Next Article in Journal
Polyelectrolyte Microcapsules: An Extended Release System for the Antiarrhythmic Complex of Allapinin with Glycyrrhizic Acid Salt
Next Article in Special Issue
Personalized, Precision Medicine to Cure Alzheimer’s Dementia: Approach #1
Previous Article in Journal
H9N2 Avian Influenza Virus Downregulates FcRY Expression in Chicken Macrophage Cell Line HD11 by Activating the JNK MAPK Pathway
Previous Article in Special Issue
Factor H’s Control of Complement Activation Emerges as a Significant and Promising Therapeutic Target for Alzheimer’s Disease Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mast Cells in Autism Spectrum Disorder—The Enigma to Be Solved?

by
Eleonora Kovacheva
1,2,
Maria Gevezova
1,2,
Michael Maes
2,3,4,5,6,7,8 and
Victoria Sarafian
1,2,*
1
Department of Medical Biology, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
2
Research Institute, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
3
Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
4
Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
5
Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
6
Cognitive Fitness and Technology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
7
Department of Psychiatry, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
8
Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2651; https://doi.org/10.3390/ijms25052651
Submission received: 15 January 2024 / Revised: 19 February 2024 / Accepted: 20 February 2024 / Published: 24 February 2024
(This article belongs to the Special Issue Neuroinflammatory Mediator in Neurodegenerative Disease)

Abstract

:
Autism Spectrum Disorder (ASD) is a disturbance of neurodevelopment with a complicated pathogenesis and unidentified etiology. Many children with ASD have a history of “allergic symptoms”, often in the absence of mast cell (MC)-positive tests. Activation of MCs by various stimuli may release molecules related to inflammation and neurotoxicity, contributing to the development of ASD. The aim of the present paper is to enrich the current knowledge on the relationship between MCs and ASD by discussing key molecules and immune pathways associated with MCs in the pathogenesis of autism. Cytokines, essential marker molecules for MC degranulation and therapeutic targets, are also highlighted. Understanding the relationship between ASD and the activation of MCs, as well as the involved molecules and interactions, are the main points contributing to solving the enigma. Key molecules, associated with MCs, may provide new insights to the discovery of drug targets for modeling inflammation in ASD.

1. Introduction

MCs are hemopoietic-derived tissue immune cells which are associated with allergies, but also participate in immunity [1] and inflammation [2]. They can produce mediators, both pro- and anti-inflammatory [3], and may have functions related to immune modulation [1,4]. In humans, two common MC phenotypes are recognized according to their protease content: MCs that contain only tryptase (MCTs) and MCs that contain both tryptase and chymase (MCTCs) [5]. MCTC predominate in the skin and connective tissue and are also present in significant numbers in the submucosal tissues of the respiratory tract. MCTs are more abundant in mucosal epithelium and in the lamina propria. Their roles are rather unclear, but their ability to release different proteases and cytokines suggests some mutually exclusive functions [5]. MCs function as protectors by responding to a wide range of “signals” (e.g., environmental antigens, allergens, invading pathogens or toxins) in a rapid and selective way [6]. For this purpose, they are equipped with a number of receptors, which induce the release of many biologically active products and may even lead to in situ proliferation of terminally differentiated MCs [4]. Furthermore, the activation of MCs by IgE-dependent triggers or other agonists is also related to significant alterations in the immunophenotypic profile of these cells. Some of the changes reflect MC degranulation, including the surface membrane expression of LAMP-1 (CD107a), LAMP-2 (CD107b) and LAMP-3 (CD63) [7].
ASD is a neurodevelopmental disease characterized by deficits in verbal and nonverbal communications, disturbance in social interactions and limited, repetitive patterns of behavior, interests and activities [8]. According to the Centers for Disease Control and Prevention, Autism and Developmental Disabilities Monitoring Network, approximately 1 in 36 children is identified with ASD (https://www.cdc.gov/ncbddd/autism/data.html accessed on 15 December 2023). Although some putative autism susceptibility genes have been identified [9] and gene interactions with environmental factors have been suspected, in the majority of cases, the cause of ASD remains unknown [10]. Some children with ASD regress at about age 3, often after a reaction to vaccination, infection [11], trauma [12], toxic exposures [13] or stress [14], implying the importance of environmental triggers [15].
Many children with ASD have a family or a personal history of “allergic symptoms”, often in the absence of positive skin or RAST (radioallergosorbent) tests. Furthermore, children with mastocytosis or MC activation syndrome (MCAS) develop ASD at a much higher rate than children of the general population. MC activation by allergic, infectious, environmental and stress-related stimuli, especially perinatally, would release molecules related to inflammation and neurotoxicity [16]. It has been suggested that mediators derived from MCs can alter the blood–brain barrier (BBB) and cause “brain allergy” [10] or “focal encephalitis” [17], thereby contributing to the pathogenesis of ASD [17,18]. Increased MC responsiveness may define at least one subset of patients with ASD who could benefit from inhibition of MC activation [16].
The aim of the present review is to summarize the current knowledge on the relationship between the activation of MCs, MC-derived molecules and mediators in the pathogenesis of ASD. Key molecules associated with MCs may provide new insights to the discovery of drug targets for modeling the inflammatory processes observed in ASD.

2. Functions of MCs

MCs have immunomodulatory and physiological functions in the epithelium, endothelium, and nervous system. These cells act as guardians of the immune system and participate in many biological processes, as well as in the maintenance of homeostasis [19]. MCs are considered crucial for tissue function and integrity [20]. A number of MC mediators, including nerve growth factor (NGF), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and fibroblast growth factor-2 (FGF-2), and also histamine and tryptase, induce epithelial cell and fibroblast proliferation [21]. Furthermore, MCs are involved in all steps of tissue repair, from the initial inflammatory reaction to extracellular matrix (ECM) remodeling [22]. Through the release of platelet-activating factor (PAF), leukotrienes, and the cytokines IL-1 and IL-8, MCs contribute to platelet activation and aggregation as well as to the extravascular deposition of fibrin [23,24]. Proteases, such as tryptase, signal nerves through protease-activated receptors (PARs) [25,26,27,28]. MCs can also be activated by substance P and endothelin-1 (ET-1) [29,30]. MC–neuron interactions also contribute to the maintenance of intestinal homeostasis by regulating ion transport, vascular permeability, the secretory activity of mucus-producing cells, and gastrointestinal motility [31]. Moreover, the role of MCs in angiogenesis is most certainly related to the release of a large spectrum of angiogenic mediators, which include angiopoietin-1, FGF-2, VEGF, IL-8, TGF-β, TNF-α, histamine, heparin, tryptase and chymase, among others [32]. Similar to dendritic cells, MCs are among the first cells of the immune system to interact with antigens, toxins, and pathogens. In addition, MCs express various surface receptors which are able to detect potentially harmful signals and enable a rapid and appropriate response through the release of pre-stored and newly synthesized mediators. MCs recognize pathogens through the direct binding of pathogens or their components to pathogen-associated molecular pattern (PAMP) receptors on an MC’s surface, binding to complement or immunoglobulin receptors, or the recognition of endogenous peptides produced by infected or injured cells [33]. In vitro studies have shown that MCs are also capable of processing and presenting antigens via MHC-I and MHC-II complexes [34,35,36]. MCs are recognized as the main effector cells responsible for IgE-mediated allergic reactions [37,38].

3. MCs and Inflammation

The activation of mammalian MCs is triggered by a variety of signals generated during innate responses. Such signals include elements of the complement system [39,40,41], agonists of Toll-like receptors (TLRs) [41,42,43], adenosine [44], corticotropin-releasing factor receptors [45], numerous endogenous peptides, including vasoactive intestinal peptide (VIP) [46,47], and substance P [46,48] and a large number of other stimuli [41]. Contrary to stimulating signals, the reported suppressing MC activation signals are quite limited.
Increasing evidence indicates that MCs are critical for the pathogenesis of inflammatory diseases [49,50], such as arthritis [51], atopic dermatitis, psoriasis [52,53], and multiple sclerosis [54]. Gene array analysis of human MCs activated by IgE showed the overexpression of numerous, mostly inflammation-related genes [55]. Moreover, proteases released by MCs could act on plasma albumin to generate histamine-releasing peptides [56,57] that would further propagate MC activation and inflammation. Proteases could also stimulate PARs, inducing microleakage and widespread inflammation [58,59]. However, unlike allergic reactions, MCs are rarely seen to degranulate during inflammatory processes. The only way to explain MC involvement in non-allergic conditions would be through “differential” or “selective” secretion of mediators without degranulation [60].

4. MCs, Neuroinflammation and ASD

The localization of MCs and, more importantly, the mediators released by both MCs and neurons collaborate in the establishment of a neuroimmune interaction between these cells. It has been shown that communication between MCs and neurons can occur through synaptic-like structures sustained by adhesion molecules such as N-cadherin or the synaptic cell adhesion molecule SynCAM [61,62].
Microglia are specialized macrophages within the central nervous system (CNS) and they have an important role in neuroinflammation [63,64] and neurodegeneration [63,65]. Microglial activation is reported in ASD [66,67] and the release of pro-inflammatory mediators IL-1β and CXCL8 [68] (Figure 1). The transition of microglia from a resting state to an activated pro-inflammatory phase in ASD is regulated by different factors. Microglia can be activated by PAMPs and endogenous damage-associated molecular patterns (DAMPs), acting on TLRs, but also in response to histamine and tryptase released by MCs [69]. Triggered brain MCs have been shown to participate in cognitive dysfunction through microglial activation and neuronal apoptosis [70].
Macrophages are polarized due to changes in their environment, which result in different subtypes, such as M1 and M2 [71]. Lipopolysaccharides can drive macrophage polarization to the M1 phenotype, while IL-4 induces macrophage polarization to M2 [72]. M1 macrophages are related to pro-inflammatory responses and produce pro-inflammatory factors, whereas M2 macrophages are capable of anti-inflammatory responses and repair damaged tissues [73]. Based on the triggers and transcriptional changes, M2 macrophages are divided into four subgroups: alternative activated macrophages (M2a), type 2 macrophages (M2b), deactivated macrophages (M2c) and M2d [73]. M2a macrophages are involved in the clearance of apoptotic cells, modulating the inflammatory response and wound healing [74]. Both IL-4 and IL-13 are capable of inducing M2a polarization [75,76].
While IL-4 activates the IL-4Rα receptor, IL-13 triggers the IL-13Rα1 receptor. The two receptors can form heterodimers that respond to both IL-4 and IL-13 [75]. IL-5 activates the IL-5Rα receptor, which induces eosinophilia [75]. In case of exposure of M2A to IL-4 or IL-13, a loss of the specific M1 marker, inducible nitric oxide synthase (iNOS), follows [76]. Typical markers for M2A polarized macrophages and microglia are the arginase enzyme Arg1, scavenger receptors and the mannose receptor [76,77]. In addition, the transcription of growth factors such as VEGF, BDNF and PDGF is increased in IL-4-induced M2A microglia [76]. In the M2A-polarized state, microglial cells also express insulin-like growth factor-1 (IGF1) [77]. As a consequence, allergy may indirectly lead to the release of many potent growth factors from microglia. Upon IL-4 or IL-13 stimulation, the expression of IGF1 by microglia [77,78] and macrophages [78] is greatly increased. IGF1 is an important cytoprotective protein with a relevant function in tissue repair after inflammation [79]. Compared to controls, blood levels of IGF1 were significantly elevated in men with ASD who had a large head circumference. Moreover, head size was also found to correlate with IGF1 blood levels [80].
MCs interact with microglia in the brain [81] and their activation [82,83], especially in the hypothalamus [84], can lead to cognitive dysfunction [85]. Microglia express receptors for CRH [86] and can be further activated by stress [87]. Microglia are also rich in receptors for NT [88], which have been reported to be increased in the serum of patients with ASD [89] and can activate microglia to secrete pro-inflammatory molecules [68]. Microglia also express neurotensin receptor 3 (NTS3), the activation of which leads to microglial proliferation [90].
Increased gene expression of the pro-inflammatory microRNA-155 (miR-155) has been also found in the amygdala of children with ASD [91], as well as decreased expression of the anti-inflammatory cytokine IL-38 [92]. A transcriptome study of 104 human brain cortical tissue samples showed a relationship between the gene expression module of M2 microglia activation and a neuronal module, suggesting dysregulated microglial responses that may lead to alterations in neuronal activity in ASD [66].

5. MC Cytokines in ASD

The increased levels of proinflammatory cytokines [93,94,95,96,97,98,99] and the decrease in the anti-inflammatory cytokine IL-10 are some of the immune hallmarks of ASD [94,100,101,102,103]. This may be important as the balance of pro- and anti-inflammatory cytokines modulates brain development. Abnormal levels of these molecules are associated with a number of complex disorders, including ASD [101]. Generally, increased levels have been recorded for IL-5, IL-8, IL-13, IL-17 [102,104,105,106,107,108], IL-12 [109], IL-21, IL-22 [107], IFNγ [98,110], TNF-α [111], TNF-receptor II17, IL-1β, and IL-6. IL-1β is associated with impairments in memory and learning [112,113], and IL-6 is linked with stereotyped behavior, and impacts synapse formation [114,115,116]. These cytokines can modulate brain function by affecting cognitive and emotional processing, mood and sleep disorders in ASD [117].
TNF-α levels are positively related to the severity of ASD symptoms [118,119]. IL-1β has been found to increase the production of IL-17, which is a mediator of IL-8 [120]. Also, higher levels of IL-8 have been associated with more deviant behavior in patients with ASD, including hyperactivity, low language and cognitive ability [102]. Once activated, IL-8 attracts neutrophils to the areas of inflammation, leading to the release of proteolytic substances [121]. Cytokines secreted by immune cells can cross the blood–brain barrier and participate in the activation of microglia. Two recent studies demonstrate altered cytokine profiles in newborn infants, suggesting early immune dysregulation. Elevated IL-4 is associated with higher severity of ASD, while excessive IL-1β is linked to a milder form of the disease [122]. Increased levels of IFNγ, IL-4 and IL-5 in maternal serum are significantly associated with an increased ASD risk in the newborn [123].
MCs also secrete transforming growth factor-β (TGF-β), which promotes the development of Th17 cells and IL-17 production [124]. Elevated IL-17 has been reported in the serum and in immune cells of children with ASD [125]. MCs also release newly synthesized lipid-derived mediators such as prostaglandin D2 [126], cytokines (IL-5, IL-6, IL31, IL-33, and TNF) and chemokines (CCL5 and CXCL8) [127]. Furthermore, MCs can be influenced by cytokines, such as IL-1b. It induces the selective release of IL-6 [128] and IL-33 [129], which are enhancers of allergic MC stimulation [129,130]. IL-33 has also been shown to significantly increase the ability of substance P to stimulate MCs’ release of VEGF [131], TNF [132] and IL-1b [133].
A number of studies report the overproduction of proinflammatory cytokines in ASD children [100,118,134,135,136,137]. Significantly increased plasma levels of several cytokines, such as IL-1β, IL-6, IL-8, and IL-12p40, were found in ASD children compared to controls [118]. Another meta-analysis revealed significantly higher concentrations of IL-1β, IL-6, IL-8, interferon-gamma (IFN-γ), eotaxin, and monocyte chemoattractant protein-1 (MCP-1) in ASD individuals compared to controls [138]. Elevated proinflammatory cytokines were also found within the CNS (specifically in brain tissues and cerebrospinal fluid) of ASD patients [98]. On the other hand, the immune system also comprises regulatory elements that counter-regulate proinflammatory effects to maintain homeostasis, including anti-inflammatory cytokines, such as the IL-1 receptor antagonist (IL-1RA), IL-4, IL-10, and TGF-β [105,139,140]. However, studies regarding anti-inflammatory cytokines in ASD, as well as attention deficit hyperactivity disorder (ADHD), reflect inconsistent findings. While one meta-analysis reported significantly decreased levels of TGF-β1 and borderline increased levels of IL-1RA [138], a more recent meta-analysis revealed decreased levels of IL-10 and IL-1RA, and slightly increased levels of IL-5 in ASD patients [141].

6. The Role of MCs in ASD

Various perinatal allergic, genetic, environmental, immune and infectious factors may increase the risk of developing ASD [142,143]. They can activate MCs containing and releasing mediators, such as preformed kinins and proteases, and de novo synthesized leukotrienes, prostaglandins, chemokines (CCXL8, CCL2), cytokines (IL-4, IL-6, IL-1, TNF) and VEGF [144], as described in Figure 1. In addition, MCs function as an “immune gate to the brain” [145].
A study of 400 patients with mastocytosis revealed 15 cases with both mastocytosis and ASD—an incidence of 3.75 per 100, or 6.75 times higher than reported for the general population (1 in 180). In other families, mothers with mastocytosis have at least one child with ASD. As both ASD and mastocytosis are rare diseases, this association seems to be impressive [146]. Cerebro-spinal-fluid (CSF) levels of TNF-α were found to be significantly higher than the corresponding serum levels in ten children with autism [111], which may affect cognitive functions [147].
MCs are also stimulated by bacteria, viruses, fungi, drugs, foods, heavy metals, organophosphates and some neuropeptides, including CRH [148], neurotensin (NT) [149] and substance P [150]. Both NT [151] and substance P [152] are known to be involved in inflammatory processes. MC-derived mediators [2,127,144] can activate microglia [68,153] and cause localized inflammation [18,154,155], leading to ASD symptoms. NT is found to be elevated in young children with autism [156] and has been proposed as a possible therapeutic target due to its ability to induce neurotoxicity [157]. NT and CRH, secreted under stress, synergistically stimulate MCs and lead to increased vascular permeability [149] and blood-brain barrier (BBB) breakdown [158]. In addition, NT has been reported to stimulate MCs’ secretion of VEGF [159]. NT also increases the expression of CRH receptor 1 (CRHR-1) [160], whose activation by CRH enhances allergic stimulation of human MCs [161]. Furthermore, NT is elevated in the skin after acute stress, stimulates cutaneous MCs and increases vascular permeability in rodents [162]. NT triggers rodent peritoneal MCs to secrete histamine, which results in high plasma histamine levels by activating a specific NT receptor (NTR) [163]. Substance P induces the ST2 receptor for IL-33 [132], which further enhances MC activation through the combined action of neuropeptides and IL-33. Other neuropeptides, such as VIP and calcitonin gene-related peptide (CGRP) have been reported to be higher in the serum of children with ASD and in those with intellectual disability without ASD. In contrast, substance P and NGF levels were similar to controls [164]. Nevertheless, the same authors later found no differences in any of these peptides between autistic and control groups [165].
MCs are located perivascularly in all tissues, including the brain, where they are adjacent to CRH-positive nerve endings [166]. They are also plentiful in the meninges [167]. Studies have found that the stimulation of MCs leads to activation of microglia, which are actually involved in ASD [66] (Figure 1). In addition, MCs are also implicated in the regulation of the hypothalamus–pituitary–adrenal (HPA) axis in the skin [168] and brain [169], as histamine, IL-6 [170] and CRH [171] can activate this axis. MCs are usually stimulated by exposure to allergens and binding of IgE to high-affinity receptors (FcERIs), whose aggregation leads to degranulation and the secretion of multiple pre-stored and newly synthesized mediators [155,172]. In addition to IgE, many substances from the environment, gut or brain can induce MC activation [173]. MCs can serve as sensors of environmental and psychological stress [174], releasing danger signals [175] such as mitochondrial DNA (mtDNA) [160], which acts as an “innate pathogen” [176], causing autoinflammatory responses [177]. It is found to be increased also in the serum of children with ASD [178]. MCs can secrete exosomes that deliver miRNAs [179] and may be involved in brain pathology [180].
MCs may be implicated in ASD because of their unique ability to respond to non-IgE stimuli and release mediators “differentially” or “selectively” without the degranulation typical of allergic or anaphylactic reactions [60]. Bacteria and viruses have the capacity to trigger MCs via TLRs, which are important in the development of innate immunity. Lipopolysaccharides induce the selective release of TNF-α without degranulation via TLR-4, while peptidoglycans cause histamine release via TLR-2 from rodent MCs [181]. Human MCs express TLR-1, -3, -5, -7 and -9 [182].
The activation of TLR-9 selectively induces the production of the pro-inflammatory cytokine IL-6, while the activation of TLR-3 generates an IFN-α in response to double-stranded RNA [182]. These findings are important as neuroenteric viruses are likely to affect children at the age of 3–5 years [183] and contribute to immune abnormalities in ASD.
Environmental toxins such as mercury [184] and polychlorinated biphenyl (PCB) [185] have been related to developmental neurotoxicity [186] and ASD. Both mercury and PCBs can stimulate MCs. Furthermore, MCs can also be activated by aluminum [187]. Both mercury [188] and aluminum [189,190] have been associated with the severity of the symptoms in children with ASD [187]. In addition, herbicides such as glyphosate and atrazine have been reported to stimulate MCs and promote inflammation and neurotoxic effects [191] (Figure 1).
The risk of developing ASD may be further increased if children have mutations leading to decreased phosphatase and tensin homolog (PTEN). It is an inhibitor of the mammalian target of rapamycin, resulting in proliferation of microglia and MCs. Activation of susceptibility genes is often used to explain ASD. Stimulation of mTOR in patients with overactive mTOR as a result of gene mutations which lower PTEN would contribute to a form of “epigenetic” signal [192].
Several studies have found that atopic diseases [156,193] are associated with an increased risk of ASD [194], which is not surprising, as inflammatory processes, and interactions between stress and the immune system have been related to the pathogenesis of ASD [69,156]. A study of 1638 pregnant women stated that high levels of stress during gestation, and especially during the second trimester, are associated with high risk of ASD developing in the newborn at 6 months of age [195]. Psychological stress during pregnancy can cause immune system dysregulation and increased IgE levels in the umbilical cord blood [196]. This in turn may enhance the development of allergic reactions or sensitization to postnatal allergens in the fetus. In fact, children with ASD cannot handle stress [197] and have an exacerbated sense of fear [69], which can only worsen their condition. Elevated serum concentrations of IL-4 and IL-5 in women during the 15th and 19th weeks of gestation have been detected. These women gave birth to children who were later diagnosed with ASD [123]. Also, increased levels of IL-4, IL-10 and TNF-α have been measured in the amniotic fluid of fetuses that later developed ASD [198]. Another study noted that while IL-1β was associated with increased ratios of mild or moderate degree of ASD, high levels of IL-4 were linked with severe ASD. These findings show that peripheral cytokine profiles at birth are not only associated with ASD later in childhood, but also differ in terms of symptom severity [122]. Mothers who suffer from asthma, allergy, atopy or eczema during gestation are more prone to have children with a higher risk of neuropsychiatric problems [199]. Studies have reported strong a connection between ASD and food allergy [200] or food intolerance [201], which can lead to brain inflammation and cognitive impairment [202]. Msallam et al. [203] showed that circulating maternal immune IgE led to the vertical transmission of atopic dermatitis in the newborn by stimulating fetal MCs. In addition, both passive and active prenatal sensitization confer allergen sensitivity [203]. This indicates that fetal MCs are functional and can be stimulated by specific IgE and allergens coming from the mother during pregnancy. Although these studies are limited to lung and skin MCs, reactivity may also extend to brain MCs [204].

7. MCs and Therapeutic Targets

MCs, their triggers and their mediators could serve as unique therapeutic targets as they are influenced by the corticotropin-releasing factor (CRF) and appear to regulate the permeability of the gut–blood–brain barrier [158,169].

7.1. Serotonin Receptor Antagonists

In a study of six children with autism, four had high urinary serotonin levels [205]. Elevated platelet serotonin has been reported in more than 40% of autistic patients [206]. In a double-blind study of 40 ASD children randomized to the antipsychotic haloperidol and cyproheptadine versus haloperidol and placebo, the combined histamine H1 and serotonin receptor antagonist cyproheptadine was associated with significant clinical improvement [207].

7.2. MC Activation Blockers

Recent evidence suggests that MC activation is regulated by several costimulatory molecules [208]. Additional proof shows that MCs can be blocked through their inhibitory receptor FcgRIIb [209]. This inhibition, however, is relevant only for the allergic stimulation of MCs. Several MC mediators can inhibit their secretion too. Chondrotin sulfate, which is abundant in MC secretory granules, restrains mucosal MC activation [210]. The compound 48/80 induces histamine release, unlike disodium cromoglycate (cromolyn), which shows rapid tachyphylaxis [211].

7.3. Cromolyn

Cromolyn is a potent inhibitor of histamine secretion by rodent MCs, but a weak inhibitor of the allergic activation of human MCs [212]. Although it is poorly absorbed orally, it appears to reduce symptoms of mastocytosis, including neurobehavioral problems, indicating that gastrointestinal MCs affect the brain. The structure of cromolyn is similar to that of some flavonoids, polyphenolic compounds present in fruits, vegetables, nuts, seeds, and red wine with potent antioxidant, anti-inflammatory, and MC-inhibiting activities [213]. However, cromolyn does not effectively inhibit neither murine [214] nor human MCs [215,216,217]. What is more, it has been reported to potentiate histamine release from MCs [218].

7.4. Quercetin

Quercetin and other flavonoids inhibit the release of histamine, IL-6, IL-8, TNF-α, and tryptase from normal human MCs [219], making them possible candidates for the treatment of ASD. Given that MCs are activated by CRF, CRF receptor antagonists have been developed for several disorders [220] such as anxiety, neuroinflammation and irritable bowel syndrome [221], and may also be useful in ASD.

7.5. Rapamycin and Luteolin

Rapamycin and its analogues are mTOR inhibitors [222] which have been tested for the treatment of ASD [223,224,225,226]. Preliminary results show that the natural flavonoid luteolin is more potent than rapamycin in its ability to inhibit TNF release from human MCs [213]. A previous report also proved that the flavonoid-related epigallocatechin gallate (EGCG) is an mTOR inhibitor [227]. Luteolin may not only suppress mTOR, but also has additional beneficial effects on brain inflammation. It inhibits oxidative stress [213], inflammation [213], MC degranulation [228], MC cytokine release [161], thimerosal-induced inflammatory mediator release [229], microglial activation and proliferation [230,231,232], and autoimmune T-cell activation [233,234]. Luteolin also acts against methylmercury-induced mitochondrial damage [235], has a neuroprotective effect [236], and mimics BDNF [237,238]. Luteolin and quercetin also inhibit the release of histamine, IL-6, IL-8, TNF, and tryptase from human MCs [219,228]. Recently, tetramethoxyluteolin was found to be a more potent inhibitor of human MCs than luteolin [217]. In addition, luteolin has broad antiviral properties [239,240,241], and inhibits COVID-19 entry into host cells [242,243,244]. Furthermore, luteolin has better brain penetration and suppresses both microglia [68,245,246,247] and MCs [150,217]. It has a neuroprotective effect [248,249,250,251], and reduces neuroinflammation [249,252,253,254] and cognitive dysfunction [155,255,256,257], especially brain fog [258,259,260].

7.6. Extracellular Mitochondrial Components

It is revealed that the activation of MCs is accompanied by mitochondrial fission and translocation to the cell surface from where they secrete at least ATP and DNA outside the cell without any cell damage [160]. These extracellular mitochondrial components are misconstrued by the body as “innate pathogens” leading to powerful autocrine and paracrine auto-immune/auto-inflammatory responses. These mitochondrial constituents could be the “missing link” in certain auto-immune and auto-inflammatory diseases, especially in ASD [176]. Therefore, the clinical manifestation could be worse in the subgroup of ASD patients with mitochondrial dysfunction [261]. Preventing the secretion of extracellular mitochondrial components may be used as a novel therapeutic approach [176].

8. Conclusions and Perspectives

Data on the association of ASD with MCs are controversial and poorly understood. The involvement of MCs in innate and acquired immunity, gastrointestinal pathology, inflammation and increased intestinal permeability may explain the observed comorbidities in ASD. Patients with mutations in ASD susceptibility genes and hypersensitive MCs may represent a unique subgroup that are more likely to respond to environmental influences and stress. Activation of brain MCs by allergic, environmental, immune, neurohormonal, stress and toxic stimuli, particularly in domains related to behavior and language, can lead to focal brain allergies and focal encephalitis. Stress can worsen the clinical condition of ASD patients by stimulating MCs, leading to increased vascular permeability and neuroinflammation. Cytokines and key molecules, markers of MC degranulation, are shown to be implicated in ASD’s development.
Understanding the relationship between ASD, the activation and regulation of MCs, as well as the participating molecules and the interactions between them are the major points contributing to solving the enigma. They are important for the clarification of ASD pathogenesis and for the implementation of effective future treatments for ASD patients by providing novel therapeutic target molecules.

Author Contributions

Conceptualization, V.S. and M.M.; writing—original draft preparation, E.K. and M.G.; writing—review and editing, V.S.; supervision, V.S.; funding acquisition, V.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0007-C01 and by National Science Fund of Bulgaria, Grant No. KΠ-06-H33/6-2019.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kalesnikoff, J.; Galli, S.J. New developments in mast cell biology. Nat. Immunol. 2008, 9, 1215–1223. [Google Scholar] [CrossRef]
  2. Theoharides, T.C.; Alysandratos, K.D.; Angelidou, A.; Delivanis, D.A.; Sismanopoulos, N.; Zhang, B.; Asadi, S.; Vasiadi, M.; Weng, Z.; Miniati, A.; et al. Mast cells and inflammation. Biochim. Biophys. Acta 2012, 1822, 21–33. [Google Scholar] [CrossRef] [PubMed]
  3. Kobayashi, H.; Ishizuka, T.; Okayama, Y. Human mast cells and basophils as sources of cytokines. Clin. Exp. Allergy 2000, 30, 1205–1212. [Google Scholar] [CrossRef]
  4. Galli, S.J.; Grimbaldeston, M.; Tsai, M. Immunomodulatory mast cells: Negative, as well as positive, regulators of immunity. Nat. Rev. Immunol. 2008, 8, 478–486. [Google Scholar] [CrossRef]
  5. Irani, A.A.; Schechter, N.M.; Craig, S.S.; DeBlois, G.; Schwartz, L.B. Two types of human mast cells that have distinct neutral protease compositions. Proc. Natl. Acad. Sci. USA 1986, 83, 4464–4468. [Google Scholar] [CrossRef]
  6. Rao, K.N.; Brown, M.A. Mast cells: Multifaceted immune cells with diverse roles in health and disease. Ann. N. Y. Acad. Sci. 2008, 1143, 83–104. [Google Scholar] [CrossRef]
  7. Valent, P.; Schernthaner, G.H.; Sperr, W.R.; Fritsch, G.; Agis, H.; Willheim, M.; Buhring, H.J.; Orfao, A.; Escribano, L. Variable expression of activation-linked surface antigens on human mast cells in health and disease. Immunol. Rev. 2001, 179, 74–81. [Google Scholar] [CrossRef] [PubMed]
  8. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association: Arlington, VA, USA, 2013. [Google Scholar]
  9. Weiss, L.A.; Arking, D.E.; Gene Discovery Project of Johns Hopkins; the Autism Consortium Writing group; the Autism, C.; Daly, M.J.; Chakravarti, A. A genome-wide linkage and association scan reveals novel loci for autism. Nature 2009, 461, 802–808. [Google Scholar] [CrossRef] [PubMed]
  10. Levy, S.E.; Mandell, D.S.; Schultz, R.T. Autism. Lancet 2009, 374, 1627–1638. [Google Scholar] [CrossRef]
  11. Hsiao, E.Y.; McBride, S.W.; Chow, J.; Mazmanian, S.K.; Patterson, P.H. Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl. Acad. Sci. USA 2012, 109, 12776–12781. [Google Scholar] [CrossRef]
  12. Blenner, S.; Reddy, A.; Augustyn, M. Diagnosis and management of autism in childhood. BMJ 2011, 343, d6238. [Google Scholar] [CrossRef] [PubMed]
  13. Deth, R.; Muratore, C.; Benzecry, J.; Power-Charnitsky, V.A.; Waly, M. How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis. Neurotoxicology 2008, 29, 190–201. [Google Scholar] [CrossRef] [PubMed]
  14. Lanni, K.E.; Schupp, C.W.; Simon, D.; Corbett, B.A. Verbal ability, social stress, and anxiety in children with autistic disorder. Autism 2012, 16, 123–138. [Google Scholar] [CrossRef]
  15. Goines, P.E.; Ashwood, P. Cytokine dysregulation in autism spectrum disorders (ASD): Possible role of the environment. Neurotoxicol. Teratol. 2013, 36, 67–81. [Google Scholar] [CrossRef]
  16. Angelidou, A.; Alysandratos, K.D.; Asadi, S.; Zhang, B.; Francis, K.; Vasiadi, M.; Kalogeromitros, D.; Theoharides, T.C. Brief report: “allergic symptoms” in children with Autism Spectrum Disorders. More than meets the eye? J. Autism Dev. Disord. 2011, 41, 1579–1585. [Google Scholar] [CrossRef] [PubMed]
  17. Theoharides, T.C.; Asadi, S.; Patel, A.B. Focal brain inflammation and autism. J. Neuroinflamm. 2013, 10, 46. [Google Scholar] [CrossRef]
  18. Theoharides, T.C.; Stewart, J.M.; Panagiotidou, S.; Melamed, I. Mast cells, brain inflammation and autism. Eur. J. Pharmacol. 2016, 778, 96–102. [Google Scholar] [CrossRef]
  19. Weller, C.L.; Collington, S.J.; Williams, T.; Lamb, J.R. Mast cells in health and disease. Clin. Sci. 2011, 120, 473–484. [Google Scholar] [CrossRef]
  20. Maurer, M.; Theoharides, T.; Granstein, R.D.; Bischoff, S.C.; Bienenstock, J.; Henz, B.; Kovanen, P.; Piliponsky, A.M.; Kambe, N.; Vliagoftis, H.; et al. What is the physiological function of mast cells? Exp. Dermatol. 2003, 12, 886–910. [Google Scholar] [CrossRef]
  21. Abe, M.; Kurosawa, M.; Ishikawa, O.; Miyachi, Y. Effect of mast cell-derived mediators and mast cell-related neutral proteases on human dermal fibroblast proliferation and type I collagen production. J. Allergy Clin. Immunol. 2000, 106 Pt 2, S78–S84. [Google Scholar] [CrossRef]
  22. Noli, C.; Miolo, A. The mast cell in wound healing. Vet. Dermatol. 2001, 12, 303–313. [Google Scholar] [CrossRef]
  23. Mekori, Y.A.; Galli, S.J. [125I]fibrin deposition occurs at both early and late intervals of IgE-dependent or contact sensitivity reactions elicited in mouse skin. Mast cell-dependent augmentation of fibrin deposition at early intervals in combined IgE-dependent and contact sensitivity reactions. J. Immunol. 1990, 145, 3719–3727. [Google Scholar]
  24. Kauhanen, P.; Kovanen, P.T.; Reunala, T.; Lassila, R. Effects of skin mast cells on bleeding time and coagulation activation at the site of platelet plug formation. Thromb. Haemost. 1998, 79, 843–847. [Google Scholar] [CrossRef]
  25. Dery, O.; Corvera, C.U.; Steinhoff, M.; Bunnett, N.W. Proteinase-activated receptors: Novel mechanisms of signaling by serine proteases. Am. J. Physiol. 1998, 274, C1429–C1452. [Google Scholar] [CrossRef]
  26. Vergnolle, N.; Wallace, J.L.; Bunnett, N.W.; Hollenberg, M.D. Protease-activated receptors in inflammation, neuronal signaling and pain. Trends Pharmacol. Sci. 2001, 22, 146–152. [Google Scholar] [CrossRef] [PubMed]
  27. Coelho, A.M.; Vergnolle, N.; Guiard, B.; Fioramonti, J.; Bueno, L. Proteinases and proteinase-activated receptor 2: A possible role to promote visceral hyperalgesia in rats. Gastroenterology 2002, 122, 1035–1047. [Google Scholar] [CrossRef] [PubMed]
  28. Cenac, N.; Coelho, A.M.; Nguyen, C.; Compton, S.; Andrade-Gordon, P.; MacNaughton, W.K.; Wallace, J.L.; Hollenberg, M.D.; Bunnett, N.W.; Garcia-Villar, R.; et al. Induction of intestinal inflammation in mouse by activation of proteinase-activated receptor-2. Am. J. Pathol. 2002, 161, 1903–1915. [Google Scholar] [CrossRef] [PubMed]
  29. Ogawa, K.; Nabe, T.; Yamamura, H.; Kohno, S. Nanomolar concentrations of neuropeptides induce histamine release from peritoneal mast cells of a substrain of Wistar rats. Eur. J. Pharmacol. 1999, 374, 285–291. [Google Scholar] [CrossRef] [PubMed]
  30. Suzuki, R.; Furuno, T.; McKay, D.M.; Wolvers, D.; Teshima, R.; Nakanishi, M.; Bienenstock, J. Direct neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J. Immunol. 1999, 163, 2410–2415. [Google Scholar] [CrossRef] [PubMed]
  31. Van Nassauw, L.; Adriaensen, D.; Timmermans, J.P. The bidirectional communication between neurons and mast cells within the gastrointestinal tract. Auton. Neurosci. 2007, 133, 91–103. [Google Scholar] [CrossRef] [PubMed]
  32. Crivellato, E.; Beltrami, C.A.; Mallardi, F.; Ribatti, D. The mast cell: An active participant or an innocent bystander? Histol. Histopathol. 2004, 19, 259–270. [Google Scholar] [CrossRef] [PubMed]
  33. Hofmann, A.M.; Abraham, S.N. New roles for mast cells in modulating allergic reactions and immunity against pathogens. Curr. Opin. Immunol. 2009, 21, 679–686. [Google Scholar] [CrossRef] [PubMed]
  34. Malaviya, R.; Twesten, N.J.; Ross, E.A.; Abraham, S.N.; Pfeifer, J.D. Mast cells process bacterial Ags through a phagocytic route for class I MHC presentation to T cells. J. Immunol. 1996, 156, 1490–1496. [Google Scholar] [CrossRef] [PubMed]
  35. Poncet, P.; Arock, M.; David, B. MHC class II-dependent activation of CD4+ T cell hybridomas by human mast cells through superantigen presentation. J. Leukoc. Biol. 1999, 66, 105–112. [Google Scholar] [CrossRef] [PubMed]
  36. Stelekati, E.; Bahri, R.; D’Orlando, O.; Orinska, Z.; Mittrucker, H.W.; Langenhaun, R.; Glatzel, M.; Bollinger, A.; Paus, R.; Bulfone-Paus, S. Mast cell-mediated antigen presentation regulates CD8+ T cell effector functions. Immunity 2009, 31, 665–676. [Google Scholar] [CrossRef] [PubMed]
  37. Eisenbarth, S.C.; Piggott, D.A.; Huleatt, J.W.; Visintin, I.; Herrick, C.A.; Bottomly, K. Lipopolysaccharide-enhanced, toll-like receptor 4-dependent T helper cell type 2 responses to inhaled antigen. J. Exp. Med. 2002, 196, 1645–1651. [Google Scholar] [CrossRef] [PubMed]
  38. Nigo, Y.I.; Yamashita, M.; Hirahara, K.; Shinnakasu, R.; Inami, M.; Kimura, M.; Hasegawa, A.; Kohno, Y.; Nakayama, T. Regulation of allergic airway inflammation through Toll-like receptor 4-mediated modification of mast cell function. Proc. Natl. Acad. Sci. USA 2006, 103, 2286–2291. [Google Scholar] [CrossRef]
  39. Ali, H. Regulation of human mast cell and basophil function by anaphylatoxins C3a and C5a. Immunol. Lett. 2010, 128, 36–45. [Google Scholar] [CrossRef]
  40. Schafer, B.; Piliponsky, A.M.; Oka, T.; Song, C.H.; Gerard, N.P.; Gerard, C.; Tsai, M.; Kalesnikoff, J.; Galli, S.J. Mast cell anaphylatoxin receptor expression can enhance IgE-dependent skin inflammation in mice. J. Allergy Clin. Immunol. 2013, 131, 541–548.e9. [Google Scholar] [CrossRef]
  41. Redegeld, F.A.; Yu, Y.; Kumari, S.; Charles, N.; Blank, U. Non-IgE mediated mast cell activation. Immunol. Rev. 2018, 282, 87–113. [Google Scholar] [CrossRef]
  42. Marshall, J.S. Mast-cell responses to pathogens. Nat. Rev. Immunol. 2004, 4, 787–799. [Google Scholar] [CrossRef] [PubMed]
  43. Abraham, S.N.; St John, A.L. Mast cell-orchestrated immunity to pathogens. Nat. Rev. Immunol. 2010, 10, 440–452. [Google Scholar] [CrossRef] [PubMed]
  44. Rudich, N.; Ravid, K.; Sagi-Eisenberg, R. Mast cell adenosine receptors function: A focus on the a3 adenosine receptor and inflammation. Front. Immunol. 2012, 3, 134. [Google Scholar] [CrossRef] [PubMed]
  45. Ayyadurai, S.; Gibson, A.J.; D’Costa, S.; Overman, E.L.; Sommerville, L.J.; Poopal, A.C.; Mackey, E.; Li, Y.; Moeser, A.J. Frontline Science: Corticotropin-releasing factor receptor subtype 1 is a critical modulator of mast cell degranulation and stress-induced pathophysiology. J. Leukoc. Biol. 2017, 102, 1299–1312. [Google Scholar] [CrossRef] [PubMed]
  46. Kulka, M.; Sheen, C.H.; Tancowny, B.P.; Grammer, L.C.; Schleimer, R.P. Neuropeptides activate human mast cell degranulation and chemokine production. Immunology 2008, 123, 398–410. [Google Scholar] [CrossRef] [PubMed]
  47. Akahoshi, M.; Song, C.H.; Piliponsky, A.M.; Metz, M.; Guzzetta, A.; Abrink, M.; Schlenner, S.M.; Feyerabend, T.B.; Rodewald, H.R.; Pejler, G.; et al. Mast cell chymase reduces the toxicity of Gila monster venom, scorpion venom, and vasoactive intestinal polypeptide in mice. J. Clin. Investig. 2011, 121, 4180–4191. [Google Scholar] [CrossRef] [PubMed]
  48. Gaudenzio, N.; Sibilano, R.; Marichal, T.; Starkl, P.; Reber, L.L.; Cenac, N.; McNeil, B.D.; Dong, X.; Hernandez, J.D.; Sagi-Eisenberg, R.; et al. Different activation signals induce distinct mast cell degranulation strategies. J. Clin. Investig. 2016, 126, 3981–3998. [Google Scholar] [CrossRef] [PubMed]
  49. Theoharides, T.C. The mast cell: A neuroimmunoendocrine master player. Int. J. Tissue React. 1996, 18, 1–21. [Google Scholar]
  50. Theoharides, T.C.; Cochrane, D.E. Critical role of mast cells in inflammatory diseases and the effect of acute stress. J. Neuroimmunol. 2004, 146, 1–12. [Google Scholar] [CrossRef]
  51. Woolley, D.E. The mast cell in inflammatory arthritis. N. Engl. J. Med. 2003, 348, 1709–1711. [Google Scholar] [CrossRef]
  52. Harvima, I.T.; Viinamaki, H.; Naukkarinen, A.; Paukkonen, K.; Neittaanmaki, H.; Harvima, R.J.; Horsmanheimo, M. Association of cutaneous mast cells and sensory nerves with psychic stress in psoriasis. Psychother. Psychosom. 1993, 60, 168–176. [Google Scholar] [CrossRef] [PubMed]
  53. Ozdamar, S.O.; Seckin, D.; Kandemir, B.; Turanli, A.Y. Mast cells in psoriasis. Dermatology 1996, 192, 190. [Google Scholar] [CrossRef] [PubMed]
  54. Theoharides, T.C. Mast cells: The immune gate to the brain. Life Sci. 1990, 46, 607–617. [Google Scholar] [CrossRef]
  55. Jayapal, M.; Tay, H.K.; Reghunathan, R.; Zhi, L.; Chow, K.K.; Rauff, M.; Melendez, A.J. Genome-wide gene expression profiling of human mast cells stimulated by IgE or FcepsilonRI-aggregation reveals a complex network of genes involved in inflammatory responses. BMC Genom. 2006, 7, 210. [Google Scholar] [CrossRef]
  56. Cochrane, D.E.; Carraway, R.E.; Feldberg, R.S.; Boucher, W.; Gelfand, J.M. Stimulated rat mast cells generate histamine-releasing peptide from albumin. Peptides 1993, 14, 117–123. [Google Scholar] [CrossRef] [PubMed]
  57. Carraway, R.E.; Cochrane, D.E.; Boucher, W.; Mitra, S.P. Structures of histamine-releasing peptides formed by the action of acid proteases on mammalian albumin(s). J. Immunol. 1989, 143, 1680–1684. [Google Scholar] [CrossRef]
  58. Schmidlin, F.; Bunnett, N.W. Protease-activated receptors: How proteases signal to cells. Curr. Opin. Pharmacol. 2001, 1, 575–582. [Google Scholar] [CrossRef]
  59. Molino, M.; Barnathan, E.S.; Numerof, R.; Clark, J.; Dreyer, M.; Cumashi, A.; Hoxie, J.A.; Schechter, N.; Woolkalis, M.; Brass, L.F. Interactions of mast cell tryptase with thrombin receptors and PAR-2. J. Biol. Chem. 1997, 272, 4043–4049. [Google Scholar] [CrossRef]
  60. Theoharides, T.C.; Kempuraj, D.; Tagen, M.; Conti, P.; Kalogeromitros, D. Differential release of mast cell mediators and the pathogenesis of inflammation. Immunol. Rev. 2007, 217, 65–78. [Google Scholar] [CrossRef]
  61. Suzuki, A.; Suzuki, R.; Furuno, T.; Teshima, R.; Nakanishi, M. N-cadherin plays a role in the synapse-like structures between mast cells and neurites. Biol. Pharm. Bull. 2004, 27, 1891–1894. [Google Scholar] [CrossRef]
  62. Furuno, T.; Ito, A.; Koma, Y.; Watabe, K.; Yokozaki, H.; Bienenstock, J.; Nakanishi, M.; Kitamura, Y. The spermatogenic Ig superfamily/synaptic cell adhesion molecule mast-cell adhesion molecule promotes interaction with nerves. J. Immunol. 2005, 174, 6934–6942. [Google Scholar] [CrossRef]
  63. Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef]
  64. Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef]
  65. Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef]
  66. Gupta, S.; Ellis, S.E.; Ashar, F.N.; Moes, A.; Bader, J.S.; Zhan, J.; West, A.B.; Arking, D.E. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat. Commun. 2014, 5, 5748. [Google Scholar] [CrossRef] [PubMed]
  67. Xu, Z.X.; Kim, G.H.; Tan, J.W.; Riso, A.E.; Sun, Y.; Xu, E.Y.; Liao, G.Y.; Xu, H.; Lee, S.H.; Do, N.Y.; et al. Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations. Nat. Commun. 2020, 11, 1797. [Google Scholar] [CrossRef] [PubMed]
  68. Patel, A.B.; Tsilioni, I.; Leeman, S.E.; Theoharides, T.C. Neurotensin stimulates sortilin and mTOR in human microglia inhibitable by methoxyluteolin, a potential therapeutic target for autism. Proc. Natl. Acad. Sci. USA 2016, 113, E7049–E7058. [Google Scholar] [CrossRef]
  69. Theoharides, T.C.; Kavalioti, M.; Tsilioni, I. Mast Cells, Stress, Fear and Autism Spectrum Disorder. Int. J. Mol. Sci. 2019, 20, 3611. [Google Scholar] [CrossRef]
  70. Zhang, X.; Dong, H.; Li, N.; Zhang, S.; Sun, J.; Zhang, S.; Qian, Y. Activated brain mast cells contribute to postoperative cognitive dysfunction by evoking microglia activation and neuronal apoptosis. J. Neuroinflamm. 2016, 13, 127. [Google Scholar] [CrossRef] [PubMed]
  71. Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef]
  72. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  73. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef]
  74. Stein, M.; Keshav, S.; Harris, N.; Gordon, S. Interleukin 4 potently enhances murine macrophage mannose receptor activity: A marker of alternative immunologic macrophage activation. J. Exp. Med. 1992, 176, 287–292. [Google Scholar] [CrossRef]
  75. Gandhi, N.A.; Bennett, B.L.; Graham, N.M.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2 inflammation in disease. Nat. Rev. Drug Discov. 2016, 15, 35–50. [Google Scholar] [CrossRef]
  76. Ghosh, M.; Xu, Y.; Pearse, D.D. Cyclic AMP is a key regulator of M1 to M2a phenotypic conversion of microglia in the presence of Th2 cytokines. J. Neuroinflammation 2016, 13, 9. [Google Scholar] [CrossRef]
  77. Chhor, V.; Le Charpentier, T.; Lebon, S.; Ore, M.V.; Celador, I.L.; Josserand, J.; Degos, V.; Jacotot, E.; Hagberg, H.; Savman, K.; et al. Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro. Brain Behav. Immun. 2013, 32, 70–85. [Google Scholar] [CrossRef] [PubMed]
  78. Suh, H.S.; Zhao, M.L.; Derico, L.; Choi, N.; Lee, S.C. Insulin-like growth factor 1 and 2 (IGF1, IGF2) expression in human microglia: Differential regulation by inflammatory mediators. J. Neuroinflamm. 2013, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  79. O’Kusky, J.; Ye, P. Neurodevelopmental effects of insulin-like growth factor signaling. Front. Neuroendocrinol. 2012, 33, 230–251. [Google Scholar] [CrossRef] [PubMed]
  80. Mills, J.L.; Hediger, M.L.; Molloy, C.A.; Chrousos, G.P.; Manning-Courtney, P.; Yu, K.F.; Brasington, M.; England, L.J. Elevated levels of growth-related hormones in autism and autism spectrum disorder. Clin. Endocrinol. 2007, 67, 230–237. [Google Scholar] [CrossRef] [PubMed]
  81. Hendriksen, E.; van Bergeijk, D.; Oosting, R.S.; Redegeld, F.A. Mast cells in neuroinflammation and brain disorders. Neurosci. Biobehav. Rev. 2017, 79, 119–133. [Google Scholar] [CrossRef] [PubMed]
  82. Kempuraj, D.; Mentor, S.; Thangavel, R.; Ahmed, M.E.; Selvakumar, G.P.; Raikwar, S.P.; Dubova, I.; Zaheer, S.; Iyer, S.S.; Zaheer, A. Mast Cells in Stress, Pain, Blood-Brain Barrier, Neuroinflammation and Alzheimer’s Disease. Front. Cell Neurosci. 2019, 13, 54. [Google Scholar] [CrossRef] [PubMed]
  83. Hansen, D.V.; Hanson, J.E.; Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 2018, 217, 459–472. [Google Scholar] [CrossRef] [PubMed]
  84. Hatziagelaki, E.; Adamaki, M.; Tsilioni, I.; Dimitriadis, G.; Theoharides, T.C. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome-Metabolic Disease or Disturbed Homeostasis due to Focal Inflammation in the Hypothalamus? J. Pharmacol. Exp. Ther. 2018, 367, 155–167. [Google Scholar] [CrossRef] [PubMed]
  85. Breach, M.R.; Dye, C.N.; Joshi, A.; Platko, S.; Gilfarb, R.A.; Krug, A.R.; Franceschelli, D.V.; Galan, A.; Dodson, C.M.; Lenz, K.M. Maternal allergic inflammation in rats impacts the offspring perinatal neuroimmune milieu and the development of social play, locomotor behavior, and cognitive flexibility. Brain Behav. Immun. 2021, 95, 269–286. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, W.; Ji, P.; Riopelle, R.J.; Dow, K.E. Functional expression of corticotropin-releasing hormone (CRH) receptor 1 in cultured rat microglia. J. Neurochem. 2002, 80, 287–294. [Google Scholar] [CrossRef]
  87. Podlesek, A.; Komidar, L.; Kavcic, V. The Relationship Between Perceived Stress and Subjective Cognitive Decline During the COVID-19 Epidemic. Front. Psychol. 2021, 12, 647971. [Google Scholar] [CrossRef]
  88. Martin, S.; Dicou, E.; Vincent, J.P.; Mazella, J. Neurotensin and the neurotensin receptor-3 in microglial cells. J. Neurosci. Res. 2005, 81, 322–326. [Google Scholar] [CrossRef]
  89. Tsilioni, I.; Dodman, N.; Petra, A.I.; Taliou, A.; Francis, K.; Moon-Fanelli, A.; Shuster, L.; Theoharides, T.C. Elevated serum neurotensin and CRH levels in children with autistic spectrum disorders and tail-chasing Bull Terriers with a phenotype similar to autism. Transl. Psychiatry 2014, 4, e466. [Google Scholar] [CrossRef]
  90. Martin, S.; Vincent, J.P.; Mazella, J. Involvement of the neurotensin receptor-3 in the neurotensin-induced migration of human microglia. J. Neurosci. 2003, 23, 1198–1205. [Google Scholar] [CrossRef] [PubMed]
  91. Almehmadi, K.A.; Tsilioni, I.; Theoharides, T.C. Increased Expression of miR-155p5 in Amygdala of Children with Autism Spectrum Disorder. Autism Res. 2020, 13, 18–23. [Google Scholar] [CrossRef] [PubMed]
  92. Tsilioni, I.; Pantazopoulos, H.; Conti, P.; Leeman, S.E.; Theoharides, T.C. IL-38 inhibits microglial inflammatory mediators and is decreased in amygdala of children with autism spectrum disorder. Proc. Natl. Acad. Sci. USA 2020, 117, 16475–16480. [Google Scholar] [CrossRef] [PubMed]
  93. Jyonouchi, H.; Geng, L.; Streck, D.L.; Toruner, G.A. Children with autism spectrum disorders (ASD) who exhibit chronic gastrointestinal (GI) symptoms and marked fluctuation of behavioral symptoms exhibit distinct innate immune abnormalities and transcriptional profiles of peripheral blood (PB) monocytes. J. Neuroimmunol. 2011, 238, 73–80. [Google Scholar] [CrossRef] [PubMed]
  94. Ashwood, P.; Anthony, A.; Torrente, F.; Wakefield, A.J. Spontaneous mucosal lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms: Mucosal immune activation and reduced counter regulatory interleukin-10. J. Clin. Immunol. 2004, 24, 664–673. [Google Scholar] [CrossRef] [PubMed]
  95. Jyonouchi, H.; Sun, S.; Le, H. Proinflammatory and regulatory cytokine production associated with innate and adaptive immune responses in children with autism spectrum disorders and developmental regression. J. Neuroimmunol. 2001, 120, 170–179. [Google Scholar] [CrossRef] [PubMed]
  96. Bilbo, S.D.; Schwarz, J.M. The immune system and developmental programming of brain and behavior. Front. Neuroendocrinol. 2012, 33, 267–286. [Google Scholar] [CrossRef] [PubMed]
  97. Molloy, C.A.; Morrow, A.L.; Meinzen-Derr, J.; Schleifer, K.; Dienger, K.; Manning-Courtney, P.; Altaye, M.; Wills-Karp, M. Elevated cytokine levels in children with autism spectrum disorder. J. Neuroimmunol. 2006, 172, 198–205. [Google Scholar] [CrossRef]
  98. 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]
  99. 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]
  100. 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]
  101. Ormstad, H.; Bryn, V.; Saugstad, O.D.; Skjeldal, O.; Maes, M. Role of the Immune System in Autism Spectrum Disorders (ASD). CNS Neurol. Disord. Drug Targets 2018, 17, 489–495. [Google Scholar] [CrossRef]
  102. Ashwood, P.; Enstrom, A.; Krakowiak, P.; Hertz-Picciotto, I.; Hansen, R.L.; Croen, L.A.; Ozonoff, S.; Pessah, I.N.; Van de Water, J. Decreased transforming growth factor beta1 in autism: A potential link between immune dysregulation and impairment in clinical behavioral outcomes. J. Neuroimmunol. 2008, 204, 149–153. [Google Scholar] [CrossRef]
  103. Croonenberghs, J.; Bosmans, E.; Deboutte, D.; Kenis, G.; Maes, M. Activation of the inflammatory response system in autism. Neuropsychobiology 2002, 45, 1–6. [Google Scholar] [CrossRef]
  104. Masi, A.; Breen, E.J.; Alvares, G.A.; Glozier, N.; Hickie, I.B.; Hunt, A.; Hui, J.; Beilby, J.; Ravine, D.; Wray, J.; et al. Cytokine levels and associations with symptom severity in male and female children with autism spectrum disorder. Mol. Autism 2017, 8, 63. [Google Scholar] [CrossRef]
  105. Masi, A.; Glozier, N.; Dale, R.; Guastella, A.J. The Immune System, Cytokines, and Biomarkers in Autism Spectrum Disorder. Neurosci. Bull. 2017, 33, 194–204. [Google Scholar] [CrossRef]
  106. Nadeem, A.; Ahmad, S.F.; Attia, S.M.; Bakheet, S.A.; Al-Harbi, N.O.; Al-Ayadhi, L.Y. Activation of IL-17 receptor leads to increased oxidative inflammation in peripheral monocytes of autistic children. Brain Behav. Immun. 2018, 67, 335–344. [Google Scholar] [CrossRef] [PubMed]
  107. Ahmad, S.F.; Nadeem, A.; Ansari, M.A.; Bakheet, S.A.; Attia, S.M.; Zoheir, K.M.; Al-Ayadhi, L.Y.; Alzahrani, M.Z.; Alsaad, A.M.; Alotaibi, M.R.; et al. Imbalance between the anti- and pro-inflammatory milieu in blood leukocytes of autistic children. Mol. Immunol. 2017, 82, 57–65. [Google Scholar] [CrossRef]
  108. Suzuki, K.; Matsuzaki, H.; Iwata, K.; Kameno, Y.; Shimmura, C.; Kawai, S.; Yoshihara, Y.; Wakuda, T.; Takebayashi, K.; Takagai, S.; et al. Plasma cytokine profiles in subjects with high-functioning autism spectrum disorders. PLoS ONE 2011, 6, e20470. [Google Scholar] [CrossRef] [PubMed]
  109. Ricci, S.; Businaro, R.; Ippoliti, F.; Lo Vasco, V.R.; Massoni, F.; Onofri, E.; Troili, G.M.; Pontecorvi, V.; Morelli, M.; Rapp Ricciardi, M.; et al. Altered cytokine and BDNF levels in autism spectrum disorder. Neurotox. Res. 2013, 24, 491–501. [Google Scholar] [CrossRef] [PubMed]
  110. Enstrom, A.M.; Lit, L.; Onore, C.E.; Gregg, J.P.; Hansen, R.L.; Pessah, I.N.; Hertz-Picciotto, I.; Van de Water, J.A.; Sharp, F.R.; Ashwood, P. Altered gene expression and function of peripheral blood natural killer cells in children with autism. Brain Behav. Immun. 2009, 23, 124–133. [Google Scholar] [CrossRef] [PubMed]
  111. Chez, M.G.; Dowling, T.; Patel, P.B.; Khanna, P.; Kominsky, M. Elevation of tumor necrosis factor-alpha in cerebrospinal fluid of autistic children. Pediatr. Neurol. 2007, 36, 361–365. [Google Scholar] [CrossRef]
  112. Goshen, I.; Kreisel, T.; Ounallah-Saad, H.; Renbaum, P.; Zalzstein, Y.; Ben-Hur, T.; Levy-Lahad, E.; Yirmiya, R. A dual role for interleukin-1 in hippocampal-dependent memory processes. Psychoneuroendocrinology 2007, 32, 1106–1115. [Google Scholar] [CrossRef]
  113. Labrousse, V.F.; Costes, L.; Aubert, A.; Darnaudery, M.; Ferreira, G.; Amedee, T.; Laye, S. Impaired interleukin-1beta and c-Fos expression in the hippocampus is associated with a spatial memory deficit in P2X(7) receptor-deficient mice. PLoS ONE 2009, 4, e6006. [Google Scholar] [CrossRef]
  114. 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. Neuroinflamm. 2011, 8, 52. [Google Scholar] [CrossRef]
  115. Patterson, P.H. Immune involvement in schizophrenia and autism: Etiology, pathology and animal models. Behav. Brain Res. 2009, 204, 313–321. [Google Scholar] [CrossRef] [PubMed]
  116. Bilbo, S.D. Early-life infection is a vulnerability factor for aging-related glial alterations and cognitive decline. Neurobiol. Learn. Mem. 2010, 94, 57–64. [Google Scholar] [CrossRef] [PubMed]
  117. Ashwood, P.; Wills, S.; Van de Water, J. The immune response in autism: A new frontier for autism research. J. Leukoc. Biol. 2006, 80, 1–15. [Google Scholar] [CrossRef] [PubMed]
  118. Ashwood, P.; Krakowiak, P.; Hertz-Picciotto, I.; Hansen, R.; Pessah, I.; Van de Water, J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav. Immun. 2011, 25, 40–45. [Google Scholar] [CrossRef] [PubMed]
  119. Xie, J.; Huang, L.; Li, X.; Li, H.; Zhou, Y.; Zhu, H.; Pan, T.; Kendrick, K.M.; Xu, W. Immunological cytokine profiling identifies TNF-alpha as a key molecule dysregulated in autistic children. Oncotarget 2017, 8, 82390–82398. [Google Scholar] [CrossRef] [PubMed]
  120. Li, L.; Kim, J.; Boussiotis, V.A. IL-1beta-mediated signals preferentially drive conversion of regulatory T cells but not conventional T cells into IL-17-producing cells. J. Immunol. 2010, 185, 4148–4153. [Google Scholar] [CrossRef] [PubMed]
  121. Bickel, M. The role of interleukin-8 in inflammation and mechanisms of regulation. J. Periodontol. 1993, 64, 456–460. [Google Scholar] [PubMed]
  122. Krakowiak, P.; Goines, P.E.; Tancredi, D.J.; Ashwood, P.; Hansen, R.L.; Hertz-Picciotto, I.; Van de Water, J. Neonatal Cytokine Profiles Associated With Autism Spectrum Disorder. Biol. Psychiatry 2017, 81, 442–451. [Google Scholar] [CrossRef] [PubMed]
  123. Goines, P.E.; Croen, L.A.; Braunschweig, D.; Yoshida, C.K.; Grether, J.; Hansen, R.; Kharrazi, M.; Ashwood, P.; Van de Water, J. Increased midgestational IFN-gamma, IL-4 and IL-5 in women bearing a child with autism: A case-control study. Mol. Autism 2011, 2, 13. [Google Scholar] [CrossRef] [PubMed]
  124. Noordenbos, T.; Blijdorp, I.; Chen, S.; Stap, J.; Mul, E.; Canete, J.D.; Lubberts, E.; Yeremenko, N.; Baeten, D. Human mast cells capture, store, and release bioactive, exogenous IL-17A. J. Leukoc. Biol. 2016, 100, 453–462. [Google Scholar] [CrossRef] [PubMed]
  125. 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]
  126. Boyce, J.A. Mast cells and eicosanoid mediators: A system of reciprocal paracrine and autocrine regulation. Immunol. Rev. 2007, 217, 168–185. [Google Scholar] [CrossRef] [PubMed]
  127. Mukai, K.; Tsai, M.; Saito, H.; Galli, S.J. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol. Rev. 2018, 282, 121–150. [Google Scholar] [CrossRef]
  128. Kandere-Grzybowska, K.; Letourneau, R.; Kempuraj, D.; Donelan, J.; Poplawski, S.; Boucher, W.; Athanassiou, A.; Theoharides, T.C. IL-1 induces vesicular secretion of IL-6 without degranulation from human mast cells. J. Immunol. 2003, 171, 4830–4836. [Google Scholar] [CrossRef]
  129. Fux, M.; Pecaric-Petkovic, T.; Odermatt, A.; Hausmann, O.V.; Lorentz, A.; Bischoff, S.C.; Virchow, J.C.; Dahinden, C.A. IL-33 is a mediator rather than a trigger of the acute allergic response in humans. Allergy 2014, 69, 216–222. [Google Scholar] [CrossRef]
  130. Piras, I.S.; Haapanen, L.; Napolioni, V.; Sacco, R.; Van de Water, J.; Persico, A.M. Anti-brain antibodies are associated with more severe cognitive and behavioral profiles in Italian children with Autism Spectrum Disorder. Brain Behav. Immun. 2014, 38, 91–99. [Google Scholar] [CrossRef]
  131. Theoharides, T.C.; Zhang, B.; Kempuraj, D.; Tagen, M.; Vasiadi, M.; Angelidou, A.; Alysandratos, K.D.; Kalogeromitros, D.; Asadi, S.; Stavrianeas, N.; et al. IL-33 augments substance P-induced VEGF secretion from human mast cells and is increased in psoriatic skin. Proc. Natl. Acad. Sci. USA 2010, 107, 4448–4453. [Google Scholar] [CrossRef]
  132. Taracanova, A.; Alevizos, M.; Karagkouni, A.; Weng, Z.; Norwitz, E.; Conti, P.; Leeman, S.E.; Theoharides, T.C. SP and IL-33 together markedly enhance TNF synthesis and secretion from human mast cells mediated by the interaction of their receptors. Proc. Natl. Acad. Sci. USA 2017, 114, E4002–E4009. [Google Scholar] [CrossRef] [PubMed]
  133. Taracanova, A.; Tsilioni, I.; Conti, P.; Norwitz, E.R.; Leeman, S.E.; Theoharides, T.C. Substance P and IL-33 administered together stimulate a marked secretion of IL-1beta from human mast cells, inhibited by methoxyluteolin. Proc. Natl. Acad. Sci. USA 2018, 115, E9381–E9390. [Google Scholar] [CrossRef] [PubMed]
  134. Inga Jacome, M.C.; Morales Chacon, L.M.; Vera Cuesta, H.; Maragoto Rizo, C.; Whilby Santiesteban, M.; Ramos Hernandez, L.; Noris Garcia, E.; Gonzalez Fraguela, M.E.; Fernandez Verdecia, C.I.; Vegas Hurtado, Y.; et al. Peripheral Inflammatory Markers Contributing to Comorbidities in Autism. Behav. Sci. 2016, 6, 29. [Google Scholar] [CrossRef] [PubMed]
  135. Eftekharian, M.M.; Ghafouri-Fard, S.; Noroozi, R.; Omrani, M.D.; Arsang-Jang, S.; Ganji, M.; Gharzi, V.; Noroozi, H.; Komaki, A.; Mazdeh, M.; et al. Cytokine profile in autistic patients. Cytokine 2018, 108, 120–126. [Google Scholar] [CrossRef] [PubMed]
  136. Wei, H.; Mori, S.; Hua, K.; Li, X. Alteration of brain volume in IL-6 overexpressing mice related to autism. Int. J. Dev. Neurosci. 2012, 30, 554–559. [Google Scholar] [CrossRef] [PubMed]
  137. Wei, H.; Chadman, K.K.; McCloskey, D.P.; Sheikh, A.M.; Malik, M.; Brown, W.T.; Li, X. Brain IL-6 elevation causes neuronal circuitry imbalances and mediates autism-like behaviors. Biochim. Biophys. Acta 2012, 1822, 831–842. [Google Scholar] [CrossRef]
  138. 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]
  139. Xu, N.; Li, X.; Zhong, Y. Inflammatory cytokines: Potential biomarkers of immunologic dysfunction in autism spectrum disorders. Mediat. Inflamm. 2015, 2015, 531518. [Google Scholar] [CrossRef]
  140. Maes, M.; Carvalho, A.F. The Compensatory Immune-Regulatory Reflex System (CIRS) in Depression and Bipolar Disorder. Mol. Neurobiol. 2018, 55, 8885–8903. [Google Scholar] [CrossRef]
  141. Saghazadeh, A.; Ataeinia, B.; Keynejad, K.; Abdolalizadeh, A.; Hirbod-Mobarakeh, A.; Rezaei, N. Anti-inflammatory cytokines in autism spectrum disorders: A systematic review and meta-analysis. Cytokine 2019, 123, 154740. [Google Scholar] [CrossRef]
  142. Rossignol, D.A.; Frye, R.E. A review of research trends in physiological abnormalities in autism spectrum disorders: Immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Mol. Psychiatry 2012, 17, 389–401. [Google Scholar] [CrossRef]
  143. Angelidou, A.; Asadi, S.; Alysandratos, K.D.; Karagkouni, A.; Kourembanas, S.; Theoharides, T.C. Perinatal stress, brain inflammation and risk of autism-review and proposal. BMC Pediatr. 2012, 12, 89. [Google Scholar] [CrossRef]
  144. Theoharides, T.C.; Valent, P.; Akin, C. Mast Cells, Mastocytosis, and Related Disorders. N. Engl. J. Med. 2015, 373, 163–172. [Google Scholar] [CrossRef] [PubMed]
  145. Ribatti, D. The crucial role of mast cells in blood-brain barrier alterations. Exp. Cell Res. 2015, 338, 119–125. [Google Scholar] [CrossRef] [PubMed]
  146. Theoharides, T.C.; Doyle, R.; Francis, K.; Conti, P.; Kalogeromitros, D. Novel therapeutic targets for autism. Trends Pharmacol. Sci. 2008, 29, 375–382. [Google Scholar] [CrossRef] [PubMed]
  147. Reichenberg, A.; Yirmiya, R.; Schuld, A.; Kraus, T.; Haack, M.; Morag, A.; Pollmacher, T. Cytokine-associated emotional and cognitive disturbances in humans. Arch. Gen. Psychiatry 2001, 58, 445–452. [Google Scholar] [CrossRef] [PubMed]
  148. Cao, J.; Papadopoulou, N.; Kempuraj, D.; Boucher, W.S.; Sugimoto, K.; Cetrulo, C.L.; Theoharides, T.C. Human mast cells express corticotropin-releasing hormone (CRH) receptors and CRH leads to selective secretion of vascular endothelial growth factor. J. Immunol. 2005, 174, 7665–7675. [Google Scholar] [CrossRef] [PubMed]
  149. Donelan, J.; Boucher, W.; Papadopoulou, N.; Lytinas, M.; Papaliodis, D.; Dobner, P.; Theoharides, T.C. Corticotropin-releasing hormone induces skin vascular permeability through a neurotensin-dependent process. Proc. Natl. Acad. Sci. USA 2006, 103, 7759–7764. [Google Scholar] [CrossRef] [PubMed]
  150. Patel, A.B.; Theoharides, T.C. Methoxyluteolin Inhibits Neuropeptide-stimulated Proinflammatory Mediator Release via mTOR Activation from Human Mast Cells. J. Pharmacol. Exp. Ther. 2017, 361, 462–471. [Google Scholar] [CrossRef]
  151. Mustain, W.C.; Rychahou, P.G.; Evers, B.M. The role of neurotensin in physiologic and pathologic processes. Curr. Opin. Endocrinol. Diabetes Obes. 2011, 18, 75–82. [Google Scholar] [CrossRef]
  152. Mashaghi, A.; Marmalidou, A.; Tehrani, M.; Grace, P.M.; Pothoulakis, C.; Dana, R. Neuropeptide substance P and the immune response. Cell Mol. Life Sci. 2016, 73, 4249–4264. [Google Scholar] [CrossRef]
  153. Zhang, X.; Wang, Y.; Dong, H.; Xu, Y.; Zhang, S. Induction of Microglial Activation by Mediators Released from Mast Cells. Cell Physiol. Biochem. 2016, 38, 1520–1531. [Google Scholar] [CrossRef]
  154. Girolamo, F.; Coppola, C.; Ribatti, D. Immunoregulatory effect of mast cells influenced by microbes in neurodegenerative diseases. Brain Behav. Immun. 2017, 65, 68–89. [Google Scholar] [CrossRef] [PubMed]
  155. Theoharides, T.C.; Stewart, J.M.; Hatziagelaki, E.; Kolaitis, G. Brain “fog”, inflammation and obesity: Key aspects of neuropsychiatric disorders improved by luteolin. Front. Neurosci. 2015, 9, 225. [Google Scholar] [CrossRef] [PubMed]
  156. Theoharides, T.C.; Tsilioni, I.; Patel, A.B.; Doyle, R. Atopic diseases and inflammation of the brain in the pathogenesis of autism spectrum disorders. Transl. Psychiatry 2016, 6, e844. [Google Scholar] [CrossRef] [PubMed]
  157. Ghanizadeh, A. Targeting neurotensin as a potential novel approach for the treatment of autism. J. Neuroinflamm. 2010, 7, 58. [Google Scholar] [CrossRef] [PubMed]
  158. Theoharides, T.C.; Konstantinidou, A.D. Corticotropin-releasing hormone and the blood-brain-barrier. Front. Biosci. 2007, 12, 1615–1628. [Google Scholar] [CrossRef] [PubMed]
  159. Vasiadi, M.; Therianou, A.; Alysandratos, K.D.; Katsarou-Katsari, A.; Petrakopoulou, T.; Theoharides, A.; Papadavid, E.; Stavrianeas, N.; Antoniou, C.; Kalogeromitros, D.; et al. Serum neurotensin (NT) is increased in psoriasis and NT induces vascular endothelial growth factor release from human mast cells. Br. J. Dermatol. 2012, 166, 1349–1352. [Google Scholar] [CrossRef] [PubMed]
  160. Zhang, B.; Asadi, S.; Weng, Z.; Sismanopoulos, N.; Theoharides, T.C. Stimulated human mast cells secrete mitochondrial components that have autocrine and paracrine inflammatory actions. PLoS ONE 2012, 7, e49767. [Google Scholar] [CrossRef]
  161. Asadi, S.; Theoharides, T.C. Corticotropin-releasing hormone and extracellular mitochondria augment IgE-stimulated human mast-cell vascular endothelial growth factor release, which is inhibited by luteolin. J. Neuroinflamm. 2012, 9, 85. [Google Scholar] [CrossRef]
  162. Singh, L.K.; Pang, X.; Alexacos, N.; Letourneau, R.; Theoharides, T.C. Acute immobilization stress triggers skin mast cell degranulation via corticotropin releasing hormone, neurotensin, and substance P: A link to neurogenic skin disorders. Brain Behav. Immun. 1999, 13, 225–239. [Google Scholar] [CrossRef] [PubMed]
  163. Barrocas, A.M.; Cochrane, D.E.; Carraway, R.E.; Feldberg, R.S. Neurotensin stimulation of mast cell secretion is receptor-mediated, pertussis-toxin sensitive and requires activation of phospholipase C. Immunopharmacology 1999, 41, 131–137. [Google Scholar] [CrossRef] [PubMed]
  164. Nelson, K.B.; Grether, J.K.; Croen, L.A.; Dambrosia, J.M.; Dickens, B.F.; Jelliffe, L.L.; Hansen, R.L.; Phillips, T.M. Neuropeptides and neurotrophins in neonatal blood of children with autism or mental retardation. Ann. Neurol. 2001, 49, 597–606. [Google Scholar] [CrossRef] [PubMed]
  165. Nelson, P.G.; Kuddo, T.; Song, E.Y.; Dambrosia, J.M.; Kohler, S.; Satyanarayana, G.; Vandunk, C.; Grether, J.K.; Nelson, K.B. Selected neurotrophins, neuropeptides, and cytokines: Developmental trajectory and concentrations in neonatal blood of children with autism or Down syndrome. Int. J. Dev. Neurosci. 2006, 24, 73–80. [Google Scholar] [CrossRef] [PubMed]
  166. Rozniecki, J.J.; Dimitriadou, V.; Lambracht-Hall, M.; Pang, X.; Theoharides, T.C. Morphological and functional demonstration of rat dura mater mast cell-neuron interactions in vitro and in vivo. Brain Res. 1999, 849, 1–15. [Google Scholar] [CrossRef] [PubMed]
  167. Polyzoidis, S.; Koletsa, T.; Panagiotidou, S.; Ashkan, K.; Theoharides, T.C. Mast cells in meningiomas and brain inflammation. J. Neuroinflamm. 2015, 12, 170. [Google Scholar] [CrossRef] [PubMed]
  168. Arck, P.C.; Slominski, A.; Theoharides, T.C.; Peters, E.M.; Paus, R. Neuroimmunology of stress: Skin takes center stage. J. Investig. Dermatol. 2006, 126, 1697–1704. [Google Scholar] [CrossRef]
  169. Theoharides, T.C.; Donelan, J.M.; Papadopoulou, N.; Cao, J.; Kempuraj, D.; Conti, P. Mast cells as targets of corticotropin-releasing factor and related peptides. Trends Pharmacol. Sci. 2004, 25, 563–568. [Google Scholar] [CrossRef]
  170. Mastorakos, G.; Chrousos, G.P.; Weber, J.S. Recombinant interleukin-6 activates the hypothalamic-pituitary-adrenal axis in humans. J. Clin. Endocrinol. Metab. 1993, 77, 1690–1694. [Google Scholar] [CrossRef]
  171. Kempuraj, D.; Papadopoulou, N.G.; Lytinas, M.; Huang, M.; Kandere-Grzybowska, K.; Madhappan, B.; Boucher, W.; Christodoulou, S.; Athanassiou, A.; Theoharides, T.C. Corticotropin-releasing hormone and its structurally related urocortin are synthesized and secreted by human mast cells. Endocrinology 2004, 145, 43–48. [Google Scholar] [CrossRef]
  172. Kraft, S.; Kinet, J.P. New developments in FcepsilonRI regulation, function and inhibition. Nat. Rev. Immunol. 2007, 7, 365–378. [Google Scholar] [CrossRef]
  173. Theoharides, T.C.; Angelidou, A.; Alysandratos, K.D.; Zhang, B.; Asadi, S.; Francis, K.; Toniato, E.; Kalogeromitros, D. Mast cell activation and autism. Biochim. Biophys. Acta 2012, 1822, 34–41. [Google Scholar] [CrossRef] [PubMed]
  174. Theoharides, T.C. Neuroendocrinology of mast cells: Challenges and controversies. Exp. Dermatol. 2017, 26, 751–759. [Google Scholar] [CrossRef] [PubMed]
  175. Theoharides, T.C. Danger Signals and Inflammation. Clin. Ther. 2016, 38, 996–999. [Google Scholar] [CrossRef] [PubMed]
  176. Theoharides, T.C.; Asadi, S.; Panagiotidou, S.; Weng, Z. The “missing link” in autoimmunity and autism: Extracellular mitochondrial components secreted from activated live mast cells. Autoimmun. Rev. 2013, 12, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
  177. Sun, S.; Sursal, T.; Adibnia, Y.; Zhao, C.; Zheng, Y.; Li, H.; Otterbein, L.E.; Hauser, C.J.; Itagaki, K. Mitochondrial DAMPs increase endothelial permeability through neutrophil dependent and independent pathways. PLoS ONE 2013, 8, e59989. [Google Scholar] [CrossRef] [PubMed]
  178. Zhang, B.; Angelidou, A.; Alysandratos, K.D.; Vasiadi, M.; Francis, K.; Asadi, S.; Theoharides, A.; Sideri, K.; Lykouras, L.; Kalogeromitros, D.; et al. Mitochondrial DNA and anti-mitochondrial antibodies in serum of autistic children. J. Neuroinflamm. 2010, 7, 80. [Google Scholar] [CrossRef] [PubMed]
  179. Bryniarski, K.; Ptak, W.; Jayakumar, A.; Pullmann, K.; Caplan, M.J.; Chairoungdua, A.; Lu, J.; Adams, B.D.; Sikora, E.; Nazimek, K.; et al. Antigen-specific, antibody-coated, exosome-like nanovesicles deliver suppressor T-cell microRNA-150 to effector T cells to inhibit contact sensitivity. J. Allergy Clin. Immunol. 2013, 132, 170–181. [Google Scholar] [CrossRef] [PubMed]
  180. Tsilioni, I.; Panagiotidou, S.; Theoharides, T.C. Exosomes in neurologic and psychiatric disorders. Clin. Ther. 2014, 36, 882–888. [Google Scholar] [CrossRef]
  181. McCurdy, J.D.; Olynych, T.J.; Maher, L.H.; Marshall, J.S. Cutting edge: Distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J. Immunol. 2003, 170, 1625–1629. [Google Scholar] [CrossRef]
  182. Kulka, M.; Alexopoulou, L.; Flavell, R.A.; Metcalfe, D.D. Activation of mast cells by double-stranded RNA: Evidence for activation through Toll-like receptor 3. J. Allergy Clin. Immunol. 2004, 114, 174–182. [Google Scholar] [CrossRef]
  183. Arvola, T.; Ruuska, T.; Keranen, J.; Hyoty, H.; Salminen, S.; Isolauri, E. Rectal bleeding in infancy: Clinical, allergological, and microbiological examination. Pediatrics 2006, 117, e760–e768. [Google Scholar] [CrossRef]
  184. Young, H.A.; Geier, D.A.; Geier, M.R. Thimerosal exposure in infants and neurodevelopmental disorders: An assessment of computerized medical records in the Vaccine Safety Datalink. J. Neurol. Sci. 2008, 271, 110–118. [Google Scholar] [CrossRef]
  185. Hertz-Picciotto, I.; Park, H.Y.; Dostal, M.; Kocan, A.; Trnovec, T.; Sram, R. Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin. Pharmacol. Toxicol. 2008, 102, 146–154. [Google Scholar] [CrossRef]
  186. Grandjean, P.; Landrigan, P.J. Developmental neurotoxicity of industrial chemicals. Lancet 2006, 368, 2167–2178. [Google Scholar] [CrossRef] [PubMed]
  187. Kempuraj, D.; Asadi, S.; Zhang, B.; Manola, A.; Hogan, J.; Peterson, E.; Theoharides, T.C. Mercury induces inflammatory mediator release from human mast cells. J. Neuroinflamm. 2010, 7, 20. [Google Scholar] [CrossRef] [PubMed]
  188. Bernard, S.; Enayati, A.; Roger, H.; Binstock, T.; Redwood, L. The role of mercury in the pathogenesis of autism. Mol. Psychiatry 2002, 7 (Suppl. 2), S42–S43. [Google Scholar] [CrossRef] [PubMed]
  189. Geier, D.A.; Kern, J.K.; King, P.G.; Sykes, L.K.; Geier, M.R. Hair toxic metal concentrations and autism spectrum disorder severity in young children. Int. J. Environ. Res. Public Health 2012, 9, 4486–4497. [Google Scholar] [CrossRef] [PubMed]
  190. Tomljenovic, L.; Shaw, C.A. Do aluminum vaccine adjuvants contribute to the rising prevalence of autism? J. Inorg. Biochem. 2011, 105, 1489–1499. [Google Scholar] [CrossRef] [PubMed]
  191. Kumar, S.; Khodoun, M.; Kettleson, E.M.; McKnight, C.; Reponen, T.; Grinshpun, S.A.; Adhikari, A. Glyphosate-rich air samples induce IL-33, TSLP and generate IL-13 dependent airway inflammation. Toxicology 2014, 325, 42–51. [Google Scholar] [CrossRef] [PubMed]
  192. Aldinger, K.A.; Plummer, J.T.; Qiu, S.; Levitt, P. SnapShot: Genetics of autism. Neuron 2011, 72, 418–418.e411. [Google Scholar] [CrossRef] [PubMed]
  193. Liao, T.C.; Lien, Y.T.; Wang, S.; Huang, S.L.; Chen, C.Y. Comorbidity of Atopic Disorders with Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder. J. Pediatr. 2016, 171, 248–255. [Google Scholar] [CrossRef] [PubMed]
  194. Theoharides, T.C. Is a subtype of autism an allergy of the brain? Clin. Ther. 2013, 35, 584–591. [Google Scholar] [CrossRef] [PubMed]
  195. Shen, Q.; Zhang, Q.; Zhao, J.; Huang, Z.; Wang, X.; Ni, M.; Tang, Z.; Liu, Z. Association Between Maternal Perceived Stress in All Trimesters of Pregnancy and Infant Atopic Dermatitis: A Prospective Birth Cohort Study. Front. Pediatr. 2020, 8, 526994. [Google Scholar] [CrossRef]
  196. Beversdorf, D.Q.; Stevens, H.E.; Margolis, K.G.; Van de Water, J. Prenatal Stress and Maternal Immune Dysregulation in Autism Spectrum Disorders: Potential Points for Intervention. Curr. Pharm. Des. 2019, 25, 4331–4343. [Google Scholar] [CrossRef] [PubMed]
  197. Cai, R.Y.; Richdale, A.L.; Uljarevic, M.; Dissanayake, C.; Samson, A.C. Emotion regulation in autism spectrum disorder: Where we are and where we need to go. Autism Res. 2018, 11, 962–978. [Google Scholar] [CrossRef]
  198. Abdallah, M.W.; Larsen, N.; Grove, J.; Norgaard-Pedersen, B.; Thorsen, P.; Mortensen, E.L.; Hougaard, D.M. Amniotic fluid inflammatory cytokines: Potential markers of immunologic dysfunction in autism spectrum disorders. World J. Biol. Psychiatry 2013, 14, 528–538. [Google Scholar] [CrossRef]
  199. Patel, S.; Cooper, M.N.; Jones, H.; Whitehouse, A.J.O.; Dale, R.C.; Guastella, A.J. Maternal immune-related conditions during pregnancy may be a risk factor for neuropsychiatric problems in offspring throughout childhood and adolescence. Psychol. Med. 2021, 51, 2904–2914. [Google Scholar] [CrossRef]
  200. Jarmolowska, B.; Bukalo, M.; Fiedorowicz, E.; Cieslinska, A.; Kordulewska, N.K.; Moszynska, M.; Swiatecki, A.; Kostyra, E. Role of Milk-Derived Opioid Peptides and Proline Dipeptidyl Peptidase-4 in Autism Spectrum Disorders. Nutrients 2019, 11, 87. [Google Scholar] [CrossRef]
  201. Li, H.; Liu, H.; Chen, X.; Zhang, J.; Tong, G.; Sun, Y. Association of food hypersensitivity in children with the risk of autism spectrum disorder: A meta-analysis. Eur. J. Pediatr. 2021, 180, 999–1008. [Google Scholar] [CrossRef]
  202. Zhou, L.; Chen, L.; Li, X.; Li, T.; Dong, Z.; Wang, Y.T. Food allergy induces alteration in brain inflammatory status and cognitive impairments. Behav. Brain Res. 2019, 364, 374–382. [Google Scholar] [CrossRef]
  203. Msallam, R.; Balla, J.; Rathore, A.P.S.; Kared, H.; Malleret, B.; Saron, W.A.A.; Liu, Z.; Hang, J.W.; Dutertre, C.A.; Larbi, A.; et al. Fetal mast cells mediate postnatal allergic responses dependent on maternal IgE. Science 2020, 370, 941–950. [Google Scholar] [CrossRef]
  204. Theoharides, T.C. Ways to Address Perinatal Mast Cell Activation and Focal Brain Inflammation, including Response to SARS-CoV-2, in Autism Spectrum Disorder. J. Pers. Med. 2021, 11, 860. [Google Scholar] [CrossRef] [PubMed]
  205. Menage, P.; Thibault, G.; Martineau, J.; Herault, J.; Muh, J.P.; Barthelemy, C.; Lelord, G.; Bardos, P. An IgE mechanism in autistic hypersensitivity? Biol. Psychiatry 1992, 31, 210–212. [Google Scholar] [CrossRef] [PubMed]
  206. Hranilovic, D.; Bujas-Petkovic, Z.; Vragovic, R.; Vuk, T.; Hock, K.; Jernej, B. Hyperserotonemia in adults with autistic disorder. J. Autism Dev. Disord. 2007, 37, 1934–1940. [Google Scholar] [CrossRef]
  207. Akhondzadeh, S.; Erfani, S.; Mohammadi, M.R.; Tehrani-Doost, M.; Amini, H.; Gudarzi, S.S.; Yasamy, M.T. Cyproheptadine in the treatment of autistic disorder: A double-blind placebo-controlled trial. J. Clin. Pharm. Ther. 2004, 29, 145–150. [Google Scholar] [CrossRef] [PubMed]
  208. Bachelet, I.; Levi-Schaffer, F. Mast cells as effector cells: A co-stimulating question. Trends Immunol. 2007, 28, 360–365. [Google Scholar] [CrossRef]
  209. Malbec, O.; Daeron, M. The mast cell IgG receptors and their roles in tissue inflammation. Immunol. Rev. 2007, 217, 206–221. [Google Scholar] [CrossRef]
  210. Theoharides, T.C.; Letourneau, R.; Patra, P.; Hesse, L.; Pang, X.; Boucher, W.; Mompoint, C.; Harrington, B. Stress-induced rat intestinal mast cell intragranular activation and inhibitory effect of sulfated proteoglycans. Dig. Dis. Sci. 1999, 44, 87S–93S. [Google Scholar]
  211. Theoharides, T.C.; Patra, P.; Boucher, W.; Letourneau, R.; Kempuraj, D.; Chiang, G.; Jeudy, S.; Hesse, L.; Athanasiou, A. Chondroitin sulphate inhibits connective tissue mast cells. Br. J. Pharmacol. 2000, 131, 1039–1049. [Google Scholar] [CrossRef]
  212. Theoharides, T.C.; Kalogeromitros, D. The critical role of mast cells in allergy and inflammation. Ann. N. Y. Acad. Sci. 2006, 1088, 78–99. [Google Scholar] [CrossRef]
  213. Middleton, E., Jr.; Kandaswami, C.; Theoharides, T.C. The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease, and cancer. Pharmacol. Rev. 2000, 52, 673–751. [Google Scholar]
  214. Oka, T.; Kalesnikoff, J.; Starkl, P.; Tsai, M.; Galli, S.J. Evidence questioning cromolyn’s effectiveness and selectivity as a ‘mast cell stabilizer’ in mice. Lab. Investig. 2012, 92, 1472–1482. [Google Scholar] [CrossRef]
  215. Weng, Z.; Zhang, B.; Asadi, S.; Sismanopoulos, N.; Butcher, A.; Fu, X.; Katsarou-Katsari, A.; Antoniou, C.; Theoharides, T.C. Quercetin is more effective than cromolyn in blocking human mast cell cytokine release and inhibits contact dermatitis and photosensitivity in humans. PLoS ONE 2012, 7, e33805. [Google Scholar] [CrossRef] [PubMed]
  216. Vieira Dos Santos, R.; Magerl, M.; Martus, P.; Zuberbier, T.; Church, M.K.; Escribano, L.; Maurer, M. Topical sodium cromoglicate relieves allergen- and histamine-induced dermal pruritus. Br. J. Dermatol. 2010, 162, 674–676. [Google Scholar] [CrossRef] [PubMed]
  217. Weng, Z.; Patel, A.B.; Panagiotidou, S.; Theoharides, T.C. The novel flavone tetramethoxyluteolin is a potent inhibitor of human mast cells. J. Allergy Clin. Immunol. 2015, 135, 1044–1052.e1045. [Google Scholar] [CrossRef] [PubMed]
  218. Church, M.K.; Gradidge, C.F. Potentiation of histamine release by sodium cromoglycate. Life Sci. 1978, 23, 1899–1903. [Google Scholar] [CrossRef]
  219. Kempuraj, D.; Madhappan, B.; Christodoulou, S.; Boucher, W.; Cao, J.; Papadopoulou, N.; Cetrulo, C.L.; Theoharides, T.C. Flavonols inhibit proinflammatory mediator release, intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast cells. Br. J. Pharmacol. 2005, 145, 934–944. [Google Scholar] [CrossRef]
  220. Gravanis, A.; Margioris, A.N. The corticotropin-releasing factor (CRF) family of neuropeptides in inflammation: Potential therapeutic applications. Curr. Med. Chem. 2005, 12, 1503–1512. [Google Scholar] [CrossRef]
  221. Martinez, V.; Tache, Y. CRF1 receptors as a therapeutic target for irritable bowel syndrome. Curr. Pharm. Des. 2006, 12, 4071–4088. [Google Scholar] [CrossRef]
  222. Zhou, H.; Luo, Y.; Huang, S. Updates of mTOR inhibitors. Anticancer. Agents Med. Chem. 2010, 10, 571–581. [Google Scholar] [CrossRef]
  223. Ehninger, D.; Silva, A.J. Rapamycin for treating Tuberous sclerosis and Autism spectrum disorders. Trends Mol. Med. 2011, 17, 78–87. [Google Scholar] [CrossRef]
  224. Sahin, M. Targeted treatment trials for tuberous sclerosis and autism: No longer a dream. Curr. Opin. Neurobiol. 2012, 22, 895–901. [Google Scholar] [CrossRef] [PubMed]
  225. de Vries, P.J. Targeted treatments for cognitive and neurodevelopmental disorders in tuberous sclerosis complex. Neurotherapeutics 2010, 7, 275–282. [Google Scholar] [CrossRef] [PubMed]
  226. Hampson, D.R.; Gholizadeh, S.; Pacey, L.K. Pathways to drug development for autism spectrum disorders. Clin. Pharmacol. Ther. 2012, 91, 189–200. [Google Scholar] [CrossRef]
  227. Van Aller, G.S.; Carson, J.D.; Tang, W.; Peng, H.; Zhao, L.; Copeland, R.A.; Tummino, P.J.; Luo, L. Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem. Biophys. Res. Commun. 2011, 406, 194–199. [Google Scholar] [CrossRef] [PubMed]
  228. Kimata, M.; Shichijo, M.; Miura, T.; Serizawa, I.; Inagaki, N.; Nagai, H. Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin. Exp. Allergy 2000, 30, 501–508. [Google Scholar] [CrossRef] [PubMed]
  229. Asadi, S.; Zhang, B.; Weng, Z.; Angelidou, A.; Kempuraj, D.; Alysandratos, K.D.; Theoharides, T.C. Luteolin and thiosalicylate inhibit HgCl(2) and thimerosal-induced VEGF release from human mast cells. Int. J. Immunopathol. Pharmacol. 2010, 23, 1015–1020. [Google Scholar] [CrossRef]
  230. Dirscherl, K.; Karlstetter, M.; Ebert, S.; Kraus, D.; Hlawatsch, J.; Walczak, Y.; Moehle, C.; Fuchshofer, R.; Langmann, T. Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype. J. Neuroinflamm. 2010, 7, 3. [Google Scholar] [CrossRef]
  231. Jang, S.; Dilger, R.N.; Johnson, R.W. Luteolin inhibits microglia and alters hippocampal-dependent spatial working memory in aged mice. J. Nutr. 2010, 140, 1892–1898. [Google Scholar] [CrossRef]
  232. Kao, T.K.; Ou, Y.C.; Lin, S.Y.; Pan, H.C.; Song, P.J.; Raung, S.L.; Lai, C.Y.; Liao, S.L.; Lu, H.C.; Chen, C.J. Luteolin inhibits cytokine expression in endotoxin/cytokine-stimulated microglia. J. Nutr. Biochem. 2011, 22, 612–624. [Google Scholar] [CrossRef] [PubMed]
  233. Kempuraj, D.; Tagen, M.; Iliopoulou, B.P.; Clemons, A.; Vasiadi, M.; Boucher, W.; House, M.; Wolfberg, A.; Theoharides, T.C. Luteolin inhibits myelin basic protein-induced human mast cell activation and mast cell-dependent stimulation of Jurkat T cells. Br. J. Pharmacol. 2008, 155, 1076–1084. [Google Scholar] [CrossRef]
  234. Verbeek, R.; Plomp, A.C.; van Tol, E.A.; van Noort, J.M. The flavones luteolin and apigenin inhibit in vitro antigen-specific proliferation and interferon-gamma production by murine and human autoimmune T cells. Biochem. Pharmacol. 2004, 68, 621–629. [Google Scholar] [CrossRef] [PubMed]
  235. Franco, J.L.; Posser, T.; Missau, F.; Pizzolatti, M.G.; Dos Santos, A.R.; Souza, D.O.; Aschner, M.; Rocha, J.B.; Dafre, A.L.; Farina, M. Structure-activity relationship of flavonoids derived from medicinal plants in preventing methylmercury-induced mitochondrial dysfunction. Environ. Toxicol. Pharmacol. 2010, 30, 272–278. [Google Scholar] [CrossRef] [PubMed]
  236. Chen, H.Q.; Jin, Z.Y.; Wang, X.J.; Xu, X.M.; Deng, L.; Zhao, J.W. Luteolin protects dopaminergic neurons from inflammation-induced injury through inhibition of microglial activation. Neurosci. Lett. 2008, 448, 175–179. [Google Scholar] [CrossRef] [PubMed]
  237. Mehterov, N.; Minchev, D.; Gevezova, M.; Sarafian, V.; Maes, M. Interactions Among Brain-Derived Neurotrophic Factor and Neuroimmune Pathways Are Key Components of the Major Psychiatric Disorders. Mol. Neurobiol. 2022, 59, 4926–4952. [Google Scholar] [CrossRef] [PubMed]
  238. Sadakata, T.; Shinoda, Y.; Oka, M.; Sekine, Y.; Sato, Y.; Saruta, C.; Miwa, H.; Tanaka, M.; Itohara, S.; Furuichi, T. Reduced axonal localization of a Caps2 splice variant impairs axonal release of BDNF and causes autistic-like behavior in mice. Proc. Natl. Acad. Sci. USA 2012, 109, 21104–21109. [Google Scholar] [CrossRef] [PubMed]
  239. Xu, L.; Su, W.; Jin, J.; Chen, J.; Li, X.; Zhang, X.; Sun, M.; Sun, S.; Fan, P.; An, D.; et al. Identification of luteolin as enterovirus 71 and coxsackievirus A16 inhibitors through reporter viruses and cell viability-based screening. Viruses 2014, 6, 2778–2795. [Google Scholar] [CrossRef]
  240. Fan, W.; Qian, S.; Qian, P.; Li, X. Antiviral activity of luteolin against Japanese encephalitis virus. Virus Res. 2016, 220, 112–116. [Google Scholar] [CrossRef]
  241. Yan, H.; Ma, L.; Wang, H.; Wu, S.; Huang, H.; Gu, Z.; Jiang, J.; Li, Y. Luteolin decreases the yield of influenza A virus in vitro by interfering with the coat protein I complex expression. J. Nat. Med. 2019, 73, 487–496. [Google Scholar] [CrossRef]
  242. Marshall, J.S.; Portales-Cervantes, L.; Leong, E. Mast Cell Responses to Viruses and Pathogen Products. Int. J. Mol. Sci. 2019, 20, 4241. [Google Scholar] [CrossRef]
  243. Russo, M.; Moccia, S.; Spagnuolo, C.; Tedesco, I.; Russo, G.L. Roles of flavonoids against coronavirus infection. Chem. Biol. Interact. 2020, 328, 109211. [Google Scholar] [CrossRef]
  244. Derosa, G.; Maffioli, P.; D’Angelo, A.; Di Pierro, F. A role for quercetin in coronavirus disease 2019 (COVID-19). Phytother. Res. 2021, 35, 1230–1236. [Google Scholar] [CrossRef] [PubMed]
  245. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Shytle, R.D. Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J. Neuroinflamm. 2008, 5, 41. [Google Scholar] [CrossRef]
  246. Jang, S.; Kelley, K.W.; Johnson, R.W. Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc. Natl. Acad. Sci. USA 2008, 105, 7534–7539. [Google Scholar] [CrossRef]
  247. Burton, M.D.; Rytych, J.L.; Amin, R.; Johnson, R.W. Dietary Luteolin Reduces Proinflammatory Microglia in the Brain of Senescent Mice. Rejuvenation Res. 2016, 19, 286–292. [Google Scholar] [CrossRef] [PubMed]
  248. Dajas, F.; Rivera-Megret, F.; Blasina, F.; Arredondo, F.; Abin-Carriquiry, J.A.; Costa, G.; Echeverry, C.; Lafon, L.; Heizen, H.; Ferreira, M.; et al. Neuroprotection by flavonoids. Braz. J. Med. Biol. Res. 2003, 36, 1613–1620. [Google Scholar] [CrossRef] [PubMed]
  249. Kempuraj, D.; Thangavel, R.; Kempuraj, D.D.; Ahmed, M.E.; Selvakumar, G.P.; Raikwar, S.P.; Zaheer, S.A.; Iyer, S.S.; Govindarajan, R.; Chandrasekaran, P.N.; et al. Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. Biofactors 2021, 47, 190–197. [Google Scholar] [CrossRef]
  250. Lin, T.Y.; Lu, C.W.; Wang, S.J. Luteolin protects the hippocampus against neuron impairments induced by kainic acid in rats. Neurotoxicology 2016, 55, 48–57. [Google Scholar] [CrossRef]
  251. Ashaari, Z.; Hadjzadeh, M.A.; Hassanzadeh, G.; Alizamir, T.; Yousefi, B.; Keshavarzi, Z.; Mokhtari, T. The Flavone Luteolin Improves Central Nervous System Disorders by Different Mechanisms: A Review. J. Mol. Neurosci. 2018, 65, 491–506. [Google Scholar] [CrossRef]
  252. Bernatoniene, J.; Kazlauskaite, J.A.; Kopustinskiene, D.M. Pleiotropic Effects of Isoflavones in Inflammation and Chronic Degenerative Diseases. Int. J. Mol. Sci. 2021, 22, 5656. [Google Scholar] [CrossRef]
  253. Theoharides, T.C.; Conti, P.; Economu, M. Brain inflammation, neuropsychiatric disorders, and immunoendocrine effects of luteolin. J. Clin. Psychopharmacol. 2014, 34, 187–189. [Google Scholar] [CrossRef]
  254. Silva Dos Santos, J.; Goncalves Cirino, J.P.; de Oliveira Carvalho, P.; Ortega, M.M. The Pharmacological Action of Kaempferol in Central Nervous System Diseases: A Review. Front. Pharmacol. 2020, 11, 565700. [Google Scholar] [CrossRef] [PubMed]
  255. Yao, Z.H.; Yao, X.L.; Zhang, Y.; Zhang, S.F.; Hu, J.C. Luteolin Could Improve Cognitive Dysfunction by Inhibiting Neuroinflammation. Neurochem. Res. 2018, 43, 806–820. [Google Scholar] [CrossRef] [PubMed]
  256. Devi, S.A.; Chamoli, A. Polyphenols as an Effective Therapeutic Intervention Against Cognitive Decline During Normal and Pathological Brain Aging. Adv. Exp. Med. Biol. 2020, 1260, 159–174. [Google Scholar] [CrossRef] [PubMed]
  257. Rezai-Zadeh, K.; Douglas Shytle, R.; Bai, Y.; Tian, J.; Hou, H.; Mori, T.; Zeng, J.; Obregon, D.; Town, T.; Tan, J. Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer’s disease beta-amyloid production. J. Cell Mol. Med. 2009, 13, 574–588. [Google Scholar] [CrossRef] [PubMed]
  258. Theoharides, T.C.; Cholevas, C.; Polyzoidis, K.; Politis, A. Long-COVID syndrome-associated brain fog and chemofog: Luteolin to the rescue. Biofactors 2021, 47, 232–241. [Google Scholar] [CrossRef] [PubMed]
  259. Gevezova, M.; Sarafian, V.; Anderson, G.; Maes, M. Inflammation and Mitochondrial Dysfunction in Autism Spectrum Disorder. CNS Neurol. Disord. Drug Targets 2020, 19, 320–333. [Google Scholar] [CrossRef]
  260. Gevezova, M.; Sbirkov, Y.; Sarafian, V.; Plaimas, K.; Suratanee, A.; Maes, M. Autistic spectrum disorder (ASD)—Gene, molecular and pathway signatures linking systemic inflammation, mitochondrial dysfunction, transsynaptic signalling, and neurodevelopment. Brain Behav. Immun. Health 2023, 30, 100646. [Google Scholar] [CrossRef] [PubMed]
  261. Gevezova, M.; Minchev, D.; Pacheva, I.; Todorova, T.; Yordanova, R.; Timova, E.; Ivanov, I.; Sarafian, V. Association of NGF and Mitochondrial Respiration with Autism Spectrum Disorder. Int. J. Mol. Sci. 2022, 23, 11917. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of ASD pathogenesis from the perspective of MCs. Legend: CRH—Corticotropin-Releasing Hormone; NT—Neurotensin; ROS—Reactive Oxygen Species; NO—Nitric Oxide; IL—Interleukins; TNF-α—Tumor Necrosis Factor alpha; VEGF—Vascular Endothelial Growth Factor; IGF—Insulin-like Growth Factor; PDGF—Platelet-derived growth factor.
Figure 1. Schematic illustration of ASD pathogenesis from the perspective of MCs. Legend: CRH—Corticotropin-Releasing Hormone; NT—Neurotensin; ROS—Reactive Oxygen Species; NO—Nitric Oxide; IL—Interleukins; TNF-α—Tumor Necrosis Factor alpha; VEGF—Vascular Endothelial Growth Factor; IGF—Insulin-like Growth Factor; PDGF—Platelet-derived growth factor.
Ijms 25 02651 g001
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

Kovacheva, E.; Gevezova, M.; Maes, M.; Sarafian, V. Mast Cells in Autism Spectrum Disorder—The Enigma to Be Solved? Int. J. Mol. Sci. 2024, 25, 2651. https://doi.org/10.3390/ijms25052651

AMA Style

Kovacheva E, Gevezova M, Maes M, Sarafian V. Mast Cells in Autism Spectrum Disorder—The Enigma to Be Solved? International Journal of Molecular Sciences. 2024; 25(5):2651. https://doi.org/10.3390/ijms25052651

Chicago/Turabian Style

Kovacheva, Eleonora, Maria Gevezova, Michael Maes, and Victoria Sarafian. 2024. "Mast Cells in Autism Spectrum Disorder—The Enigma to Be Solved?" International Journal of Molecular Sciences 25, no. 5: 2651. https://doi.org/10.3390/ijms25052651

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