IL-6 upregulates the IDO1 expression in chorionic villi and decidua by enhancing SC-43-activated SH2-domain-containing phosphatase (SHP)-1/2 expression via signal transducers and activators of transcription (STAT) 3 and phosphorylated-STAT3 [57], and high expression of IL-6 may help promote immunological tolerance and a successful pregnancy. However, Th2 cells may participate in autoantibody production and enhance autoimmunity. In addition, overly increased tolerogenic signals from Th2 cells may induce uncontrolled viral infections [54]. The fetal ZIKA virus infection causes microcephaly, and an association is reported with ASD, along with the discovery of the predominant expression of Th2 cytokines, including IL-6, in the meninges, perivascular space, and parenchyma [54,58]. These findings show that adequate timing and properly immuno-activated Th2 are important for immunotolerance and fetus protection from infection.
Tryptophan 2,3-dioxygenase (TDO) is the other key enzyme in the KP, and it is mainly expressed in the liver and maintains systemic Trp levels by degrading excess dietary Trp under normal physiological conditions [59,60]. TDO is expressed pathologically in various tumors [61]. Kyn, catabolized from Trp by IDO1 and TDO, suppresses antitumor immune responses and promotes tumor-cell survival [62]. TDO was also identified in pericytes and interstitial syncytiotrophoblasts in the human placenta [63], and continuous TDO expression was observed in mouse decidual stromal cells, starting at E3.5 until gestation end. Despite the highly expressed TDO in decidual stromal cells, TDO-deficient and IDO1/TDO-double-deficient female mice did not show increased rates of miscarriage because of an absence of an increased immune attack against allogenic fetuses [60]. These results show that TDO may not be a dominant mechanism of maternal immunotolerance able to compensate for the absence of IDO1. IL-1β is a multifunctional cytokine and one of the first cytokines released by macrophages, monocytes, and dendritic cells during an infection. In addition, IL-1β is essential for efficient innate and adaptive immune responses [64], and previous research shows that IL-1β stimulates KP and regulates the production of IL-6 secretion by increasing the TDO expression [65]. However, little information is available on regulating IL-1β and TDO in normal pregnancy. A few reports on tumor immunology indicate a relationship between inflammation and TDO induction, and TDO has immunomodulatory functions in promoting tumor resistance and proliferation [61,62,65,66,67].
In the peripheral blood of healthy pregnant women, Th17 cells are rare among CD4-positive T cells (0.64–1.4%), and the number of circulating Th17 cells does not change during pregnancy [68]. Another study reported that pregnant women in the third trimester presented with a decreased proportion of Th17 cells compared to non-pregnant women [69]. The decidua contains a higher density of Th17 cells than the peripheral blood [68]. The decidual IL-17a-positive cell count was consistent with the neutrophil count, showing that IL-17a-positive cells are intimately involved in neutrophil infiltration [70] and induce protective immunity against extracellular microbes in the uterus. The paradoxical association between IL-17a and the induction of IDO1 in opportunistic infections is well known. In a fungal infection with physiological conditions, the IL-17a pathway downregulated the Trp catabolism and completely antagonized the induction IDO1 by interferon-gamma (IFN-γ) in neutrophils [71]. Although little information is available on the regulation of IL-17a and KP in normal pregnancy, Krause et al. demonstrated that candidemic patients with antibiotic therapy had significantly higher IL-17a and Kyn levels than non-candidemic patients [72]. In addition, we demonstrated that maternal overexpression of IL-17a induced significantly elevated levels of Kyn and KP metabolites in maternal serum and fetal plasma [3].
Cytokines and KP metabolites are closely associated with mediating CNS and immune system communication. Both molecules regulate not only neuronal cells, such as neurons and in glia activity, but also several immune cells, such as in leukocytes activity. Stone et al. show that cytokines and KP metabolites perform complementary functions, generating an integrated network related to some neuroimmune communications [73]. The KP is a key to understanding how systemic inflammation can affect brain function. Additionally, the KP metabolites can influence the surveillance, defensive, and tolerance activities of the immune system. Overall, KP and its metabolites may play key roles in the pathogenesis of normal and abnormal pregnancies. The induction of KP by inflammation may influence the neurodevelopment of the fetus.

3. The Metabolism of Trp in the Intestine and the Role of Microbiota in ASD

3.1. Intestinal Trp Metabolism Pathways and Physiological Roles

Trp metabolism follows three major pathways in the gastrointestinal (GI) tract: (1) the KP in both host immune cells and epithelial cells via IDO1 (details in next part) [74]; (2) the serotonin (5-hydroxytryptamine; 5-HT) production pathway in host enterochromaffin cells via Trp hydroxylase (THP) 1 [75]; and the direct transformation of Trp into several molecules, including aryl hydrocarbon receptor (AhR) ligands, by the gut microbiota [76,77,78].
TPH2 produces the neurotransmitter 5-HT, which plays an important role in the brain (see the other part). More than 90% of the body’s 5-HT is produced in the intestine through TPH1. Peripheral 5-HT triggers various functions in the GI tract and is implicated in a wide range of physiological functions through its activation of specific 5-HT receptors [79,80]. Intestinal 5-HT has been found to modulate intrinsic or extrinsic neurons and to influence intestinal peristalsis and the motility, secretion, vasodilation, and absorption of nutrients [79,81,82]. Additionally, intestinal 5-HT was shown to have a hormonal role in bone formation by studies of the low-density receptor-related protein (LRP) 5, which works with its co-receptors to activate the Wnt-β-catenin signaling pathway [83]. The gut microbiota also produces major intestinal 5-HT [75]. Germ-free mice exhibit significantly decreased colonic and fecal 5-HT production levels and low blood concentration of 5-HT [75,84]. The mechanisms of how the gut microbiota regulates 5-HT production remain unknown. However, a few studies demonstrated that some metabolites, such as short-chain fatty acids (SCFAs) or secondary bile acids, can modulate 5-HT biosynthesis [75,85].
The degradation of dietary proteins leads to the release of Trp, which is directly converted by gut microorganisms into various metabolites, such as indole and its derivatives. Many indole derivatives, including indole acrylic acid (IA), indole-3-acetaaldehyde (IAAld), indole-3-acetic acid (IAA), indole-3-propionic acid (IPA), and indole-3-aldehyde (IAld), are ligands for AhR [86,87]. Only a few commensal species can produce AhR ligands, such as Lactobacillus spp and Peptostreptococcus russellii [76,88,89]. AhR is a key receptor, which regulates the immune response at barrier sites, barrier integrity, and homeostasis by acting on epithelial renewal and many immune cells’ differentiation [90]. Especially, Trp metabolites play an important role in the differentiation and function of several immune cells, such as T-regulatory cells (Tregs), B-regulatory cells (Bregs), IL-22-producing innate lymphocyte cells 3 (ILC3s), and anti-inflammatory macrophages in the gut, because Th17 cells, Tregs, B cells, and APCs express AhR [91]. Trp metabolizing pathways have been identified in other species, such as Clostridium sporogenes, Clostridium limosum, Escherichia coli (E. coli), Enterococcus faecalis, and Bacteroides ovatus [78,89,92]. Clostridium sporogenes can decarboxylate Trp to produce the neurotransmitter tryptamine [92]. In the brain, tryptamine has been shown to activate trace dopaminergic, serotonergic, and glutamatergic systems [93]. In the GI tract, tryptamine activates the 5-HT4 receptor and regulates GI motility [94]. Clostridium sporogenes also oxidate and reduce to produce IAA and IPA, which are known to affect intestinal permeability and host immunity [90,95,96]. Clostridium limosum, E. coli, Enterococcus faecalis, and Bacteroides ovatus can convert Trp into indole [78,97], an interspecies signaling molecule that can control aspects of bacterial physiology, such as sporulation, antibiotic resistance, and biofilm formation.

3.2. The Regulation of Trp Metabolism by Gut Microbiota

Gut microbiota can influence the KP via the alteration of Trp availability or the regulation of the immune system, which can affect the activity of IDO1. In germ-free mice or mice with an altered microbial composition due to antibiotics, the plasma levels of Trp were increased, while the levels of KP metabolites and the peripheral 5-HT levels were decreased. Consistent with these changes, the Kyn/Trp ratio was reduced, which indicated the lower activity of IDO1 and TDO. Notably, the induction of some gut microbiotas, such as Bifidobacterium infantis, could restore the normal activity of these two enzymes [74,75,98,99,100,101]. The gut microorganism can degrade Trp into several metabolites that consequently limit the availability of Trp for the KP and 5-HT pathways, as shown in the previous paragraph.
Gut microbiota and IDO1 also have feedback control over each other. IDO1 can induce an immunosuppressive response in the GI tract by regulating immune reactivity and microbial metabolism. Contrarily, gut microbiota can change the amount of KP metabolites and IDO1 activity by limiting Trp usage. Additionally, some metabolites derived from microbiota have anti-inflammatory effects and modulate the immune system and KP via the regulation of IDO1. SCFAs are one of the major metabolites from microbiota and play a significant role in intestinal homeostasis. It has been demonstrated that butyrate (one of the SCFAs) can downregulate the expression of STAT1, which is one of the main mediators of IDO1 expression [102]. Decreased STAT1 expression inhibits IFN-γ-dependent STAT1 phosphorylation and subsequently reduces the STAT-1-dependent transcriptional activity of IDO1. SCFAs also can inhibit histone deacetylase (HDAC) [103], and the downregulation of HDAC could suppress the production of several proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, IFN-γ, and IL-6 [104,105]. Therefore, SCFAs could inhibit IDO1 activity in an indirect manner.

3.3. Dysbiosis in ASD

Among several comorbidities in ASD, GI distress is reportedly related to the prevalence of and a correlation with symptom severity. Dysbiosis of the microbiota is implicated not only in the pathogenesis of ASD, but also in several chronic diseases, such as IBD, allergy, asthma, cardiovascular disease, obesity, and diabetes mellitus. Especially, evidence of microbial dysbiosis in ASD has been growing in the last two decades [106,107,108,109,110,111,112,113,114,115]. A recent systematic review shows that ASD patients had an elevated abundance Proteobacteria, Clostridium, and Bacteroides, while they had lower levels of Bifidobacterium, Prevotella, and Blautia, compared to healthy controls [116]. The elevated abundance of Proteobacteria is associated with host inflammation because this microbe can produce lipopolysaccharide (LPS). Clostridium also can produce pro-inflammatory toxins and propionic acid that may be related to the severity of ASD symptoms [117,118]. Sandler et al. showed that oral vancomycin treatment improved behavioral and GI symptoms in ASD children by reducing Clostridium [106]. Bacteroides produce SCFAs, especially propionic acid, and SCFAs are involved in the proper function of the gut immune system through the modulation of gene expression. Therefore, an imbalance in the concentration of SCFAs can alter the gut homeostasis and trigger peripheral inflammation. SCFAs can cross the blood–brain barrier (BBB) via monocarboxylate transporters located on endothelial cells and influence brain development by modulation of 5-HT and dopamine production [119,120]. Contrarily, some Bifidobacterium species synthesize gamma-amino butyric acid (GABA), which is found in lower concentrations in ASD children. Prevotella species have essential genes for the biosynthesis of vitamin B1, which was reported to palliate ASD symptoms [121,122]. Blautia species have roles in bile acid and Trp metabolism in the intestine, which is related to 5-HT synthesis and accelerating GI motility [75,123].
In addition to immune and GI dysfunction that may be linked to dysbiosis, there is some evidence that altering the microbiota can modulate ASD behaviors in mice [124,125,126]. Hsiao et al. found that GI barrier defects and microbiota alterations in the MIA offspring and oral treatment of MIA offspring with the human commensal Bacteroides fragilis improved their gut permeability and altered their microbial composition. Additionally, this treatment ameliorates defects in anxiety-like behavior, ultrasonic vocalization, the marble burying test with stereotyped behavior, and the pre-pulse inhibition (PPI) test with sensorimotor gating [125]. They also demonstrated that specific metabolites are altered in MIA offspring and normalized by Bacteroides fragilis treatment, with at least two molecules (4-ethylphenylsulfate and indole pyruvate) having potential relevance to ASD. In addition, reflecting another alteration in Trp metabolites, serum 5-HT was also increased. Sharon et al. demonstrated that transferring human ASD gut microbiota into germ-free mice is sufficient to induce hallmark ASD-like behaviors [126]. They also find that microbiota from ASD and typically developing (TD) individuals produce differential metabolome profiles in mice. Especially, 5-aminovaleric acid (5AV) and taurine, which are weak GABAA receptor agonists, are significantly decreased in ASD offspring. Lower levels of GABA agonists show that gut microbes may impact inhibitory GABA signaling in the brain, which is related to ASD behaviors.
All accumulated evidence clarifies that the gut microbiota is either directly or indirectly associated with the pathogenesis of ASD. Microbial balance may influence brain development through the neuroendocrine, neuroimmune, and autonomic nervous systems. Additionally, the changes of Trp-related metabolites in the intestine may significantly impact the host’s physiological conditions and brain function.

4. Perinatal KP Metabolism

4.1. Neurodevelopment and KP Metabolites

KP metabolite levels in the fetal brain are higher during the perinatal period [127], decrease in the immediate postnatal period, and remain lower in adulthood under physiological conditions [128]. A recent systematic review also indicated that physiological pregnancy requires a tight balance of KP metabolites [129]. A major source of these KP metabolites is provided from the mother to the fetus via transplacental transfer and Trp degradation in the placenta [128]. As mentioned in previous parts, the placenta expresses IDO1/TDO, and also other KP enzymes, such as kynureninase, kynurenine aminotransferase (KAT), kynurenine 3-monooxygenase (KMO), and quinolic acid phosphoribosyltransferase (QPRT) (Figure 1). In line with the expression of KP enzymes, its metabolites, Kyna, 3-hydrooxygense (3-HK), and quinolinic acid (QUIN), have been detected in the placenta [128]. Prenatal administration of Kyn and KMO inhibitors leads to biochemical and structural abnormalities in the rat hippocampus [130,131]. Previous studies showed that prolonged administration of a high concentration of Kyn to pregnant mice or rats results in elevated Kyn and Kyna (but not 3-HK) levels in specific areas of the fetal brain, parallel with distinct abnormal social behaviors and cognitive abnormalities in adult offspring [3,23,132]. Kyna, which is produced primarily by irreversible enzymatic transamination of Kyn, is an endogenous antagonist of the N-methyl-D-aspartate (NMDA) receptor [133] and α7 nicotinic acetylcholine receptor (α7nAChR) [134], and the levels of Kyna in the fetal brain under physiological conditions are high in several species, including mammalians [135]. Although Kyna does not cross the BBB in adulthood [136], it can directly transfer to the fetal or neonatal brain from circulation [137]. In the postnatal brain, Kyna levels immediately decline after birth. However, elevated Kyna levels have been found in the postmortem brain and cerebrospinal fluid of individuals with psychiatric disorders. One is SCZ, a severe mental disease from early neurodevelopment [11]. In experimental animals, increased brain Kyna concentrations during the perinatal period cause several cognitive impairments, consistent with that reported in SCZ patients [3,23,132]. Additionally, endogenous Kyn and Kyna levels are markedly increased in KMO-gene-deficient mice brains, and offspring of KMO-gene-deficient mice exhibit anxiety- and depression-like behavior [138] and several ASD-like behaviors [139]. Therefore, high Kyn, Kyna, or both levels in the brain may have a specific role in the normal and abnormal neurodevelopment of the fetus. However, little is still known about the roles of KP metabolites during the neurodevelopment process, and further investigation under physiological/pathological conditions is required to understand the ASD etiology.

4.2. Relation between Key Receptors and KP Metabolites in Developing Brain

During early brain development, NMDA, α7nACh, and aryl hydrocarbon receptors are key receptors targeted by KP metabolites. NMDA receptors are one of the glutamate receptors and are involved in neuronal cell migration [140], neurogenesis [141], axon guidance, synapse formation [142], and spine density [143]. Clinical studies on ASDs have identified genetic variants of NMDA receptor subunit genes. Specifically, de novo mutations have been identified in the GRIN2B gene, encoding the GluN2B subunit [144,145,146,147,148]. Additionally, many single nucleotide polymorphisms (SNPs) of GRIN2A (GluN2A subunit) and GRIN2B are linked with ASDs [149]. Pharmacological research shows that NMDA receptor agonist [D-cycloserine (DCS)] or antagonists (Memantine) can modulate ASD-related symptoms, including social deficits, stereotypy, and cognitive impairments [150,151,152,153]. Furthermore, animal studies have supported the contribution of NMDA receptor dysfunction to ASDs. In parallel with human research, positive or negative modulation of NMDA receptors can also normalize animal ASD-like behavior [154,155,156,157,158]. Generally, with clinical and animal studies, these results indicated that the optimal range of NMDA receptor function is important, and deviations in either direction can lead to shared behavioral impairments [159].
The homomeric α7nAChR subtype is abundantly present in the CNS/peripheral tissues and plays a key role in synaptic plasticity and various disease pathogenesis [160,161]. Several studies have shown a highly regulated expression of α7nAChRs in the developing brain during periods critical for establishing synaptic plasticity. A widespread distribution of α7nAChR mRNA is reported throughout the embryonic mouse nervous systems, highlighting the ubiquitous expression of α7nAChR mRNA in the central, peripheral, and enteric nervous systems during embryonic development [162]. In addition, the role of α7nAChRs in the pathogenesis of ASD has been investigated by several experimental and clinical studies. A larger and increased number of neurons were reported in the basal forebrain, a site of origin of cholinergic projections in the CNS, in children with ASD. In contrast, smaller and fewer neurons were reported in adults with ASD than in controls, indicating a functional disruption of cholinergic transmission in patients with ASD [163]. Pharmacological administration of a selective α7nAChR agonist (choline) from the beginning of pregnancy throughout lactation attenuated some of the deleterious ASD-like behaviors following MIA on the development of the offspring [164]. Additionally, the BTBR T+Itpr3tf/J mouse (BTBR), identified only a decade ago as displaying strong and consistent ASD-relevant behaviors, has shown decreased ACh levels and increased levels of Kyna in the medial prefrontal cortex [165]. Pharmacological administration of α7nAChR agonist (ALV-3288 or nicotine) significantly attenuated the deleterious ASD-like behaviors in BTBR mice [166,167]. Administering the acetylcholinesterase inhibitor or positive allosteric modulator of α7nAChR to children with ASD showed beneficial effects in clinical trials [168,169]. Moreover, it is well known that the mutations of the human chromosome 15q13.3 have been identified in the context of multiple neurological and psychiatric disorders, such as ASD and SCZ [170]. One of the striking genes in 15q13.3 is CHRNA7, which encodes α7nAChR [171]. In a clinical setting, significantly decreased levels of expression of CHRNA7 have been revealed in the frontal cortex of patients with Rett syndrome, one of the neurodevelopment disorders strongly associated with ASD [172]. All these findings indicate that regulating α7nAChR activity during neuronal development is important, and endogenous Kyna may help balance the activation of these receptors.
Kyna, an astrocyte-derived product of the KP, is well-known for its neuroprotective and neuroinhibitory properties, which have been attributed to its action as a competitive antagonist at the glycine site on NMDA receptors with higher concentrations [173] and the allosteric site on the α7nAChRs at physiological levels [134]. Even though the sensitivity to inhibition of α7nAChR by Kyna is age-dependent [174], several electrophysiological and animal experiments show that α7nAChRs are the preferential target of endogenous Kyna in the brain during neurodevelopment [134,175,176]. α7nAChR is abundantly expressed in the CNS and located at pre- and postsynaptic sites [177], and neuronal α7nAChRs appear early during brain development [162]. Additionally, functional nAChRs responses can be found not only in neurons, but also in non-excitable cells, including microglia [178], astrocytes [179], Schwann cells [180], and other non-neuronal tissues [181], and these responses are often mediated by α7nAChRs. Notably, even relatively modest increased levels of Kyna in the brain negatively modulate the release of several neurotransmitters, such as glutamate [182,183,184], GABA [185], dopamine [186], and ACh [187]. These neurotransmitters are essential to the function of complex neural systems. Furthermore, there is evidence that α7nAChRs regulate the GABAA receptor function and the developmental GABAergic switch from excitation to inhibition in ganglia and hippocampal neurons [188,189,190]. During critical phases of brain development, impaired neurotransmitter functions disrupt the maturation of the excitatory/inhibitory balance in cortical transmission, resulting in cognitive impairments and social abnormalities in ASD, SCZ, or both. Therefore, excessive blockage of α7nAChRs by abnormally elevated Kyna may be related to these behavioral deficits [5,11,176]. Further research will be needed to investigate the effect of Kyna on the maturation of the excitatory/inhibitory balance and other synaptic transmission systems during neuronal development.
AhR is a ligand-activated transcription factor that regulates cell differentiation, proliferation, and cancer imitation. Therefore, activation of AhR is related to the pathogenesis of several diseases, such as cancer, cardiovascular disease, inflammatory diseases, atherosclerosis, and neurodegenerative diseases [191]. In adults, AhR is widely distributed and expressed in various tissues and regions of the brain [192]. However, expression of AhR during fetal development is very limited in the placenta and epithelial cells of the fetus in physiological conditions. An experimental study shows that excessive activation of AhR signaling in neurons during embryonic development disrupts neuronal migration in the hippocampus [193]. It shows that AhR overactivation impairs neuronal growth and the neuronal circuit structure. In addition, during the developmental period, mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which is the strong ligand for AhR, were shown to reveal AhR in neurons of the locus coeruleus (LC) and the island of Calleja major (ICjM) [194]. Kyn and Kyna are well-known to have AhR-ligand activity [62,195,196]. AhR-gene-deficient mice demonstrated increased levels of Kyna in specific brain areas associated with higher expression of KAT II. Furthermore, these animals were protected against neurological damage of excitotoxic QUIN by high levels of Kyna [197]. Generally, these studies disclose the multiplicity of biological actions of Kyna in the CNS, depending on the neuronal physiological condition.

4.3. Kynurenine Profile in ASD

There is increasing evidence that altered immune responses play a role in the pathogenesis of ASD, together with dysfunction of the glutamatergic and serotonergic systems. A few studies investigated the pro-inflammatory cytokine profile in patients with ASD [43,198]. In contrast, others focused on glutamatergic imbalance and toxicity as neuroinflammation markers [8] and dysfunction of the serotoninergic systems as diagnostic markers [199,200]. Among the KP metabolites, QUIN is involved in neurotoxicity during several inflammatory neuronal diseases because QUIN can activate NMDA receptors, increase neuronal activity, and elevate the intracellular calcium concentration [133,201]. Excessive activation of NMDA receptors leads to consequent impairment of the cytoskeleton homeostasis, with mitochondrial dysfunction and cell death induction [202]. High cerebral levels of QUIN, working as an NMDA agonist, can alter the excitation/inhibition ratio of the NMDA receptor and increase neuronal glutamate release, inhibiting its reuptake by astrocytes, and blocking astroglial glutamine synthetase, leading to excessive micro-environmental glutamate concentrations [203]. Lim et al. reported that an increased Kyn/Trp ratio and the production of Kyn and QUIN in children with ASD were consistent with increased levels of several pro-inflammatory cytokines, including IL-6 and IL-17a [8]. However, there were no significant changes in the concentration of Kyna compared to healthy children. However, other research shows no changes in Kyn and QUIN and lower levels of Kyna in ASD children [1].
Additionally, Carpita et al. investigated the changes in KP metabolites in ASD adults, and they found significantly lower levels of Trp and QUIN and no changes in Kyn and Kyna [2]. Recently, a meta-analysis comprising all KP metabolites and keywords related to maternal pregnancy and the fetal outcome showed that an altered KP metabolite concentration is significantly related to a high risk of preeclampsia, fetal growth restriction, and preterm birth [129]. These are all considered risk factors for ASD [204,205,206]. Even though single compound changes of KP are still controversial in each clinical study (Table 1) [1,2,8,207,208,209,210,211,212,213,214,215,216,217,218,219], it is clear that an abnormal balance of KP metabolites may be associated with adverse fetal neurodevelopment and the pathogenesis of ASD.
In addition to altered KP metabolites, it is well-known that ASD patients display high 5-HT blood levels [200,220,221] and decreased levels of central 5-HT [222]. The gene that encodes TPH, a rate-limiting enzyme for 5-HT synthesis, has been associated with ASD and other neurological disorders [223,224]. Moreover, a systematic review of common genetic variation in ASD showed that enriched pathways in the over-representation analysis are mostly associated with neurotransmitter receptors and their subunits, including 5-HT, GABA, and glutamate receptors [225]. During brain development, 5-HT has been shown to modulate numerous events, including cell division, neuronal migration, cell differentiation, and synaptogenesis, even before it becomes important as a neurotransmitter [226,227]. A pharmacological study showed that the administration of the 5-HT-depleting drug (p-chlorophenylalanine) to pregnant rats delays the onset of the differentiation of 5-HT neurons [228]. Consistently, human ASD model mice with 15q11.13 duplication (15q dup mice) show lower levels of 5-HT in all brain regions during developmental stages, and pharmacologically restoring 5-HT levels in 15q dup offspring improves the cortical excitation/inhibition balance and rescues the impaired social behavior in adulthood [222]. Metabolized from Trp, 5-HT has been long since described as reducing 5-HT levels caused by Trp depletion [229,230]. Most dietary Trp (~95%) is metabolized via KP in the liver, and only a small fraction of the Trp pool is converted to 5-HT. In addition, a systematic review of the effects of acute Trp depletion on human brain function shows that acute Trp depletion impairs the consolidation of episodic memory for verbal information involved in ASD pathology [231]. Therefore, even small changes in the activity of the KP can significantly impact the Trp pool and normal levels of 5-HT in the brain during neuronal development.

5. Experimental Animal Models with Pre- and Postnatal KP Confusion to Study Neuropathological and Behavioral Deficits

As described in previous parts, the levels of KP metabolite in the developing brain are altered by environmental stimuli, including maternal infection and stress-causing MIA. It is well-known that maternal infections or stress are the most influential environmental risk factors for the offspring to develop psychiatric disorders, such as SCZ, depression, and ASD [6,232,233]. Furthermore, in experimental animal models, it is shown that MIA or maternal stress during pregnancy altered maternal and fetal KP metabolites [3,234,235]. Recent evidence has been accumulated on the impact of long-lasting changes of KP metabolites in the offspring involved in neuropathological and behavioral deficits (Table 2) [3,5,23,130,138,139,165,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252].
Impaired KMO function has been implicated in the pathophysiology of SCZ [9,10,11,257] because decreased KMO activity is directly related to elevated levels of Kyna. Either pharmacological KMO inhibition or genomic deletion of the KMO gene in mice demonstrated reduced KMO activity and induced a shift in KP metabolism toward increasing the levels of Kyna. KMO-gene-deficit mice demonstrate increased basal Kyna from late gestation at E17-18 [128]. Conversely, 3-HK concentrations are almost undetectable in the placenta and fetal brain [138,258]. KMO-gene-deficient mice exhibited depressive-like behaviors, such as decreased sucrose preference and increased immobility in the forced swimming test, and the administration of antidepressants, selective serotonin reuptake inhibitors (SSRIs), can reverse these depressive-like behaviors [138]. In addition, these mice showed impairments in contextual memory, spent less time than controls interacting with an unfamiliar mouse in a social interaction paradigm, and showed increased anxiety-like behavior in the elevated plus maze and a light-dark box test. However, KMO-gene-deficit mice show an abnormally large increase in locomotor activity, compared to wild-type mice, when challenged with D-amphetamine and do not display disruption in PPI [139]. Pharmacological inhibition of KMO using 3,4-dimethoxy-N-[4-(3-nitrophenyl)thiazol-2-yl]benzenesulfonamide 16 (Ro 61-8048) during the pregnancy also resulted in distinct and long-lasting increased levels of Kyna, and significantly decreased expression of GluN2A and increased expression of GluN2B in the embryo brain [242]. The adolescent offspring at postnatal day (PND) 21 exhibited increased neuronal excitability, with increased levels of the Glu2A/Glu2B subunit of NMDA receptor and postsynaptic density protein (PDS)-95 [242,243]. Additionally, adult offspring (PND60) exhibited decreased overall numbers and lengths of hippocampal dendrites, together with fewer dendritic spines and less dendritic complexity, and disruptions in long-term potentiation (LTP) [130,131]. The number of neuron terminals staining for vesicular glutamate transporter (VGLUT)-1 and VGLUT-2 was significantly increased by Ro 61-8048 treatment, with no changes in the expression of vesicular GABA transporter, showing that prenatal inhibition of the KP produces marked effects on neuronal structure and the excitatory/inhibitory balance.
A different approach is directly increasing Kyn levels during pre- and postnatal development by feeding Kyn-laced chow or administrating Kyn to the pregnant dam continuously. Pocivavsek et al. demonstrated that continuous feeding of Kyn-laced chow from E15 to 22 elevated the levels of Kyna in the rat brain during the entire treatment period. Biochemical and behavioral tests in adulthood showed distinct changes, such as decreased expression levels of GluN2A and a trend toward decreased α7nAChR expression, and lower performance in trace fear conditioning tests [245]. The adult offspring also displayed behavioral impairments in hippocampus-related cognitive tasks, such as a lower passive avoidance perform and the Morris water maze [23]. Embryonic Kyn-exposure male offspring also displayed reduced rapid eye movement (REM) sleep, indicating prenatal Kyn elevation impairs sleeping behavior in rats [246], and ASD people are often reported to have sleeping troubles. In addition, they found time-of-day- and sex-dependent alterations in the levels of Kyna, glutamate, and GABA in the hippocampus, indicating that these hippocampal neuromodulations may be related to regulating memory consolidation, retrieval, and locomotor activity [247]. As a follow-up to these studies, continuous maternal Kyn administration in pregnant mice from E12.5 to 19 resulted in behavioral abnormalities, including social and cognitive defects. Kyn-injected adult offspring exhibited higher Kyn and Kyna in the fetal brain and lower performance on social recognition tests and novel object recognition tasks [3].
Additionally, neonatal Kyn-administered mice (PND7-16) showed an enhanced sensitivity to a D-amphetamine-induced increase in locomotor activity, mild impairment in PPI, and lower performance on the trace fear conditioning test, as shown in KMO-gene-deficient mice [251]. Furthermore, neonatal rats (PND7-10) administered Kyn exhibited decreased social interaction and locomotor activity, with long-lasting high concentrations of Kyna and QUIN in the brain [250]. Systemic administration of a high dose of Kyn in adult mice also disrupts their object recognition memory and decreases their locomotor activity [132]. Even a low dose of systemic Kyn injections in adult mice induces depression-like behavior [259].
However, KAT-II-gene-deficient mice show lower Kyna levels in the hippocampus and striatum and higher spontaneous locomotor activity, compared to wild-type mice during the early postnatal time-point (PND14 and 21), but no changes at PND60 [253,254]. At this age, KAT-II-gene-deficient mice exhibited a significantly increased performance in object exploration and recognition tasks, the passive avoidance test, and the spatial discrimination test, reflecting partly on the hippocampal function. Additionally, hippocampal slices from KAT-II-deficient mice showed a significant increase in the amplitude of LTP in vitro compared to wild-type controls [183]. Moreover, the α7nAChR activity induced by exogenous application of agonists to hippocampal stratum radiatum interneurons was extremely higher in KAT-II-gene-deficient mice than that in wild-type mice [253]. Pharmacological administration of selective KAT II inhibitor raised extracellular dopamine levels in the striatum [260], inhibited the firing rate and burst activity of dopamine neurons in the midbrain area, and reduced the number of spontaneously active dopamine cells. Pretreatment with an agonist of the NMDA receptor prevented the inhibitory action, and pretreatment with an antagonist of the GABAA receptor partially prevented the inhibitory effect on the KAT II inhibitor’s firing rate and burst firing activity. Therefore, the effect of KAT II inhibitor appears to be specifically executed by NMDA receptors and mediated indirectly via GABAB-receptor-induced disinhibition of dopamine neurons [261]. However, intrastriatal infusions of neurotoxic QUIN resulted in dose-dependent lesions, and the striatal damages is larger in KAT-II-gene-deficient mice than in wildtype mice in PND14 when the Kyna levels are lower, but no difference in the lesion volume at PND60 has been reported [254].
Generally, all studies show that time- and dose-dependent optimal levels of KP metabolites during critical neurodevelopmental periods are essential for normal brain development. Additionally, an imbalance of KP metabolites induces long-lasting changes relevant to various psychiatric diseases.

6. Conclusions

The immune system plays an important role in neurodevelopment and multiple neurobiological functions. Exposure to maternal immune activation during early pregnancy has been identified as the most influential environmental risk factor for ASD. Recent evidence shows that a responsible inflammatory pathway in MIA-associated ASD is related to the activity of Th17 lymphocytes and their effector, cytokine IL-17a, among immunological factors. The association of IL-17a in the etiology of ASD has been found not only in human clinical studies of patients with ASD, but also in MIA model experimental research. Therefore, there is no doubt that IL-17a dysregulation may play a causal role in the development of ASD. However, it is still controversial whether IL-17a can pass through the placental barrier and directly affect fetal brain development.
However, it is clearly shown that abnormal levels of KP metabolites, especially Kyn and Kyna, during neurodevelopment have been related to several neurobiochemical and behavioral impairments associated with depression, anxiety, ASD, and SCZ phenotypes (Figure 2). During early pre- or postnatal fetal development, activation of the KP, with an increase in neurotoxic metabolites and excitotoxicity, causing long-term changes in glutamatergic/GABAergic functions, trophic support, and synaptic function, may be linked to various psychiatric disorders. Yet, to validate the direct connection between neuroinflammation and KP metabolites in the pathology of ASD, some information is missing, and wide translational research is necessary. All recent research on the imbalance of KP metabolites during brain development show that interventions aimed at directly reducing KP metabolites or their sites of actions at critical periods may shed light on novel therapeutic strategies not only for neurodevelopmental disorders, but also to prevent the manifestation of neuropsychiatric and other CNS disorders.

Author Contributions

Conceptualization, Y.M. (Yuki Murakami), Y.I. and Y.K. (Yukuo Konishi); writing-original draft preparation, Y.M. (Yuki Murakami); writing-review and editing, Y.M. (Yuki Murakami), Y.I., Y.K. (Yoshiyuki Kasahara), C.Y., Y.M. (Yuta Momono), K.F., D.S. and T.N.; visualization, Y.M. (Yuki Murakami); project administration, Y.K. (Yukuo Konishi); funding acquisition, Y.M. (Yuki Murakami), Y.I. and Y.K. (Yukuo Konishi). All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a Grant-in-Aid for Scientific Research (16K08948 and 20K21587) from the Japan Society for Promotion of Science (JSPS). Additionally, this work was supported in part by the RIKEN Healthcare and Medical Data Platform Project and the Developmental Disorder Data Multi-level Integration Unit of the Medical Sciences Innovation Hub Program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest regarding this review article.

Abbreviations

ABCAberrant Behavior Checklist
ADDMAutism Developmental Disabilities Monitoring
ADI-RAutism Diagnostic Interview-Revised
ADOSAutism Diagnostic Observation Schedule
AhRaryl hydrocarbon receptor
APCsantigen-presenting cells
ASDautism spectrum disorder
5AV5-aminovaleric acid
BregB-regulatory cell
BBBblood-brain barrier
CARSChildhood Autism Rating Scale
CNScentral nervous system
DCSD-cycloserine
DCXdoublecortin
DSMDiagnostic and Statistical Manual of Mental Disorder
Eembryonic day
ELISAenzyme-linked immune sorbent assay
E. coliEscherichia coli
15q dup15q11.13 duplication
GABAgamma-aminobutyric acid
GS-MSgas chromatography-mass spectrometry
GIgastrointestinal
HDAChistone deacetylase
HILIChydrophilic interaction chromatography
3-HK3-hydroxy-L-kynurenine
HPLChigh performance liquid chromatography
5-HT5-hydroxytryptamine
IAindole acrylic acid
IAAindole-3-acetic acid
IAAldindole-3acetaaldehyde
IAldindole-3-aldehyde
IBDinflammatory bowel disease
ICD-10International Statistical Classification of Diseases and Related Health Problems-10
ICjMisland of Calleja major
IDO1indoleamine 2,3-dioxygenase1
IFN-γinterferon-gamma
IL-1βinterleukin-1 beta
IL-6interleukin-6
IL-17interleukin-17
IL-21interlekin-21
ILC3innate lymphocyte cells 3
IPAindole-3-propionic acid
KATkynurenine aminotransferase
KMOkynurenine 3-monooxygenase
KPkynurenine pathway
Kynkynurenine
Kynakynurenic acid
LClocus coeruleus
LC-MS/MSliquid chromatography-tandem mass spectrometry
LPSlipopolysaccharide
LRPlow-density receptor-related protein
LTPlong-term potentiation
MIAmaternal immune activation
nAChRnicotinic acetylcholine receptor
NMDAN-methyl-D-aspartate
PDS-95postsynaptic density protein-95
PNDpostnatal day
poly(I:C)polyinosinic:polycytidylic acid
PPIpre-pulse inhibition
PV+parvalbumin positive
QPRTquinolinic phosphoribosyltransferase
QUINquinolinic acid
RAADS-14Ritovo Autism & Asperger Diagnostic Scale-14
REMrapid eye movement
Ro 61-80483,4-dimethoxy-N-[4-(3-nitrophenyl)thiazol-2-yl]benzenesulfonamide 16
RRSRuminative Response Scale
SCFAshort-chain fatty acid
SCZschizophrenia
SERTserotonin transporter
SFBsegmented filamentous bacteria
SHPSC-43 activated SH2 domain-containing phosphatase
SNPsingle nucleotide polymorphism
SSRIsselective serotonin reuptake inhibitors
STATsignal transducers and activators of transcription
TCDD2,3,7,8-tetrachlorodibenzo-p-dioxin
TDtypically developing
TDOtryptophan 2,3-dioxygenase
TGF-®transforming growth factor-beta
ThT helper
TMS/MStandem mass spectrometry
TNFtumor necrosis factor
TPHtryptophan hydroxylase
TregT-regulatory cell
Trptryptophan
UHPLCultra-high performance liquid chromatography
VGLUTvesicular glutamate transporter
WSASWork and Social Adjustment Scale

References

  1. Bryn, V.; Verkerk, R.; Skjeldal, O.H.; Saugstad, O.D.; Ormstad, H. Kynurenine Pathway in Autism Spectrum Disorders in Children. Neuropsychobiology 2017, 76, 82–88. [Google Scholar] [CrossRef] [PubMed]
  2. Carpita, B.; Nardi, B.; Palego, L.; Cremone, I.M.; Massimetti, G.; Carmassi, C.; Betti, L.; Giannaccini, G.; Dell’Osso, L. Kynurenine pathway and autism spectrum phenotypes: An investigation among adults with autism spectrum disorder and their first-degree relatives. CNS Spectr. 2022, 1–12. [Google Scholar] [CrossRef] [PubMed]
  3. Murakami, Y.; Imamura, Y.; Kasahara, Y.; Yoshida, C.; Momono, Y.; Fang, K.; Nishiyama, T.; Sakai, D.; Konishi, Y. The Effects of Maternal Interleukin-17A on Social Behavior, Cognitive Function, and Depression-Like Behavior in Mice with Altered Kynurenine Metabolites. Int. J. Tryptophan Res. 2021, 14, 11786469211026639. [Google Scholar] [CrossRef]
  4. Hahn, B.; Reneski, C.H.; Pocivavsek, A.; Schwarcz, R. Prenatal kynurenine treatment in rats causes schizophrenia-like broad monitoring deficits in adulthood. Psychopharmacology 2018, 235, 651–661. [Google Scholar] [CrossRef]
  5. Pershing, M.L.; Bortz, D.M.; Pocivavsek, A.; Fredericks, P.J.; Jorgensen, C.V.; Vunck, S.A.; Leuner, B.; Schwarcz, R.; Bruno, J.P. Elevated levels of kynurenic acid during gestation produce neurochemical, morphological, and cognitive deficits in adulthood: Implications for schizophrenia. Neuropharmacology 2015, 90, 33–41. [Google Scholar] [CrossRef] [Green Version]
  6. Murphy, S.K.; Fineberg, A.M.; Maxwell, S.D.; Alloy, L.B.; Zimmermann, L.; Krigbaum, N.Y.; Cohn, B.A.; Drabick, D.A.G.; Ellman, L.M. Maternal infection and stress during pregnancy and depressive symptoms in adolescent offspring. Psychiatry Res. 2017, 257, 102–110. [Google Scholar] [CrossRef]
  7. Sha, Q.; Madaj, Z.; Keaton, S.; Escobar Galvis, M.L.; Smart, L.; Krzyzanowski, S.; Fazleabas, A.T.; Leach, R.; Postolache, T.T.; Achtyes, E.D.; et al. Cytokines and tryptophan metabolites can predict depressive symptoms in pregnancy. Transl. Psychiatry 2022, 12, 35. [Google Scholar] [CrossRef] [PubMed]
  8. Lim, C.K.; Essa, M.M.; de Paula Martins, R.; Lovejoy, D.B.; Bilgin, A.A.; Waly, M.I.; Al-Farsi, Y.M.; Al-Sharbati, M.; Al-Shaffae, M.A.; Guillemin, G.J. Altered kynurenine pathway metabolism in autism: Implication for immune-induced glutamatergic activity. Autism Res. 2016, 9, 621–631. [Google Scholar] [CrossRef]
  9. Wonodi, I.; Schwarcz, R. Cortical kynurenine pathway metabolism: A novel target for cognitive enhancement in Schizophrenia. Schizophr. Bull. 2010, 36, 211–218. [Google Scholar] [CrossRef] [Green Version]
  10. Sathyasaikumar, K.V.; Stachowski, E.K.; Wonodi, I.; Roberts, R.C.; Rassoulpour, A.; McMahon, R.P.; Schwarcz, R. Impaired kynurenine pathway metabolism in the prefrontal cortex of individuals with schizophrenia. Schizophr. Bull. 2011, 37, 1147–1156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Plitman, E.; Iwata, Y.; Caravaggio, F.; Nakajima, S.; Chung, J.K.; Gerretsen, P.; Kim, J.; Takeuchi, H.; Chakravarty, M.M.; Remington, G.; et al. Kynurenic Acid in Schizophrenia: A Systematic Review and Meta-analysis. Schizophr. Bull. 2017, 43, 764–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ogyu, K.; Kubo, K.; Noda, Y.; Iwata, Y.; Tsugawa, S.; Omura, Y.; Wada, M.; Tarumi, R.; Plitman, E.; Moriguchi, S.; et al. Kynurenine pathway in depression: A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2018, 90, 16–25. [Google Scholar] [CrossRef]
  13. Yun, Y.; Zhang, Q.; Zhao, W.; Ma, T.; Fan, H.; Bai, L.; Ma, B.; Qi, S.; Wang, Z.; An, H.; et al. Relationship between the tryptophan-kynurenine pathway and painful physical symptoms in patients with major depressive disorder. J. Psychosom. Res. 2022, 163, 111069. [Google Scholar] [CrossRef] [PubMed]
  14. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Text Revision DSM-5-TR, 5th ed.; Text Revision (DSM-5-TR); American Psychiatric Association Publishing Inc.: Washington, DC, USA, 2022. [Google Scholar]
  15. Maenner, M.J.; Shaw, K.A.; Bakian, A.V.; Bilder, D.A.; Durkin, M.S.; Esler, A.; Furnier, S.M.; Hallas, L.; Hall-Lande, J.; Hudson, A.; et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2018. MMWR Surveill. Summ. 2021, 70, 1–16. [Google Scholar] [CrossRef]
  16. Cogswell, M.E.; Coil, E.; Tian, L.H.; Tinker, S.C.; Ryerson, A.B.; Maenner, M.J.; Rice, C.E.; Peacock, G. Health Needs and Use of Services Among Children with Developmental Disabilities—United States, 2014–2018. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 453–458. [Google Scholar] [CrossRef]
  17. Liu, X.; Takumi, T. Genomic and genetic aspects of autism spectrum disorder. Biochem. Biophys. Res. Commun. 2014, 452, 244–253. [Google Scholar] [CrossRef] [Green Version]
  18. Tick, B.; Bolton, P.; Happe, F.; Rutter, M.; Rijsdijk, F. Heritability of autism spectrum disorders: A meta-analysis of twin studies. J. Child Psychol. Psychiatry 2016, 57, 585–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Patterson, P.H. Maternal infection and immune involvement in autism. Trends Mol. Med. 2011, 17, 389–394. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, B.K.; Magnusson, C.; Gardner, R.M.; Blomstrom, A.; Newschaffer, C.J.; Burstyn, I.; Karlsson, H.; Dalman, C. Maternal hospitalization with infection during pregnancy and risk of autism spectrum disorders. Brain Behav. Immun. 2015, 44, 100–105. [Google Scholar] [CrossRef] [Green Version]
  21. Han, V.X.; Patel, S.; Jones, H.F.; Dale, R.C. Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nat. Rev. Neurol. 2021, 17, 564–579. [Google Scholar] [CrossRef]
  22. 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]
  23. Pocivavsek, A.; Thomas, M.A.; Elmer, G.I.; Bruno, J.P.; Schwarcz, R. Continuous kynurenine administration during the prenatal period, but not during adolescence, causes learning and memory deficits in adult rats. Psychopharmacology 2014, 231, 2799–2809. [Google Scholar] [CrossRef] [Green Version]
  24. 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]
  25. Morgan, J.T.; Chana, G.; Pardo, C.A.; Achim, C.; Semendeferi, K.; Buckwalter, J.; Courchesne, E.; Everall, I.P. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol. Psychiatry 2010, 68, 368–376. [Google Scholar] [CrossRef] [PubMed]
  26. Pardo, C.A.; Vargas, D.L.; Zimmerman, A.W. Immunity, neuroglia and neuroinflammation in autism. Int. Rev. Psychiatry 2005, 17, 485–495. [Google Scholar] [CrossRef] [Green Version]
  27. Vargas, D.L.; Nascimbene, C.; Krishnan, C.; Zimmerman, A.W.; Pardo, C.A. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann. Neurol. 2005, 57, 67–81. [Google Scholar] [CrossRef]
  28. Wong, H.; Hoeffer, C. Maternal IL-17A in autism. Exp. Neurol. 2018, 299, 228–240. [Google Scholar] [CrossRef]
  29. Jiang, H.Y.; Xu, L.L.; Shao, L.; Xia, R.M.; Yu, Z.H.; Ling, Z.X.; Yang, F.; Deng, M.; Ruan, B. Maternal infection during pregnancy and risk of autism spectrum disorders: A systematic review and meta-analysis. Brain Behav. Immun. 2016, 58, 165–172. [Google Scholar] [CrossRef]
  30. Chen, S.W.; Zhong, X.S.; Jiang, L.N.; Zheng, X.Y.; Xiong, Y.Q.; Ma, S.J.; Qiu, M.; Huo, S.T.; Ge, J.; Chen, Q. Maternal autoimmune diseases and the risk of autism spectrum disorders in offspring: A systematic review and meta-analysis. Behav. Brain Res. 2016, 296, 61–69. [Google Scholar] [CrossRef]
  31. Gong, T.; Lundholm, C.; Rejno, G.; Bolte, S.; Larsson, H.; D’Onofrio, B.M.; Lichtenstein, P.; Almqvist, C. Parental asthma and risk of autism spectrum disorder in offspring: A population and family-based case-control study. Clin. Exp. Allergy 2019, 49, 883–891. [Google Scholar] [CrossRef] [Green Version]
  32. Wei, H.; Zou, H.; Sheikh, A.M.; Malik, M.; Dobkin, C.; Brown, W.T.; Li, X. IL-6 is increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and synaptic formation. J. Neuroinflammation 2011, 8, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. 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]
  34. Rudolph, M.D.; Graham, A.M.; Feczko, E.; Miranda-Dominguez, O.; Rasmussen, J.M.; Nardos, R.; Entringer, S.; Wadhwa, P.D.; Buss, C.; Fair, D.A. Maternal IL-6 during pregnancy can be estimated from newborn brain connectivity and predicts future working memory in offspring. Nat. Neurosci. 2018, 21, 765–772. [Google Scholar] [CrossRef]
  35. 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] [Green Version]
  36. Yasuda, K.; Takeuchi, Y.; Hirota, K. Correction to: The pathogenicity of Th17 cells in autoimmune diseases. Semin. Immunopathol. 2019, 41, 299. [Google Scholar] [CrossRef] [Green Version]
  37. 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] [PubMed]
  38. Zhou, L.; Ivanov, I.I.; Spolski, R.; Min, R.; Shenderov, K.; Egawa, T.; Levy, D.E.; Leonard, W.J.; Littman, D.R. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. 2007, 8, 967–974. [Google Scholar] [CrossRef]
  39. Mangan, P.R.; Harrington, L.E.; O’Quinn, D.B.; Helms, W.S.; Bullard, D.C.; Elson, C.O.; Hatton, R.D.; Wahl, S.M.; Schoeb, T.R.; Weaver, C.T. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 2006, 441, 231–234. [Google Scholar] [CrossRef] [PubMed]
  40. Akintunde, M.E.; Rose, M.; Krakowiak, P.; Heuer, L.; Ashwood, P.; Hansen, R.; Hertz-Picciotto, I.; Van de Water, J. Increased production of IL-17 in children with autism spectrum disorders and co-morbid asthma. J. Neuroimmunol. 2015, 286, 33–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Al-Ayadhi, L.Y.; Mostafa, G.A. Elevated serum levels of interleukin-17A in children with autism. J. Neuroinflammation 2012, 9, 158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. 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]
  43. Garay, P.A.; Hsiao, E.Y.; Patterson, P.H.; McAllister, A.K. Maternal immune activation causes age- and region-specific changes in brain cytokines in offspring throughout development. Brain Behav. Immun. 2013, 31, 54–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Solek, C.M.; Farooqi, N.; Verly, M.; Lim, T.K.; Ruthazer, E.S. Maternal immune activation in neurodevelopmental disorders. Dev. Dyn. 2018, 247, 588–619. [Google Scholar] [CrossRef] [PubMed]
  45. Sasaki, T.; Tome, S.; Takei, Y. Intraventricular IL-17A administration activates microglia and alters their localization in the mouse embryo cerebral cortex. Mol. Brain 2020, 13, 93. [Google Scholar] [CrossRef] [PubMed]
  46. Gumusoglu, S.B.; Hing, B.W.Q.; Chilukuri, A.S.S.; Dewitt, J.J.; Scroggins, S.M.; Stevens, H.E. Correction: Chronic maternal interleukin-17 and autism-related cortical gene expression, neurobiology, and behavior. Neuropsychopharmacology 2020, 45, 1588. [Google Scholar] [CrossRef]
  47. Li, Z.; Li, K.; Zhu, L.; Kan, Q.; Yan, Y.; Kumar, P.; Xu, H.; Rostami, A.; Zhang, G.X. Inhibitory effect of IL-17 on neural stem cell proliferation and neural cell differentiation. BMC Immunol. 2013, 14, 20. [Google Scholar] [CrossRef] [Green Version]
  48. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549, 528–532. [Google Scholar] [CrossRef] [Green Version]
  49. Honig, A.; Rieger, L.; Kapp, M.; Sutterlin, M.; Dietl, J.; Kammerer, U. Indoleamine 2,3-dioxygenase (IDO) expression in invasive extravillous trophoblast supports role of the enzyme for materno-fetal tolerance. J. Reprod. Immunol. 2004, 61, 79–86. [Google Scholar] [CrossRef]
  50. Sedlmayr, P.; Blaschitz, A.; Stocker, R. The role of placental tryptophan catabolism. Front. Immunol. 2014, 5, 230. [Google Scholar] [CrossRef] [Green Version]
  51. Munn, D.H.; Zhou, M.; Attwood, J.T.; Bondarev, I.; Conway, S.J.; Marshall, B.; Brown, C.; Mellor, A.L. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 1998, 281, 1191–1193. [Google Scholar] [CrossRef] [PubMed]
  52. Munn, D.H.; Sharma, M.D.; Baban, B.; Harding, H.P.; Zhang, Y.; Ron, D.; Mellor, A.L. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 2005, 22, 633–642. [Google Scholar] [CrossRef] [Green Version]
  53. Kudo, Y.; Boyd, C.A.; Sargent, I.L.; Redman, C.W. Decreased tryptophan catabolism by placental indoleamine 2,3-dioxygenase in preeclampsia. Am. J. Obstet. Gynecol. 2003, 188, 719–726. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, W.; Sung, N.; Gilman-Sachs, A.; Kwak-Kim, J. T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells. Front. Immunol. 2020, 11, 2025. [Google Scholar] [CrossRef]
  55. Wang, R.; Weng, Y.; Zhao, S.; Li, S.; Wen, X.; Huang, G. [IL-6 up-regulates indoleamine 2, 3-dioxygenase (IDO) expression in chorionic villi and decidua]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2021, 37, 158–163. [Google Scholar] [PubMed]
  56. Cheng, H.; Huang, Y.; Huang, G.; Chen, Z.; Tang, J.; Pan, L.; Lv, J.; Long, A.; Wang, R.; Chen, Z.; et al. Effect of the IDO Gene on Pregnancy in Mice with Recurrent Pregnancy Loss. Reprod. Sci. 2021, 28, 52–59. [Google Scholar] [CrossRef]
  57. Wang, R.; Zhao, S.; Chen, X.; Xiao, Z.; Wen, X.; Zhong, X.; Li, S.; Cheng, H.; Huang, G. Molecular mechanisms involved in the IL-6-mediated upregulation of indoleamine 2,3-dioxygenase 1 (IDO1) expression in the chorionic villi and decidua of women in early pregnancy. BMC Pregnancy Childbirth 2022, 22, 983. [Google Scholar] [CrossRef] [PubMed]
  58. Azevedo, R.S.S.; de Sousa, J.R.; Araujo, M.T.F.; Martins Filho, A.J.; de Alcantara, B.N.; Araujo, F.M.C.; Queiroz, M.G.L.; Cruz, A.C.R.; Vasconcelos, B.H.B.; Chiang, J.O.; et al. In situ immune response and mechanisms of cell damage in central nervous system of fatal cases microcephaly by Zika virus. Sci. Rep. 2018, 8, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Kanai, M.; Funakoshi, H.; Takahashi, H.; Hayakawa, T.; Mizuno, S.; Matsumoto, K.; Nakamura, T. Tryptophan 2,3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice. Mol. Brain 2009, 2, 8. [Google Scholar] [CrossRef] [Green Version]
  60. Hoffmann, D.; Dvorakova, T.; Schramme, F.; Stroobant, V.; Van den Eynde, B.J. Tryptophan 2,3-Dioxygenase Expression Identified in Murine Decidual Stromal Cells Is Not Essential for Feto-Maternal Tolerance. Front. Immunol. 2020, 11, 601759. [Google Scholar] [CrossRef]
  61. Pilotte, L.; Larrieu, P.; Stroobant, V.; Colau, D.; Dolusic, E.; Frederick, R.; De Plaen, E.; Uyttenhove, C.; Wouters, J.; Masereel, B.; et al. Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc. Natl. Acad. Sci. USA 2012, 109, 2497–2502. [Google Scholar] [CrossRef] [Green Version]
  62. Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef] [Green Version]
  63. Hoffmann, D.; Dvorakova, T.; Stroobant, V.; Bouzin, C.; Daumerie, A.; Solvay, M.; Klaessens, S.; Letellier, M.C.; Renauld, J.C.; van Baren, N.; et al. Tryptophan 2,3-Dioxygenase Expression Identified in Human Hepatocellular Carcinoma Cells and in Intratumoral Pericytes of Most Cancers. Cancer Immunol. Res. 2020, 8, 19–31. [Google Scholar] [CrossRef]
  64. Bent, R.; Moll, L.; Grabbe, S.; Bros, M. Interleukin-1 Beta-A Friend or Foe in Malignancies? Int. J. Mol. Sci. 2018, 19, 2155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Urata, Y.; Koga, K.; Hirota, Y.; Akiyama, I.; Izumi, G.; Takamura, M.; Nagai, M.; Harada, M.; Hirata, T.; Yoshino, O.; et al. IL-1beta increases expression of tryptophan 2,3-dioxygenase and stimulates tryptophan catabolism in endometrioma stromal cells. Am. J. Reprod. Immunol. 2014, 72, 496–503. [Google Scholar] [CrossRef] [PubMed]
  66. Pantouris, G.; Loudon-Griffiths, J.; Mowat, C.G. Insights into the mechanism of inhibition of tryptophan 2,3-dioxygenase by isatin derivatives. J. Enzym. Inhib. Med. Chem. 2016, 31, 70–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zador, F.; Joca, S.; Nagy-Grocz, G.; Dvoracsko, S.; Szucs, E.; Tomboly, C.; Benyhe, S.; Vecsei, L. Pro-Inflammatory Cytokines: Potential Links between the Endocannabinoid System and the Kynurenine Pathway in Depression. Int. J. Mol. Sci. 2021, 22, 5903. [Google Scholar] [CrossRef]
  68. Nakashima, A.; Ito, M.; Yoneda, S.; Shiozaki, A.; Hidaka, T.; Saito, S. Circulating and decidual Th17 cell levels in healthy pregnancy. Am. J. Reprod. Immunol. 2010, 63, 104–109. [Google Scholar] [CrossRef]
  69. Santner-Nanan, B.; Peek, M.J.; Khanam, R.; Richarts, L.; Zhu, E.; Fazekas de St Groth, B.; Nanan, R. Systemic increase in the ratio between Foxp3+ and IL-17-producing CD4+ T cells in healthy pregnancy but not in preeclampsia. J. Immunol. 2009, 183, 7023–7030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Cua, D.J.; Tato, C.M. Innate IL-17-producing cells: The sentinels of the immune system. Nat. Rev. Immunol. 2010, 10, 479–489. [Google Scholar] [CrossRef]
  71. Romani, L.; Zelante, T.; De Luca, A.; Fallarino, F.; Puccetti, P. IL-17 and therapeutic kynurenines in pathogenic inflammation to fungi. J. Immunol. 2008, 180, 5157–5162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Krause, R.; Zollner-Schwetz, I.; Salzer, H.J.; Valentin, T.; Rabensteiner, J.; Pruller, F.; Raggam, R.; Meinitzer, A.; Prattes, J.; Rinner, B.; et al. Elevated levels of interleukin 17A and kynurenine in candidemic patients, compared with levels in noncandidemic patients in the intensive care unit and those in healthy controls. J. Infect. Dis. 2015, 211, 445–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Stone, T.W.; Clanchy, F.I.L.; Huang, Y.S.; Chiang, N.Y.; Darlington, L.G.; Williams, R.O. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front. Neurosci. 2022, 16, 1002004. [Google Scholar] [CrossRef] [PubMed]
  74. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
  75. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [Green Version]
  78. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef] [Green Version]
  79. Mawe, G.M.; Hoffman, J.M. Serotonin signalling in the gut—Functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473–486. [Google Scholar] [CrossRef] [Green Version]
  80. Terry, N.; Margolis, K.G. Serotonergic Mechanisms Regulating the GI Tract: Experimental Evidence and Therapeutic Relevance. Handb. Exp. Pharmacol. 2017, 239, 319–342. [Google Scholar] [CrossRef] [Green Version]
  81. Heredia, D.J.; Gershon, M.D.; Koh, S.D.; Corrigan, R.D.; Okamoto, T.; Smith, T.K. Important role of mucosal serotonin in colonic propulsion and peristaltic reflexes: In vitro analyses in mice lacking tryptophan hydroxylase 1. J. Physiol. 2013, 591, 5939–5957. [Google Scholar] [CrossRef]
  82. Margolis, K.G.; Li, Z.; Stevanovic, K.; Saurman, V.; Israelyan, N.; Anderson, G.M.; Snyder, I.; Veenstra-VanderWeele, J.; Blakely, R.D.; Gershon, M.D. Serotonin transporter variant drives preventable gastrointestinal abnormalities in development and function. J. Clin. Invest. 2016, 126, 2221–2235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Baron, R.; Rawadi, G. Wnt signaling and the regulation of bone mass. Curr. Osteoporos. Rep. 2007, 5, 73–80. [Google Scholar] [CrossRef] [PubMed]
  84. Hata, T.; Asano, Y.; Yoshihara, K.; Kimura-Todani, T.; Miyata, N.; Zhang, X.T.; Takakura, S.; Aiba, Y.; Koga, Y.; Sudo, N. Regulation of gut luminal serotonin by commensal microbiota in mice. PLoS ONE 2017, 12, e0180745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. 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 J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
  86. Alexeev, E.E.; Lanis, J.M.; Kao, D.J.; Campbell, E.L.; Kelly, C.J.; Battista, K.D.; Gerich, M.E.; Jenkins, B.R.; Walk, S.T.; Kominsky, D.J.; et al. Microbiota-Derived Indole Metabolites Promote Human and Murine Intestinal Homeostasis through Regulation of Interleukin-10 Receptor. Am. J. Pathol. 2018, 188, 1183–1194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef] [Green Version]
  88. Wlodarska, M.; Luo, C.; Kolde, R.; d’Hennezel, E.; Annand, J.W.; Heim, C.E.; Krastel, P.; Schmitt, E.K.; Omar, A.S.; Creasey, E.A.; et al. Indoleacrylic Acid Produced by Commensal Peptostreptococcus Species Suppresses Inflammation. Cell Host Microbe 2017, 22, 25–37.e26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Su, X.; Gao, Y.; Yang, R. Gut Microbiota-Derived Tryptophan Metabolites Maintain Gut and Systemic Homeostasis. Cells 2022, 11, 2296. [Google Scholar] [CrossRef]
  90. Lamas, B.; Natividad, J.M.; Sokol, H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol. 2018, 11, 1024–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Stockinger, B.; Di Meglio, P.; Gialitakis, M.; Duarte, J.H. The aryl hydrocarbon receptor: Multitasking in the immune system. Annu. Rev. Immunol. 2014, 32, 403–432. [Google Scholar] [CrossRef] [PubMed]
  92. Williams, B.B.; Van Benschoten, A.H.; Cimermancic, P.; Donia, M.S.; Zimmermann, M.; Taketani, M.; Ishihara, A.; Kashyap, P.C.; Fraser, J.S.; Fischbach, M.A. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 2014, 16, 495–503. [Google Scholar] [CrossRef] [Green Version]
  93. Juorio, A.V.; Paterson, I.A. Tryptamine may couple dopaminergic and serotonergic transmission in the brain. Gen. Pharmacol. 1990, 21, 613–616. [Google Scholar] [CrossRef] [PubMed]
  94. Bhattarai, Y.; Williams, B.B.; Battaglioli, E.J.; Whitaker, W.R.; Till, L.; Grover, M.; Linden, D.R.; Akiba, Y.; Kandimalla, K.K.; Zachos, N.C.; et al. Gut Microbiota-Produced Tryptamine Activates an Epithelial G-Protein-Coupled Receptor to Increase Colonic Secretion. Cell Host Microbe 2018, 23, 775–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Galligan, J.J. Beneficial actions of microbiota-derived tryptophan metabolites. Neurogastroenterol. Motil. 2018, 30, e13283. [Google Scholar] [CrossRef] [PubMed]
  96. Dodd, D.; Spitzer, M.H.; Van Treuren, W.; Merrill, B.D.; Hryckowian, A.J.; Higginbottom, S.K.; Le, A.; Cowan, T.M.; Nolan, G.P.; Fischbach, M.A.; et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Lee, J.H.; Lee, J. Indole as an intercellular signal in microbial communities. FEMS Microbiol. Rev. 2010, 34, 426–444. [Google Scholar] [CrossRef] [PubMed]
  98. 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] [PubMed] [Green Version]
  99. Lukic, I.; Getselter, D.; Koren, O.; Elliott, E. Role of Tryptophan in Microbiota-Induced Depressive-Like Behavior: Evidence from Tryptophan Depletion Study. Front. Behav. Neurosci. 2019, 13, 123. [Google Scholar] [CrossRef] [Green Version]
  100. Dehhaghi, M.; Kazemi Shariat Panahi, H.; Guillemin, G.J. Microorganisms, Tryptophan Metabolism, and Kynurenine Pathway: A Complex Interconnected Loop Influencing Human Health Status. Int. J. Tryptophan Res. 2019, 12, 1178646919852996. [Google Scholar] [CrossRef] [Green Version]
  101. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
  102. He, Y.W.; Wang, H.S.; Zeng, J.; Fang, X.; Chen, H.Y.; Du, J.; Yang, X.Y. Sodium butyrate inhibits interferon-gamma induced indoleamine 2,3-dioxygenase expression via STAT1 in nasopharyngeal carcinoma cells. Life Sci. 2013, 93, 509–515. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  104. Li, G.; Lin, J.; Zhang, C.; Gao, H.; Lu, H.; Gao, X.; Zhu, R.; Li, Z.; Li, M.; Liu, Z. Microbiota metabolite butyrate constrains neutrophil functions and ameliorates mucosal inflammation in inflammatory bowel disease. Gut Microbes 2021, 13, 1968257. [Google Scholar] [CrossRef] [PubMed]
  105. Glauben, R.; Batra, A.; Fedke, I.; Zeitz, M.; Lehr, H.A.; Leoni, F.; Mascagni, P.; Fantuzzi, G.; Dinarello, C.A.; Siegmund, B. Histone hyperacetylation is associated with amelioration of experimental colitis in mice. J. Immunol. 2006, 176, 5015–5022. [Google Scholar] [CrossRef] [Green Version]
  106. Sandler, R.H.; Finegold, S.M.; Bolte, E.R.; Buchanan, C.P.; Maxwell, A.P.; Vaisanen, M.L.; Nelson, M.N.; Wexler, H.M. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J. Child Neurol. 2000, 15, 429–435. [Google Scholar] [CrossRef]
  107. Finegold, S.M.; Molitoris, D.; Song, Y.; Liu, C.; Vaisanen, M.L.; Bolte, E.; McTeague, M.; Sandler, R.; Wexler, H.; Marlowe, E.M.; et al. Gastrointestinal microflora studies in late-onset autism. Clin. Infect. Dis. 2002, 35, S6–S16. [Google Scholar] [CrossRef] [PubMed]
  108. Song, Y.; Liu, C.; Finegold, S.M. Real-time PCR quantitation of clostridia in feces of autistic children. Appl. Environ. Microbiol. 2004, 70, 6459–6465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Parracho, H.M.; Bingham, M.O.; Gibson, G.R.; McCartney, A.L. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 2005, 54, 987–991. [Google Scholar] [CrossRef] [PubMed]
  110. Finegold, S.M.; Dowd, S.E.; Gontcharova, V.; Liu, C.; Henley, K.E.; Wolcott, R.D.; Youn, E.; Summanen, P.H.; Granpeesheh, D.; Dixon, D.; et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010, 16, 444–453. [Google Scholar] [CrossRef] [PubMed]
  111. Williams, B.L.; Hornig, M.; Buie, T.; Bauman, M.L.; Cho Paik, M.; Wick, I.; Bennett, A.; Jabado, O.; Hirschberg, D.L.; Lipkin, W.I. Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances. PLoS ONE 2011, 6, e24585. [Google Scholar] [CrossRef] [Green Version]
  112. Adams, J.B.; Johansen, L.J.; Powell, L.D.; Quig, D.; Rubin, R.A. Gastrointestinal flora and gastrointestinal status in children with autism--comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011, 11, 22. [Google Scholar] [CrossRef] [Green Version]
  113. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Increased abundance of Sutterella spp. and Ruminococcus torques in feces of children with autism spectrum disorder. Mol. Autism 2013, 4, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kushak, R.I.; Winter, H.S.; Buie, T.M.; Cox, S.B.; Phillips, C.D.; Ward, N.L. Analysis of the Duodenal Microbiome in Autistic Individuals: Association with Carbohydrate Digestion. J. Pediatr. Gastroenterol. Nutr. 2017, 64, e110–e116. [Google Scholar] [CrossRef] [PubMed]
  115. Luna, R.A.; Oezguen, N.; Balderas, M.; Venkatachalam, A.; Runge, J.K.; Versalovic, J.; Veenstra-VanderWeele, J.; Anderson, G.M.; Savidge, T.; Williams, K.C. Distinct Microbiome-Neuroimmune Signatures Correlate with Functional Abdominal Pain in Children with Autism Spectrum Disorder. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 218–230. [Google Scholar] [CrossRef] [Green Version]
  116. Liu, F.; Li, J.; Wu, F.; Zheng, H.; Peng, Q.; Zhou, H. Altered composition and function of intestinal microbiota in autism spectrum disorders: A systematic review. Transl. Psychiatry 2019, 9, 43. [Google Scholar] [CrossRef] [Green Version]
  117. Frye, R.E.; Rose, S.; Slattery, J.; MacFabe, D.F. Gastrointestinal dysfunction in autism spectrum disorder: The role of the mitochondria and the enteric microbiome. Microb. Ecol. Health Dis. 2015, 26, 27458. [Google Scholar] [CrossRef] [PubMed]
  118. Argou-Cardozo, I.; Zeidan-Chulia, F. Clostridium Bacteria and Autism Spectrum Conditions: A Systematic Review and Hypothetical Contribution of Environmental Glyphosate Levels. Med. Sci. 2018, 6, 29. [Google Scholar] [CrossRef] [Green Version]
  119. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Srikantha, P.; Mohajeri, M.H. The Possible Role of the Microbiota-Gut-Brain-Axis in Autism Spectrum Disorder. Int. J. Mol. Sci. 2019, 20, 2115. [Google Scholar] [CrossRef] [Green Version]
  121. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [Green Version]
  122. Kang, D.W.; Park, J.G.; Ilhan, Z.E.; Wallstrom, G.; Labaer, J.; Adams, J.B.; Krajmalnik-Brown, R. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS ONE 2013, 8, e68322. [Google Scholar] [CrossRef] [Green Version]
  123. 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]
  124. Kang, D.W.; Adams, J.B.; Gregory, A.C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S.; et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study. Microbiome 2017, 5, 10. [Google Scholar] [CrossRef]
  125. 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]
  126. 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] [PubMed] [Green Version]
  127. Notarangelo, F.M.; Beggiato, S.; Schwarcz, R. Assessment of Prenatal Kynurenine Metabolism Using Tissue Slices: Focus on the Neosynthesis of Kynurenic Acid in Mice. Dev. Neurosci. 2019, 41, 102–111. [Google Scholar] [CrossRef]
  128. Notarangelo, F.M.; Pocivavsek, A. Elevated kynurenine pathway metabolism during neurodevelopment: Implications for brain and behavior. Neuropharmacology 2017, 112, 275–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Van Zundert, S.K.; Broekhuizen, M.; Smit, A.J.; van Rossem, L.; Mirzaian, M.; Willemsen, S.P.; Danser, A.J.; De Rijke, Y.B.; Reiss, I.K.; Merkus, D.; et al. The Role of the Kynurenine Pathway in the (Patho) physiology of Maternal Pregnancy and Fetal Outcomes: A Systematic Review. Int. J. Tryptophan Res. 2022, 15, 11786469221135545. [Google Scholar] [CrossRef]
  130. Khalil, O.S.; Pisar, M.; Forrest, C.M.; Vincenten, M.C.; Darlington, L.G.; Stone, T.W. Prenatal inhibition of the kynurenine pathway leads to structural changes in the hippocampus of adult rat offspring. Eur. J. Neurosci. 2014, 39, 1558–1571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Pisar, M.; Forrest, C.M.; Khalil, O.S.; McNair, K.; Vincenten, M.C.; Qasem, S.; Darlington, L.G.; Stone, T.W. Modified neocortical and cerebellar protein expression and morphology in adult rats following prenatal inhibition of the kynurenine pathway. Brain Res. 2014, 1576, 1–17. [Google Scholar] [CrossRef] [PubMed]
  132. Varga, D.; Heredi, J.; Kanvasi, Z.; Ruszka, M.; Kis, Z.; Ono, E.; Iwamori, N.; Iwamori, T.; Takakuwa, H.; Vecsei, L.; et al. Systemic L-Kynurenine sulfate administration disrupts object recognition memory, alters open field behavior and decreases c-Fos immunopositivity in C57Bl/6 mice. Front. Behav. Neurosci. 2015, 9, 157. [Google Scholar] [CrossRef] [Green Version]
  133. Perkins, M.N.; Stone, T.W. An iontophoretic investigation of the actions of convulsant kynurenines and their interaction with the endogenous excitant quinolinic acid. Brain Res. 1982, 247, 184–187. [Google Scholar] [CrossRef]
  134. Hilmas, C.; Pereira, E.F.; Alkondon, M.; Rassoulpour, A.; Schwarcz, R.; Albuquerque, E.X. The brain metabolite kynurenic acid inhibits alpha7 nicotinic receptor activity and increases non-alpha7 nicotinic receptor expression: Physiopathological implications. J. Neurosci. 2001, 21, 7463–7473. [Google Scholar] [CrossRef] [PubMed]
  135. Ceresoli-Borroni, G.; Schwarcz, R. Perinatal kynurenine pathway metabolism in the normal and asphyctic rat brain. Amino Acids 2000, 19, 311–323. [Google Scholar] [CrossRef] [PubMed]
  136. Fukui, S.; Schwarcz, R.; Rapoport, S.I.; Takada, Y.; Smith, Q.R. Blood-brain barrier transport of kynurenines: Implications for brain synthesis and metabolism. J. Neurochem. 1991, 56, 2007–2017. [Google Scholar] [CrossRef]
  137. Goeden, N.; Notarangelo, F.M.; Pocivavsek, A.; Beggiato, S.; Bonnin, A.; Schwarcz, R. Prenatal Dynamics of Kynurenine Pathway Metabolism in Mice: Focus on Kynurenic Acid. Dev. Neurosci. 2017, 39, 519–528. [Google Scholar] [CrossRef]
  138. Tashiro, T.; Murakami, Y.; Mouri, A.; Imamura, Y.; Nabeshima, T.; Yamamoto, Y.; Saito, K. Kynurenine 3-monooxygenase is implicated in antidepressants-responsive depressive-like behaviors and monoaminergic dysfunctions. Behav. Brain Res. 2017, 317, 279–285. [Google Scholar] [CrossRef] [PubMed]
  139. Erhardt, S.; Pocivavsek, A.; Repici, M.; Liu, X.C.; Imbeault, S.; Maddison, D.C.; Thomas, M.A.R.; Smalley, J.L.; Larsson, M.K.; Muchowski, P.J.; et al. Adaptive and Behavioral Changes in Kynurenine 3-Monooxygenase Knockout Mice: Relevance to Psychotic Disorders. Biol. Psychiatry 2017, 82, 756–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Namba, T.; Ming, G.L.; Song, H.; Waga, C.; Enomoto, A.; Kaibuchi, K.; Kohsaka, S.; Uchino, S. NMDA receptor regulates migration of newly generated neurons in the adult hippocampus via Disrupted-In-Schizophrenia 1 (DISC1). J. Neurochem. 2011, 118, 34–44. [Google Scholar] [CrossRef] [PubMed]
  141. Nacher, J.; McEwen, B.S. The role of N-methyl-D-asparate receptors in neurogenesis. Hippocampus 2006, 16, 267–270. [Google Scholar] [CrossRef]
  142. Colonnese, M.T.; Zhao, J.P.; Constantine-Paton, M. NMDA receptor currents suppress synapse formation on sprouting axons in vivo. J. Neurosci. 2005, 25, 1291–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ultanir, S.K.; Kim, J.E.; Hall, B.J.; Deerinck, T.; Ellisman, M.; Ghosh, A. Regulation of spine morphology and spine density by NMDA receptor signaling in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 19553–19558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Lee, E.J.; Choi, S.Y.; Kim, E. NMDA receptor dysfunction in autism spectrum disorders. Curr. Opin. Pharmacol. 2015, 20, 8–13. [Google Scholar] [CrossRef]
  145. O’Roak, B.J.; Vives, L.; Girirajan, S.; Karakoc, E.; Krumm, N.; Coe, B.P.; Levy, R.; Ko, A.; Lee, C.; Smith, J.D.; et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 2012, 485, 246–250. [Google Scholar] [CrossRef] [Green Version]
  146. O’Roak, B.J.; Vives, L.; Fu, W.; Egertson, J.D.; Stanaway, I.B.; Phelps, I.G.; Carvill, G.; Kumar, A.; Lee, C.; Ankenman, K.; et al. Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science 2012, 338, 1619–1622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Tarabeux, J.; Kebir, O.; Gauthier, J.; Hamdan, F.F.; Xiong, L.; Piton, A.; Spiegelman, D.; Henrion, E.; Millet, B.; S2D Team; et al. Rare mutations in N-methyl-D-aspartate glutamate receptors in autism spectrum disorders and schizophrenia. Transl. Psychiatry 2011, 1, e55. [Google Scholar] [CrossRef] [Green Version]
  148. Hu, C.; Chen, W.; Myers, S.J.; Yuan, H.; Traynelis, S.F. Human GRIN2B variants in neurodevelopmental disorders. J. Pharmacol. Sci. 2016, 132, 115–121. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, J.; Ma, Y.; Fan, R.; Yang, Z.; Li, M.D. Implication of Genes for the N-Methyl-D-Aspartate (NMDA) Receptor in Substance Addictions. Mol. Neurobiol. 2018, 55, 7567–7578. [Google Scholar] [CrossRef]
  150. Wink, L.K.; Minshawi, N.F.; Shaffer, R.C.; Plawecki, M.H.; Posey, D.J.; Horn, P.S.; Adams, R.; Pedapati, E.V.; Schaefer, T.L.; McDougle, C.J.; et al. d-Cycloserine enhances durability of social skills training in autism spectrum disorder. Mol. Autism 2017, 8, 2. [Google Scholar] [CrossRef] [Green Version]
  151. Wu, H.F.; Chen, P.S.; Hsu, Y.T.; Lee, C.W.; Wang, T.F.; Chen, Y.J.; Lin, H.C. (D)-Cycloserine Ameliorates Autism-Like Deficits by Removing GluA2-Containing AMPA Receptors in a Valproic Acid-Induced Rat Model. Mol. Neurobiol. 2018, 55, 4811–4824. [Google Scholar] [CrossRef]
  152. Hosenbocus, S.; Chahal, R. Memantine: A review of possible uses in child and adolescent psychiatry. J. Can. Acad. Child Adolesc. Psychiatry 2013, 22, 166–171. [Google Scholar]
  153. Soorya, L.V.; Fogg, L.; Ocampo, E.; Printen, M.; Youngkin, S.; Halpern, D.; Kolevzon, A.; Lee, S.; Grodberg, D.; Anagnostou, E. Neurocognitive Outcomes from Memantine: A Pilot, Double-Blind, Placebo-Controlled Trial in Children with Autism Spectrum Disorder. J. Child Adolesc. Psychopharmacol. 2021, 31, 475–484. [Google Scholar] [CrossRef] [PubMed]
  154. Budreck, E.C.; Kwon, O.B.; Jung, J.H.; Baudouin, S.; Thommen, A.; Kim, H.S.; Fukazawa, Y.; Harada, H.; Tabuchi, K.; Shigemoto, R.; et al. Neuroligin-1 controls synaptic abundance of NMDA-type glutamate receptors through extracellular coupling. Proc. Natl. Acad. Sci. USA 2013, 110, 725–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Berkel, S.; Marshall, C.R.; Weiss, B.; Howe, J.; Roeth, R.; Moog, U.; Endris, V.; Roberts, W.; Szatmari, P.; Pinto, D.; et al. Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat. Genet. 2010, 42, 489–491. [Google Scholar] [CrossRef] [PubMed]
  156. Won, H.; Lee, H.R.; Gee, H.Y.; Mah, W.; Kim, J.I.; Lee, J.; Ha, S.; Chung, C.; Jung, E.S.; Cho, Y.S.; et al. Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature 2012, 486, 261–265. [Google Scholar] [CrossRef] [PubMed]
  157. Silverman, J.L.; Smith, D.G.; Rizzo, S.J.; Karras, M.N.; Turner, S.M.; Tolu, S.S.; Bryce, D.K.; Smith, D.L.; Fonseca, K.; Ring, R.H.; et al. Negative allosteric modulation of the mGluR5 receptor reduces repetitive behaviors and rescues social deficits in mouse models of autism. Sci. Transl. Med. 2012, 4, 131ra151. [Google Scholar] [CrossRef] [Green Version]
  158. Benson, A.D.; Burket, J.A.; Deutsch, S.I. Balb/c mice treated with D-cycloserine arouse increased social interest in conspecifics. Brain Res. Bull. 2013, 99, 95–99. [Google Scholar] [CrossRef]
  159. Auerbach, B.D.; Osterweil, E.K.; Bear, M.F. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 2011, 480, 63–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Albuquerque, E.X.; Pereira, E.F.; Alkondon, M.; Rogers, S.W. Mammalian nicotinic acetylcholine receptors: From structure to function. Physiol. Rev. 2009, 89, 73–120. [Google Scholar] [CrossRef] [Green Version]
  161. Bouzat, C.; Lasala, M.; Nielsen, B.E.; Corradi, J.; Esandi, M.D.C. Molecular function of alpha7 nicotinic receptors as drug targets. J. Physiol. 2018, 596, 1847–1861. [Google Scholar] [CrossRef] [Green Version]
  162. Broide, R.S.; Winzer-Serhan, U.H.; Chen, Y.; Leslie, F.M. Distribution of alpha7 Nicotinic Acetylcholine Receptor Subunit mRNA in the Developing Mouse. Front. Neuroanat. 2019, 13, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Eissa, N.; Al-Houqani, M.; Sadeq, A.; Ojha, S.K.; Sasse, A.; Sadek, B. Current Enlightenment About Etiology and Pharmacological Treatment of Autism Spectrum Disorder. Front. Neurosci. 2018, 12, 304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Wu, W.L.; Adams, C.E.; Stevens, K.E.; Chow, K.H.; Freedman, R.; Patterson, P.H. The interaction between maternal immune activation and alpha 7 nicotinic acetylcholine receptor in regulating behaviors in the offspring. Brain Behav. Immun. 2015, 46, 192–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. McTighe, S.M.; Neal, S.J.; Lin, Q.; Hughes, Z.A.; Smith, D.G. The BTBR mouse model of autism spectrum disorders has learning and attentional impairments and alterations in acetylcholine and kynurenic acid in prefrontal cortex. PLoS ONE 2013, 8, e62189. [Google Scholar] [CrossRef] [PubMed]
  166. Yoshimura, R.F.; Tran, M.B.; Hogenkamp, D.J.; Ayala, N.L.; Johnstone, T.; Dunnigan, A.J.; Gee, T.K.; Gee, K.W. Allosteric modulation of nicotinic and GABA(A) receptor subtypes differentially modify autism-like behaviors in the BTBR mouse model. Neuropharmacology 2017, 126, 38–47. [Google Scholar] [CrossRef]
  167. Mahmood, H.M.; Aldhalaan, H.M.; Alshammari, T.K.; Alqasem, M.A.; Alshammari, M.A.; Albekairi, N.A.; AlSharari, S.D. The Role of Nicotinic Receptors in the Attenuation of Autism-Related Behaviors in a Murine BTBR T + tf/J Autistic Model. Autism Res. 2020, 13, 1311–1334. [Google Scholar] [CrossRef]
  168. Ghaleiha, A.; Ghyasvand, M.; Mohammadi, M.R.; Farokhnia, M.; Yadegari, N.; Tabrizi, M.; Hajiaghaee, R.; Yekehtaz, H.; Akhondzadeh, S. Galantamine efficacy and tolerability as an augmentative therapy in autistic children: A randomized, double-blind, placebo-controlled trial. J. Psychopharmacol. 2014, 28, 677–685. [Google Scholar] [CrossRef]
  169. Buckley, A.W.; Sassower, K.; Rodriguez, A.J.; Jennison, K.; Wingert, K.; Buckley, J.; Thurm, A.; Sato, S.; Swedo, S. An open label trial of donepezil for enhancement of rapid eye movement sleep in young children with autism spectrum disorders. J. Child Adolesc. Psychopharmacol. 2011, 21, 353–357. [Google Scholar] [CrossRef] [PubMed]
  170. Chamberlain, S.J.; Lalande, M. Neurodevelopmental disorders involving genomic imprinting at human chromosome 15q11-q13. Neurobiol. Dis. 2010, 39, 13–20. [Google Scholar] [CrossRef]
  171. Ziats, M.N.; Goin-Kochel, R.P.; Berry, L.N.; Ali, M.; Ge, J.; Guffey, D.; Rosenfeld, J.A.; Bader, P.; Gambello, M.J.; Wolf, V.; et al. The complex behavioral phenotype of 15q13.3 microdeletion syndrome. Genet. Med. 2016, 18, 1111–1118. [Google Scholar] [CrossRef] [Green Version]
  172. Yasui, D.H.; Scoles, H.A.; Horike, S.; Meguro-Horike, M.; Dunaway, K.W.; Schroeder, D.I.; Lasalle, J.M. 15q11.2-13.3 chromatin analysis reveals epigenetic regulation of CHRNA7 with deficiencies in Rett and autism brain. Hum. Mol. Genet. 2011, 20, 4311–4323. [Google Scholar] [CrossRef] [PubMed]
  173. Stone, T.W. Neuropharmacology of quinolinic and kynurenic acids. Pharmacol. Rev. 1993, 45, 309–379. [Google Scholar]
  174. Alkondon, M.; Pereira, E.F.; Eisenberg, H.M.; Kajii, Y.; Schwarcz, R.; Albuquerque, E.X. Age dependency of inhibition of alpha7 nicotinic receptors and tonically active N-methyl-D-aspartate receptors by endogenously produced kynurenic acid in the brain. J. Pharmacol. Exp. Ther. 2011, 337, 572–582. [Google Scholar] [CrossRef] [Green Version]
  175. Alkondon, M.; Pereira, E.F.; Albuquerque, E.X. Endogenous activation of nAChRs and NMDA receptors contributes to the excitability of CA1 stratum radiatum interneurons in rat hippocampal slices: Effects of kynurenic acid. Biochem. Pharmacol. 2011, 82, 842–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Albuquerque, E.X.; Schwarcz, R. Kynurenic acid as an antagonist of alpha7 nicotinic acetylcholine receptors in the brain: Facts and challenges. Biochem. Pharmacol. 2013, 85, 1027–1032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Tribollet, E.; Bertrand, D.; Marguerat, A.; Raggenbass, M. Comparative distribution of nicotinic receptor subtypes during development, adulthood and aging: An autoradiographic study in the rat brain. Neuroscience 2004, 124, 405–420. [Google Scholar] [CrossRef]
  178. Shytle, R.D.; Mori, T.; Townsend, K.; Vendrame, M.; Sun, N.; Zeng, J.; Ehrhart, J.; Silver, A.A.; Sanberg, P.R.; Tan, J. Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J. Neurochem. 2004, 89, 337–343. [Google Scholar] [CrossRef] [PubMed]
  179. Papouin, T.; Dunphy, J.M.; Tolman, M.; Dineley, K.T.; Haydon, P.G. Septal Cholinergic Neuromodulation Tunes the Astrocyte-Dependent Gating of Hippocampal NMDA Receptors to Wakefulness. Neuron 2017, 94, 840–854. [Google Scholar] [CrossRef] [Green Version]
  180. Petrov, K.A.; Girard, E.; Nikitashina, A.D.; Colasante, C.; Bernard, V.; Nurullin, L.; Leroy, J.; Samigullin, D.; Colak, O.; Nikolsky, E.; et al. Schwann cells sense and control acetylcholine spillover at the neuromuscular junction by alpha7 nicotinic receptors and butyrylcholinesterase. J. Neurosci. 2014, 34, 11870–11883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Sharma, G.; Vijayaraghavan, S. Nicotinic receptor signaling in nonexcitable cells. J. Neurobiol. 2002, 53, 524–534. [Google Scholar] [CrossRef] [PubMed]
  182. Konradsson-Geuken, A.; Wu, H.Q.; Gash, C.R.; Alexander, K.S.; Campbell, A.; Sozeri, Y.; Pellicciari, R.; Schwarcz, R.; Bruno, J.P. Cortical kynurenic acid bi-directionally modulates prefrontal glutamate levels as assessed by microdialysis and rapid electrochemistry. Neuroscience 2010, 169, 1848–1859. [Google Scholar] [CrossRef] [Green Version]
  183. Potter, M.C.; Elmer, G.I.; Bergeron, R.; Albuquerque, E.X.; Guidetti, P.; Wu, H.Q.; Schwarcz, R. Reduction of endogenous kynurenic acid formation enhances extracellular glutamate, hippocampal plasticity, and cognitive behavior. Neuropsychopharmacology 2010, 35, 1734–1742. [Google Scholar] [CrossRef] [PubMed]
  184. Wu, H.Q.; Pereira, E.F.; Bruno, J.P.; Pellicciari, R.; Albuquerque, E.X.; Schwarcz, R. The astrocyte-derived alpha7 nicotinic receptor antagonist kynurenic acid controls extracellular glutamate levels in the prefrontal cortex. J. Mol. Neurosci. 2010, 40, 204–210. [Google Scholar] [CrossRef] [Green Version]
  185. Beggiato, S.; Antonelli, T.; Tomasini, M.C.; Tanganelli, S.; Fuxe, K.; Schwarcz, R.; Ferraro, L. Kynurenic acid, by targeting alpha7 nicotinic acetylcholine receptors, modulates extracellular GABA levels in the rat striatum in vivo. Eur. J. Neurosci. 2013, 37, 1470–1477. [Google Scholar] [CrossRef]
  186. Rassoulpour, A.; Wu, H.Q.; Ferre, S.; Schwarcz, R. Nanomolar concentrations of kynurenic acid reduce extracellular dopamine levels in the striatum. J. Neurochem. 2005, 93, 762–765. [Google Scholar] [CrossRef]
  187. Zmarowski, A.; Wu, H.Q.; Brooks, J.M.; Potter, M.C.; Pellicciari, R.; Schwarcz, R.; Bruno, J.P. Astrocyte-derived kynurenic acid modulates basal and evoked cortical acetylcholine release. Eur. J. Neurosci. 2009, 29, 529–538. [Google Scholar] [CrossRef]
  188. Liu, Z.; Zhang, J.; Berg, D.K. Role of endogenous nicotinic signaling in guiding neuronal development. Biochem. Pharmacol. 2007, 74, 1112–1119. [Google Scholar] [CrossRef] [Green Version]
  189. Liu, Z.; Otsu, Y.; Vasuta, C.; Nawa, H.; Murphy, T.H. Action-potential-independent GABAergic tone mediated by nicotinic stimulation of immature striatal miniature synaptic transmission. J. Neurophysiol. 2007, 98, 581–593. [Google Scholar] [CrossRef]
  190. Lozada, A.F.; Wang, X.; Gounko, N.V.; Massey, K.A.; Duan, J.; Liu, Z.; Berg, D.K. Glutamatergic synapse formation is promoted by alpha7-containing nicotinic acetylcholine receptors. J. Neurosci. 2012, 32, 7651–7661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Dhulkifle, H.; Agouni, A.; Zeidan, A.; Al-Kuwari, M.S.; Parray, A.; Tolefat, M.; Korashy, H.M. Influence of the Aryl Hydrocarbon Receptor Activating Environmental Pollutants on Autism Spectrum Disorder. Int. J. Mol. Sci. 2021, 22, 9258. [Google Scholar] [CrossRef] [PubMed]
  192. Schultz, R.; Suominen, J.; Varre, T.; Hakovirta, H.; Parvinen, M.; Toppari, J.; Pelto-Huikko, M. Expression of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator messenger ribonucleic acids and proteins in rat and human testis. Endocrinology 2003, 144, 767–776. [Google Scholar] [CrossRef]
  193. Kimura, E.; Kubo, K.I.; Endo, T.; Nakajima, K.; Kakeyama, M.; Tohyama, C. Excessive activation of AhR signaling disrupts neuronal migration in the hippocampal CA1 region in the developing mouse. J. Toxicol. Sci. 2017, 42, 25–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Kimura, E.; Kohda, M.; Maekawa, F.; Fujii-Kuriyama, Y.; Tohyama, C. Correction to: Neurons expressing the aryl hydrocarbon receptor in the locus coeruleus and island of Calleja major are novel targets of dioxin in the mouse brain. Histochem. Cell. Biol. 2021, 156, 293. [Google Scholar] [CrossRef] [PubMed]
  195. Wang, W.; Huang, L.; Jin, J.Y.; Jolly, S.; Zang, Y.; Wu, H.; Yan, L.; Pi, W.; Li, L.; Mellor, A.L.; et al. IDO Immune Status after Chemoradiation May Predict Survival in Lung Cancer Patients. Cancer Res. 2018, 78, 809–816. [Google Scholar] [CrossRef] [Green Version]
  196. DiNatale, B.C.; Murray, I.A.; Schroeder, J.C.; Flaveny, C.A.; Lahoti, T.S.; Laurenzana, E.M.; Omiecinski, C.J.; Perdew, G.H. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol. Sci. 2010, 115, 89–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Garcia-Lara, L.; Perez-Severiano, F.; Gonzalez-Esquivel, D.; Elizondo, G.; Segovia, J. Absence of aryl hydrocarbon receptors increases endogenous kynurenic acid levels and protects mouse brain against excitotoxic insult and oxidative stress. J. Neurosci. Res. 2015, 93, 1423–1433. [Google Scholar] [CrossRef]
  198. Gomez-Fernandez, A.; de la Torre-Aguilar, M.J.; Gil-Campos, M.; Flores-Rojas, K.; Cruz-Rico, M.D.; Martin-Borreguero, P.; Perez-Navero, J.L. Children with Autism Spectrum Disorder with Regression Exhibit a Different Profile in Plasma Cytokines and Adhesion Molecules Compared to Children Without Such Regression. Front. Pediatr. 2018, 6, 264. [Google Scholar] [CrossRef] [Green Version]
  199. Yoshimura, Y.; Kikuchi, M.; Saito, D.N.; Hirosawa, T.; Takahashi, T.; Munesue, T.; Kosaka, H.; Naito, N.; Ouchi, Y.; Minabe, Y. Markers for the central serotonin system correlate to verbal ability and paralinguistic social voice processing in autism spectrum disorder. Sci. Rep. 2020, 10, 14558. [Google Scholar] [CrossRef]
  200. Muller, C.L.; Anacker, A.M.J.; Veenstra-VanderWeele, J. The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 2016, 321, 24–41. [Google Scholar] [CrossRef] [Green Version]
  201. Wang, X.D.; Notarangelo, F.M.; Wang, J.Z.; Schwarcz, R. Kynurenic acid and 3-hydroxykynurenine production from D-kynurenine in mice. Brain Res. 2012, 1455, 1–9. [Google Scholar] [CrossRef] [Green Version]
  202. Gonzalez Esquivel, D.; Ramirez-Ortega, D.; Pineda, B.; Castro, N.; Rios, C.; Perez de la Cruz, V. Kynurenine pathway metabolites and enzymes involved in redox reactions. Neuropharmacology 2017, 112, 331–345. [Google Scholar] [CrossRef] [PubMed]
  203. Maddison, D.C.; Giorgini, F. The kynurenine pathway and neurodegenerative disease. Semin. Cell Dev. Biol. 2015, 40, 134–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Jenabi, E.; Karami, M.; Khazaei, S.; Bashirian, S. The association between preeclampsia and autism spectrum disorders among children: A meta-analysis. Korean J. Pediatr. 2019, 62, 126–130. [Google Scholar] [CrossRef] [PubMed]
  205. Lampi, K.M.; Lehtonen, L.; Tran, P.L.; Suominen, A.; Lehti, V.; Banerjee, P.N.; Gissler, M.; Brown, A.S.; Sourander, A. Risk of autism spectrum disorders in low birth weight and small for gestational age infants. J. Pediatr. 2012, 161, 830–836. [Google Scholar] [CrossRef] [Green Version]
  206. Crump, C.; Sundquist, J.; Sundquist, K. Preterm or Early Term Birth and Risk of Autism. Pediatrics 2021, 148, e2020032300. [Google Scholar] [CrossRef] [PubMed]
  207. Ormstad, H.; Bryn, V.; Verkerk, R.; Skjeldal, O.H.; Halvorsen, B.; Saugstad, O.D.; Isaksen, J.; Maes, M. Serum Tryptophan, Tryptophan Catabolites and Brain-derived Neurotrophic Factor in Subgroups of Youngsters with Autism Spectrum Disorders. CNS Neurol. Disord. Drug Targets 2018, 17, 626–639. [Google Scholar] [CrossRef] [Green Version]
  208. Bilgic, A.; Abusoglu, S.; Sadic Celikkol, C.; Oflaz, M.B.; Akca, O.F.; Sivrikaya, A.; Baysal, T.; Unlu, A. Altered kynurenine pathway metabolite levels in toddlers and preschool children with autism spectrum disorder. Int. J. Neurosci. 2022, 132, 826–834. [Google Scholar] [CrossRef] [PubMed]
  209. Yu, X.; Qian-Qian, L.; Cong, Y.; Xiao-Bing, Z.; Hong-Zhu, D. Reduction of essential amino acid levels and sex-specific alterations in serum amino acid concentration profiles in children with autism spectrum disorder. Psychiatry Res. 2021, 297, 113675. [Google Scholar] [CrossRef] [PubMed]
  210. Tu, W.J.; Chen, H.; He, J. Application of LC-MS/MS analysis of plasma amino acids profiles in children with autism. J. Clin. Biochem. Nutr. 2012, 51, 248–249. [Google Scholar] [CrossRef] [Green Version]
  211. Naushad, S.M.; Jain, J.M.; Prasad, C.K.; Naik, U.; Akella, R.R. Autistic children exhibit distinct plasma amino acid profile. Indian J. Biochem. Biophys. 2013, 50, 474–478. [Google Scholar]
  212. Chen, W.X.; Chen, Y.R.; Peng, M.Z.; Liu, X.; Cai, Y.N.; Huang, Z.F.; Yang, S.Y.; Huang, J.Y.; Wang, R.H.; Yi, P.; et al. Plasma Amino Acid Profile in Children with Autism Spectrum Disorder in Southern China: Analysis of 110 Cases. J. Autism Dev. Disord. 2023. [Google Scholar] [CrossRef]
  213. Adams, J.B.; Audhya, T.; McDonough-Means, S.; Rubin, R.A.; Quig, D.; Geis, E.; Gehn, E.; Loresto, M.; Mitchell, J.; Atwood, S.; et al. Nutritional and metabolic status of children with autism vs. neurotypical children, and the association with autism severity. Nutr. Metab. 2011, 8, 34. [Google Scholar] [CrossRef] [Green Version]
  214. Noto, A.; Fanos, V.; Barberini, L.; Grapov, D.; Fattuoni, C.; Zaffanello, M.; Casanova, A.; Fenu, G.; De Giacomo, A.; De Angelis, M.; et al. The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J. Matern.-Fetal Neonatal Med. 2014, 27, 46–52. [Google Scholar] [CrossRef]
  215. Kaluzna-Czaplinska, J.; Zurawicz, E.; Struck, W.; Markuszewski, M. Identification of organic acids as potential biomarkers in the urine of autistic children using gas chromatography/mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2014, 966, 70–76. [Google Scholar] [CrossRef] [PubMed]
  216. Gevi, F.; Zolla, L.; Gabriele, S.; Persico, A.M. Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism. Mol. Autism 2016, 7, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Timperio, A.M.; Gevi, F.; Cucinotta, F.; Ricciardello, A.; Turriziani, L.; Scattoni, M.L.; Persico, A.M. Urinary Untargeted Metabolic Profile Differentiates Children with Autism from Their Unaffected Siblings. Metabolites 2022, 12, 797. [Google Scholar] [CrossRef] [PubMed]
  218. Harutyunyan, A.A.; Harutyunyan, H.A.; Yenkoyan, K.B. Novel Probable Glance at Inflammatory Scenario Development in Autistic Pathology. Front. Psychiatry 2021, 12, 788779. [Google Scholar] [CrossRef] [PubMed]
  219. Zimmerman, A.W.; Jyonouchi, H.; Comi, A.M.; Connors, S.L.; Milstien, S.; Varsou, A.; Heyes, M.P. Cerebrospinal fluid and serum markers of inflammation in autism. Pediatr. Neurol. 2005, 33, 195–201. [Google Scholar] [CrossRef] [PubMed]
  220. Yang, C.J.; Liu, C.L.; Sang, B.; Zhu, X.M.; Du, Y.J. The combined role of serotonin and interleukin-6 as biomarker for autism. Neuroscience 2015, 284, 290–296. [Google Scholar] [CrossRef] [PubMed]
  221. Gabriele, S.; Sacco, R.; Persico, A.M. Blood serotonin levels in autism spectrum disorder: A systematic review and meta-analysis. Eur. Neuropsychopharmacol. 2014, 24, 919–929. [Google Scholar] [CrossRef]
  222. Nakai, N.; Nagano, M.; Saitow, F.; Watanabe, Y.; Kawamura, Y.; Kawamoto, A.; Tamada, K.; Mizuma, H.; Onoe, H.; Watanabe, Y.; et al. Serotonin rebalances cortical tuning and behavior linked to autism symptoms in 15q11-13 CNV mice. Sci. Adv. 2017, 3, e1603001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Rind, H.B.; Russo, A.F.; Whittemore, S.R. Developmental regulation of tryptophan hydroxylase messenger RNA expression and enzyme activity in the raphe and its target fields. Neuroscience 2000, 101, 665–677. [Google Scholar] [CrossRef]
  224. Yang, S.Y.; Yoo, H.J.; Cho, I.H.; Park, M.; Kim, S.A. Association with tryptophan hydroxylase 2 gene polymorphisms and autism spectrum disorders in Korean families. Neurosci. Res. 2012, 73, 333–336. [Google Scholar] [CrossRef]
  225. Rodriguez-Gomez, D.A.; Garcia-Guaqueta, D.P.; Charry-Sanchez, J.D.; Sarquis-Buitrago, E.; Blanco, M.; Velez-van-Meerbeke, A.; Talero-Gutierrez, C. A systematic review of common genetic variation and biological pathways in autism spectrum disorder. BMC Neurosci. 2021, 22, 60. [Google Scholar] [CrossRef]
  226. Saitow, F.; Takumi, T.; Suzuki, H. Change in serotonergic modulation contributes to the synaptic imbalance of neuronal circuit at the prefrontal cortex in the 15q11-13 duplication mouse model of autism. Neuropharmacology 2020, 165, 107931. [Google Scholar] [CrossRef] [PubMed]
  227. Takumi, T.; Tamada, K.; Hatanaka, F.; Nakai, N.; Bolton, P.F. Behavioral neuroscience of autism. Neurosci. Biobehav. Rev. 2020, 110, 60–76. [Google Scholar] [CrossRef]
  228. Lauder, J.M.; Krebs, H. Serotonin as a differentiation signal in early neurogenesis. Dev. Neurosci. 1978, 1, 15–30. [Google Scholar] [CrossRef]
  229. Salter, M.; Knowles, R.G.; Pogson, C.I. How does displacement of albumin-bound tryptophan cause sustained increases in the free tryptophan concentration in plasma and 5-hydroxytryptamine synthesis in brain? Biochem. J. 1989, 262, 365–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Nishizawa, S.; Benkelfat, C.; Young, S.N.; Leyton, M.; Mzengeza, S.; de Montigny, C.; Blier, P.; Diksic, M. Differences between males and females in rates of serotonin synthesis in human brain. Proc. Natl. Acad. Sci. USA 1997, 94, 5308–5313. [Google Scholar] [CrossRef] [Green Version]
  231. Mendelsohn, D.; Riedel, W.J.; Sambeth, A. Effects of acute tryptophan depletion on memory, attention and executive functions: A systematic review. Neurosci. Biobehav. Rev. 2009, 33, 926–952. [Google Scholar] [CrossRef]
  232. Robinson, N.; Bergen, S.E. Environmental Risk Factors for Schizophrenia and Bipolar Disorder and Their Relationship to Genetic Risk: Current Knowledge and Future Directions. Front. Genet. 2021, 12, 686666. [Google Scholar] [CrossRef] [PubMed]
  233. Lyall, K.; Schmidt, R.J.; Hertz-Picciotto, I. Maternal lifestyle and environmental risk factors for autism spectrum disorders. Int. J. Epidemiol. 2014, 43, 443–464. [Google Scholar] [CrossRef] [Green Version]
  234. Notarangelo, F.M.; Schwarcz, R. Restraint Stress during Pregnancy Rapidly Raises Kynurenic Acid Levels in Mouse Placenta and Fetal Brain. Dev. Neurosci. 2016, 38, 458–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Notarangelo, F.M.; Schwarcz, R. A single prenatal lipopolysaccharide injection has acute, but not long-lasting, effects on cerebral kynurenine pathway metabolism in mice. Eur. J. Neurosci. 2021, 54, 5968–5981. [Google Scholar] [CrossRef]
  236. Holloway, T.; Moreno, J.L.; Umali, A.; Rayannavar, V.; Hodes, G.E.; Russo, S.J.; Gonzalez-Maeso, J. Prenatal stress induces schizophrenia-like alterations of serotonin 2A and metabotropic glutamate 2 receptors in the adult offspring: Role of maternal immune system. J. Neurosci. 2013, 33, 1088–1098. [Google Scholar] [CrossRef] [PubMed]
  237. MacDowell, K.S.; Munarriz-Cuezva, E.; Meana, J.J.; Leza, J.C.; Ortega, J.E. Paliperidone Reversion of Maternal Immune Activation-Induced Changes on Brain Serotonin and Kynurenine Pathways. Front. Pharmacol. 2021, 12, 682602. [Google Scholar] [CrossRef] [PubMed]
  238. Canetta, S.; Bolkan, S.; Padilla-Coreano, N.; Song, L.J.; Sahn, R.; Harrison, N.L.; Gordon, J.A.; Brown, A.; Kellendonk, C. Maternal immune activation leads to selective functional deficits in offspring parvalbumin interneurons. Mol. Psychiatry 2016, 21, 956–968. [Google Scholar] [CrossRef] [PubMed]
  239. Khalil, O.S.; Forrest, C.M.; Pisar, M.; Smith, R.A.; Darlington, L.G.; Stone, T.W. Prenatal activation of maternal TLR3 receptors by viral-mimetic poly(I:C) modifies GluN2B expression in embryos and sonic hedgehog in offspring in the absence of kynurenine pathway activation. Immunopharmacol. Immunotoxicol. 2013, 35, 581–593. [Google Scholar] [CrossRef] [PubMed]
  240. Zavitsanou, K.; Lim, C.K.; Purves-Tyson, T.; Karl, T.; Kassiou, M.; Banister, S.D.; Guillemin, G.J.; Weickert, C.S. Effect of maternal immune activation on the kynurenine pathway in preadolescent rat offspring and on MK801-induced hyperlocomotion in adulthood: Amelioration by COX-2 inhibition. Brain Behav. Immun. 2014, 41, 173–181. [Google Scholar] [CrossRef]
  241. Forrest, C.M.; Kennedy, P.G.; Rodgers, J.; Dalton, R.N.; Turner, C.; Darlington, L.G.; Cobb, S.R.; Stone, T.W. Kynurenine pathway metabolism following prenatal KMO inhibition and in Mecp2(+/−) mice, using liquid chromatography-tandem mass spectrometry. Neurochem. Int. 2016, 100, 110–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Forrest, C.M.; Khalil, O.S.; Pisar, M.; Darlington, L.G.; Stone, T.W. Prenatal inhibition of the tryptophan-kynurenine pathway alters synaptic plasticity and protein expression in the rat hippocampus. Brain Res. 2013, 1504, 1–15. [Google Scholar] [CrossRef] [Green Version]
  243. Forrest, C.M.; Khalil, O.S.; Pisar, M.; McNair, K.; Kornisiuk, E.; Snitcofsky, M.; Gonzalez, N.; Jerusalinsky, D.; Darlington, L.G.; Stone, T.W. Changes in synaptic transmission and protein expression in the brains of adult offspring after prenatal inhibition of the kynurenine pathway. Neuroscience 2013, 254, 241–259. [Google Scholar] [CrossRef] [Green Version]
  244. Forrest, C.M.; McNair, K.; Pisar, M.; Khalil, O.S.; Darlington, L.G.; Stone, T.W. Altered hippocampal plasticity by prenatal kynurenine administration, kynurenine-3-monoxygenase (KMO) deletion or galantamine. Neuroscience 2015, 310, 91–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Pershing, M.L.; Phenis, D.; Valentini, V.; Pocivavsek, A.; Lindquist, D.H.; Schwarcz, R.; Bruno, J.P. Prenatal kynurenine exposure in rats: Age-dependent changes in NMDA receptor expression and conditioned fear responding. Psychopharmacology 2016, 233, 3725–3735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Rentschler, K.M.; Baratta, A.M.; Ditty, A.L.; Wagner, N.T.J.; Wright, C.J.; Milosavljevic, S.; Mong, J.A.; Pocivavsek, A. Prenatal Kynurenine Elevation Elicits Sex-Dependent Changes in Sleep and Arousal During Adulthood: Implications for Psychotic Disorders. Schizophr. Bull. 2021, 47, 1320–1330. [Google Scholar] [CrossRef]
  247. Wright, C.J.; Rentschler, K.M.; Wagner, N.T.J.; Lewis, A.M.; Beggiato, S.; Pocivavsek, A. Time of Day-Dependent Alterations in Hippocampal Kynurenic Acid, Glutamate, and GABA in Adult Rats Exposed to Elevated Kynurenic Acid During Neurodevelopment. Front. Psychiatry 2021, 12, 734984. [Google Scholar] [CrossRef] [PubMed]
  248. Pocivavsek, A.; Wu, H.Q.; Elmer, G.I.; Bruno, J.P.; Schwarcz, R. Pre- and postnatal exposure to kynurenine causes cognitive deficits in adulthood. Eur. J. Neurosci. 2012, 35, 1605–1612. [Google Scholar] [CrossRef] [Green Version]
  249. Alexander, K.S.; Pocivavsek, A.; Wu, H.Q.; Pershing, M.L.; Schwarcz, R.; Bruno, J.P. Early developmental elevations of brain kynurenic acid impair cognitive flexibility in adults: Reversal with galantamine. Neuroscience 2013, 238, 19–28. [Google Scholar] [CrossRef] [Green Version]
  250. Iaccarino, H.F.; Suckow, R.F.; Xie, S.; Bucci, D.J. The effect of transient increases in kynurenic acid and quinolinic acid levels early in life on behavior in adulthood: Implications for schizophrenia. Schizophr. Res. 2013, 150, 392–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Liu, X.C.; Holtze, M.; Powell, S.B.; Terrando, N.; Larsson, M.K.; Persson, A.; Olsson, S.K.; Orhan, F.; Kegel, M.; Asp, L.; et al. Behavioral disturbances in adult mice following neonatal virus infection or kynurenine treatment--role of brain kynurenic acid. Brain Behav. Immun. 2014, 36, 80–89. [Google Scholar] [CrossRef] [Green Version]
  252. Tufvesson-Alm, M.; Schwieler, L.; Schwarcz, R.; Goiny, M.; Erhardt, S.; Engberg, G. Importance of kynurenine 3-monooxygenase for spontaneous firing and pharmacological responses of midbrain dopamine neurons: Relevance for schizophrenia. Neuropharmacology 2018, 138, 130–139. [Google Scholar] [CrossRef]
  253. Alkondon, M.; Pereira, E.F.; Yu, P.; Arruda, E.Z.; Almeida, L.E.; Guidetti, P.; Fawcett, W.P.; Sapko, M.T.; Randall, W.R.; Schwarcz, R.; et al. Targeted deletion of the kynurenine aminotransferase ii gene reveals a critical role of endogenous kynurenic acid in the regulation of synaptic transmission via alpha7 nicotinic receptors in the hippocampus. J. Neurosci. 2004, 24, 4635–4648. [Google Scholar] [CrossRef] [Green Version]
  254. Sapko, M.T.; Guidetti, P.; Yu, P.; Tagle, D.A.; Pellicciari, R.; Schwarcz, R. Endogenous kynurenate controls the vulnerability of striatal neurons to quinolinate: Implications for Huntington’s disease. Exp. Neurol. 2006, 197, 31–40. [Google Scholar] [CrossRef] [PubMed]
  255. Meyza, K.Z.; Blanchard, D.C. The BTBR mouse model of idiopathic autism—Current view on mechanisms. Neurosci. Biobehav. Rev. 2017, 76, 99–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Akaba, Y.; Shiohama, T.; Komaki, Y.; Seki, F.; Ortug, A.; Sawada, D.; Uchida, W.; Kamagata, K.; Shimoji, K.; Aoki, S.; et al. Comprehensive Volumetric Analysis of Mecp2-Null Mouse Model for Rett Syndrome by T2-Weighted 3D Magnetic Resonance Imaging. Front. Neurosci. 2022, 16, 885335. [Google Scholar] [CrossRef] [PubMed]
  257. Almulla, A.F.; Vasupanrajit, A.; Tunvirachaisakul, C.; Al-Hakeim, H.K.; Solmi, M.; Verkerk, R.; Maes, M. The tryptophan catabolite or kynurenine pathway in schizophrenia: Meta-analysis reveals dissociations between central, serum, and plasma compartments. Mol. Psychiatry 2022, 27, 3679–3691. [Google Scholar] [CrossRef] [PubMed]
  258. Giorgini, F.; Huang, S.Y.; Sathyasaikumar, K.V.; Notarangelo, F.M.; Thomas, M.A.; Tararina, M.; Wu, H.Q.; Schwarcz, R.; Muchowski, P.J. Targeted deletion of kynurenine 3-monooxygenase in mice: A new tool for studying kynurenine pathway metabolism in periphery and brain. J. Biol. Chem. 2013, 288, 36554–36566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  259. Walker, A.K.; Wing, E.E.; Banks, W.A.; Dantzer, R. Leucine competes with kynurenine for blood-to-brain transport and prevents lipopolysaccharide-induced depression-like behavior in mice. Mol. Psychiatry 2019, 24, 1523–1532. [Google Scholar] [CrossRef]
  260. Amori, L.; Wu, H.Q.; Marinozzi, M.; Pellicciari, R.; Guidetti, P.; Schwarcz, R. Specific inhibition of kynurenate synthesis enhances extracellular dopamine levels in the rodent striatum. Neuroscience 2009, 159, 196–203. [Google Scholar] [CrossRef] [Green Version]
  261. Linderholm, K.R.; Alm, M.T.; Larsson, M.K.; Olsson, S.K.; Goiny, M.; Hajos, M.; Erhardt, S.; Engberg, G. Inhibition of kynurenine aminotransferase II reduces activity of midbrain dopamine neurons. Neuropharmacology 2016, 102, 42–47. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Simplified diagram of the Tryptophan (Trp) metabolism through serotonin (5-HT) and kynurenine (Kyn) pathways and direct metabolism by microorganisms. Pro-inflammatory cytokines, which upregulate enzyme activity of indoleamine 2,3-dioxygenase1 (IDO1) and tryptophan 2,3-dioxygenase (TDO), are highlighted by “+”. The triangle shows enzymes. Trapezoid with dotted line shows intestinal microbial pathway. TPH, tryptophan hydroxylase; KMO, kynurenine 3-monooxygenase; KATs, kynurenine aminotransferases; KYNU, kynureninase; QPRT, quinolinic phosphoribosyltransferase; TNA, tryptophanase; AraT, aromatic amino acid aminotransferase; TMO, tryptophan 2-monooxygenase. * Aryl hydrocarbon receptor (AhR) ligands, ** potential AhR ligand.
Figure 1. Simplified diagram of the Tryptophan (Trp) metabolism through serotonin (5-HT) and kynurenine (Kyn) pathways and direct metabolism by microorganisms. Pro-inflammatory cytokines, which upregulate enzyme activity of indoleamine 2,3-dioxygenase1 (IDO1) and tryptophan 2,3-dioxygenase (TDO), are highlighted by “+”. The triangle shows enzymes. Trapezoid with dotted line shows intestinal microbial pathway. TPH, tryptophan hydroxylase; KMO, kynurenine 3-monooxygenase; KATs, kynurenine aminotransferases; KYNU, kynureninase; QPRT, quinolinic phosphoribosyltransferase; TNA, tryptophanase; AraT, aromatic amino acid aminotransferase; TMO, tryptophan 2-monooxygenase. * Aryl hydrocarbon receptor (AhR) ligands, ** potential AhR ligand.
Cells 12 01087 g001
Figure 2. Summary of the review.
Figure 2. Summary of the review.
Cells 12 01087 g002
Table 1. Changes of kynurenine (Kyn) metabolites’ profile in ASD patients.
Table 1. Changes of kynurenine (Kyn) metabolites’ profile in ASD patients.
Age
(Mean ± SD, Range)
CountrySpecimenDiagnostic
Criteria
Detection MethodObservationRef.
11.2 ± 2.02 yNorwaySerumADI-R, ADOS, ICD-10HPLCKyna (⇈), QUIN (⇈)[1]
[207]
8.47 ± 2.36 y, 3-10 yAustralia DSM-IV, CARSUHPLCKyn (⇈), QUIN (⇈)[8]
35.5 ± 9.9 mTurkey DSM-5, CARSHPLC3-HK (⇈), Kyna (⇈)[208]
42.86 ± 11.03 mChina DSM-IV, ADI-R, ADOSTMS/MSTrp (⇊)[209]
3.46 ±0.56 y, 2–6 yChinaPlasmaDSM-IVLC-MS/MSTrp (⇊)[210]
4.4 ± 1.7 yIndia DSM-IV, ABCReverse-phase HPLCTrp (⇊)[211]
3.22 ± 1.18 y, 1–14 yChina DSM-5, ADI-R, ADOSLC-MS/MSTrp (⇊)[212]
10.0 ± 3.1 y, 5–16 yUSA DSM-IVLC-MS/MSTrp (⇊)[213]
27.75 ± 6.97 yItaly DSM-5,RAADS-14,RRS, WSAS ELISATrp (⇊), Kyna (⇊)[2]
4–16 yItalyUrineADOSGC-MSTrp (⇈)[214]
4–10 yPoland DSM-IVGC-MSTrp (⇊)[215]
4.83 ± 2.40 y, 3–7 yItaly DSM-IV, ADI-R, ADOS, CARSHILIC-UHPLCKyn (⇊), Kyna (⇊), QUIN (⇈), 5-HT (⇊)[216]
7.06 ± 0.96 yItaly DSM-IV, DSM-5, ADI-R, ADOSUHPLCKyn (⇊)[217]
3–6 yArmenia ADI-R, ADOS, DSM-IVLC-MS/MSQUIN (⇈)[218]
6.1 y, 33 m-10 yUSACSFDSM-IV, ADI-RGC-MSQUIN (⇊)[219]
Significantly decreased (⇊), significantly increased (⇈). y, years old; m, months old; ADI-R, Autism Diagnostic Interview-Revised; ADOS, Autism Diagnostic Observation Schedule; ICD-10, International Statistical Classification of Diseases and Related Health Problems-10; HPLC, high-performance liquid chromatography; DSM-IV, Diagnostic and Statistical Manual of Mental Disorder, Fourth Edition; CARS, Childhood Autism Rating Scale; UHPLC, ultra-high-performance liquid chromatography; DSM-5, Diagnostic and Statistical Manual of Mental Disorder, Fifth Edition; TMS/MS, tandem mass spectrometry; LC-MS/MS, liquid chromatography—tandem mass spectrometry; ABC, Aberrant Behavior Checklist; RAADS-14, Ritvo Autism & Asperger Diagnostic Scale-14; RRS, Ruminative Response Scale; WSAS, the Work and Social Adjustment Scale; ELISA, enzyme-linked immunosorbent assay; GC-MS, gas chromatography—mass spectrometry; HILIC, hydrophilic interaction chromatography.
Table 2. Effect of disrupted Kynurenine (Kyn) metabolites during the neurodevelopmental period in animal models.
Table 2. Effect of disrupted Kynurenine (Kyn) metabolites during the neurodevelopmental period in animal models.
Study
Model
SpeciesChanges of Kyn Metabolites
in Offspring
Behavioral ChangesEffect on BrainRef.
Poly(I:C) i.p.
at E9.0–9.5
MiceTrp (⇊), Kyn (⇊), QUIN (⇈), KMO (⇈), 5-HT (⇊), QUIN/Kyna ratio (⇈)
after PND80
Impaired working memory
Increased anxiety-like behavior
Impaired attentional set shifting after PND80
5-HT2A (⇈), SERT (⇈), mGlu2 (⇊)
Functional GABAergic transmission (⇊)
Decreased PV+ interneuron transmission after PND80
[236]
[237]
[238]
Poly(I:C) i.p.
at E14, 16, 18
RatKyn (↑)
5 h after injection
GluN2A (↓), GluN2B (⇈),
DCX (⇊)
5 h after injection
[239]
Poly(I:C) i.p.
at E9.0–9.5
MiceTrp (⇊), Kyn (⇊), QUIN (⇈), KMO (⇈), 5-HT (⇊), QUIN/Kyna ratio (⇈)
after PND80
Impaired working memory
Increased anxiety-like behavior
Impaired attentional set shifting after PND80
5-HT2A (⇈), SERT (⇈), mGlu2 (⇊)
Functional GABAergic transmission (⇊)
Decreased PV+ interneuron transmission after PND80
[240]
KMO inhibitor i.p. at E14, 16,18MiceKyn (⇈), Kyna (⇈)
5 and 24 h after injection
[241]
KMO inhibitor i.p. at E14, 16, 18RatKyn (⇈), Kyna (⇈)
5 and 24 h after injection
Does not affect the initial acquisition nor the subsequent memory consolidation
at PND60
GluN2A (⇈), GluN2B (⇈), PSD-95 (⇈)
Increased LTP at PND21
GluN2A (⇊), GluN2B (↓), PSD-95 (↓), VGLUT-1(⇈), VGLUT-2 (⇈),
Decreased LTP and basal spine densities in CA1 at PND60
[130]
[242]
[243]
IL-17a high expression at E12.5-PND0
Kyn i.p.
at E12.5–19
MiceKyn (⇈), Kyna (⇈)
Kyn/Trp ratio (⇈)
at E18.5
TRP (⇊), Kyn/Trp ratio (⇈)
at PND77
Impaired cognitive function
Lower social behavior and social cognition
Depressive-like behavior
at PND 56-70
[3]
Kyn i.p.
at E14,16,18
Rat GluN1 (↑), GluN2A (↑), GluN2B (↑)
Decreased LPT
[244]
Fed high Kyn diet at E15–22RatKyn (⇈), Kyna (⇈) at E22
Kyna (⇈) at PND56
Impaired spatial learning, reference memory, and contextual memory
Impaired attentional set-shifting
at PND 56-85
Reduced rapid eye
movement sleep at PND85
mGluR2 (⇊) at E21
mChrna7 (⇊) at PND2
Decreased dendritic spine density at PND56-80
mGrin1 (⇊), mGrin2a (⇊)
at PND70
Time- and sex-dependent alterations in the levels of GABA and glutamate at PND56
[5]
[23]
[245]
[246]
[247]
Fed high Kyn diet at E15–PND21RatKyn (⇈), Kyna (⇈), 3-HK (⇈) at PND21Impaired spatial learning, reference memory, and contextual memory
Impaired attentional set-shifting at PND56
Decreased extracellular glutamate[248]
[249]
Kyn i.p. at PND7–10RatKyna (⇈), QUIN (⇈)
at PND10
Kyna (↑), QUIN (↑)
at PND70
Impaired social behavior
Decreased locomotor activity
[250]
Kyn i.p. at PND7–16MiceKyna (⇈) at PND16Decreased amphetamine-induced locomotor activity
Impaired PPI, working memory
Enhanced amphetamine-induced dopamine release[251]
KMO gene- deficitMiceKyn (⇈), Kyna (⇈)Impaired contextual memory
Decreased social interaction
Increased anxiety- and depressive-like behavior
Increased amphetamine-induced locomotor activity
Increased spontaneous ventral tegmental area dopamine neuron activity
Decreased LPT
[138]
[139]
[244]
[252]
KAT II gene-deficitMiceKyna (⇊), QUIN(↓) at PND14 and 21
No significant changes
at PND60
Increased performance in object exploration and recognition tasks, passive avoidance test, spatial discrimination test, and spontaneous locomotor activity at PND21
Increased LPT
Increased extracellular glutamate concentrations and endogenous α7nAChR activity
[183]
[253]
[254]
BTBR T+Itpr3tf/J MiceKyna (⇈)
Display autism-relevant behaviorsAbsence of the corpus
Severely reduced hippocampal commissure
[165]
[255]
Mecp2+/−MiceNo significant changesExhibit hindlimb clasping
and uneven breathing
Model of human Rett Syndrome
Reduced in global and all local volumes in the brain[241]
[256]
Significantly decreased (⇊), significantly increased(⇈), decreased (↓), increased (↑) but not statically significant. SERT, serotonin transporter; PV+, parvalbumin positive; DCX, doublecortin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Murakami, Y.; Imamura, Y.; Kasahara, Y.; Yoshida, C.; Momono, Y.; Fang, K.; Sakai, D.; Konishi, Y.; Nishiyama, T. Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder. Cells 2023, 12, 1087. https://doi.org/10.3390/cells12071087

AMA Style

Murakami Y, Imamura Y, Kasahara Y, Yoshida C, Momono Y, Fang K, Sakai D, Konishi Y, Nishiyama T. Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder. Cells. 2023; 12(7):1087. https://doi.org/10.3390/cells12071087

Chicago/Turabian Style

Murakami, Yuki, Yukio Imamura, Yoshiyuki Kasahara, Chihiro Yoshida, Yuta Momono, Ke Fang, Daisuke Sakai, Yukuo Konishi, and Toshimasa Nishiyama. 2023. "Maternal Inflammation with Elevated Kynurenine Metabolites Is Related to the Risk of Abnormal Brain Development and Behavioral Changes in Autism Spectrum Disorder" Cells 12, no. 7: 1087. https://doi.org/10.3390/cells12071087

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