Next Article in Journal
Internal Ophthalmoplegic Migraine During Pregnancy: A Clinical Case
Next Article in Special Issue
Therapeutic Role of Heterocyclic Compounds in Neurodegenerative Diseases: Insights from Alzheimer’s and Parkinson’s Diseases
Previous Article in Journal
Management of Post-Traumatic Pseudomeningocele as Consequence of Root Nerve Avulsion: Case Report and Review of the Literature
Previous Article in Special Issue
Survival After Shunt Therapy in Normal-Pressure Hydrocephalus: A Meta-Analysis of 1614 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut–Brain Axis and Helicobacter pylori Infection

by
Marina Boziki
1,*,
Paschalis Theotokis
1,
Evangelia Kesidou
1,
Maria Nella
1,
Christos Bakirtzis
1,
Eleni Karafoulidou
1,
Maria Tzitiridou-Chatzopoulou
2,3,
Michael Doulberis
2,4,5,
Evangelos Kazakos
2,
Georgia Deretzi
2,6,
Nikolaos Grigoriadis
1 and
Jannis Kountouras
2,*
1
Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
2
Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
3
Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
4
Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
5
Division of Gastroenterology and Hepatology, Medical University Department, 5001 Aarau, Switzerland
6
Department of Neurology, Papageorgiou General Hospital, 54629 Thessaloniki, Greece
*
Authors to whom correspondence should be addressed.
Neurol. Int. 2024, 16(6), 1750-1778; https://doi.org/10.3390/neurolint16060127
Submission received: 20 August 2024 / Revised: 8 November 2024 / Accepted: 3 December 2024 / Published: 6 December 2024
(This article belongs to the Collection Advances in Neurodegenerative Diseases)

Abstract

:
Background: The innate immune response aims to prevent pathogens from entering the organism and/or to facilitate pathogen clearance. Innate immune cells, such as macrophages, mast cells (MCs), natural killer cells and neutrophils, bear pattern recognition receptors and are thus able to recognize common molecular patterns, such as pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), the later occurring in the context of neuroinflammation. An inflammatory component in the pathology of otherwise “primary cerebrovascular and neurodegenerative” disease has recently been recognized and targeted as a means of therapeutic intervention. Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. Methods: This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and multiple sclerosis. Results: In view of recent evidence that the gut–brain axis may be implicated in the pathogenesis of neurodegenerative diseases and the characterization of the neuroinflammatory component in the pathology of these diseases, this review also focuses on MCs as potential mediators in the gut–brain axis bi-directional communication and the possible role of Helicobacter pylori, a gastric pathogen known to alter the gut–brain axis homeostasis towards local and systemic pro-inflammatory responses. Conclusion: As MCs and Helicobacter pylori infection may offer targets of intervention with potential therapeutic implications for neurodegenerative disease, more clinical and translational evidence is needed to elucidate this field.

1. Introduction

Neurodegenerative diseases are a heterogenous group of disorders characterized by irreversible neuron loss and the accumulation of progressive neurological disability. What is more, therapeutic approaches utilizing agents with a potential for neuroprotection have, to the most part, failed to tackle disease mechanisms and to ameliorate disease evolution. Although neurodegenerative diseases exhibit heterogeneity regarding clinical presentation, genetic predisposing and/or causative factors, as well as aspects of the underlying pathology, they also share several common characteristics. Cognitive decline, for instance, characterizes Alzheimer’s disease (AD), dementia with Lewy bodies (LBD), Parkinson’s disease (PD) and Huntington’s disease (HD). Motor deficits are prominent in amyotrophic lateral sclerosis (ALS), PD, HD and multiple sclerosis (MS) [1]. Moreover, aging is a major risk factor common in all neurodegenerative diseases [2]. In terms of pathology, neurodegenerative diseases are characterized by the central nervous system (CNS) aggregation of proteins, such as the aggregation of amyloid beta (Aβ) and hyperphosphorylated tau in AD, α-synuclein in PD and in LBD, TAR DNA-binding protein (TDP)-43 in ALS and fronto-temporal dementia (FTD) and Cytosine – Adenine - Guanine (CAG)-polyglutamine protein in HD [3]. The composition of these aggregates is genetically determined, especially in familial cases [4]. Overall, in the context of neurodegenerative disease, the aggregation of these misfolded proteins leads to the dysregulation of several pathways that primarily affect neurons, such as mitochondrial damage, energy defect, production of reactive oxygen species and oxidative stress, synaptic toxicity and, eventually, synapse and neuron loss. The gut–brain axis (GBA) is thought to affect neurodegenerative diseases by several pathways, such as alterations in the immune system via the influence of the gut microbiota, the vagus nerve and the enteric nervous system (ENS), the neuroendocrine system, as well as direct metabolic pathways and hormones [5]. Of note, alterations in the composition of gut commensal microbiota, referred to as “gut dysbiosis” have been reported in patients with AD, PD, ALS and MS, to name few of the neurodegenerative diseases [6,7,8]. Experimental studies also indicate pathways by which the GBA affects the pathogenesis of neurodegenerative diseases, such as CNS innate immune system alterations, microglial activation and propagation of neuroinflammatory responses [9,10].
Recently, a large body of evidence supports neuroinflammation as a crucial component of the pathology that underlies the otherwise regarded as “primary neurodegenerative disease such as AD, PD, ALS and HD. Conversely, for MS, traditionally regarded as a disease of neuroinflammatory origin, with secondary neurodegeneration occurring later in the disease’s course, it is nowadays widely acknowledged that neurodegeneration is also present at the initial disease stages, to a lesser degree, compared to the later stages. These observations suggest that neuroinflammation and neurodegeneration are both crucial players in these pathological processes. Moreover, due to the lack of successful treatments for neurodegenerative diseases so far, modulation of the immune responses involved in neurodegeneration appears to be a promising therapeutic strategy, further supported by evidence stemming from experimental and clinical studies. Several of these approaches attempt to modulate signaling pathways and cellular components of the innate immune system. Mast cells (MCs) are multifunctional effector cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. When MCs are activated, depending on either physiological and/or pathological conditions, they degranulate and release several mediating molecules. For instance, MCs participate in the regulation of vasodilation and the overall vascular homeostasis, and they are bearing potential to trigger responses of the innate and adaptive immune system. Moreover, MCs are cellular mediators of several pathologies, such as allergic reactions, asthma, gastrointestinal (GI) diseases, cancer and cardiovascular disease (CVD). MCs are normally located in mucosal and epithelial tissues, as well as in other vascularized tissues, with the possible exception of the CNS and the retina [11]; in normal human brains, MCs are not numerous [12]. MCs are cellular components of the innate immune system and, as such, they are abundant at sites of the body where the host and the external environment interact, as well as at sites that often serve as entry points for antigens (the GI tract, the skin and/or the respiratory epithelium) [13]. Human MCs exhibit two phenotypes: the mucosal MCs, producing tryptase, and the connective tissue MCs, producing additional mediators, such as chymase and carboxypeptidases [14,15]. In this respect, MCs are a component of the innate immune system that may bear potential as a possible target of therapeutic intervention for disease. This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, AD, PD, ALS, HD and MS. In view of recent evidence that the GBA may be implicated the pathogenesis of neurodegenerative diseases, this review also focuses on the MCs as potential mediators in the GBA bi-directional communication and the possible role of Helicobacter pylori (Hp), a gastric pathogen known to alter the GBA homeostasis towards local and systemic pro-inflammatory responses. As MCs and Hp infection may offer targets of intervention with potential therapeutic implications for neurodegenerative diseases, more clinical and translational evidence is needed to elucidate in depth this field.

2. Methodology

Regarding the strategy of sources and methods used, an online literature search was conducted in PubMed, EMBASE and Web of science databases, without time restrictions. Search phrases included the keywords or MESH (mast cells and central nervous system or neurodegenerative disease or neuroinflammation or intestine or immune system or Alzheimer’s disease or Parkinson’s disease or Amyotrophic Lateral Sclerosis or Huntington’s disease or Multiple Sclerosis, mast cells and intestine or gut–brain axis or Helicobacter pylori). Research and clinical studies in humans, animals, tissues and cell cultures were included. Original articles, patient series and reviews, systematic and meta-analyses and book chapters were retrieved. The literature list of the reviews was manually searched and additional studies that met the selection criteria of this review were selected. With respect to the study selection criteria, although no limitation regarding the year of publication was used in the search, studies and articles from the last five years were preferred, with a particular focus on clinical and laboratory experimental studies. However, several older studies were also included, for reasons of completion. Articles written or translated into English were included. Reviews were also considered appropriate for inclusion as sources for cited studies that were not retrieved in the initial literature search with the respective keywords.

3. Neuroinflammation and Neurodegenerative Diseases

Inflammatory response may be either acute, developing from seconds to hours and frequently acting as a beneficial mechanism, or chronic, with a longer duration that may cause tissue destruction. Chronic inflammation develops either due to the failing of the several inflammatory response’s regulatory mechanisms, or in the presence of over-activation of the immune system [16,17]. The innate immune response aims at preventing pathogens from entering the organism and/or to facilitate pathogen clearance via a non-specific mechanism in terms of antigen recognition. Non-specific defense mechanisms towards pathogens include physical and chemical barriers. Innate immune cells, such as macrophages, MCs, natural killer (NK) cells and neutrophils bear pattern recognition receptors (PRRs) and are thus able to recognize molecular patterns, common across a wide variety of pathogens, namely, pathogen-associated molecular patterns (PAMPs), as well as molecules derived from tissue destruction, namely, damage-associated molecular patterns (DAMPs) as it occurs in the context of so-called neuroinflammation [18].
Inflammation in the nervous system, termed as aforementioned “neuroinflammation”, can be particularly detrimental [17]. Microglia are resident cells of the innate immune system of the CNS. Upon chronic inflammation microglia secretes pro-inflammatory cytokines that exert tissue detrimental effect, whereas, upon the initial immune response, its activation is considered mostly beneficial [17,19,20,21,22]. Microglia, scavenger cells of the CNS, MCs and astrocytes comprise the main sources in the CNS milieu of pro-inflammatory cytokines [17,23]. Chronic disorders, such as cerebrovascular disease, type 2 diabetes mellitus (T2DM) and rheumatoid arthritis, are subjected to the effect of systemic inflammation [19]. Moreover, the Western diet and overall lifestyle is linked to obesity, insulin resistance (IR), hypercholesterolemia and metabolic (dysfunction) associated steatotic disease (MASLD), [24,25,26] contributing factors to metabolic syndrome (MetS), a common denominator for several CNS pathologies, such as cerebrovascular and neurodegenerative diseases [27]. An inflammatory component in the pathology of otherwise “primary cerebrovascular and neurodegenerative” disease has recently been recognized and targeted as a means of therapeutic intervention [28].

4. Mast Cells (MCs)

Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens [29,30]. MCs differentiate in the bone marrow in the presence of c-kit ligand and interleukin 3 (IL-3) [31] and are subsequently inserted in the blood circulation [29,30]. Their maturation occurs following to their migration to various tissues and organs in the presence of the local tissue microenvironment [32,33]. MCs are prevalent in host/environment interfaces such as the skin, lung tissue, GI and urogenital tracts, as well as the connective tissue adjacent to blood and lymphatic vessels. Conversely, they are absent from avascular tissues (cornea, mineralized bone and cartilage) [11]. Furthermore, MCs circulate from the periphery towards the CNS upon neonatal development and in the adult [34,35,36]. MCs are also found in small numbers in the healthy brain, in perivascular areas in the meninges [37] and other parts of the CNS (i.e., dorsal medulla, choroid plexus, thalami and hypothalamus) [38,39]. Upon injury, illness or stress, additional MCs are recruited in the CNS [40,41,42]. In the spinal cord, MCs are mainly distributed in the dura, where they secrete immune mediating molecules that regulate synaptic transmission in the proximal CNS regions [43,44]. MCs are also prevalent in several tissues, in close proximity to peripheral nerves [45,46,47,48].
MCs are thought to be implicated in physiological homeostatic processes and immune defense mechanisms with immediate and/or delayed effects [49,50]. Specifically, MCs can rapidly, within minutes, secrete cytokines, leukotrienes and other inflammatory mediators in response to allergens, antigens, complement factors, neuropeptides, medications, and trauma [29,34]. Moreover, MCs bear the ability to interact with surrounding cells and structures, such as arteries and nerve fibers, via the secretion of extracellular vesicles, traps and nanotubes [46,51]. MCs in the CNS exhibit the potential of antigen presentation, pro-inflammatory molecule release and phagocytosis. Lastly, the degranulation of MCs has been shown to increase neuronal excitotoxicity [52] and to enhance the homeostatic responses to trauma and stress [53,54].

5. Mast Cells (MCs) and the Gut–Brain Axis (GBA)

The GBA is a complex network that describes the bilateral interaction between the CNS and the gut, including the ENS, through various mechanisms that consist of nervous, hormonal, metabolic and immunological signaling [55]. Consequently, the GBA plays a pivotal role in regulating multiple functions such as the CNS function, emotional regulation, neuromuscular regulation and immunity. Current research increasingly focuses on unveiling the connection between CNS higher functions and GI processes, including gut microbiota symbiosis [55,56]. Because of the established function of the gut microbiome in the maintenance of a physiological brain–gut interaction and its participation in the pathogenesis of various disorders, this relationship has recently been termed the “microbiota—gut—brain axis” (Figure 1) [57].
Gut microbiota consists of more than 100 trillion microorganisms that inhabit the human digestive tract [58]; their number is approximately 10 times that of total human cells, with above 1000 diverse species [59]. Not only do the gut microbiota serve as a natural protective barrier against pathogens, but they also participate in the regulation of the neurotransmission and in neuromodulation pathways via the regulation, at least in part, of the biosynthesis as well as the production of serotonin, aminobutyric acid, dopamine and short chain fatty acids (SCFAs) [60]. The intestinal epithelial barrier is a key player in maintaining tissue homeostasis through immune responses which are coordinated via internal and external signals. MCs are abundant in the intestinal mucosa, where they pose both defense- and immune-regulatory role. MC dysregulation may disrupt GI and gut microbiota homeostasis, which induces pro-inflammatory conditions and/or autoimmunity, locally in the intestinal mucosa [61]. Increasing evidence recently established the critical importance of the interplay among gut microbiota, CNS and MCs in neurodegenerative disorders [62]. The rising incidence of neurodegenerative disorders may be promoted through CNS–MCs triggering leading to blood–brain barrier (BBB) disruption, neuro-immune activation and synaptic dysfunction. In the case of neuroinflammatory conditions, chemokines and other immune mediators are produced and migrate through the GBA causing intestinal MCs degranulation which subsequently leads to gut-barrier breakdown.

6. Helicobacter pylori and Neurodegenerative Diseases

Helicobacter pylori (Hp) is a Gram-negative, microaerophilic spiral-shaped and flagellated [63] bacterium, prevalent in about 58% of the world’s population [64], being implicated in the pathogenesis of duodenal and gastric ulcer disease, as well as of gastric malignancies [65]. The main natural permanent reservoirs for Hp include the gastric epithelium and the oral cavity [66,67,68]. The gut appears to be a secondary location for Hp [69,70]. When left without treatment, Hp may cause a chronic infection and histological inflammation (gastritis) that can endure for many years, if not throughout lifetime. Hp is one of the most widespread human infectious agents [71,72,73], and affects above 4.4 billion individuals worldwide [74]. Hp infection is facilitated by genetic diversity, as well as the effective evasion of the host innate and adaptive immune response. Hp genetic diversity results from a high mutation rate, which is aided by the absence of a DNA mismatch repair mechanism and the acquisition of DNA from other strains [75]. By eliminating phosphate groups from the lipid A, the Hp lipopolysaccharide evades recognition by antimicrobial peptides and Toll-like receptors (TLRs) [76]. Vacuolating cytotoxin A (VacA), which is secreted by Hp, inhibits nuclear translocation of the nuclear factor of activated T-cells (NF-AT), which regulates T cell proliferation [77]. Beyond its impact on the GI tract, many investigations have implicated Hp infection in a variety of diseases, including neurological and neurodegenerative disease [65,78,79,80,81].

6.1. Alzheimer’s Disease (AD)

A link between Hp infection and AD has previously been described [82,83,84,85,86]. We previously [87] hypothesized that Hp acts through immune-mediated mechanisms according to which secreted interleukins, interferons and other inflammatory mediators orchestrate the effect of the acute and chronic phase of the inflammatory response towards Hp. In this respect, mononuclear cells may be stimulated to create a tissue factor-like procoagulant which converts fibrinogen to fibrin. A detailed review of the current findings on humoral and cellular immune dysregulation elucidated a potential relationship between Hp-infection and neurodegenerative conditions, including AD [85,87]. We and others also proposed that cognitive impairment may be precipitated by Hp infection in the context of anti-Hp IgG that targets the brain milieu [87,88]. Furthermore, experimental data indicate the abundance of IL-1 pro-inflammatory cytokine, secreted by microglia and astrocytes, in amyloid plaques, as a common trait in human AD brains as well as in animal models of AD [89,90,91]. IL-1 synthesis is based on MAPK activation and the NF-κB signaling cascade and overexpressed IL-1 enhances phosphorylation of tau protein and the development of neurofibrillary tangles due to GSK-3 activation [92]. The B subunit of Hp-Urease (HPU) activates the NOD-like receptor protein 3 (NLRP3) inflammasome, a mechanism upstream of the IL-1 production, by a TLR2-dependent process [93,94].
Hp infection appears to be involved in AD pathophysiology by several additional mechanisms. For instance, Hp infection may be associated with the brain pericyte dysfunction [95], which could contribute to the pathophysiology of neurodegenerative disorders, such as AD [96]. Likewise, Hp infection-related hyperhomocysteinemia appears to contribute to atherosclerosis, which is connected with systemic disorders, including cardio-cerebrovascular and neurodegenerative diseases, such as AD [86]. Hyperhomocysteinemia is an active player implicated in the pathophysiology of mild cognitive impairment, a prodromal condition strongly predictive of subsequent AD development [80], and AD sequence by provoking Aβ and tau pathologies, along with synaptic dysfunction, neuroinflammatory process and memory decline. This suggests a new therapeutic strategy for patients exhibiting this risk factor [97]. Related studies provide evidence of the connection between Hp infection and AD-like Aβ and phospho-tau pathology, signifying that Hp eradication may offer benefits in the prevention of tauopathy [98]. Moreover, Hp infection is associated with atrial fibrillation (AF), Hp is an independent predictor for long-lasting AF [99], and AF is closely linked to AD and cognitive decline [100]. Beyond catheter ablation for AF management [100], Hp eradication may also reduce the risk of AF-related AD, and further investigation is required. Additionally, Hp is related to galectin-3, which is significantly associated with the severity of memory loss and AD stage [84]. Galectin-3 inhibitors appear to suppress microglial activation [101], suggesting an exciting therapeutic target to impede neurodegenerative disorders, including AD. Furthermore, Hp, by promoting gut dysbiosis, might lead to the brain disorders such as AD [102]. Gut dysbiosis plays an important role in the pathophysiology of AD, by influencing neuroinflammation and disease progression. Related therapeutic approaches, including probiotics, prebiotics and fecal microbiota transplantation, offer benefits in AD pathology [103]. Hp eradication may also positively influence AD manifestations at 2- and 5-year clinical endpoints, thereby contributing to the long-term survival rate of this disorder [104].

6.2. Parkinson’s Disease (PD)

Increasing evidence indicates higher prevalence of Hp infection in people with PD, compared to the general population, though, the exact cause for this association remains to be elucidated. Hp toxins, gut dysbiosis [105], excess of pro-inflammatory molecules [106,107], Hp antigens and molecular mimicry towards the host’s neuronal proteins [108] as well as a possible effect of Hp on levodopa pharmacokinetics [109], are all advocated as possible pathogenic mechanisms. Moreover, the emergence of motor symptoms in the context of PD may be precipitated by mechanisms acting on the bidirectional micobiome—GBA [110,111]. For instance, patients with PD exhibit lower levels of gut beneficial fecal of SCFA, as well as lower relative abundance of SCFA-producing bacteria, compared to healthy controls [112]. Lack of bacteria-produced SCFA, namely, butyrate, has also been implicated in the progression of PD [113]. Ιn a single-center, double-blinded, randomized placebo-controlled study, Hp eradication did not improve clinical outcomes in PD, suggesting no potential benefit from routine Hp-screening or Hp-eradication in the management of PD [114]. Nevertheless, Hp eradication in patients with PD might improve the bioavailability of L-3,4-dihydroxyphenylalanine (L-dopa), a precursor of dopamine used as a treatment for PD, and reduce motor fluctuations [115]. Moreover, Hp, which colonizes predominantly the gastric epithelium and oral cavity, is a risk factor for periodontitis [116], which, beyond AD [117], is also linked to PD [118]. Hp eradication and treatment of periodontitis are beneficial for the prevention of PD and dementia [118]. Thus, further related prospective studies are warranted to clarify this critical issue.

6.3. Amyotrophic Lateral Sclerosis (ALS)

Helicobacter spp., together with Pasteurella pneumotropica, Tritrichomonas muris and murine norovirus were significantly more abundant in C9orf72Harvard mice than in C9orf72Broad mice [10]. This increase in Helicobacter spp. relative abundance suggested a possible link with increased mortality and inflammation in the C9orf72Harvard mice, whereas administration of broad-spectrum antibiotics lead to a reduction in inflammation and improved clinical outcomes. To our knowledge, however, no clinical evidence of an association between Hp and/or other Helicobacter species with ALS in humans has been reported.

6.4. Huntington’s Disease (HD)

In a retrospective study, the results of 105 esophagogastroduodenoscopies, performed in 68 patients with HD, showed that Hp was positive in 4 of 19 patients investigated (21.1%). A high prevalence of gastritis or esophagitis was reported [119]. Although this finding indicates an association of HD with dysregulation of the GI system, thus posing potential implication for the role of the GBA in HD, this assumption warrants for a more detailed investigation. To our knowledge, however, additional clinical and/or experimental evidence regarding potential association between Hp with HD is currently lacking.

6.5. Multiple Sclerosis (MS)

While a robust link between Hp-infection and MS was not demonstrated in initial several observational studies, a noteworthy occurrence of concomitant autoimmune disorders among MS patients was indicated [78]. Patients with MS also exhibited atrophic gastritis evidenced by histology. Molecular mimicry is an intriguing assumption for this association: Hp-infection may induce immune responses following to an erroneous cross-reaction of Hp antigens with antigens of the peripheral and the CNS, thus potentially initiating and perpetuating nervous tissue damage [78]. Another study identified galectin-3 as a potential antigen recognized by antibodies present in the peripheral blood of individuals with secondary progressive MS (SPMS) [120]. Although these antibodies were advocated to serve as potential novel marker for SPMS diagnosis and as a potential target for therapeutic agents [120], the exact implication that these antibodies pose for SPMS remains to be elucidated. Galectins, being highly conserved molecules across a wide range of epithelial and immune cells, may explain some of the phenotypic and functional variation in the otherwise well-characterized TLR-dependent signaling, induced by several microbes and/or pathogens, including Hp [121]. Specifically, galectin-3 increased expression is reported in active injured regions in MS patients [122], and in postmortem MS human brain tissues [123], thereby appearing to be an important therapeutic target in neurodegenerative disorders including MS [124]. Greek and other data indicate that active Hp infection, apart from MS [78,125,126], is also common in patients with clinically isolated syndrome (CIS), the prodromal phase of consequent development of MS, accompanied by the mentioned hyperhomocysteinemia [79]. Hp eradication could delay the progression of CIS to the development of MS [79].
Finally, Hp-induced vacuolating cytotoxin A (VacA), displays chemotactic activities toward bone marrow-derived MCs (BMD-MCs) and induces them to produce pro-inflammatory cytokines that, in addition to causing local pathologies [127], lead to disruptions of the blood–brain barrier (BBB) and the blood-ocular barrier (BOB), resulting in neuropathies such as MS, AD and glaucoma, the latter referred to as ‘ocular AD’ [128]. Likewise, Hp-related VacA appears to promote the intracellular survival of Hp, and activated monocytes (possibly infected with Hp due to defective autophagy) could access the brain (Trojan horse theory) through BBB/BOB disruption, thereby triggering the development and progression of neurodegenerative disorders [129], possibly by inducing additional defensin abnormal expression. When Hp accesses the brain, it may trigger the maturation and activation of defensin-related dendritic cells leading to the release of pro-inflammatory cytokine by effector T cells, thereby promoting neuronal cell injury and death [130]. Interestingly, defensin-related activation of MCs [131] may also trigger neurogenic disorders, and thus further research is needed.

7. Mast Cells (MCs) and Mechanisms of Neuroinflammation in Neurodegenerative Diseases

Evidence of neuroinflammation in the context of neurodegenerative disease has been the focus of extensive research and MCs, as a cellular component of the innate immune system, have been shown to participate inflammatory responses in the CNS in several neurodegenerative diseases (Table 1)

7.1. Mast Cells (MCs) in Alzheimer’s Disease (AD)

AD is a prevalent neurological disorder [174,175]. Memory loss is a common symptom of the disease, but there is also a considerable reduction in other cognitive domains (language, visual–spatial skills, praxis skills, reasoning and judgment) [176]. It is estimated that more than 100 million individuals will have dementia by 2050, placing a significant economic impact on societies and families [177]. The main AD pathophysiological characteristics in the CNS are Aβ accumulation, formation of neurofibrillary tangles and abnormal phosphorylation of tau protein, resulting in impaired synaptic function and cognition [178,179]. The primary basis of the amyloid hypothesis of AD is based on the notion that AD pathology is characterized by the accumulation of Aβ in brain senile plaques. However, the prevailing viewpoint for AD has evolved, for AD to be considered as a condition in which soluble oligomeric Aβ forms play a crucial role in disrupting synaptic plasticity and cognition [17]. Although in the normal aging brain, evidence of limited neuroinflammation exists, the critical role of neuroinflammation in the onset and progression of AD, based on the abundance of inflammatory cytokines and microglia in amyloid plaques has long been suspected [138]. Increased exposure to continuous stimuli produces a phenotypic transformation in microglia into a dysfunctional senescent state with limited capacity for phagocytosis and immune tolerance, thus further promoting brain tissue pathology [180].
Neuroinflammation enhances the pathology of AD via several mechanisms including oxidative stress [132], hyperphosphorylation of tau protein [136], Aβ buildup [138] and the impairment of cholinergic transmission [139]. Neuroinflammatory key players such as IL-1, IL-6, and IFN-γ recruit macrophages and other immune cells, such as MCs, into the amyloid plaques, thus further contributing to AD pathogenesis [133,134,135]. Conversely, phagocyted amyloid particles inside MCs were found in skin and gastric samples, where they may trigger histamine release [137]. In addition to Aβ fragments, the amyloid A protein precursor serum amyloid A (SAA), is increased in AD brains [140]. SAA causes MCs degranulation, cytokine release, especially the pro-inflammatory tumor necrosis factor alpha (TNF-α) and IL-1, and chemotaxis [137]. MC granule material causes SAA degradation into proto-fibrillar intermediates [140]. Furthermore, exposure to amyloid peptide fragment 25–35 causes histamine release by BMD-MCs, via a mechanism involving pannexin 1 (panx1) hemichannel (HC) [135]. Panx1 organizes a membrane channel that serves as a conduit for ATP release extracellularly [137]. This channel’s activity can be enhanced with an increase in intracellular [Ca2+], extracellular [K+], and alkaline pH, whereas protein kinase A activation reduces the activity of this channel [135,181]. Additionally, it has been noted that Aβ also contributes to the degranulation of prefrontal cortical MCs, via the enhancement of connexin-43 (Cx43) and Panx1 HC flux [137]. MC abundance in the brains of amyloid precursor protein (APP) and presenilin 1 (PS1) APPswe/PS1dE9 mice significantly surpassed MC abundance in control mice, more notably in the hippocampus and the cortex [137]. Moreover, in APPswe/PS1dE9 mice, prefrontal cortex MCs exhibited basal ethidium bromide uptake, which was reduced significantly by Panx1 and Cx43 HC inhibitors, further suggesting concurrent involvement of these channels in MC activation [137].
During the evolution of AD, especially prior to the amyloid plaque formation, MCs are reported to detect low-solubility amyloid particles and to migrate to plaque sites, where they trigger the release of inflammatory mediators and disrupt the BBB [137]. Upon activation, MCs release protease gelatinases (metalloproteinases 2 and 9) and vascular endothelial growth factor (VEGF), leading to vascular leakage, CNS leukocyte infiltration and edema [141]. Upon the development of amyloid plaques, reactive microglia release pro-inflammatory cytokines and glutamate/ATP through HCs, possibly continuously activating Cx43 HCs in nearby microglia and astrocytes [182]. Also in other pathologies with inflammatory components, such as cerebral ischemia [141], traumatic brain injury (TBI) [183], experimental autoimmune encephalomyelitis (EAE) [50] and stress [184], MC activation was shown to exacerbate BBB permeabilization.
MCs can also detect IL-33, a cytokine released by damaged cells. Moreover, MCs reciprocally induce glia and neurons to further produce IL-33, thus further activating MCs [142,146]. These MCs act as early responders to brain injury, as they release pre-stored TNF and proteases and recruit immune cells, such as neutrophils, within hours form the initial injury [142]. TNF, released by MCs, upregulates adhesion molecules in vascular endothelial cells, thus promoting leukocyte recruitment [143,144]. VEGFs released at injury sites stimulate MC accumulation through VEGF receptors (VEGFR) [145]. Conversely, transforming growth factor beta (TGF-β) derived from MC granules suppresses MC activity and bears role in fibrosis, angiogenesis, tissue repair and the inhibition of immune response. As a response to traumatic brain injury (TBI), MCs were shown to infiltrate the brain tissue and to degranulate, thus releasing pre-stored neuroprotective mediators [185]. However, in severe injuries, the innate immune response leads to chronic neuroinflammation, continuous production of pro-inflammatory mediators, sustained tissue damage and neurobehavioral abnormalities associated with neuronal loss. In this respect, TBI and stress have been advocated as factors to increase AD risk in susceptible individuals, accelerating AD onset and progression. While some studies report no direct link between TBI and AD, the majority support such association [142].
Chronic stress may also accelerate AD progression by the promotion of inflammation and the enhancement of Aβ accumulation, tau hyperphosphorylation, oxidative stress, mitochondrial dysfunction and glucose metabolism dysregulation [186]. In this respect, MCs play a critical role in stress-induced inflammation. MCs release the corticotropin-releasing hormone (CRH), which further activates neighboring MCs and microglia [147]. Chronic stress triggers MC degranulation via the hypothalamic–pituitary–adrenal (HPA) axis, causing excessive pro-inflammatory mediator release, synaptic loss, BBB disruption and neuroinflammation in affected brain regions [146]. Evidence suggests that MC-derived inflammatory mediators may hasten AD progression [187].

7.2. Mast Cells (MCs) in Parkinson’s Disease (PD)

PD, the second most prevalent age-related neurodegenerative disorder globally, is characterized clinically by typical motor symptoms, including resting tremor, bradykinesia, rigidity, as well as non-motor symptoms [149]. Key pathological features of PD encompass the loss of dopaminergic neurons in the substantia nigra (SN) and ventral tegmental area (VTA), leading to reduced dopaminergic neurotransmission to the striatum (STR), and the formation of α-synuclein bodies. Severe motor deterioration typically manifests in the middle to late stages of the disease [148]. However, the precise pathogenic mechanisms responsible for PD remain elusive. Emerging evidence also links PD progression to intracellular mitochondrial dysfunction-induced reactive oxygen species generation, neuroinflammation, dysfunction in the ubiquitin–proteasomal and autophagy–lysosomal systems, as well as the accumulation of α-synuclein [152].
Neuroinflammation has been reportedly linked with all aspects of PD pathology, from α-synuclein aggregation to dopaminergic cell loss and the development of PD symptoms. Activated microglia and astrocytes that accompany midbrain dopaminergic neuron loss are the hallmarks of neuroinflammation in patients with PD and in PD animal models [149]. PD-associated factors, including key gene mutations [α-synuclein (SNCA), Parkin, protein deglycase DJ-1, Leucine-rich repeat kinase 2 (LRRK2)] and neurotoxins (rotenone, methyl-4-phenyl-1,2,3,6-tetrahydropyridine) can contribute in microglia and astrocyte activation [149]. Neuroinflammation plays pivotal role in PD pathogenic processes, contributing to and aggravated by protein aggregation, oxidative stress, mitochondrial dysfunction, calcium homeostasis disruption, and iron deposition [188]. Accumulation of misfolded α-synuclein is linked to dysregulation of immune response in the CNS [151]. Moreover, genome-wide association studies support the neuroinflammatory hypothesis for sporadic PD, identifying PD-associated polymorphisms in the human leukocyte antigen (HLA) region, particularly the HLA-DR gene [151]. Lastly, processes such as neurotoxic substance release by reactive microglia and astrocytes, brain infiltration and activation of inflammatory MCs and T lymphocytes, increased oxidative stress, and increased expression of inflammatory signaling molecules were shown to contribute to PD-related neuronal death and neurodegeneration [151,189].
In neuroinflammatory conditions, glial cells, neurons and MCs communicate via various signaling and mediating molecules [CD40 ligand (CD40L), CD40, TLR2, TLR4, protease-activated receptor-2 (PAR2), chemokine C-X-C receptors 4/12 (CXCR4/CXCL12), C5a receptor], promoting glial activation and further inflammatory mediator release [150]. MCs are advocated as pivotal intermediaries that bridge neurons and neuroinflammation [150]. Specifically, CD40-CD40L interaction triggers pro-inflammatory mediator production; 1-methyl-4-phenylpyridinium (MPP+), a metabolite of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) elevates CD40L expression in BMD-MCs, highlighting MCs’ role in neuroinflammation [150]. In the context of PD, brain MCs exert neurotoxic effects, thus contributing to neurodegeneration particularly in the SN [150]. Of note, administration of MPTP after MC reconstitution was shown to increase oxidative stress and altered levels of malondialdehyde, Glutathione, Superoxide dismutase, and Glutathione peroxidase [152]. Lastly, the upregulation of calpain 1 and intercellular Adhesion Molecule 1 (ICAM 1) expression in response to MPTP after MC reconstitution was shown to result into astroglia activation-dependent dopaminergic cell death and motor behavior deterioration in vivo, highlighting the potential therapeutic benefit of GMF inhibition in the context of neuroinflammation that accompanies neurodegeneration [152].
These results are further supported by the report that MCs are attracted and activated in the SN region of MPTP-induced mice through C-C motif chemokine ligand 2/C-C chemokine receptor type 2 (CCL2/CCR2) up-regulation and the expression of adhesion molecules [vascular cell adhesion molecule 1 (VCAM-1), platelet endothelial cell adhesion molecule-1 (PECAM-1), and ICAM-1) [153]. NF-κB activation induces transglutaminase 2 (TG2) expression in activated MCs, releasing inflammatory mediators like histamine, leukotrienes, TNF-α, and IL-1, potentially contributing to tyrosine hydroxylase (TH) + dopaminergic (DA) neuron loss in the SN region. While a similar process may occur in human PD patients, more research is needed to confirm these experimental results [153]. Moreover, MC proteases, namely mouse mast cell protease 6 (MMCP-6) and mouse mast cell protease 7 (MMCP-7), were also shown to stimulate astrocytes and neurons [146]. More specifically, MPP+ triggers MCs to release IL-33 by activating extracellular signal-regulated kinase ½ (ERK1/2), p38 mitogen-activated protein kinase (MAPKs), and NF-κB. Furthermore, in PD brains, particularly in the midbrain and striatum regions, there is an increase in MC numbers, activation status, and IL-33 expression linked to cell injury. The latter suggests that targeting glia-neuron and MC activation pathways may represent a novel therapeutic approach for PD [146].
The NLRP3 inflammasome’s significant involvement in PD has recently come to light. Cleaved caspase-1, IL-1, NLRP3 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-specks were detected in postmortem PD brain tissues and plasma from PD patients, indicating an overall increase in inflammasome activity compared to age-matched controls [132,154,155]. Moreover, heightened systemic NLRP3 activation has been linked to declining motor function and PD progression [190]. In experimental disease, particularly the MPTP mouse model, a strong association between α-synuclein levels, microglial NLRP3 inflammasome activation and dopaminergic neuron toxicity has been shown [132]. While CNS MC expression of the NLRP3 inflammasome has not been directly studied, peripheral tissue MCs in cryopyrin-associated periodic syndrome patients evidently express functional inflammasomes and produce IL-1 [156]. With respect to the CNS MCs, MC inflammasome was shown to play a pivotal role in the meningeal inflammation that, at least in part, determines disease severity in a rodent model of MS [171]. Taken together, these findings emphasize the central role of MCs as primary instigators of inflammatory responses and suggest that, although direct evidence of NLRP3 inflammasome involvement in brain MC activation and function in neurodegenerative diseases, such as PD and AD is currently lacking, a potential role for CNS MCs in the pathogenesis of these diseases is reasonable to be hypothesized and further investigated.
Chronic stress can trigger the HPA axis activation, thus potentially contributing to the loss of dopaminergic neurons in the SN in PD [191]. The expression of corticotropin-releasing hormone receptor 1 (CRH-R1) receptors in dopaminergic neurons, depending on their brain location, may exert anxiolytic effects [192]. Microglia and astrocytes surround MCs and can become activated in PD, releasing inflammatory cytokines, CRH, neurotensin, and neuromodulators that stimulate bidirectional communication capable of damaging dopaminergic neurons [150]. Further research is needed in order to better understand the molecular and cellular communication pathways that exist between MCs, CRH neurons and glial cells in the etiology of PD [178].

7.3. Mast Cells (MCs) in Amyotrophic Lateral Sclerosis (ALS)

ALS is a heterogenous disease with genetic predisposition, also thought to have multigenetic and possibly environmental causes [159]. Autosomal dominant mutations have been primarily, but not exclusively, related to ALS, namely, the C9orf72, superoxide dismutase 1 (SOD1), transactive response DNA binding protein (TARDBP) and fused in sarcoma/translocated in liposarcoma (FUS/TLS) mutation, among others [193]. ALS diagnosis is challenging, particularly at early stages. Typically, more common diseases are initially investigated, thus resulting in significant diagnostic delay [194]. The pathophysiological hallmarks of ALS are degeneration of motor neurons and distal motor axonopathy. Motor axon degeneration in the context of ALS has been linked to impaired axonal transport and mitochondrial function defect [157].
Reactive immune cells have been identified in postmortem tissue of patients with familial and sporadic ALS. More specifically, degenerating motor neuron cell bodies and neuronal axons were reportedly surrounded by microglial cells and/or CNS macrophages. Neuroinflammation has been described as a pathogenic process in ALS. For instance, degranulating MCs have been reported in the quadriceps muscle of patients with ALS but not in controls. In this context, MCs were shown to interact with myofibers and motor endplates. MCs and neutrophils are reportedly surrounding motor axons in SOD1G93A rats with ALS, thus indicating that infiltration by immune cells in ALS extends across the peripheral motor pathway. Moreover, administration of masitinib, a tyrosine kinase inhibitor, resulted in reduced infiltration by MCs and neutrophils, ameliorated axonal pathology and secondary demyelination, and hindered the loss of type 2B myofibers in SOD1G93A rats [158,159]. The tyrosine kinase receptor c-Kit (KIT or CD117) is a member of the transmembrane growth factor receptor family [164], also known as MC growth factor [164]. MCs express c-Kit throughout their lifetime [164] and the stem cell factor (SCF) /c-Kit pathway is essential for MCs in order to survive and to differentiate [164]. Moreover, it has been shown to be necessary in order for MCs to infiltrate nerves and skeletal muscles in patients and transgenic rodents with ALS [160]. In contrast to macrophages for which existing evidence on their potential beneficial or detrimental role in ALS is conflicting [157], MCs have recently been demonstrated to be located in degenerating motor nerve endplates in SOD1G93A mice, and the degree of MC degranulation was shown to correspond to clinical disease endpoints [160]. Masitinib administration was shown to diminish NMJ denervation and motor impairment in SOD1G93A mice [160], thus providing evidence of the role of MCs in ALS. The exact mechanism is unknown; however, MCs have been hypothesized to enhance vascular permeability in the context of neuroinflammation, thus triggering further neutrophil recruitment, clustering and activation [157,161,162,163]. Kovacs et al. [164] verified the presence of MCs and many c-Kit+ progenitors in the motor neuron-vascular niche, thus further supporting MC pathogenic potential in the context of ALS.

7.4. Mast Cells (MCs) in Huntington’s Disease (HD)

HD is an inherited autosomal dominant neurodegenerative disease. HD is caused by, to various degrees, amplification of a CAG triplet in the HTT gene, thus resulting in the production of a mutant Huntingtin protein (mHTT). Excitatory neurotoxicity is caused by mHTT in the striatum and cortex, linked to movement disorder with chorea and/or dystonia accompanied by cognitive, and neuropsychiatric symptoms [195]. Reactive astrocytes have been reported in pre-clinical stages of HD and their presence further correlate with later disease severity [165]. Moreover, reactive microglia have been reported in the striatum and cortex of HD, accompanying neuron loss, in postmortem brains [167]. Although a neuroinflammatory component has been identified in HD pathogenesis, the relative contribution of MCs in HD pathology remains to be elucidated. MCs express TLR-4, a receptor recognizing PAMPs and DAMPs, such as the Gram-negative bacteria-derived lipopolysaccharide (LPS) [166]. TLR-4 activation in MCs results in TNF production, thus promoting mouse survival following systemic administration of LPS [166]. Perez-Rodriguez et al. [166] discovered that mHTT promotes TLR-4 receptor internalization, an important step in the signaling axis that affects MC activation. Thus, mHTT expression inhibits TLR4-dependent signaling, resulting in impaired innate immune responses and altering MC-dependent inflammatory response in vivo [166].

7.5. Mast Cells (MCs) in Multiple Sclerosis (MS)

MS is a multifactorial chronic demyelinating disorder of the CNS that displays inflammatory and neurodegenerative origins. It affects approximately 2.9 million people worldwide and is considered to be the most common cause of neurologic disability in young adults [196]. MS is frequently diagnosed between the ages of 20 and 50 years, with females experiencing it more often than males [197]; nevertheless, cases of pediatric and late onset MS are not rare [198]. In Greece, it is estimated that approximately 21,000 people are affected by MS [199].
MS has traditionally been regarded as a disease primarily of neuroinflammatory origin, with secondary neurodegeneration occurring later in the disease course. In MS, auto-activated T cell recognizing myelin antigens infiltrate the CNS, thus causing multi-focal demyelination in the context of neuroinflammation [200]. The majority of MS cases are diagnosed with the relapsing–remitting form of MS, a disease form with evident disease activity due to neuroinflammation. However, following several years, these patients are expected to develop secondary progressive disease, as neurodegeneration becomes increasingly evident as a result of accumulating age in combination with long-term disease pathology [201,202]. Of note, biomarkers that depict disease pathology and thus exhibiting potential to facilitate disease management and/or to estimate prognosis are currently lacking and are the focus of extensive research [203,204,205]. According to the latest international classification, there are three main types of MS: relapsing-remitting MS (RRMS), characterized by sub-acute episodes of neurological disability and intermittent periods of remission; primary progressive MS (PPMS), characterized by gradual accumulation of neurological disability from the disease onset; and SPMS, characterized by gradual accumulation of neurological disability following to the RRMS form [206]. The diversity in the pathophysiology of the two latter courses is still uncertain. The underlying mechanisms of MS are yet to be clarified [207,208], and the management relies on immune-modifying agents [204,209,210]. About 50% of individuals with MS display impairments in a wide variety of cognitive domains; among them, deficits in cognitive processing speed and working memory are the most frequently observed [211,212]. Additional cognitive domains that appear to be regularly affected by MS are diverse other aspects of memory, attention and executive functions [213,214]. Cortical inflammatory process, neurodegeneration, demyelination, axonal damage, axonal loss, oligodendrocytes, mitochondrial disfunction, microglia stimulation, oxidative and nitrosative stress are underlying processes of MS [200,215]. Myelinated axons in the CNS are the target of MS attacks, which can induce varying degrees of damage to both myelin and axon [216]. Reactive astrocytes sustained multifaceted contribution to neuroinflammatory outcomes and oligodendrocyte and neuronal function through chronicity, imply that they could be an integral cell type that can govern the pathophysiology of MS [217].
Beyond symptoms directly related to MS, typical comorbid conditions often diagnosed in people with MS include hyperlipidemia, hypertension, GI diseases, chronic lung disease, thyroid disease, obesity and neuropsychiatric disorders [218,219,220]. Comorbid conditions impact the disease course and affect treatment selection, adherence and outcome. In this sense, several intertwined issues emerge concerning diagnostic delay, degree of disability progression and additive effect on mortality [221]. Early diagnosis of comorbid conditions in clinical practice is of great importance to prevent a negative effect on MS [220]. However, little is recognized regarding the mechanisms of neurodegeneration in this disorder, a limitation that declines the development of more effective managements for MS [222].
Recently, the neurodegenerative component of MS has been the focus of extensive research, as it has been acknowledged that neurodegeneration contributes to the disease pathology across the disease course [200]. Moreover, although the activation of the adaptive immune system has been thoroughly interrogated in MS, the relative contribution of innate immune responses, especially in association to the chronic, more subjective to the role of neurodegeneration, disease stages, remains less elucidated [168]. MCs have been reported to contribute to MS pathogenesis by mediating inflammation and demyelination, acting as antigen-presenting cells towards T cells and/or by disrupting the BBB (reviewed in [169] and in [147]). With respect to the CNS MCs, MC inflammasome was shown to play a pivotal role in the meningeal inflammation that, at least in part, determines disease severity in a rodent model of MS [171]. The development of EAE reportedly involves MCs accumulation in the CNS, as shown in rats and mice [172,173]. What is more, evidence suggest that perivascular MCs may pose potential to regulate BBB permeability, an aspect important for MS pathogenesis [170].
Activated MCs appear to be involved in both innate and specific immunity, and their contribution to the diverse stages of MS and the development of EAE, the major animal model of MS, is significant [50,223]. MCs inhabitant in the dura mater and pia mater also aggravate EAE, through inducing CNS inflammatory cell influx [163]. According to Christy and Brown (2007), the most detrimental effects of MC occurrence in the CNS through MS/EAE development include the BBB permeability and a subsequent increase in the number of cells that access the CNS, which is mediated by histamine, chemokines, and leukotrienes. MCs could also contribute to epitope spreading through tissue damage provoked by proteases, T helper (Th) 1 and Th17 differentiation by releasing polarizing cytokines (IL-12 for Th1 and IL-6 plus TGF-β for Th17), and a reduction in the number of Tr1 regulatory cells by a direct interaction involving the OX40L/OX40 pair [163]. The contribution of MCs in MS is shown by gene overexpression of chemical mediators and inflammatory cytokines. MCs mediate inflammatory process and demyelization through presenting myelin antigens to T cells and/or disrupting the BBB, thereby allowing entry of inflammatory cells and cytokines in the brain, leading to MS neuropathy [169]. In the CNS, MCs are critically positioned in the perivascular area and produce numerous pro-inflammatory and vasoactive molecules that can disturb the BBB, an effect antecedent any clinical or pathological marks of MS [224]. In MS, brain MCs seem to be activated by neural agents, such as substance P, myelin basic protein, the principal MS antigen, and corticotropin-releasing hormone, caused by acute stress, which induce release of several inflammatory mediators involved in the pathophysiology of MS [224]. MCs are frequently also located near to MS plaques, and the myelin basic protein can trigger human cultured MCs to induce IL-8, TNF-α, tryptase, and histamine, contributing to the pathophysiology of MS. MCs might provide T cell activation because addition of MCs to anti-CD3/anti-CD28 activated T cells rises T cell activation above 30-fold, leading to MS neuropathy [225]. Therefore, the option to employ MC-targeted drugs to control this disease appears to be a promising therapeutic strategy. In this respect, for instance, pretreatment with the flavone luteolin completely inhibit MCs s and T cell activations, thus constitute a new therapeutic option for MS [225]. Moreover, ketotifen fumarate, a second-generation antihistamine agent, inhibits MCs exocytosis among other effects. Its early intervention seems to be very effective in controlling both the prevalence and severity of MS [226].

8. Impact of Gut–Brain Axis (GBA) and the Activation of Mast Cells (MCs) on Neurodegeneration: A Potential Role for Hp Infection

The GBA represents an important regulator of the brain’s immune responses with implication for neuroinflammation, thus bearing potential as a therapeutic target against the development and progression of neurodegenerative diseases [227]. The gut microbiome provides stimuli to the CNS innate immune cells, namely, the microglia, as well as to astrocytes and oligodendrocytes, via metabolites, gut microbiota-related neurotransmitters and gut hormones [228,229,230,231]. Growing evidence elucidates aspects of the GBA subjected to potential modifications, such as the intestinal barrier [232], the BBB [233], the meninges [234] and the peripheral immune system [235], as measures to ameliorate neuroinflammation in the context of neurodegeneration. Moreover, pre-clinical and clinical evidence on the beneficial application of probiotics [236], prebiotics [237] and fecal microbiota transplantation [238] in neurodegenerative diseases further underline the potential of the GBA to affect brain pathology. MCs are mediators of stress and neuroinflammation, as they have been shown to release CRH with systemic effects [239]. In the context of the GBA, MCs have been advocated as a cellular population with immune-modulatory potential [32], as MCs are abundant in the gut and present, to a lesser degree, in the brain upon neuroinflammation, shown to be capable of interacting with resident innate immune cells of the CNS, namely, the microglia [240]. Moreover, intestinal MCs have been shown to participate neuronal and endocrine signaling, thereby modulating neuroinflammation in the context of neurodegeneration [241]. Whereas MCs have been advocated to participate in neuroimmune signaling in the GBA [242], thorough mechanistic insight regarding the underlying mechanisms of this interaction is currently lacking.
An association between Hp infection and neurodegenerative diseases has been previously well described. MetS may be one of several common denominators in this association. MetS has been related to several brain neurodegenerative disorders. For instance, patients with HD exhibit peripheral IR, and a high risk of T2DM. Cellular cholesterol metabolism may be disrupted in HD, and it is advocated that the mHTT protein impacts cholesterol transport within the cell. Furthermore, patients with HD exhibit cerebral cortical hypoperfusion, even prior to the manifestation of cognitive symptoms and increased regional BBB permeability in the caudate nucleus, similarly to AD and PD [27,243].
IR is considered as a key factor contributing to MetS-linked pathology, in addition to well-described genetic and environmental factors [244]. In the context of MetS, glucose, lipid, and protein metabolism are impaired and ultimately lead in IR, obesity and CVD [245]. Growing evidence links Hp infection and IR/MetS, as well as T2DM, dyslipidemia, hypertension, MASLD and neurodegenerative and cardio-cerebrovascular diseases [69,246,247,248]. Underlying mechanisms by which MetS may be associated with brain pathology include for instance, but not exclusively, the disruption of the BBB, the triggering of neuroinflammation, the disruption of amyloid clearance and of the homeostasis of the cerebral blood vessels, thus resulting reduced cerebral blood flow, brain microenvironment alterations and disruption of the neurovascular unit [249,250,251,252]. Hp, as mentioned, is a common pathogen with its global prevalence approximating 58% [253]. Hp infection is associated with MetS-related systemic pathologies, including cardio-cerebrovascular and neurodegenerative disorders [86,87,115,128,246]. Moreover, Hp bacterial virulent factors may activate MC migration and the production of pro-inflammatory cytokines in the context of innate immunity. Hp-neutrophil-activating protein has been shown to induce degranulation of stored chemical mediators from MCs [254]. Oral administration of the Hp-related VacA in mice causes MC accumulation in the gastric mucosa [255] and the density of MC has been advocated as means of evaluating gastritis activity, in terms of histopathology, in patients with Hp infection [256]. However, the exact implications of MC–gastric epithelial cell interaction upon Hp infection in humans remain to be elucidated [257]. Recently, gastric IL-33 was found to be increased in the gastric epithelium of patients and mice with Hp infection, a result associated with the degree of bacterial expansion and the severity of gastritis [258]. Gastric IL-33 resulted in increased TNF-α production from MCs in vitro, and in vivo and inhibited gastric epithelial cell proliferation, thus further promoting Hp-associated gastritis and bacteria proliferation. The study provided evidence of Hp, gastric epithelial cells, IL-33, MCs, and TNF-α forming a complex regulatory network that, when disrupted, contributes to Hp infection-associated gastritis [258]. Eosinophilic esophagitis (EoE), an allergen/immune-mediated disease, also provides a model for MC association with Hp infection-related pathology. Recent evidence suggests a relatively protective effect of Hp infection in EoE, an observation that was not, however, further verified [259]. Although EoE is characterized by allergic reaction, thus typically Th2 immune response, a subset of older patients who exhibit intense allergic reaction at onset, also show activation of Th1 and Th17 pro-inflammatory responses [260], as in the case of Hp infection [261,262,263]. In the context of EoE, MCs mediate inflammation and fibrosis via the production of TGF-β and tryptase, thus further inducing smooth muscle contraction, esophageal spasm and collagen secretion [264,265]. In addition, TGF-β [266] and TGF-β-inducing protein from Hp mediate the pathogenesis of Hp infection [267]. Hp cytotoxin VacA induces the production of pro-inflammatory cytokines, such as TNF-α from the mentioned BMD-MCs, a cytokine reportedly overexpressed in esophageal epithelial cells in the context of EoE [268,269]. TNF-α stimulates MCs [270,271], a mechanism linked with Hp infection -associated chronic urticaria [272]. Interestingly, EoE-related activated MCs, beyond achalasia, have been implicated in the pathogenesis of neurodegenerative disorders such as AD and PD [273]. In this respect, Hp infection may contribute to the pathophysiology of achalasia, which, apart from AD and PD, is also associated with Guillain-Barré syndrome, by inducing autoimmunity and apoptosis. Thus, Hp eradication may offer benefit through amelioration the apoptotic loss of the esophageal wall ganglion cells and their axons [274].
MC activation in the context of Hp infection and the related MetS may contribute to the induction of CNS pathology via several mechanisms: Hp-induced MCs alter the balance of the GBA. As indicated by several CNS pathologies, such as infection, trauma and/or stress [42], MCs are contributors in the communication between the CNS and the systemic circulation, an observation with implication for neuroinflammation and neurodegeneration. For instance, as Hp-associated VacA shifts BMD-MCs towards the production of including TNF-α and other pro-inflammatory cytokines [275] and BMD-MCs are in proximity to the components of BBB, MCs may alter BBB permeability by affecting the microenvironment in terms of produced cytokines and chemokines. Moreover, Hp induces, via stress and/or increased gastrin production, local and systemic activation of MCs with the expression of VEGF, IL-8, chymase or tryptase, thus further disrupting the BBB and promoting immune cell infiltration towards the CNS. An additional mechanism is the activation of microglia in response to production of tryptase and histamine by MCs. Microglia secrete cytokines and/or chemokines into the microenvironment and further recruit MCs, eosinophils, monocytes and neutrophils, thus exacerbating neuroinflammation [276,277]. Hp and the associated activation of MCs in the context of Hp infection are, therefore, potential contributors of pro-inflammatory processes. MCs, in the presence of activated microglia, exhibit upregulated expression of TLR-2 and -4, both also expressed by Hp, thus rendered more responsive to endotoxin stimuli and microbial LPS [278].
These observations underline the complex interaction and the potential of MCs, in the context of Hp infection and in relation to microglia and other immune cells in the CNS, to alter the tissue microenvironment and to contribute to several neurodegenerative pathologies. In view of the recent recognition that neuroinflammatory processes have received in the context of diseases previously regarded as “primary neurodegenerative”, MCs and Hp infection may offer targets of intervention with potential therapeutic implications. More clinical and translational evidence is needed to elucidate in depth this field.

9. Conclusions

In conclusion, this review underscores the complex interplay between Hp infection, MCs activation, and neurodegenerative diseases, particularly within the context of MetS. The findings suggest that Hp may play a significant role in promoting neuroinflammation and neurodegeneration through mechanisms such as IR, compromised BBB integrity, and altered immune responses. MCs emerge as key mediators in these processes, as their activation can lead to the release of pro-inflammatory cytokines and contribute to an unfavorable microenvironment for neuronal health. Importantly, the evidence linking Hp infection with MetS highlights a critical area for further exploration. Future research should focus on elucidating the specific molecular pathways that connect Hp to neurodegenerative pathologies, examining the potential therapeutic benefits of MCs modulation in neuroprotection, and investigating the bidirectional influences between gut health and brain function. Additionally, clinical studies are warranted to evaluate the impact of Hp eradication on neurodegenerative risk in at-risk populations, providing insights into the GBA’s role in overall neurological health and opening avenues for novel preventative and therapeutic strategies. This multifaceted approach could significantly advance our understanding of neurodegeneration and foster the development of targeted interventions to mitigate its progression.

Author Contributions

Conceptualization, J.K.; methodology, M.B., P.T., E.K. (Evangelia Kesidou), M.N., C.B., E.K. (Eleni Karafoulidou), M.T.-C., M.D., E.K. (Evangelos Kazakos), G.D. and N.G.; investigation, M.B., P.T., E.K. (Evangelia Kesidou), M.N., C.B., E.K. (Eleni Karafoulidou), M.T.-C., M.D., G.D., N.G. and J.K.; writing—original draft preparation, M.B.; writing—review and editing, M.B., P.T., E.K. (Evangelia Kesidou), M.N., C.B., E.K. (Eleni Karafoulidou), M.T.-C., M.D., E.K. (Evangelos Kazakos), G.D., N.G. and J.K.; supervision, J.K.; project administration, J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cruciani, A.; Santoro, F.; Pozzilli, V.; Todisco, A.; Pilato, F.; Motolese, F.; Celani, L.M.; Pantuliano, M.C.; Tortorella, C.; Haggiag, S.; et al. Neurophysiological methods for assessing and treating cognitive impairment in multiple sclerosis: A scoping review of the literature. Mult. Scler. Relat. Disord. 2024, 91, 105892. [Google Scholar] [CrossRef] [PubMed]
  2. Prince, M.; Bryce, R.; Albanese, E.; Wimo, A.; Ribeiro, W.; Ferri, C.P. The global prevalence of dementia: A systematic review and metaanalysis. Alzheimer’s Dement. 2013, 9, 63–75.e2. [Google Scholar] [CrossRef] [PubMed]
  3. Goedert, M.; Eisenberg, D.S.; Crowther, R.A. Propagation of Tau Aggregates and Neurodegeneration. Annu. Rev. Neurosci. 2017, 40, 189–210. [Google Scholar] [CrossRef] [PubMed]
  4. Taylor, J.P.; Hardy, J.; Fischbeck, K.H. Toxic proteins in neurodegenerative disease. Science 2002, 296, 1991–1995. [Google Scholar] [CrossRef]
  5. Morais, L.H.; Schreiber, H.L.t.; Mazmanian, S.K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
  6. Ferreiro, A.L.; Choi, J.; Ryou, J.; Newcomer, E.P.; Thompson, R.; Bollinger, R.M.; Hall-Moore, C.; Ndao, I.M.; Sax, L.; Benzinger, T.L.S.; et al. Gut microbiome composition may be an indicator of preclinical Alzheimer’s disease. Sci. Transl. Med. 2023, 15, eabo2984. [Google Scholar] [CrossRef]
  7. Huang, B.; Chau, S.W.H.; Liu, Y.; Chan, J.W.Y.; Wang, J.; Ma, S.L.; Zhang, J.; Chan, P.K.S.; Yeoh, Y.K.; Chen, Z.; et al. Gut microbiome dysbiosis across early Parkinson’s disease, REM sleep behavior disorder and their first-degree relatives. Nat. Commun. 2023, 14, 2501. [Google Scholar] [CrossRef] [PubMed]
  8. Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef] [PubMed]
  9. Dodiya, H.B.; Lutz, H.L.; Weigle, I.Q.; Patel, P.; Michalkiewicz, J.; Roman-Santiago, C.J.; Zhang, C.M.; Liang, Y.; Srinath, A.; Zhang, X.; et al. Gut microbiota-driven brain Abeta amyloidosis in mice requires microglia. J. Exp. Med. 2022, 219, e20200895. [Google Scholar] [CrossRef]
  10. Burberry, A.; Wells, M.F.; Limone, F.; Couto, A.; Smith, K.S.; Keaney, J.; Gillet, G.; Van Gastel, N.; Wang, J.Y.; Pietilainen, O.; et al. C9orf72 suppresses systemic and neural inflammation induced by gut bacteria. Nature 2020, 582, 89–94. [Google Scholar] [CrossRef]
  11. da Silva, E.Z.; Jamur, M.C.; Oliver, C. Mast cell function: A new vision of an old cell. J. Histochem. Cytochem. 2014, 62, 698–738. [Google Scholar] [CrossRef] [PubMed]
  12. Maslinska, D.; Wozniak, R.; Kaliszek, A.; Schmidt-Sidor, B.; Lipska, A.; Woolley, D.E. Phenotype of mast cells in the brain tumor. Capillary hemangioblastoma. Folia Neuropathol. 1999, 37, 138–142. [Google Scholar] [PubMed]
  13. Galli, S.J.; Tsai, M. Mast cells in allergy and infection: Versatile effector and regulatory cells in innate and adaptive immunity. Eur. J. Immunol. 2010, 40, 1843–1851. [Google Scholar] [CrossRef] [PubMed]
  14. 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]
  15. Krystel-Whittemore, M.; Dileepan, K.N.; Wood, J.G. Mast Cell: A Multi-Functional Master Cell. Front. Immunol. 2015, 6, 620. [Google Scholar] [CrossRef]
  16. Castellheim, A.; Brekke, O.L.; Espevik, T.; Harboe, M.; Mollnes, T.E. Innate immune responses to danger signals in systemic inflammatory response syndrome and sepsis. Scand J. Immunol. 2009, 69, 479–491. [Google Scholar] [CrossRef]
  17. Skaper, S.D.; Facci, L.; Zusso, M.; Giusti, P. An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front. Cell. Neurosci. 2018, 12, 72. [Google Scholar] [CrossRef]
  18. Presta, I.; Vismara, M.; Novellino, F.; Donato, A.; Zaffino, P.; Scali, E.; Pirrone, K.C.; Spadea, M.F.; Malara, N.; Donato, G. Innate Immunity Cells and the Neurovascular Unit. Int. J. Mol. Sci. 2018, 19, 3856. [Google Scholar] [CrossRef] [PubMed]
  19. Edison, P. Neuroinflammation, microglial activation, and glucose metabolism in neurodegenerative diseases. Int. Rev. Neurobiol. 2020, 154, 325–344. [Google Scholar]
  20. Le Thuc, O.; Blondeau, N.; Nahon, J.L.; Rovere, C. The complex contribution of chemokines to neuroinflammation: Switching from beneficial to detrimental effects. Ann. N. Y. Acad. Sci. 2015, 1351, 127–140. [Google Scholar] [CrossRef]
  21. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2014, 79, 1–12. [Google Scholar] [CrossRef] [PubMed]
  22. Piirainen, S.; Youssef, A.; Song, C.; Kalueff, A.V.; Landreth, G.E.; Malm, T.; Tian, L. Psychosocial stress on neuroinflammation and cognitive dysfunctions in Alzheimer’s disease: The emerging role for microglia? Neurosci. Biobehav. Rev. 2017, 77, 148–164. [Google Scholar] [CrossRef] [PubMed]
  23. Thacker, M.A.; Clark, A.K.; Marchand, F.; McMahon, S.B. Pathophysiology of peripheral neuropathic pain: Immune cells and molecules. Anesth Analg. 2007, 105, 838–847. [Google Scholar] [CrossRef] [PubMed]
  24. Ali, A.; Ahmad, W.; Ahmad, N.; Khan, S.; Nuruddin, S.M.; Husain, I. Deciphering the Role of WNT Signaling in Metabolic Syndrome-Linked Alzheimer’s Disease. Mol. Neurobiol. 2020, 57, 302–314. [Google Scholar] [CrossRef]
  25. Dabke, K.; Hendrick, G.; Devkota, S. The gut microbiome and metabolic syndrome. J. Clin. Investig. 2019, 129, 4050–4057. [Google Scholar] [CrossRef]
  26. Wieckowska-Gacek, A.; Mietelska-Porowska, A.; Wydrych, M.; Wojda, U. Western diet as a trigger of Alzheimer’s disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Res. Rev. 2021, 70, 101397. [Google Scholar] [CrossRef]
  27. Fakih, W.; Zeitoun, R.; AlZaim, I.; Eid, A.H.; Kobeissy, F.; Abd-Elrahman, K.S.; El-Yazbi, A.F. Early metabolic impairment as a contributor to neurodegenerative disease: Mechanisms and potential pharmacological intervention. Obesity 2022, 30, 982–993. [Google Scholar] [CrossRef]
  28. Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
  29. 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]
  30. Nelissen, S.; Lemmens, E.; Geurts, N.; Kramer, P.; Maurer, M.; Hendriks, J.; Hendrix, S. The role of mast cells in neuroinflammation. Acta Neuropathol. 2013, 125, 637–650. [Google Scholar] [CrossRef]
  31. Galli, S.J.; Nakae, S.; Tsai, M. Mast cells in the development of adaptive immune responses. Nat. Immunol. 2005, 6, 135–142. [Google Scholar] [CrossRef]
  32. 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]
  33. Kitamura, Y.; Ito, A. Mast cell-committed progenitors. Proc. Natl. Acad. Sci. USA 2005, 102, 11129–11130. [Google Scholar] [CrossRef]
  34. Jones, M.K.; Nair, A.; Gupta, M. Mast Cells in Neurodegenerative Disease. Front. Cell. Neurosci. 2019, 13, 171. [Google Scholar] [CrossRef]
  35. Nautiyal, K.M.; Liu, C.; Dong, X.; Silver, R. Blood-borne donor mast cell precursors migrate to mast cell-rich brain regions in the adult mouse. J. Neuroimmunol. 2011, 240–241, 142–146. [Google Scholar] [CrossRef]
  36. Skaper, S.D.; Facci, L.; Giusti, P. Mast cells, glia and neuroinflammation: Partners in crime? Immunology 2014, 141, 314–327. [Google Scholar] [CrossRef]
  37. Banuelos-Cabrera, I.; Valle-Dorado, M.G.; Aldana, B.I.; Orozco-Suarez, S.A.; Rocha, L. Role of histaminergic system in blood-brain barrier dysfunction associated with neurological disorders. Arch. Med. Res. 2014, 45, 677–686. [Google Scholar] [CrossRef]
  38. Polyzoidis, S.; Koletsa, T.; Panagiotidou, S.; Ashkan, K.; Theoharides, T.C. Mast cells in meningiomas and brain inflammation. J. Neuroinflam. 2015, 12, 170. [Google Scholar] [CrossRef]
  39. Ribatti, D. The crucial role of mast cells in blood-brain barrier alterations. Exp. Cell Res. 2015, 338, 119–125. [Google Scholar] [CrossRef] [PubMed]
  40. Bugajski, A.J.; Chlap, Z.; Gadek, M.; Bugajski, J. Effect of isolation stress on brain mast cells and brain histamine levels in rats. Inflamm. Res. 1994, 41, C75–C76. [Google Scholar] [CrossRef] [PubMed]
  41. Maslinska, D.; Laure-Kamionowska, M.; Gujski, M.; Ciurzynska, G.; Wojtecka-Lukasik, E. Post-infectional distribution and phenotype of mast cells penetrating human brains. Inflamm. Res. 2005, 54 (Suppl. S1), S15–S16. [Google Scholar] [CrossRef]
  42. Silver, R.; Curley, J.P. Mast cells on the mind: New insights and opportunities. Trends Neurosci. 2013, 36, 513–521. [Google Scholar] [CrossRef]
  43. Michaloudi, H.; Batzios, C.; Chiotelli, M.; Grivas, I.; Papadopoulos, G.C. Mast cells populations fluctuate along the spinal dura mater of the developing rat. Brain Res. 2008, 1226, 8–17. [Google Scholar] [CrossRef]
  44. Xanthos, D.N.; Gaderer, S.; Drdla, R.; Nuro, E.; Abramova, A.; Ellmeier, W.; Sandkühler, J. Central nervous system mast cells in peripheral inflammatory nociception. Mol. Pain 2011, 7, 42. [Google Scholar] [CrossRef]
  45. Forsythe, P. The parasympathetic nervous system as a regulator of mast cell function. Methods Mol. Biol. 2015, 1220, 141–154. [Google Scholar]
  46. Gupta, K.; Harvima, I.T. Mast cell-neural interactions contribute to pain and itch. Immunol. Rev. 2018, 282, 168–187. [Google Scholar] [CrossRef] [PubMed]
  47. Kritas, S.K.; Caraffa, A.; Antinolfi, P.; Saggini, A.; Pantalone, A.; Rosati, M.; Tei, M.; Speziali, A.; Saggini, R.; Pandolfi, F.; et al. Nerve growth factor interactions with mast cells. Int. J. Immunopathol. Pharmacol. 2014, 27, 15–19. [Google Scholar] [CrossRef]
  48. Schemann, M.; Camilleri, M. Functions and imaging of mast cell and neural axis of the gut. Gastroenterology 2013, 144, 698–704. [Google Scholar] [CrossRef]
  49. Bachelet, I.; Levi-Schaffer, F.; Mekori, Y.A. Mast cells: Not only in allergy. Immunol. Allergy Clin. N. Am. 2006, 26, 407–425. [Google Scholar] [CrossRef]
  50. Pinke, K.H.; Zorzella-Pezavento, S.F.G.; Lara, V.S.; Sartori, A. Should mast cells be considered therapeutic targets in multiple sclerosis? Neural Regen. Res. 2020, 15, 1995–2007. [Google Scholar] [CrossRef]
  51. Weng, Z.; Zhang, B.; Tsilioni, I.; Theoharides, T.C. Nanotube Formation: A Rapid Form of “Alarm Signaling”? Clin. Ther. 2016, 38, 1066–1072. [Google Scholar] [CrossRef]
  52. Patkai, J.; Mesples, B.; Dommergues, M.A.; Fromont, G.; Thornton, E.M.; Renauld, J.C.; Evrard, P.; Gressens, P. Deleterious effects of IL-9-activated mast cells and neuroprotection by antihistamine drugs in the developing mouse brain. Pediatr. Res. 2001, 50, 222–230. [Google Scholar] [CrossRef]
  53. Hendrix, S.; Warnke, K.; Siebenhaar, F.; Peters, E.M.; Nitsch, R.; Maurer, M. The majority of brain mast cells in B10.PL mice is present in the hippocampal formation. Neurosci. Lett. 2006, 392, 174–177. [Google Scholar] [CrossRef]
  54. Taiwo, O.B.; Kovacs, K.J.; Larson, A.A. Chronic daily intrathecal injections of a large volume of fluid increase mast cells in the thalamus of mice. Brain Res. 2005, 1056, 76–84. [Google Scholar] [CrossRef]
  55. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  56. Sinagra, E.; Utzeri, E.; Morreale, G.C.; Fabbri, C.; Pace, F.; Anderloni, A. Microbiota-gut-brain axis and its affect inflammatory bowel disease: Pathophysiological concepts and insights for clinicians. World J. Clin. Cases 2020, 8, 1013–1025. [Google Scholar] [CrossRef]
  57. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  58. Asseri, A.H.; Bakhsh, T.; Abuzahrah, S.S.; Ali, S.; Rather, I.A. The gut dysbiosis-cancer axis: Illuminating novel insights and implications for clinical practice. Front. Pharmacol. 2023, 14, 1208044. [Google Scholar] [CrossRef]
  59. Zhang, M.; Qin, Z.; Huang, C.; Liang, B.; Zhang, X.; Sun, W. The gut microbiota modulates airway inflammation in allergic asthma through the gut-lung axis related immune modulation: A review. Biomol. Biomed. 2024. [Google Scholar] [CrossRef]
  60. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693 Pt B, 128–133. [Google Scholar] [CrossRef]
  61. Mukhtar, K.; Nawaz, H.; Abid, S. Functional gastrointestinal disorders and gut-brain axis: What does the future hold? World J. Gastroenterol. 2019, 25, 552–566. [Google Scholar] [CrossRef] [PubMed]
  62. Conte, C.; Sichetti, M.; Traina, G. Gut–Brain Axis: Focus on Neurodegeneration and Mast Cells. Appl. Sci. 2020, 10, 1828. [Google Scholar] [CrossRef]
  63. Baj, J.; Forma, A.; Flieger, W.; Morawska, I.; Michalski, A.; Buszewicz, G.; Sitarz, E.; Portincasa, P.; Garruti, G.; Flieger, M.; et al. Helicobacter pylori Infection and Extragastric Diseases-A Focus on the Central Nervous System. Cells 2021, 10, 2191. [Google Scholar] [CrossRef]
  64. Kountouras, J.; Papaefthymiou, A. Letter: Higher severe outcomes among Helicobacter pylori-related lean patients with non-alcoholic fatty liver disease and metabolic comorbidities. Aliment. Pharmacol. Ther. 2023, 57, 1186–1187. [Google Scholar] [CrossRef] [PubMed]
  65. Piekut, T.; Hurla, M.; Banaszek, N.; Szejn, P.; Dorszewska, J.; Kozubski, W.; Prendecki, M. Infectious agents and Alzheimer’s disease. J. Integr. Neurosci. 2022, 21, 73. [Google Scholar] [CrossRef]
  66. Zavos, C.; Kountouras, J.; Sakkias, G.; Venizelos, I.; Deretzi, G.; Arapoglou, S. Histological presence of Helicobacter pylori bacteria in the trabeculum and iris of patients with primary open-angle glaucoma. Ophthalmic Res. 2012, 47, 150–156. [Google Scholar] [CrossRef] [PubMed]
  67. Adler, I.; Muiño, A.; Aguas, S.; Harada, L.; Diaz, M.; Lence, A.; Labbrozzi, M.; Muiño, J.M.; Elsner, B.; Avagnina, A.; et al. Helicobacter pylori and oral pathology: Relationship with the gastric infection. World J. Gastroenterol. 2014, 20, 9922–9935. [Google Scholar] [CrossRef]
  68. Liu, Y.; Yue, H.; Li, A.; Wang, J.; Jiang, B.; Zhang, Y.; Bai, Y. An epidemiologic study on the correlation between oral Helicobacter pylori and gastric H. pylori. Curr. Microbiol. 2009, 58, 449–453. [Google Scholar] [CrossRef]
  69. Kountouras, J.; Polyzos, S.A.; Doulberis, M.; Zeglinas, C.; Artemaki, F.; Vardaka, E.; Deretzi, G.; Giartza-Taxidou, E.; Tzivras, D.; Vlachaki, E.; et al. Potential impact of Helicobacter pylori-related metabolic syndrome on upper and lower gastrointestinal tract oncogenesis. Metab. Clin. Exp. 2018, 87, 18–24. [Google Scholar] [CrossRef]
  70. Kountouras, J.; Papaefthymiou, A. Influence of Helicobacter pylori-connected metabolic syndrome on non-alcoholic fatty liver disease and its related colorectal neoplasm high risk. Liver Int. 2020, 40, 475–476. [Google Scholar] [CrossRef]
  71. Cuomo, P.; Papaianni, M.; Fulgione, A.; Guerra, F.; Capparelli, R.; Medaglia, C. An Innovative Approach to Control H. pylori-Induced Persistent Inflammation and Colonization. Microorganisms 2020, 8, 1214. [Google Scholar] [CrossRef] [PubMed]
  72. Cuomo, P.; Papaianni, M.; Sansone, C.; Iannelli, A.; Iannelli, D.; Medaglia, C.; Paris, D.; Motta, A.; Capparelli, R. An In Vitro Model to Investigate the Role of Helicobacter pylori in Type 2 Diabetes, Obesity, Alzheimer’s Disease and Cardiometabolic Disease. Int. J. Mol. Sci. 2020, 21, 8369. [Google Scholar] [CrossRef] [PubMed]
  73. Salama, N.R.; Hartung, M.L.; Muller, A. Life in the human stomach: Persistence strategies of the bacterial pathogen Helicobacter pylori. Nat. Rev. Microbiol. 2013, 11, 385–399. [Google Scholar] [CrossRef] [PubMed]
  74. Lin, M.M.; Yang, S.S.; Huang, Q.Y.; Cui, G.H.; Jia, X.F.; Yang, Y.; Shi, Z.-M.; Ye, H.; Zhang, X.-Z. Effect and mechanism of Qingre Huashi decoction on drug-resistant Helicobacter pylori. World J. Gastroenterol. 2024, 30, 3086–3105. [Google Scholar] [CrossRef]
  75. Cooke, C.L.; Huff, J.L.; Solnick, J.V. The role of genome diversity and immune evasion in persistent infection with Helicobacter pylori. FEMS Immunol. Med. Microbiol. 2005, 45, 11–23. [Google Scholar] [CrossRef]
  76. Cullen, T.W.; Giles, D.K.; Wolf, L.N.; Ecobichon, C.; Boneca, I.G.; Trent, M.S. Helicobacter pylori versus the host: Remodeling of the bacterial outer membrane is required for survival in the gastric mucosa. PLoS Pathog. 2011, 7, e1002454. [Google Scholar] [CrossRef]
  77. Gebert, B.; Fischer, W.; Weiss, E.; Hoffmann, R.; Haas, R. Helicobacter pylori vacuolating cytotoxin inhibits T lymphocyte activation. Science 2003, 301, 1099–1102. [Google Scholar] [CrossRef]
  78. Kountouras, J.; Papaefthymiou, A.; Gavalas, E.; Polyzos, S.A.; Boziki, M.; Kyriakou, P.; Katsinelos, P.; Zavos, C.; Liatsos, C.; Tzivras, D.; et al. Helicobacter pylori infection as a potential risk factor for multiple sclerosis. Med. Hypotheses 2020, 143, 110135. [Google Scholar] [CrossRef]
  79. Deretzi, G.; Gavalas, E.; Boziki, M.; Tsiptsios, D.; Polyzos, S.A.; Venizelos, I.; Zavos, C.; Koutlas, E.; Tsiptsios, I.; Katsinelos, P.; et al. Impact of Helicobacter pylori on multiple sclerosis-related clinically isolated syndrome. Acta Neurol. Scand. 2016, 133, 268–275. [Google Scholar] [CrossRef]
  80. Kountouras, J.; Tsolaki, M.; Boziki, M.; Gavalas, E.; Zavos, C.; Stergiopoulos, C.; Kapetanakis, N.; Chatzopoulos, D.; Venizelos, I. Association between Helicobacter pylori infection and mild cognitive impairment. Eur. J. Neurol. 2007, 14, 976–982. [Google Scholar] [CrossRef]
  81. Kountouras, J.; Deretzi, G.; Grigoriadis, N.; Zavos, C.; Boziki, M.; Gavalas, E.; Katsinelos, P.; Tzilves, D.; Giouleme, O.; Lazaraki, G. Guillain-Barre syndrome. Lancet Neurol. 2008, 7, 1080–1081, author reply 3–5. [Google Scholar] [CrossRef] [PubMed]
  82. Kountouras, J.; Tsolaki, M.; Gavalas, E.; Boziki, M.; Zavos, C.; Karatzoglou, P.; Chatzopoulos, D.; Venizelos, I. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology 2006, 66, 938–940. [Google Scholar] [CrossRef] [PubMed]
  83. Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Grigoriadis, N.; Tsolaki, M.; Chatzopoulos, D.; Katsinelos, P.; Tzilves, D.; et al. Increased cerebrospinal fluid Helicobacter pylori antibody in Alzheimer’s disease. Int. J. Neurosci. 2009, 119, 765–777. [Google Scholar] [CrossRef] [PubMed]
  84. Boziki, M.; Polyzos, S.A.; Papaefthymiou, A.; Doulberis, M.; Bakirtzis, C.; Sintila, S.A.; Touloumtzi, M.; Grigoriadis, N.; Kountouras, J. Potential impact of Helicobacter pylori-related metabolic syndrome and Galectin-3 on liver, chronic kidney and brain disorders. Metab. Clin. Exp. 2021, 118, 154736. [Google Scholar] [CrossRef]
  85. Doulberis, M.; Kotronis, G.; Thomann, R.; Polyzos, S.A.; Boziki, M.; Gialamprinou, D.; Deretzi, G.; Katsinelos, P.; Kountouras, J. Review: Impact of Helicobacter pylori on Alzheimer’s disease: What do we know so far? Helicobacter 2018, 23, e12454. [Google Scholar] [CrossRef]
  86. Kountouras, J.; Doulberis, M.; Polyzos, S.A.; Katsinelos, T.; Vardaka, E.; Kountouras, C.; Arapoglou, S.; Exadaktylos, A.K.; Deretzi, G.; Tsolaki, M.; et al. Impact of Helicobacter pylori and/or Helicobacter pylori-related metabolic syndrome on incidence of all-cause and Alzheimer’s dementia. Alzheimer’s Dement. 2019, 15, 723–725. [Google Scholar] [CrossRef]
  87. Doulberis, M.; Kotronis, G.; Gialamprinou, D.; Polyzos, S.A.; Papaefthymiou, A.; Katsinelos, P.; Kountouras, J. Alzheimer’s disease and gastrointestinal microbiota; impact of Helicobacter pylori infection involvement. Int. J. Neurosci. 2021, 131, 289–301. [Google Scholar] [CrossRef]
  88. Malaguarnera, M.; Bella, R.; Alagona, G.; Ferri, R.; Carnemolla, A.; Pennisi, G. Helicobacter pylori and Alzheimer’s disease: A possible link. Eur. J. Intern. Med. 2004, 15, 381–386. [Google Scholar] [CrossRef]
  89. Boutajangout, A.; Wisniewski, T. The innate immune system in Alzheimer’s disease. Int. J. Cell Biol. 2013, 2013, 576383. [Google Scholar] [CrossRef]
  90. Hunter, J.M.; Kwan, J.; Malek-Ahmadi, M.; Maarouf, C.L.; Kokjohn, T.A.; Belden, C.; Sabbagh, M.N.; Beach, T.G.; Roher, A.E. Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer’s disease. PLoS ONE 2012, 7, e36893. [Google Scholar] [CrossRef]
  91. Uberti, A.F.; Callai-Silva, N.; Grahl, M.V.C.; Piovesan, A.R.; Nachtigall, E.G.; Furini, C.R.G.; Carlini, C.R. Helicobacter pylori Urease: Potential Contributions to Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 3091. [Google Scholar] [CrossRef] [PubMed]
  92. Sheng, J.G.; Zhu, S.G.; Jones, R.A.; Griffin, W.S.; Mrak, R.E. Interleukin-1 promotes expression and phosphorylation of neurofilament and tau proteins in vivo. Exp. Neurol. 2000, 163, 388–391. [Google Scholar] [CrossRef] [PubMed]
  93. Koch, K.N.; Hartung, M.L.; Urban, S.; Kyburz, A.; Bahlmann, A.S.; Lind, J.; Backert, S.; Taube, C.; Müller, A. Helicobacter urease-induced activation of the TLR2/NLRP3/IL-18 axis protects against asthma. J. Clin. Investig. 2015, 125, 3297–3302. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, X.L.; Zeng, J.; Yang, Y.; Xiong, Y.; Zhang, Z.H.; Qiu, M.; Yan, X.; Sun, X.Y.; Tuo, Q.Z.; Liu, R.; et al. Helicobacter pylori filtrate induces Alzheimer-like tau hyperphosphorylation by activating glycogen synthase kinase-3beta. J. Alzheimer’s Dis. 2015, 43, 153–165. [Google Scholar] [CrossRef]
  95. Kazakos, E.I.; Kountouras, J.; Polyzos, S.A.; Deretzi, G. Novel aspects of defensins’ involvement in virus-induced autoimmunity in the central nervous system. Med. Hypotheses 2017, 102, 33–36. [Google Scholar] [CrossRef]
  96. Pugazhenthi, S. Metabolic Syndrome and the Cellular Phase of Alzheimer’s Disease. Prog. Mol. Biol. Transl. Sci. 2017, 146, 243–258. [Google Scholar]
  97. Di Meco, A.; Li, J.G.; Pratico, D. Dissecting the Role of 5-Lipoxygenase in the Homocysteine-Induced Alzheimer’s Disease Pathology. J. Alzheimer’s Dis. 2018, 62, 1337–1344. [Google Scholar] [CrossRef]
  98. Katsinelos, T.; Doulberis, M.; Polyzos, S.A.; Papaefthymiou, A.; Katsinelos, P.; Kountouras, J. Molecular Links Between Alzheimer’s Disease and Gastrointestinal Microbiota: Emphasis on Helicobacter pylori Infection Involvement. Curr. Mol. Med. 2019, 20, 3–12. [Google Scholar] [CrossRef]
  99. Kountouras, J.; Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A. Impact of Helicobacter pylori-linked metabolic syndrome on non-alcoholic fatty liver disease and its connected atrial fibrillation risk. Liver Int. 2020, 40, 2036–2037. [Google Scholar] [CrossRef]
  100. Passey, S.; Patel, J.; Patail, H.; Aronow, W. Association of Atrial Fibrillation and Cognitive Dysfunction: A Comprehensive Narrative Review of Current Understanding and Recent Updates. J. Clin. Med. 2024, 13, 5581. [Google Scholar] [CrossRef]
  101. Boza-Serrano, A.; Reyes, J.F.; Rey, N.L.; Leffler, H.; Bousset, L.; Nilsson, U.; Brundin, P.; Venero, J.L.; Burguillos, M.A.; Deierborg, T. The role of Galectin-3 in alpha-synuclein-induced microglial activation. Acta Neuropathol. Commun. 2014, 2, 156. [Google Scholar]
  102. Boziki, M.; Grigoriadis, N.; Papaefthymiou, A.; Doulberis, M.; Polyzos, S.A.; Gavalas, E.; Deretzi, G.; Karafoulidou, E.; Kesidou, E.; Taloumtzis, C.; et al. The trimebutine effect on Helicobacter pylori-related gastrointestinal tract and brain disorders: A hypothesis. Neurochem. Int. 2021, 144, 104938. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, J.; Liang, J.; Hu, N.; He, N.; Liu, B.; Liu, G.; Qin, Y. The Gut Microbiota Modulates Neuroinflammation in Alzheimer’s Disease: Elucidating Crucial Factors and Mechanistic Underpinnings. CNS Neurosci. Ther. 2024, 30, e70091. [Google Scholar] [CrossRef] [PubMed]
  104. Kountouras, J.; Boziki, M.; Gavalas, E.; Zavos, C.; Deretzi, G.; Chatzigeorgiou, S.; Katsinelos, P.; Grigoriadis, N.; Giartza-Taxidou, E.; Venizelos, I. Five-year survival after Helicobacter pylori eradication in Alzheimer disease patients. Cogn. Behav. Neurol. 2010, 23, 199–204. [Google Scholar] [CrossRef] [PubMed]
  105. Noto, J.M.; Peek, R.M., Jr. The gastric microbiome, its interaction with Helicobacter pylori, and its potential role in the progression to stomach cancer. PLoS Pathog. 2017, 13, e1006573. [Google Scholar] [CrossRef] [PubMed]
  106. Deretzi, G.; Kountouras, J.; Polyzos, S.A.; Zavos, C.; Giartza-Taxidou, E.; Gavalas, E.; Deretzi, G.; Karafoulidou, E.; Kesidou, E.; Taloumtzis, C.; et al. Gastrointestinal immune system and brain dialogue implicated in neuroinflammatory and neurodegenerative diseases. Curr. Mol. Med. 2011, 11, 696–707. [Google Scholar] [CrossRef]
  107. Kountouras, J.; Zavos, C.; Polyzos, S.A.; Deretzi, G.; Vardaka, E.; Giartza-Taxidou, E.; Katsinelos, P.; Rapti, E.; Chatzopoulos, D.; Tzilves, D.; et al. Helicobacter pylori infection and Parkinson’s disease: Apoptosis as an underlying common contributor. Eur. J. Neurol. 2012, 19, e56. [Google Scholar] [CrossRef]
  108. Suwarnalata, G.; Tan, A.H.; Isa, H.; Gudimella, R.; Anwar, A.; Loke, M.F.; Mahadeva, S.; Lim, S.Y.; Vadivelu, J. Augmentation of Autoantibodies by Helicobacter pylori in Parkinson’s Disease Patients May Be Linked to Greater Severity. PLoS ONE 2016, 11, e0153725. [Google Scholar] [CrossRef]
  109. Lahner, E.; Annibale, B.; Delle Fave, G. Systematic review: Helicobacter pylori infection and impaired drug absorption. Aliment. Pharmacol. Ther. 2009, 29, 379–386. [Google Scholar] [CrossRef]
  110. Sian-Hulsmann, J. Wilful pathogens provoke a gut feeling in Parkinson’s disease. J. Neural Transm. 2022, 129, 557–562. [Google Scholar] [CrossRef]
  111. Zhao, Z.; Li, F.; Ning, J.; Peng, R.; Shang, J.; Liu, H.; Shang, M.; Bao, X.Q.; Zhang, D. Novel compound FLZ alleviates rotenone-induced PD mouse model by suppressing TLR4/MyD88/NF-kappaB pathway through microbiota-gut-brain axis. Acta Pharm Sin B 2021, 11, 2859–2879. [Google Scholar] [CrossRef] [PubMed]
  112. Nuzum, N.D.; Loughman, A.; Szymlek-Gay, E.A.; Hendy, A.; Teo, W.P.; Macpherson, H. Gut microbiota differences between healthy older adults and individuals with Parkinson’s disease: A systematic review. Neurosci. Biobehav. Rev. 2020, 112, 227–241. [Google Scholar] [CrossRef] [PubMed]
  113. Shen, S.; Zhang, C.; Xu, Y.M.; Shi, C.H. The Role of Pathogens and Anti-Infective Agents in Parkinson’s Disease, from Etiology to Therapeutic Implications. J. Park. Dis. 2022, 12, 27–44. [Google Scholar] [CrossRef]
  114. Tan, A.H.; Lim, S.Y.; Mahadeva, S.; Loke, M.F.; Tan, J.Y.; Ang, B.H.; Chin, K.P.; Adnan, A.F.M.; Ong, S.M.C.; Ibrahim, A.I.; et al. Helicobacter pylori Eradication in Parkinson’s Disease: A Randomized Placebo-Controlled Trial. Mov. Disord. 2020, 35, 2250–2260. [Google Scholar] [CrossRef]
  115. Franceschi, F.; Gasbarrini, A.; Polyzos, S.A.; Kountouras, J. Extragastric Diseases and Helicobacter pylori. Helicobacter 2015, 20 (Suppl. S1), 40–46. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, Y.; Li, R.; Xue, X.; Xu, T.; Luo, Y.; Dong, Q.; Liu, J.; Liu, J.; Pan, Y.; Zhang, D. Periodontal disease and Helicobacter pylori infection in oral cavity: A meta-analysis of 2727 participants mainly based on Asian studies. Clin. Oral Investig. 2020, 24, 2175–2188. [Google Scholar] [CrossRef]
  117. Alvarenga, M.O.P.; Frazao, D.R.; de Matos, I.G.; Bittencourt, L.O.; Fagundes, N.C.F.; Rosing, C.K.; Maia, L.C.; Lima, R.R. Is There Any Association Between Neurodegenerative Diseases and Periodontitis? A Systematic Review. Front. Aging Neurosci. 2021, 13, 651437. [Google Scholar] [CrossRef]
  118. Hu, S.; Li, S.; Ning, W.; Huang, X.; Liu, X.; Deng, Y.; Franceschi, D.; Ogbuehi, A.C.; Lethaus, B.; Savkovic, V.; et al. Identifying crosstalk genetic biomarkers linking a neurodegenerative disease, Parkinson’s disease, and periodontitis using integrated bioinformatics analyses. Front. Aging Neurosci. 2022, 14, 1032401. [Google Scholar] [CrossRef]
  119. Andrich, J.E.; Wobben, M.; Klotz, P.; Goetze, O.; Saft, C. Upper gastrointestinal findings in Huntington’s disease: Patients suffer but do not complain. J. Neural Transm. 2009, 116, 1607–1611. [Google Scholar] [CrossRef]
  120. Nishihara, H.; Shimizu, F.; Kitagawa, T.; Yamanaka, N.; Akada, J.; Kuramitsu, Y.; Sano, Y.; Takeshita, Y.; Maeda, T.; Abe, M.; et al. Identification of galectin-3 as a possible antibody target for secondary progressive multiple sclerosis. Mult. Scler. 2017, 23, 382–394. [Google Scholar] [CrossRef]
  121. Boziki, M.; Polyzos, S.A.; Deretzi, G.; Kazakos, E.; Katsinelos, P.; Doulberis, M.; Kotronis, G.; Giartza-Taxidou, E.; Laskaridis, L.; Tzivras, D.; et al. A potential impact of Helicobacter pylori-related galectin-3 in neurodegeneration. Neurochem. Int. 2018, 113, 137–151. [Google Scholar] [CrossRef] [PubMed]
  122. Stancic, M.; van Horssen, J.; Thijssen, V.L.; Gabius, H.J.; van der Valk, P.; Hoekstra, D.; Baron, W. Increased expression of distinct galectins in multiple sclerosis lesions. Neuropathol. Appl. Neurobiol. 2011, 37, 654–671. [Google Scholar] [CrossRef] [PubMed]
  123. Haines, J.D.; Vidaurre, O.G.; Zhang, F.; Riffo-Campos, A.L.; Castillo, J.; Casanova, B.; Casaccia, P.; Lopez-Rodas, G. Multiple sclerosis patient-derived CSF induces transcriptional changes in proliferating oligodendrocyte progenitors. Mult. Scler. 2015, 21, 1655–1669. [Google Scholar] [CrossRef] [PubMed]
  124. Srejovic, I.; Selakovic, D.; Jovicic, N.; Jakovljevic, V.; Lukic, M.L.; Rosic, G. Galectin-3: Roles in Neurodevelopment, Neuroinflammation, and Behavior. Biomolecules 2020, 10, 798. [Google Scholar] [CrossRef] [PubMed]
  125. Gavalas, E.; Kountouras, J.; Boziki, M.; Zavos, C.; Polyzos, S.A.; Vlachaki, E.; Venizelos, I.; Tsiptsios, D.; Deretzi, G. Relationship between Helicobacter pylori infection and multiple sclerosis. Ann. Gastroenterol. 2015, 28, 353–356. [Google Scholar]
  126. Arjmandi, D.; Abdollahi, A.; Ardekani, A.; Razavian, I.; Razavian, E.; Sartip, B.; Mahjour, S.; Parsa, H.; Kyvanani, N.A.; Marhoommirzabak, E.; et al. Helicobacter pylori infection and risk of multiple sclerosis: An updated meta-analysis. Helicobacter 2022, 27, e12927. [Google Scholar] [CrossRef]
  127. Shishkina, V.V.; Klochkova, S.V.; Alexeeva, N.T.; Samodurova, N.Y.; Nikityuk, D.V. [Discussion of the immunomorphological role of interactions between mast cells and Helicobacter pylori in the gastric mucosa]. Probl. Nutr. 2022, 91, 98–108. [Google Scholar] [CrossRef]
  128. Kountouras, J.; Boziki, M.; Polyzos, S.A.; Katsinelos, P.; Gavalas, E.; Zeglinas, C.; Tzivras, D.; Romiopoulos, I.; Giorgakis, N.; Anastasiadou, K.; et al. The Emerging Role of Helicobacter Pylori-Induced Metabolic Gastrointestinal Dysmotility and Neurodegeneration. Curr. Mol. Med. 2017, 17, 389–404. [Google Scholar] [CrossRef]
  129. Kountouras, J.; Doulberis, M.; Papaefthymiou, A.; Polyzos, S.A.; Zavos, C.; Kazakos, E.; Arapoglou, S.; Kyrailidi, F.; Mouratidou, M.C.; Boziki, M.; et al. Controlling the Impact of Helicobacter pylori-Related Hyperhomocysteinemia on Neurodegeneration. Medicina 2023, 59, 504. [Google Scholar] [CrossRef]
  130. Kountouras, J.; Zavos, C.; Polyzos, S.A.; Deretzi, G. Potential impact of Helicobacter pylori-related human beta-defensin-1 on hepatic encephalopathy and neurodegeneration. Ann. Gastroenterol. 2016, 29, 99. [Google Scholar]
  131. Strajtenberger, M.; Stipic-Markovic, A.; Barac, E.; Artukovic, M.; Lugovic-Mihic, L. Human beta-defensin 2: A connection between infections and allergic skin diseases. Acta Dermatovenerol. Alp. Pannonica Adriat. 2024, 33, 135–139. [Google Scholar]
  132. Gordon, R.; Albornoz, E.A.; Christie, D.C.; Langley, M.R.; Kumar, V.; Mantovani, S.; Robertson, A.A.B.; Butler, M.S.; Rowe, D.B.; O’neill, L.A.; et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci. Transl. Med. 2018, 10, eaah4066. [Google Scholar] [CrossRef] [PubMed]
  133. Dandrea, M.R.; Reiser, P.A.; Gumula, N.A.; Hertzog, B.M.; Andrade-Gordon, P. Application of triple immunohistochemistry to characterize amyloid plaque-associated inflammation in brains with Alzheimer’s disease. Biotech. Histochem. 2001, 76, 97–106. [Google Scholar] [CrossRef]
  134. Mehlhorn, G.; Hollborn, M.; Schliebs, R. Induction of cytokines in glial cells surrounding cortical beta-amyloid plaques in transgenic Tg2576 mice with Alzheimer pathology. Int. J. Dev. Neurosci. 2000, 18, 423–431. [Google Scholar] [CrossRef]
  135. Harcha, P.A.; Vargas, A.; Yi, C.; Koulakoff, A.A.; Giaume, C.; Saez, J.C. Hemichannels Are Required for Amyloid beta-Peptide-Induced Degranulation and Are Activated in Brain Mast Cells of APPswe/PS1dE9 Mice. J. Neurosci. 2015, 35, 9526–9538. [Google Scholar] [CrossRef]
  136. Laurent, C.; Buee, L.; Blum, D. Tau and neuroinflammation: What impact for Alzheimer’s Disease and Tauopathies? Biomed. J. 2018, 41, 21–33. [Google Scholar] [CrossRef] [PubMed]
  137. Harcha, P.A.; Garces, P.; Arredondo, C.; Fernandez, G.; Saez, J.C.; van Zundert, B. Mast Cell and Astrocyte Hemichannels and Their Role in Alzheimer’s Disease, ALS, and Harmful Stress Conditions. Int. J. Mol. Sci. 2021, 22, 1924. [Google Scholar] [CrossRef]
  138. Tuppo, E.E.; Arias, H.R. The role of inflammation in Alzheimer’s disease. Int. J. Biochem. Cell Biol. 2005, 37, 289–305. [Google Scholar] [CrossRef] [PubMed]
  139. Giovannini, M.G.; Scali, C.; Prosperi, C.; Bellucci, A.; Vannucchi, M.G.; Rosi, S.; Pepeu, G.; Casamenti, F. Beta-amyloid-induced inflammation and cholinergic hypofunction in the rat brain in vivo: Involvement of the p38MAPK pathway. Neurobiol. Dis. 2002, 11, 257–274. [Google Scholar] [CrossRef]
  140. Liang, H.Y.; Chen, Y.; Wei, X.; Ma, G.G.; Ding, J.; Lu, C.; Zhou, R.-P.; Hu, W. Immunomodulatory functions of TRPM7 and its implications in autoimmune diseases. Immunology 2022, 165, 3–21. [Google Scholar] [CrossRef]
  141. Mattila, O.S.; Strbian, D.; Saksi, J.; Pikkarainen, T.O.; Rantanen, V.; Tatlisumak, T.; Lindsberg, P.J. Cerebral mast cells mediate blood-brain barrier disruption in acute experimental ischemic stroke through perivascular gelatinase activation. Stroke 2011, 42, 3600–3605. [Google Scholar] [CrossRef] [PubMed]
  142. Kempuraj, D.; Ahmed, M.E.; Selvakumar, G.P.; Thangavel, R.; Dhaliwal, A.S.; Dubova, I.; Mentor, S.; Premkumar, K.; Saeed, D.; Zahoor, H.; et al. Brain Injury-Mediated Neuroinflammatory Response and Alzheimer’s Disease. Neuroscientist 2020, 26, 134–155. [Google Scholar] [CrossRef] [PubMed]
  143. 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] [PubMed]
  144. Meyer, P.F.; Savard, M.; Poirier, J.; Labonte, A.; Rosa-Neto, P.; Weitz, T.M.; Town, T.; Breitner, J. Bi-directional Association of Cerebrospinal Fluid Immune Markers with Stage of Alzheimer’s Disease Pathogenesis. J. Alzheimer’s Dis. 2018, 63, 577–590. [Google Scholar] [CrossRef]
  145. Varricchi, G.; Loffredo, S.; Galdiero, M.R.; Marone, G.; Cristinziano, L.; Granata, F. Innate effector cells in angiogenesis and lymphangiogenesis. Curr. Opin Immunol. 2018, 53, 152–160. [Google Scholar] [CrossRef]
  146. Kempuraj, D.; Thangavel, R.; Selvakumar, G.P.; Ahmed, M.E.; Zaheer, S.; Raikwar, S.P.; Zahoor, H.; Saeed, D.; Dubova, I.; Giler, G.; et al. Mast Cell Proteases Activate Astrocytes and Glia-Neurons and Release Interleukin-33 by Activating p38 and ERK1/2 MAPKs and NF-kappaB. Mol. Neurobiol. 2019, 56, 1681–1693. [Google Scholar] [CrossRef]
  147. Karagkouni, A.; Alevizos, M.; Theoharides, T.C. Effect of stress on brain inflammation and multiple sclerosis. Autoimmun. Rev. 2013, 12, 947–953. [Google Scholar] [CrossRef]
  148. Selvakumar, G.P.; Manivasagam, T.; Rekha, K.R.; Jayaraj, R.L.; Elangovan, N. Escin, a novel triterpene, mitigates chronic MPTP/p-induced dopaminergic toxicity by attenuating mitochondrial dysfunction, oxidative stress, and apoptosis. J. Mol. Neurosci. 2015, 55, 184–197. [Google Scholar] [CrossRef]
  149. Zhang, X.; Shao, Z.; Xu, S.; Liu, Q.; Liu, C.; Luo, Y.; Jin, L.; Li, S. Immune Profiling of Parkinson’s Disease Revealed Its Association With a Subset of Infiltrating Cells and Signature Genes. Front. Aging Neurosci. 2021, 13, 605970. [Google Scholar] [CrossRef]
  150. Kempuraj, D.; Selvakumar, G.P.; Zaheer, S.; Thangavel, R.; Ahmed, M.E.; Raikwar, S.; Govindarajan, R.; Iyer, S.; Zaheer, A. Cross-Talk between Glia, Neurons and Mast Cells in Neuroinflammation Associated with Parkinson’s Disease. J. Neuroimmune Pharmacol. 2018, 13, 100–112. [Google Scholar] [CrossRef]
  151. Fathi, M.; Vakili, K.; Yaghoobpoor, S.; Qadirifard, M.S.; Kosari, M.; Naghsh, N.; Taei, A.A.; Klegeris, A.; Dehghani, M.; Bahrami, A.; et al. Pre-clinical Studies Identifying Molecular Pathways of Neuroinflammation in Parkinson’s Disease: A Systematic Review. Front. Aging Neurosci. 2022, 14, 855776. [Google Scholar] [CrossRef] [PubMed]
  152. Selvakumar, G.P.; Ahmed, M.E.; Thangavel, R.; Kempuraj, D.; Dubova, I.; Raikwar, S.P.; Zaheer, S.; Iyer, S.S.; Zaheer, A. A role for glia maturation factor dependent activation of mast cells and microglia in MPTP induced dopamine loss and behavioural deficits in mice. Brain Behav. Immun. 2020, 87, 429–443. [Google Scholar] [CrossRef] [PubMed]
  153. Hong, G.U.; Cho, J.W.; Kim, S.Y.; Shin, J.H.; Ro, J.Y. Inflammatory mediators resulting from transglutaminase 2 expressed in mast cells contribute to the development of Parkinson’s disease in a mouse model. Toxicol. Appl. Pharmacol. 2018, 358, 10–22. [Google Scholar] [CrossRef]
  154. von Herrmann, K.M.; Salas, L.A.; Martinez, E.M.; Young, A.L.; Howard, J.M.; Feldman, M.S.; Christensen, B.C.; Wilkins, O.M.; Lee, S.L.; Hickey, W.F.; et al. NLRP3 expression in mesencephalic neurons and characterization of a rare NLRP3 polymorphism associated with decreased risk of Parkinson’s disease. npj Park. Dis. 2018, 4, 24. [Google Scholar] [CrossRef]
  155. Chatterjee, K.; Roy, A.; Banerjee, R.; Choudhury, S.; Mondal, B.; Halder, S.; Basu, P.; Shubham, S.; Dey, S.; Kumar, H. Inflammasome and alpha-synuclein in Parkinson’s disease: A cross-sectional study. J. Neuroimmunol. 2020, 338, 577089. [Google Scholar] [CrossRef]
  156. Nakamura, Y.; Kambe, N.; Saito, M.; Nishikomori, R.; Kim, Y.G.; Murakami, M.; Núñez, G.; Matsue, H. Mast cells mediate neutrophil recruitment and vascular leakage through the NLRP3 inflammasome in histamine-independent urticaria. J. Exp. Med. 2009, 206, 1037–1046. [Google Scholar] [CrossRef]
  157. Trias, E.; King, P.H.; Si, Y.; Kwon, Y.; Varela, V.; Ibarburu, S.; Kovacs, M.; Moura, I.C.; Beckman, J.S.; Hermine, O.; et al. Mast cells and neutrophils mediate peripheral motor pathway degeneration in ALS. JCI Insight 2018, 3, e123249. [Google Scholar] [CrossRef]
  158. Engelhardt, J.I.; Appel, S.H. IgG reactivity in the spinal cord and motor cortex in amyotrophic lateral sclerosis. Arch. Neurol. 1990, 47, 1210–1216. [Google Scholar] [CrossRef] [PubMed]
  159. Chiot, A.; Lobsiger, C.S.; Boillee, S. New insights on the disease contribution of neuroinflammation in amyotrophic lateral sclerosis. Curr. Opin. Neurol. 2019, 32, 764–770. [Google Scholar] [CrossRef]
  160. Trias, E.; Ibarburu, S.; Barreto-Nunez, R.; Varela, V.; Moura, I.C.; Dubreuil, P.; Hermine, O.; Beckman, J.S.; Barbeito, L. Evidence for mast cells contributing to neuromuscular pathology in an inherited model of ALS. JCI Insight 2017, 2, e95934. [Google Scholar] [CrossRef]
  161. Ashina, K.; Tsubosaka, Y.; Nakamura, T.; Omori, K.; Kobayashi, K.; Hori, M.; Ozaki, H.; Murata, T. Histamine Induces Vascular Hyperpermeability by Increasing Blood Flow and Endothelial Barrier Disruption In Vivo. PLoS ONE 2015, 10, e0132367. [Google Scholar] [CrossRef] [PubMed]
  162. He, S.; Walls, A.F. Human mast cell chymase induces the accumulation of neutrophils, eosinophils and other inflammatory cells in vivo. Br. J. Pharmacol. 1998, 125, 1491–1500. [Google Scholar] [CrossRef] [PubMed]
  163. Christy, A.L.; Walker, M.E.; Hessner, M.J.; Brown, M.A. Mast cell activation and neutrophil recruitment promotes early and robust inflammation in the meninges in EAE. J. Autoimmun. 2013, 42, 50–61. [Google Scholar] [CrossRef]
  164. Kovacs, M.; Alamon, C.; Maciel, C.; Varela, V.; Ibarburu, S.; Tarrago, L.; King, P.H.; Si, Y.; Kwon, Y.; Hermine, O.; et al. The pathogenic role of c-Kit+ mast cells in the spinal motor neuron-vascular niche in ALS. Acta Neuropathol. Commun. 2021, 9, 136. [Google Scholar] [CrossRef]
  165. Faideau, M.; Kim, J.; Cormier, K.; Gilmore, R.; Welch, M.; Auregan, G.; Dufour, N.; Guillermier, M.; Brouillet, E.; Hantraye, P.; et al. In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: A correlation with Huntington’s disease subjects. Hum. Mol. Genet. 2010, 19, 3053–3067. [Google Scholar] [CrossRef]
  166. Perez-Rodriguez, M.J.; Ibarra-Sanchez, A.; Roman-Figueroa, A.; Perez-Severiano, F.; Gonzalez-Espinosa, C. Mutant Huntingtin affects toll-like receptor 4 intracellular trafficking and cytokine production in mast cells. J. Neuroinflamm. 2020, 17, 95. [Google Scholar] [CrossRef] [PubMed]
  167. Sapp, E.; Kegel, K.B.; Aronin, N.; Hashikawa, T.; Uchiyama, Y.; Tohyama, K.; Bhide, P.G.; Vonsattel, J.P.; Difiglia, M. Early and progressive accumulation of reactive microglia in the Huntington disease brain. J. Neuropathol. Exp. Neurol. 2001, 60, 161–172. [Google Scholar] [CrossRef]
  168. Gandhi, R.; Laroni, A.; Weiner, H.L. Role of the innate immune system in the pathogenesis of multiple sclerosis. J. Neuroimmunol. 2010, 221, 7–14. [Google Scholar] [CrossRef]
  169. Conti, P.; Kempuraj, D. Important role of mast cells in multiple sclerosis. Mult. Scler. Relat. Disord. 2016, 5, 77–80. [Google Scholar] [CrossRef]
  170. Silver, R.; Silverman, A.J.; Vitkovic, L.; Lederhendler, I.I. Mast cells in the brain: Evidence and functional significance. Trends Neurosci. 1996, 19, 25–31. [Google Scholar] [CrossRef]
  171. Russi, A.E.; Walker-Caulfield, M.E.; Brown, M.A. Mast cell inflammasome activity in the meninges regulates EAE disease severity. Clin. Immunol. 2018, 189, 14–22. [Google Scholar] [CrossRef]
  172. Dimitriadou, V.; Pang, X.; Theoharides, T.C. Hydroxyzine inhibits experimental allergic encephalomyelitis (EAE) and associated brain mast cell activation. Int. J. Immunopharmacol. 2000, 22, 673–684. [Google Scholar] [CrossRef]
  173. Secor, V.H.; Secor, W.E.; Gutekunst, C.A.; Brown, M.A. Mast cells are essential for early onset and severe disease in a murine model of multiple sclerosis. J. Exp. Med. 2000, 191, 813–822. [Google Scholar] [CrossRef]
  174. Jack, C.R., Jr.; Knopman, D.S.; Jagust, W.J.; Shaw, L.M.; Aisen, P.S.; Weiner, M.W.; Petersen, R.C.; Trojanowski, J.Q. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9, 119–128. [Google Scholar] [CrossRef]
  175. Zhang, H.; Cao, S.; Xu, Y.; Sun, X.; Fei, M.; Jing, Q.; Xu, X.; Tang, J.; Niu, B.; Li, C. Landscape of immune infiltration in entorhinal cortex of patients with Alzheimer’s disease. Front. Pharmacol. 2022, 13, 941656. [Google Scholar]
  176. Novellino, F.; Sacca, V.; Donato, A.; Zaffino, P.; Spadea, M.F.; Vismara, M.; Arcidiacono, B.; Malara, N.; Presta, I.; Donato, G. Innate Immunity: A Common Denominator between Neurodegenerative and Neuropsychiatric Diseases. Int. J. Mol. Sci. 2020, 21, 1115. [Google Scholar] [CrossRef]
  177. Jing, Q.; Zhang, H.; Sun, X.; Xu, Y.; Cao, S.; Fang, Y.; Zhao, X.; Li, C. A Comprehensive Analysis Identified Hub Genes and Associated Drugs in Alzheimer’s Disease. Biomed. Res. Int. 2021, 2021, 8893553. [Google Scholar] [CrossRef]
  178. Bhuiyan, P.; Wang, Y.W.; Sha, H.H.; Dong, H.Q.; Qian, Y.N. Neuroimmune connections between corticotropin-releasing hormone and mast cells: Novel strategies for the treatment of neurodegenerative diseases. Neural Regen. Res. 2021, 16, 2184–2197. [Google Scholar]
  179. Soria Lopez, J.A.; Gonzalez, H.M.; Leger, G.C. Alzheimer’s disease. Handb. Clin. Neurol. 2019, 167, 231–255. [Google Scholar] [PubMed]
  180. Salani, F.; Sterbini, V.; Sacchinelli, E.; Garramone, M.; Bossu, P. Is Innate Memory a Double-Edge Sword in Alzheimer’s Disease? A Reappraisal of New Concepts and Old Data. Front. Immunol. 2019, 10, 1768. [Google Scholar] [CrossRef] [PubMed]
  181. Lopez, X.; Escamilla, R.; Fernandez, P.; Duarte, Y.; Gonzalez-Nilo, F.; Palacios-Prado, N.; Martinez, A.D.; Sáez, J.C. Stretch-Induced Activation of Pannexin 1 Channels Can Be Prevented by PKA-Dependent Phosphorylation. Int. J. Mol. Sci. 2020, 21, 9180. [Google Scholar] [CrossRef]
  182. Orellana, J.A.; Shoji, K.F.; Abudara, V.; Ezan, P.; Amigou, E.; Saez, P.J.; Giaume, C. Amyloid beta-induced death in neurons involves glial and neuronal hemichannels. J. Neurosci. 2011, 31, 4962–4977. [Google Scholar] [CrossRef] [PubMed]
  183. Stokely, M.E.; Orr, E.L. Acute effects of calvarial damage on dural mast cells, pial vascular permeability, and cerebral cortical histamine levels in rats and mice. J. Neurotrauma 2008, 25, 52–61. [Google Scholar] [CrossRef] [PubMed]
  184. Esposito, P.; Gheorghe, D.; Kandere, K.; Pang, X.; Connolly, R.; Jacobson, S.; Theoharides, T.C. Acute stress increases permeability of the blood-brain-barrier through activation of brain mast cells. Brain Res. 2001, 888, 117–127. [Google Scholar] [CrossRef] [PubMed]
  185. Moretti, R.; Chhor, V.; Bettati, D.; Banino, E.; De Lucia, S.; Le Charpentier, T.; Lebon, S.; Schwendimann, L.; Pansiot, J.; Rasika, S.; et al. Contribution of mast cells to injury mechanisms in a mouse model of pediatric traumatic brain injury. J. Neurosci. Res. 2016, 94, 1546–1560. [Google Scholar] [CrossRef]
  186. Machado, A.; Herrera, A.J.; de Pablos, R.M.; Espinosa-Oliva, A.M.; Sarmiento, M.; Ayala, A.; Venero, J.L.; Santiago, M.; Villarán, R.F.; Delgado-Cortés, M.J.; et al. Chronic stress as a risk factor for Alzheimer’s disease. Rev. Neurosci. 2014, 25, 785–804. [Google Scholar] [CrossRef]
  187. Shaik-Dasthagirisaheb, Y.B.; Conti, P. The Role of Mast Cells in Alzheimer’s Disease. Adv. Clin. Exp. Med. 2016, 25, 781–787. [Google Scholar] [CrossRef]
  188. Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction, Oxidative Stress, and Neuroinflammation: Intertwined Roads to Neurodegeneration. Antioxidants 2020, 9, 647. [Google Scholar] [CrossRef]
  189. Jarrott, B.; Williams, S.J. Chronic Brain Inflammation: The Neurochemical Basis for Drugs to Reduce Inflammation. Neurochem. Res. 2016, 41, 523–533. [Google Scholar] [CrossRef]
  190. Fan, Z.; Pan, Y.T.; Zhang, Z.Y.; Yang, H.; Yu, S.Y.; Zheng, Y.; Ma, J.-H.; Wang, X.-M. Systemic activation of NLRP3 inflammasome and plasma alpha-synuclein levels are correlated with motor severity and progression in Parkinson’s disease. J. Neuroinflamm. 2020, 17, 11. [Google Scholar] [CrossRef]
  191. Mpofana, T.; Daniels, W.M.; Mabandla, M.V. Exposure to Early Life Stress Results in Epigenetic Changes in Neurotrophic Factor Gene Expression in a Parkinsonian Rat Model. Park. Dis. 2016, 2016, 6438783. [Google Scholar] [CrossRef] [PubMed]
  192. Refojo, D.; Schweizer, M.; Kuehne, C.; Ehrenberg, S.; Thoeringer, C.; Vogl, A.M.; Dedic, N.; Schumacher, M.; von Wolff, G.; Avrabos, C.; et al. Glutamatergic and dopaminergic neurons mediate anxiogenic and anxiolytic effects of CRHR1. Science 2011, 333, 1903–1907. [Google Scholar] [CrossRef] [PubMed]
  193. Brown, R.H.; Al-Chalabi, A. Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2017, 377, 162–172. [Google Scholar] [CrossRef] [PubMed]
  194. Feldman, E.L.; Goutman, S.A.; Petri, S.; Mazzini, L.; Savelieff, M.G.; Shaw, P.J.; Sobue, G. Amyotrophic lateral sclerosis. Lancet 2022, 400, 1363–1380. [Google Scholar] [CrossRef]
  195. Zuccato, C.; Cattaneo, E. Huntington’s disease. Handb. Exp. Pharmacol. 2014, 220, 357–409. [Google Scholar]
  196. Federation, M.I. Atlas of MS 2023. Available online: https://www.atlasofms.org/map/global/epidemiology/number-of-people-with-ms (accessed on 1 August 2024).
  197. McGinley, M.P.; Goldschmidt, C.H.; Rae-Grant, A.D. Diagnosis and Treatment of Multiple Sclerosis: A Review. JAMA 2021, 325, 765–779. [Google Scholar] [CrossRef]
  198. Rolak, L.A.; Fleming, J.O. The differential diagnosis of multiple sclerosis. Neurologist 2007, 13, 57–72. [Google Scholar] [CrossRef]
  199. Bakirtzis, C.; Grigoriadou, E.; Boziki, M.K.; Kesidou, E.; Siafis, S.; Moysiadis, T.; Tsakona, D.; Thireos, E.; Nikolaidis, I.; Pourzitaki, C.; et al. The Administrative Prevalence of Multiple Sclerosis in Greece on the Basis of a Nationwide Prescription Database. Front. Neurol. 2020, 11, 1012. [Google Scholar] [CrossRef]
  200. Lassmann, H. Multiple Sclerosis Pathology. Cold Spring Harb. Perspect. Med. 2018, 8, a028936. [Google Scholar] [CrossRef]
  201. Kesidou, E.; Theotokis, P.; Damianidou, O.; Boziki, M.; Konstantinidou, N.; Taloumtzis, C.; Sintila, S.-A.; Grigoriadis, P.; Evangelopoulos, M.E.; Bakirtzis, C.; et al. CNS Ageing in Health and Neurodegenerative Disorders. J. Clin. Med. 2023, 12, 2255. [Google Scholar] [CrossRef]
  202. Planas-Fontánez, T.M.; Sainato, D.M.; Sharma, I.; Dreyfus, C.F. Roles of astrocytes in response to aging, Alzheimer’s disease and multiple sclerosis. Brain Res. 2021, 1764, 147464. [Google Scholar] [CrossRef] [PubMed]
  203. Boziki, M.; Pechlivanis, A.; Virgiliou, C. Blood Metabolomics May Discriminate a Sub-Group of Patients with First Demyelinating Episode in the Context of RRMS with Increased Disability and MRI Characteristics Indicative of Poor Prognosis. Int. J. Mol. Sci. 2022, 23, 14578. [Google Scholar] [CrossRef]
  204. Boziki, M.; Bakirtzis, C. Ocrelizumab in Patients with Active Primary Progressive Multiple Sclerosis: Clinical Outcomes and Immune Markers of Treatment Response. Cells 2022, 11, 1959. [Google Scholar] [CrossRef] [PubMed]
  205. Pousinis, P.; Begou, O. Recent Advances in Metabolomics and Lipidomics Studies in Human and Animal Models of Multiple Sclerosis. Metabolites 2024, 14, 545. [Google Scholar] [CrossRef] [PubMed]
  206. Lublin, F.D.; Reingold, S.C.; Cohen, J.A.; Cutter, G.R.; Sørensen, P.S.; Thompson, A.J.; Wolinsky, J.S.; Balcer, L.J.; Banwell, B.; Barkhof, F.; et al. Defining the clinical course of multiple sclerosis: The 2013 revisions. Neurology 2014, 83, 278–286. [Google Scholar] [CrossRef]
  207. Boziki, M.; Theotokis, P.; Kesidou, E.; Karafoulidou, E.; Konstantinou, C.; Michailidou, I.; Bahar, Y.; Altintas, A.; Grigoriadis, N. Sex, aging and immunity in multiple sclerosis and experimental autoimmune encephalomyelitis: An intriguing interaction. Front. Neurol. 2022, 13, 1104552. [Google Scholar] [CrossRef]
  208. Dermitzakis, I.; Manthou, M.E.; Meditskou, S.; Miliaras, D.; Kesidou, E.; Boziki, M.; Petratos, S.; Grigoriadis, N.; Theotokis, P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr. Issues Mol. Biol. 2022, 44, 3208–3237. [Google Scholar] [CrossRef]
  209. Boziki, M.; Bakirtzis, C.; Giantzi, V.; Sintila, S.A.; Kallivoulos, S.; Afrantou, T.; Nikolaidis, I.; Ioannidis, P.; Karapanayiotides, T.; Koutroulou, I.; et al. Long-Term Efficacy Outcomes of Natalizumab vs. Fingolimod in Patients With Highly Active Relapsing-Remitting Multiple Sclerosis: Real-World Data From a Multiple Sclerosis Reference Center. Front. Neurol. 2021, 12, 699844. [Google Scholar] [CrossRef]
  210. Boziki, M.; Lagoudaki, R.; Melo, P.; Kanidou, F.; Bakirtzis, C.; Grigoriadou, E.; Afrantou, T.; Tatsi, T.; Matsi, S.; Grigoriadis, N. Induction of apoptosis in CD4(+) T-cells is linked with optimal treatment response in patients with relapsing-remitting multiple sclerosis treated with Glatiramer acetate. J. Neurol. Sci. 2019, 401, 43–50. [Google Scholar] [CrossRef]
  211. Bakirtzis, C.; Konstantinopoulou, E.; Langdon, D.W.; Grigoriadou, E.; Minti, F.; Nikolaidis, I.; Boziki, M.K.; Tatsi, T.; Ioannidis, P.; Karapanayiotides, T.; et al. Long-term effects of prolonged-release fampridine in cognitive function, fatigue, mood and quality of life of MS patients: The IGNITE study. J. Neurol. Sci. 2018, 395, 106–112. [Google Scholar] [CrossRef]
  212. Polychroniadou, E.; Bakirtzis, C.; Langdon, D.; Lagoudaki, R.; Kesidou, E.; Theotokis, P.; Tsalikakis, D.; Poulatsidou, K.; Kyriazis, O.; Boziki, M.; et al. Validation of the Brief International Cognitive Assessment for Multiple Sclerosis (BICAMS) in Greek population with multiple sclerosis. Mult. Scler. Relat. Disord. 2016, 9, 68–72. [Google Scholar] [CrossRef] [PubMed]
  213. De Meo, E.; Portaccio, E.; Giorgio, A.; Ruano, L.; Goretti, B.; Niccolai, C.; Patti, F.; Chisari, C.G.; Gallo, P.; Grossi, P.; et al. Identifying the Distinct Cognitive Phenotypes in Multiple Sclerosis. JAMA Neurol. 2021, 78, 414–425. [Google Scholar] [CrossRef] [PubMed]
  214. Grigoriadis, P.; Bakirtzis, C. Morphosyntactic Abilities and Cognitive Performance in Multiple Sclerosis. Brain Sci. 2024, 14, 237. [Google Scholar] [CrossRef] [PubMed]
  215. Arya, A.; Dhall, M.; Mittal, V.; Kaushik, D.; Mudgal, P.; Kumar, T.; Pandey, M.; Kadian, R.; Sharma, P.; Rani, N.; et al. Multiple Sclerosis: Pathogenesis Mechanism and Biomarkers. Cent. Nerv. Syst. Agents Med. Chem. 2024, 24, 1–14. [Google Scholar] [CrossRef]
  216. Haase, S.; Linker, R.A. Inflammation in multiple sclerosis. Ther. Adv. Neurol. Disord. 2021, 14, 17562864211007687. [Google Scholar] [CrossRef]
  217. Barmpagiannos, K.; Theotokis, P. The Diversity of Astrocyte Activation during Multiple Sclerosis: Potential Cellular Targets for Novel Disease Modifying Therapeutics. Healthcare 2023, 11, 1585. [Google Scholar] [CrossRef]
  218. Bakirtzis, C.; Karakasi, M.V.; Boziki, M.K.; Moysiadis, T.; Tsakona, D.; Fyntanidou, B.; Papazisis, G.; Thireos, E.; Grigoriadis, N. Psychopharmacology of patients with multiple sclerosis in Greece during the period 2017–2019. Psychiatr. Psychiatr. 2022, 33, 65–71. [Google Scholar] [CrossRef]
  219. Bakirtzis, C.; Nikolaidis, I.; Boziki, M.K.; Grigoriadou, E.; Karakasi, M.V.; Moysiadis, T.; Kesidou, E.; Papazisis, G.; Grigoriadis, N. Epidemiological Insights on Medication Concurrency and Polypharmacy in People With Multiple Sclerosis in Greece. Int. J. MS Care 2023, 25, 140–144. [Google Scholar] [CrossRef]
  220. Marrie, R.A. Comorbidity in multiple sclerosis: Implications for patient care. Nat. Rev. Neurol. 2017, 13, 375–382. [Google Scholar] [CrossRef]
  221. Bede, P.; Finegan, E.; Chipika, R.H.; Li Hi Shing, S.; Lambe, J.; Meaney, J.; Redmond, J. Occulomotor Neural Integrator Dysfunction in Multiple Sclerosis: Insights From Neuroimaging. Front. Neurol. 2018, 9, 691. [Google Scholar] [CrossRef]
  222. Michailidou, I.; Fluiter, K.; Boziki, M.; Grigoriadis, N.; Baas, F. Editorial: Complement in nervous system disease. Front. Cell. Neurosci. 2023, 17, 1268023. [Google Scholar] [CrossRef] [PubMed]
  223. Zhang, S.; Ma, Y.; Luo, X.; Xiao, H.; Cheng, R.; Jiang, A.; Qin, X. Integrated Analysis of Immune Infiltration and Hub Pyroptosis-Related Genes for Multiple Sclerosis. J. Inflamm. Res. 2023, 16, 4043–4059. [Google Scholar] [CrossRef] [PubMed]
  224. Theoharides, T.C.; Kempuraj, D.; Kourelis, T.; Manola, A. Human mast cells stimulate activated T cells: Implications for multiple sclerosis. Ann. N. Y. Acad. Sci. 2008, 1144, 74–82. [Google Scholar] [CrossRef] [PubMed]
  225. Theoharides, T.C.; Kempuraj, D.; Iliopoulou, B.P. Mast cells, T cells, and inhibition by luteolin: Implications for the pathogenesis and treatment of multiple sclerosis. Adv. Exp. Med. Biol. 2007, 601, 423–430. [Google Scholar]
  226. Pinke, K.H.; Zorzella-Pezavento, S.F.G.; de Campos Fraga-Silva, T.F.; Mimura, L.A.N.; de Oliveira, L.R.C.; Ishikawa, L.L.W.; Fernandes, A.A.H.; Lara, V.S.; Sartori, A. Calming Down Mast Cells with Ketotifen: A Potential Strategy for Multiple Sclerosis Therapy? Neurotherapeutics 2020, 17, 218–234. [Google Scholar] [CrossRef]
  227. Loh, J.S.; Mak, W.Q.; Tan, L.K.S.; Ng, C.X.; Chan, H.H.; Yeow, S.H.; Foo, J.B.; Ong, Y.S.; How, C.W.; Khaw, K.Y. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal transduction and targeted therapy. Signal Transduct. Target. Ther. 2024, 9, 37. [Google Scholar] [CrossRef] [PubMed]
  228. Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome-microglia connections via the gut-brain axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef]
  229. Krautkramer, K.A.; Fan, J.; Backhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 2021, 19, 77–94. [Google Scholar] [CrossRef]
  230. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  231. Gribble, F.M.; Reimann, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat. Rev. Endocrinol. 2019, 15, 226–237. [Google Scholar] [CrossRef]
  232. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Park. Dis. 2021, 7, 27. [Google Scholar] [CrossRef] [PubMed]
  233. Hoyles, L.; Pontifex, M.G.; Rodriguez-Ramiro, I.; Anis-Alavi, M.A.; Jelane, K.S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A.L.; Carding, S.R.; et al. Regulation of blood-brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235. [Google Scholar] [CrossRef] [PubMed]
  234. Zhang, Y.; Bailey, J.T.; Xu, E.; Singh, K.; Lavaert, M.; Link, V.M.; D’souza, S.; Hafiz, A.; Cao, J.; Cao, G.; et al. Mucosal-associated invariant T cells restrict reactive oxidative damage and preserve meningeal barrier integrity and cognitive function. Nat. Immunol. 2022, 23, 1714–1725. [Google Scholar] [CrossRef]
  235. Ramanan, D.; Pratama, A.; Zhu, Y.; Venezia, O.; Sassone-Corsi, M.; Chowdhary, K.; Galván-Peña, S.; Sefik, E.; Brown, C.; Gélineau, A.; et al. Regulatory T cells in the face of the intestinal microbiota. Nat. Rev. Immunol. 2023, 23, 749–762. [Google Scholar] [CrossRef] [PubMed]
  236. Lee, H.J.; Lee, K.E.; Kim, J.K.; Kim, D.H. Suppression of gut dysbiosis by Bifidobacterium longum alleviates cognitive decline in 5XFAD transgenic and aged mice. Sci. Rep. 2019, 9, 11814. [Google Scholar] [CrossRef]
  237. Fu, J.; Li, J.; Sun, Y.; Liu, S.; Song, F.; Liu, Z. In-depth investigation of the mechanisms of Schisandra chinensis polysaccharide mitigating Alzheimer’s disease rat via gut microbiota and feces metabolomics. Int. J. Biol. Macromol. 2023, 232, 123488. [Google Scholar] [CrossRef]
  238. Sun, J.; Xu, J.; Ling, Y.; Wang, F.; Gong, T.; Yang, C.; Ye, S.; Ye, K.; Wei, D.; Song, Z.; et al. Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Transl. Psychiatry 2019, 9, 189. [Google Scholar] [CrossRef]
  239. Kritas, S.K.; Saggini, A.; Cerulli, G.; Caraffa, A.; Antinolfi, P.; Pantalone, A.; Rosati, M.; Tei, M.; Speziali, A.; Saggini, R.; et al. Corticotropin-releasing hormone, microglia and mental disorders. Int. J. Immunopathol. Pharmacol. 2014, 27, 163–167. [Google Scholar] [CrossRef]
  240. Sandhu, J.K.; Kulka, M. Decoding Mast Cell-Microglia Communication in Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 1093. [Google Scholar] [CrossRef]
  241. Traina, G. Mast cells in the brain—Old cells, new target. J. Integr. Neurosci. 2017, 16, S69–S83. [Google Scholar] [CrossRef]
  242. Forsythe, P. Mast Cells in Neuroimmune Interactions. Trends Neurosci. 2019, 42, 43–55. [Google Scholar] [CrossRef] [PubMed]
  243. Drouin-Ouellet, J.; Sawiak, S.J.; Cisbani, G.; Lagace, M.; Kuan, W.L.; Saint-Pierre, M.; Dury, R.J.; Alata, W.; St-Amour, I.; Mason, S.L.; et al. Cerebrovascular and blood-brain barrier impairments in Huntington’s disease: Potential implications for its pathophysiology. Ann. Neurol. 2015, 78, 160–177. [Google Scholar] [CrossRef]
  244. Grundy, S.M.; Hansen, B.; Smith, S.C., Jr.; Cleeman, J.I.; Kahn, R.A. Clinical management of metabolic syndrome: Report of the American Heart Association/National Heart, Lung, and Blood Institute/American Diabetes Association conference on scientific issues related to management. Arter. Thromb. Vasc. Biol. 2004, 24, e19–e24. [Google Scholar]
  245. Jornayvaz, F.R.; Samuel, V.T.; Shulman, G.I. The role of muscle insulin resistance in the pathogenesis of atherogenic dyslipidemia and nonalcoholic fatty liver disease associated with the metabolic syndrome. Annu. Rev. Nutr. 2010, 30, 273–290. [Google Scholar] [CrossRef] [PubMed]
  246. Kountouras, J.; Polyzos, S.A.; Katsinelos, P.; Zeglinas, C.; Artemaki, F.; Tzivras, D.; Vardaka, E.; Gavalas, E.; Romiopoulos, I.; Simeonidou, C.; et al. Cardio-cerebrovascular disease and Helicobacter pylori-related metabolic syndrome: We consider eradication therapy as a potential cardio-cerebrovascular prevention strategy. Int. J. Cardiol. 2017, 229, 17–18. [Google Scholar] [CrossRef]
  247. Wang, A.; Su, Z.; Liu, X.; Yang, Y.; Chen, S.; Wang, S.; Luo, Y.; Guo, X.; Zhao, X.; Wu, S. Brachial-ankle pulse wave velocity and metabolic syndrome in general population: The APAC study. BMC Cardiovasc. Disord. 2016, 16, 228. [Google Scholar] [CrossRef] [PubMed]
  248. Ojala, J.O.; Sutinen, E.M. The Role of Interleukin-18, Oxidative Stress and Metabolic Syndrome in Alzheimer’s Disease. J. Clin. Med. 2017, 6, 55. [Google Scholar] [CrossRef] [PubMed]
  249. Ungvari, Z.; Toth, P.; Tarantini, S.; Prodan, C.I.; Sorond, F.; Merkely, B.; Csiszar, A. Hypertension-induced cognitive impairment: From pathophysiology to public health. Nat. Rev. Nephrol. 2021, 17, 639–654. [Google Scholar] [CrossRef]
  250. Jennings, J.R.; Muldoon, M.F.; Ryan, C.; Price, J.C.; Greer, P.; Sutton-Tyrrell, K.; van der Veen, F.M.; Meltzer, C.C. Reduced cerebral blood flow response and compensation among patients with untreated hypertension. Neurology 2005, 64, 1358–1365. [Google Scholar] [CrossRef]
  251. Muller, M.; van der Graaf, Y.; Visseren, F.L.; Mali, W.P.; Geerlings, M.I. Hypertension and longitudinal changes in cerebral blood flow: The SMART-MR study. Ann. Neurol. 2012, 71, 825–833. [Google Scholar] [CrossRef]
  252. Immink, R.V.; van den Born, B.J.; van Montfrans, G.A.; Koopmans, R.P.; Karemaker, J.M.; van Lieshout, J.J. Impaired cerebral autoregulation in patients with malignant hypertension. Circulation 2004, 110, 2241–2245. [Google Scholar] [CrossRef]
  253. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Helicobacter pylori infection and nonalcoholic fatty liver disease: Are the four meta-analyses favoring an intriguing association pointing to the right direction? Metab. Clin. Exp. 2019, 96, iii. [Google Scholar] [CrossRef] [PubMed]
  254. Montemurro, P.; Nishioka, H.; Dundon, W.G.; de Bernard, M.; Del Giudice, G.; Rappuoli, R.; Montecucco, C. The neutrophil-activating protein (HP-NAP) of Helicobacter pylori is a potent stimulant of mast cells. Eur. J. Immunol. 2002, 32, 671–676. [Google Scholar] [CrossRef] [PubMed]
  255. Supajatura, V.; Ushio, H.; Wada, A.; Yahiro, K.; Okumura, K.; Ogawa, H.; Hirayama, T.; Ra, C. Cutting edge: VacA, a vacuolating cytotoxin of Helicobacter pylori, directly activates mast cells for migration and production of proinflammatory cytokines. J. Immunol. 2002, 168, 2603–2607. [Google Scholar] [CrossRef]
  256. Hofman, V.; Lassalle, S.; Selva, E.; Kalem, K.; Steff, A.; Hebuterne, X.; Sicard, D.; Auberger, P.; Hofman, P. Involvement of mast cells in gastritis caused by Helicobacter pylori: A potential role in epithelial cell apoptosis. J. Clin. Pathol. 2007, 60, 600–607. [Google Scholar] [CrossRef] [PubMed]
  257. Nakajima, S.; Krishnan, B.; Ota, H.; Segura, A.M.; Hattori, T.; Graham, D.Y.; Genta, R. Mast cell involvement in gastritis with or without Helicobacter pylori infection. Gastroenterology 1997, 113, 746–754. [Google Scholar] [CrossRef] [PubMed]
  258. Lv, Y.P.; Teng, Y.S.; Mao, F.Y.; Peng, L.S.; Zhang, J.Y.; Cheng, P.; Liu, Y.-G.; Kong, H.; Wang, T.-T.; Wu, X.-L.; et al. Helicobacter pylori-induced IL-33 modulates mast cell responses, benefits bacterial growth, and contributes to gastritis. Cell Death Dis. 2018, 9, 457. [Google Scholar] [CrossRef]
  259. Talley, N.J.; Walker, M.M. The Rise and Rise of Eosinophilic Gut Diseases Including Eosinophilic Esophagitis Is Probably Not Explained by the Disappearance of Helicobacter pylori, so Who or What’s to Blame? Am. J. Gastroenterol. 2018, 113, 941–944. [Google Scholar] [CrossRef]
  260. Lianto, P.; Zhang, Y.; Che, H. Signals from the various immune cells in promoting food allergy-induced eosinophilic esophagitis like disease. Asia Pac. Allergy 2019, 9, e28. [Google Scholar] [CrossRef]
  261. Shi, Y.; Liu, X.F.; Zhuang, Y.; Zhang, J.Y.; Liu, T.; Yin, Z.; Wu, C.; Mao, X.H.; Jia, K.R.; Wang, F.J.; et al. Helicobacter pylori-induced Th17 responses modulate Th1 cell responses, benefit bacterial growth, and contribute to pathology in mice. J. Immunol. 2010, 184, 5121–5129. [Google Scholar] [CrossRef]
  262. Johnson, K.S.; Ottemann, K.M. Colonization, localization, and inflammation: The roles of H. pylori chemotaxis in vivo. Curr. Opin. Microbiol. 2018, 41, 51–57. [Google Scholar] [CrossRef] [PubMed]
  263. D’Elios, M.M.; Amedei, A.; Benagiano, M.; Azzurri, A.; Del Prete, G. Helicobacter pylori, T cells and cytokines: The “dangerous liaisons”. FEMS Immunol. Med. Microbiol. 2005, 44, 113–119. [Google Scholar] [CrossRef] [PubMed]
  264. Rothenberg, M.E. Molecular, genetic, and cellular bases for treating eosinophilic esophagitis. Gastroenterology 2015, 148, 1143–1157. [Google Scholar] [CrossRef] [PubMed]
  265. Cianferoni, A.; Spergel, J.M.; Muir, A. Recent advances in the pathological understanding of eosinophilic esophagitis. Expert. Rev. Gastroenterol. Hepatol. 2015, 9, 1501–1510. [Google Scholar] [CrossRef]
  266. Li, N.; Xie, C.; Lu, N.H. Transforming growth factor-beta: An important mediator in Helicobacter pylori-associated pathogenesis. Front. Cell Infect. Microbiol. 2015, 5, 77. [Google Scholar] [CrossRef]
  267. Wu, M.S.; Lin, J.T.; Hsu, P.N.; Lin, C.Y.; Hsieh, Y.T.; Chiu, Y.H.; Hsueh, P.R.; Liao, K.W. Preferential induction of transforming growth factor-beta production in gastric epithelial cells and monocytes by Helicobacter pylori soluble proteins. J. Infect. Dis. 2007, 196, 1386–1393. [Google Scholar] [CrossRef]
  268. Kountouras, J. Helicobacter pylori: An intruder involved in conspiring glaucomatous neuropathy. Br. J. Ophthalmol. 2009, 93, 1413–1415. [Google Scholar] [CrossRef]
  269. Ko, E.; Chehade, M. Biological Therapies for Eosinophilic Esophagitis: Where Do We Stand? Clin. Rev. Allergy Immunol. 2018, 55, 205–216. [Google Scholar] [CrossRef]
  270. Gao, Y.; Xu, B.; Zhang, P.; He, Y.; Liang, X.; Liu, J.; Li, J. TNF-alpha Regulates Mast Cell Functions by Inhibiting Cell Degranulation. Cell Physiol. Biochem. 2017, 44, 751–762. [Google Scholar] [CrossRef]
  271. Gu, Y.; Yang, D.K.; Spinas, E.; Kritas, S.K.; Saggini, A.; Caraffa, A.; Antinolfi, P.; Saggini, R.; Conti, P. Role of TNF in mast cell neuroinflammation and pain. J. Biol. Regul. Homeost. Agents 2015, 29, 787–791. [Google Scholar]
  272. Liutu, M.; Kalimo, K.; Kalimo, H.; Uksila, J.; Leino, R. Mast cells and IgE-containing cells in gastric mucosa of Helicobacter pylori infected and non-infected patients with chronic urticaria. J. Eur. Acad. Dermatol. Venereol. 2004, 18, 69–72. [Google Scholar] [CrossRef] [PubMed]
  273. Nelson, M.; Zhang, X.; Pan, Z.; Spechler, S.J.; Souza, R.F. Mast cell effects on esophageal smooth muscle and their potential role in eosinophilic esophagitis and achalasia. Am. J. Physiol. Liver Physiol. 2021, 320, G319–G327. [Google Scholar] [CrossRef] [PubMed]
  274. Kountouras, J.; Zavos, C.; Chatzopoulos, D. Apoptosis and autoimmunity as proposed pathogenetic links between Helicobacter pylori infection and idiopathic achalasia. Med. Hypotheses 2004, 63, 624–629. [Google Scholar] [CrossRef] [PubMed]
  275. Konturek, P.C.; Brzozowski, T.; Konturek, S.J. Stress and the gut: Pathophysiology, clinical consequences, diagnostic approach and treatment options. J. Physiol. Pharmacol. 2011, 62, 591–599. [Google Scholar]
  276. Vukman, K.V.; Forsonits, A.; Oszvald, A.; Toth, E.A.; Buzas, E.I. Mast cell secretome: Soluble and vesicular components. Semin. Cell Dev. Biol. 2017, 67, 65–73. [Google Scholar] [CrossRef]
  277. Jolly, P.S.; Bektas, M.; Olivera, A.; Gonzalez-Espinosa, C.; Proia, R.L.; Rivera, J.; Milstien, S.; Spiegel, S. Transactivation of sphingosine-1-phosphate receptors by FcepsilonRI triggering is required for normal mast cell degranulation and chemotaxis. J. Exp. Med. 2004, 199, 959–970. [Google Scholar] [CrossRef]
  278. Pietrzak, A.; Wierzbicki, M.; Wiktorska, M.; Brzezinska-Blaszczyk, E. Surface TLR2 and TLR4 expression on mature rat mast cells can be affected by some bacterial components and proinflammatory cytokines. Mediat. Inflamm. 2011, 2011, 427473. [Google Scholar] [CrossRef]
Figure 1. The gut–brain axis (GBA) as a bidirectional network. The figure illustrates the gut–brain axis as a bidirectional network controlled by hormonal, nervous, immunological, and metabolic signaling pathways. It shows how dysregulated mast cells disrupt the gut microbiome, leading to inflammation and gut barrier breakdown. The role of Helicobacter pylori is highlighted, showing its contribution to neurodegenerative diseases through immune mechanisms, including defective monocytes and molecular mimicry, which may lead to neuroinflammation and blood–brain barrier disruption. single-headed yellow arrows denote result; double-headed yellow arrows denote bi-directional interaction/communication; yellow lightning signs denote pathology.
Figure 1. The gut–brain axis (GBA) as a bidirectional network. The figure illustrates the gut–brain axis as a bidirectional network controlled by hormonal, nervous, immunological, and metabolic signaling pathways. It shows how dysregulated mast cells disrupt the gut microbiome, leading to inflammation and gut barrier breakdown. The role of Helicobacter pylori is highlighted, showing its contribution to neurodegenerative diseases through immune mechanisms, including defective monocytes and molecular mimicry, which may lead to neuroinflammation and blood–brain barrier disruption. single-headed yellow arrows denote result; double-headed yellow arrows denote bi-directional interaction/communication; yellow lightning signs denote pathology.
Neurolint 16 00127 g001
Table 1. Mast cells and mechanisms of neuroinflammation in neurodegenerative diseases.
Table 1. Mast cells and mechanisms of neuroinflammation in neurodegenerative diseases.
DiseaseMajor Disease
Mechanisms
Evidence of
Neuroinflammation
/Key Players
Evidence of MC Involvement
ADoxidative stress [132]IL-1, IL-6, IFNγ [133,134,135]MCs recruited into amyloid plaques [133,134,135]
hyperphosphorylation of tau protein [136] phagocyted amyloid particles in MCs in skin and
gastric samples—histamine release [137]
Aβ accumulation [138] SAA causes MCs degranulation, cytokine release (TNF-α and IL-1) and chemotaxis [137]
impairment of cholinergic transmission [139] MC granule material causes SAA degradation into proto-fibrillar intermediates [140].
Aβ contributes to the degranulation of prefrontal
cortical MCs, via the enhancement of Cx43 and Panx1 HC flux [137].
MCs detect low-solubility amyloid particles and to migrate to plaque sites, where they trigger the release of inflammatory mediators and disrupt the BBB [137].
MCs release MMP 2 and 9 and VEGF, leading to
vascular leakage, CNS leukocyte infiltration, and edema [141].
MCs detect IL-33 and act as early responders to brain injury via the release of pre-stored TNF [142] and VEGFs thus further recruiting leukocytes and MCs [143,144,145].
MCs in stress-induced inflammation: release of CRH and further activation of MCs and microglia. Chronic stress triggers MC degranulation causing pro-
inflammatory mediator release, synaptic loss, BBB disruption and neuroinflammation [146,147].
PDloss of DA neurons in the SN and VTA [148]Activated microglia and
astrocytes present in areas with midbrain DA neuron loss [149]
Brain MCs neurotoxicity in the SN [150].
reduced DA
transmission to STR [148]
Accumulation of misfolded α-synuclein linked to dysregulated immune
responses in the CNS [151].
Administration of MPTP after MC reconstitution in rats shown to increase oxidative stress and to alter levels of MDA, GSH, SOD and GPx [152].
accumulation of α-synuclein [152]PD-associated
polymorphisms in the
HLA-DR gene [151].
MCs are attracted and activated in the SN region of MPTP-induced mice: releasing inflammatory
mediators (histamine, LTs, TNF-α, and IL-1, thus
contributing to TH+ DA neuron loss in the SN [153]
intracellular
mitochondrial
dysfunction-induced ROS generation [152]
Increased activated MCs in PD brains, particularly in the midbrain and the striatum and increased
IL-33 expression linked to cell injury [146].
dysfunction in the
ubiquitin-proteasomal and autophagy-
lysosomal system [152]
NLRP3 inflammasome’s involvement in Parkinson’s disease [132,154,155]. peripheral tissue MCs in CAPS patients express inflammasomes and produce IL-1 [156]—potential role for CNS MCs
Limitation: CNS MC expression of the NLRP3 inflammasome has not been directly studied
ALSdegeneration of motor neurons [157]Reactive immune cells in postmortem tissue of
patients with familial and sporadic ALS [158,159].
Degranulating MCs present in the quadriceps muscle of patients with ALS [158,159]
distal motor
axonopathy [157]
MCs and neutrophils surrounding motor axons in SOD1G93A rats with ALS [158,159]
impaired axonal transport [157] Masitinib results in reduced infiltration by MCs and neutrophils, ameliorates axonal pathology and
secondary demyelination, and hinders the loss of type 2B myofibers in SOD1G93A rats [158,159]
mitochondrial function defect [157] The degree of MC degranulation corresponds to
clinical disease endpoints in SOD1G93A mice [160].
MCs are regarded to enhance vascular permeability in the context of neuroinflammation, thus triggering
further neutrophil recruitment, clustering and
activation [157,161,162,163].
Presence of MCs and many c-Kit+ progenitors in the motor neuron-vascular niche [164]
HDproduction of a mHTTReactive astrocytes have been reported in pre-clinical stages of HD. Their presence correlates with later disease severity [165]. mHTT promotes TLR-4 receptor internalization, thus affecting MC activation [166]
Excitatory neurotoxicity caused by mHTT in the striatum and cortexReactive microglia have been reported in the
striatum and cortex of HD, accompanying neuron loss, in postmortem brains [167].
MSauto-reactive T cells
recognizing CNS
myelin antigens
Activation of the adaptive immune system [168]MCs may act as antigen-presenting cells towards T cells [147,169]
multi-focal
demyelination
Microglia activation [168]MCs disrupt the blood–brain barrier [170]
neurodegeneration MC inflammasome plays pivotal role in the
meningeal inflammation in EAE [171].
EAE development involves MC accumulation in the CNS [172,173].
Alzheimer’s disease (AD); amyloid beta (Aβ); interleukin-1 (IL-1); interleukin-6 (IL-6); interferon gamma(IFNγ); mast cells (MCs); amyloid A protein precursor serum amyloid A (SAA); tumor necrosis factor alpha (TNF-α); connexin-43 (Cx43); pannexin 1 (panx1); hemichannel (HC); blood–brain barrier (BBB); metalloproteinase 2 (MMP2); metalloproteinase 9 (MMP9); vascular-endothelial growth factor (VEGF); central nervous system (CNS); interleukin-33 (IL-33); corticotropin releasing hormone (CRH); Parkinson’s disease (PD); dopaminergic (DA) neurons; substantia nigra (SN); ventral tegmental area (VTA); striatum (STR); methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP); malondialdehyde (MDA); glutathione (GSH); superoxide dismutase (SOD); glutathione peroxidase (GPx); leukotrienes (LTs); human leukocyte antigen—DR isotype (HLA–DR); tyrosine hydroxylase (TH); dopaminergic (DA); reactive oxygen species (ROS); nucleotide-binding domain, leucine-rich-containing family, NOD-like receptor protein 3 (NLRP3); cryopyrin-associated periodic syndrome (CAPS); amyotrophic lateral sclerosis (ALS); superoxide dismutase (SOD); Huntington’s disease (HD); mutant Huntingtin protein (mHTT); toll-like receptor 4 (TLR-4); multiple sclerosis (MS); experimental autoimmune encephalomyelitis (EAE).
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

Boziki, M.; Theotokis, P.; Kesidou, E.; Nella, M.; Bakirtzis, C.; Karafoulidou, E.; Tzitiridou-Chatzopoulou, M.; Doulberis, M.; Kazakos, E.; Deretzi, G.; et al. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut–Brain Axis and Helicobacter pylori Infection. Neurol. Int. 2024, 16, 1750-1778. https://doi.org/10.3390/neurolint16060127

AMA Style

Boziki M, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Tzitiridou-Chatzopoulou M, Doulberis M, Kazakos E, Deretzi G, et al. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut–Brain Axis and Helicobacter pylori Infection. Neurology International. 2024; 16(6):1750-1778. https://doi.org/10.3390/neurolint16060127

Chicago/Turabian Style

Boziki, Marina, Paschalis Theotokis, Evangelia Kesidou, Maria Nella, Christos Bakirtzis, Eleni Karafoulidou, Maria Tzitiridou-Chatzopoulou, Michael Doulberis, Evangelos Kazakos, Georgia Deretzi, and et al. 2024. "Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut–Brain Axis and Helicobacter pylori Infection" Neurology International 16, no. 6: 1750-1778. https://doi.org/10.3390/neurolint16060127

APA Style

Boziki, M., Theotokis, P., Kesidou, E., Nella, M., Bakirtzis, C., Karafoulidou, E., Tzitiridou-Chatzopoulou, M., Doulberis, M., Kazakos, E., Deretzi, G., Grigoriadis, N., & Kountouras, J. (2024). Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut–Brain Axis and Helicobacter pylori Infection. Neurology International, 16(6), 1750-1778. https://doi.org/10.3390/neurolint16060127

Article Metrics

Back to TopTop