1. Introduction
Currently, there are more and more data supporting the participation of reactive oxygen species (ROS) in the initiation and development of inflammation during the disease process at different age periods of an individual [
1,
2,
3,
4,
5]. Excessive production of ROS results in oxidative stress, which contributes to the progression of the inflammatory process [
6,
7,
8,
9]. In a specific tissue microenvironment, the role of mast cells (MCs) in regulation of local homeostasis and the integrated-buffer metabolic environment is critical. On the one hand, MCs express a wide range of receptors that provide high sensitivity mechanisms to form a selective response to external and internal challenges. On the other hand, MCs can selectively secrete various classes of mediators, alternative profiles of cytokines and chemokines, thereby providing targeted effects on the immune and stromal landscapes of a specific tissue microenvironment. MC tools are three basic classes of mediators - preformed mediators, lipid-derived mediators and multiple cytokines, chemokines, growth factors formed after MC stimulation for the requisite modification of physiological responses and immune functions [
10,
11]. MCs are of special significance in the development of a pro-inflammatory background, regulating the state of numerous cells of the immune and stromal landscape, and the extracellular matrix of connective tissue [
12,
13,
14,
15,
16,
17,
18,
19,
20,
21]. ROS potential to modify and activate MC secretory activity is well known [
22,
23,
24,
25,
26,
27,
28,
29,
30,
31,
32]. Under oxidative stress, MCs become an essential structural and functional component, and regulation of this component can affect the integral state of the local tissue microenvironment and its resistance to various challenges. Therefore, it is of interest to search for new molecular agents that can use MC properties to manipulate local inflammation, one of these agents - H
2 - being of special significance. Mechanisms underlying the biological effects of H
2, including anti-inflammatory, antiapoptotic, neuroprotective, radioprotective, adaptive, homeostatic, etc. have been elucidated, but insufficiently [
33,
34,
35,
36,
37,
38,
39,
40].
In the present study, MCs are considered as a potential H2 target in regulating homeostasis of the integrated-buffer metabolic environment of the extracellular matrix and the immune landscape of a specific tissue microenvironment. Enzymes released by MCs during inflammation, which may be a point of impact for H2, are attractive points of discussion. MC activation is accompanied by increased levels of ROS. Considering that MCs or macrophages produce ROS at an early stage of the inflammatory response, H2 can be used at the earliest stages of the local inflammatory foci formation. Thus, H2 is completely integrated into the list of agents that can influence the activity of MCs indirectly through the effect on ROS.
To date, there are only a few research studies related to the effect of H
2 on MC biology. H
2 exposure resulted in a decreased migration and secretory activity of MCs, and a reduced scope of inflammatory reactions in several organs [
41,
42,
43]. In addition, the experiment demonstrated positive effects of H
2 on the skin dermis remodeling, due to the state of MCs [
44]. However, this is clearly insufficient to reveal the actual potential of MCs in developing biological outcomes of H
2. This review paper is aimed at focusing attention of numerous specialists on MCs as a potential target of H
2 under its therapeutic effects on the pathogenesis of conditions accompanied by acute and chronic inflammation.
2. Molecular Hydrogen as a Promising Agent for Regulating the State of the Integrated-Buffer Metabolic Environment of the Local Tissue Microenvironment
H
2 is a diatomic gas consisting of two hydrogen atoms covalently bonded to each other. Cellular bioavailability of molecular hydrogen is extremely high due to its unique physicochemical properties. Its small size, low mass, neutral charge, and nonpolar nature, combined with a high diffusion rate, allow it to freely penetrate cell membranes and diffuse into mitochondria and the nucleus [
38,
39,
45]. The therapeutic and prophylactic effects of H
2 on various organs have been demonstrated. H
2 has antioxidant properties, as it immediately counteracts hydroxyl radicals [
46] and decreases peroxynitrite levels [
37]. Due to its antioxidant action, H
2 maintains stability of the genome, by a number of markers slowing down the processes of cellular aging, and provides in histone modification and telomere maintenance [
37]. Apart from this, H
2 can inhibit inflammatory processes and control the immune system, cell death mechanisms (apoptosis, autophagy, and pyroptosis), the mTOR regulatory pathway, autophagy, apoptosis, and mitochondrial health [
37,
41,
47,
48,
49,
50,
51]
Inflammation is induced by the release of pro-inflammatory cytokines, which are produced in the greatest amount by immunocompetent cells, including macrophages, MCs and neutrophils. The resulting uncontrolled cytokine storm can result in severe conditions with acute or chronic inflammation. Mitochondria generating profuse amounts of the most potential hydroxyl radical •OH appear to be one of the main sources of reactive oxygen species (ROS). H
2 can selectively neutralise •OH formed in mitochondria and thereby produce its proper effects [
52,
53,
54,
55]. As demonstrated, ROS provides NLRP3 inflammasome activation and this, in turns, triggers the pro-inflammatory cytokine production. H
2 contributes to inhibition of NLRP3 inflammasome activation by suppressing oxidative stress. This fact is associated with its prophylactic effect related to inflammatory diseases, including the ones in a presymptomatic state [
56,
57]. Recently, interesting research results have emerged supporting the effective H
2 application as a new antitumor agent, and evidencing its effect on mitigating side effects of cancer treatment. Notably, mechanisms for the formation of H
2 effects are not only due to the leveling of •OH, but also through the regulation of gene expression [
55]. It is reported about the radioprotective effects of H
2 based on the elimination of •OH due to ionizing radiation [
58]. The neuroprotective effects of H
2 due to antioxidant, anti-inflammatory, anti-apoptotic effects, regulation of autophagy, modulation of mitochondrial function and the blood-brain barrier have been described as well. As demonstrated, H
2 has the protective effect on ischemic damage to the nervous system, traumatic injuries, subarachnoid hemorrhages, neuropathic pain, neurodegenerative diseases, cognitive dysfunctions, depression, etc. [
59]. H
2 is stated to have diverse options of indirect action, which are mediated by the production of various biologically active substances to form chronic effects [
35]. However, the main mechanisms of H
2 are still far from being revealed, and further search for tissue and intracellular H
2 targets is currently required.
3. Mast Cells Are Key Regulatory Players in the Organ-Specific Tissue Microenvironment
The state of a specific tissue microenvironment represented by vessels, a cellular component, and an extracellular matrix, is critical in the formation of the pathological focus. Each organ has specialised cellular clusters that use proper regulatory mechanisms to maintain local homeostasis. MCs actively participate in the management of cellular cooperations, monitoring most of the key parameters of the cellular microenvironment [
10,
60,
61]. The heterogeneity of MCs determines their specific role for each state of the organ-specific tissue microenvironment. The phenotypic plasticity of MCs creates a great potential for the formation of subpopulations with specialized properties due to epigenetic mechanisms at the level of the local microenvironment. Notably, a balanced regulation of MC adaptation to a specific tissue microenvironment is achieved at each specific temporal point due to various mechanisms, including the effects of tryptase on DNA [
62]. The uniqueness of MCs lies in the extraordinary combination, on the one hand, of an adapted sensory apparatus to informatively meaningful signs of the integrated-buffer metabolic environment, and, on the other hand, of a multifunctional effector apparatus represented by a secretome. The changes existing in the tissue microenvironment are recorded by MCs using multiple receptors, including surface IgG receptors, toll-like receptors, C-type lectin receptor, retinoic acid-inducible gene-I - like receptors, nucleotide oligomerisation domain-like receptors, siglecs, G-protein-coupled receptors, lipid mediator receptors, alarmin receptors, leukocyte Ig-like receptors, cytokine receptors, integrins, tetraspanins, nuclear receptors and a lot more [
63,
64].
In case of non-allergic activation of MCs adequately to the challenges of the cellular microenvironment, MCs are able to secrete with high selectivity a variety of biologically active substances, which can be classified as preformed mediators and mediators that resynthesize during the process of MC activation. Pre-accumulated secretome products are represented by biogenic amines (histamine, polyamines, dopamine, serotonin), proteases (chymase, carboxypeptidase A, trypase, cathepsin G, granzyme B, metalloproteinases), enzymes (kinogenases, heparanase, angiogenin, active caspase-3), including lysosome enzymes (β-hexosaminidase, β-glucuronidase, β-D-galactosidase, arylsulfatase A, cathepsins), proteoglycans (heparin, chondroitin sulfate), cytokines (TNF, IL-4, IL-15, etc.), chemokines ( RANTES, eotaxin, IL-8, MCP-1, etc.), growth factors (TGF-β, bFGF, EGF, VEGF, NGF, FGF-2, SCF, PDGF), as well as numerous regulatory peptides (corticoliberin, endorphin, endothelin-1, substance P, vasoactive intestinal peptide, angiogenin, bradykinin, leptin, renin, somatostatin, etc.). Resynthetic products include cell-derived cytokines, growth factors, and mitogens, MC-derived chemokines, and various lipid metabolites, in particular prostaglandins and leukotrienes [
65,
66] (
Figure 1).
MC phenotypic plasticity is affected by reactions to specific inflammatory stimuli, this results in their long-lasting polarity effects on adaptive immune responses. Considering the effect of ROS on DNA methylation [
67], a pro-inflammatory phenotype of MCs is likely to be formed under certain conditions. Since MCs are long-lived and can be exposed to multiple cycles of activation, they are likely to gain experience in response to repeated stimuli. Such "trained immunity" after exposure to a pathogen has been demonstrated among various immunocompetent cells [
68]. The ability of MCs to re-granulate contrasts with the activity of the most other immune cells, which undergo apoptosis upon activation [
69]. Therefore, MCs have a great potential for tissue remodeling through induced collagen fibrillogenesis, angiogenesis, wound healing, etc., without reaching high quantitative indices per unit area of tissue.
Being immediate integral part of the developing adaptive and pathological mechanisms through secretome components, MCs serve as an informative marker of disease progression, and represent a promising therapeutic target. Of particular importance are specific MC proteases – tryptase, chymase, and carboxypeptidase A3 [
70,
71,
72]. The secretory mechanisms of proteases and different secretome components provide diverse options for excreting substances having high selectivity into the extracellular matrix, thereby forming a wide range of biological effects [
73,
74,
75,
76].
4. Mast Cells and Inflammation
MCs play a central topic in the initiation, enhancement, and regulation of inflammation. A significantly increased number of MCs and activation of degranulation mechanisms in tissues with inflammatory phenomena makes it promising to develop new therapeutic algorithms aimed at targeted regulation of the release of specific mediators. Under inflammation, the number of MCs in the tissue increases dramatically. Activated MCs promptly degranulate, releasing bioactive molecules from secretory granules via numerous mechanisms that ensure selectivity of their entry into the extracellular matrix [
20,
75,
76]. Activated MCs react during acute and chronic inflammation, in addition to secretome components, and release ROS. Further, MC phenotype in tissues with inflammation is characterized by an increased density of mas-related G protein-coupled receptor X2 (MRGPRX2). MC activation results in the release of many bioactive compounds, which takes into account spatial, temporal and chemical-physical properties of the local tissue microenvironment, correcting local homeostasis both during the development of inflammation and subsequent restoration of homeostasis to normal values. Thus, each of the points of intracellular processing of inflammatory mediators in the MCs and post-secretory metabolism are effective targets for the modulation of inflammation by tropic agents. H
2 can be one of such agents.
MC activation is controlled by several receptors: FcɛRI, Toll-like receptors (TLR), KIT receptor, complement receptors C5a and C3a, MRGPRX2, etc. [
77,
78,
79]
. Under MC degranulation, tissues release mediators with high pro-inflammatory activity, including specific proteases (tryptase, chymase and carboxypeptidases); pro-inflammatory cytokines (interleukin (IL)-4,5,6,15), tumor necrosis factor – α (TNF- α); vascular endothelial growth factors (VEGF); biogenic amines (histamine); hydrolases that neutralise pathogens (β-hexosaminidase), etc. [
20,
70,
71,
72,
80,
81]. Notably, MC activation is characterised by an increase in intra- and extracellular ROS [
82,
83]. Importantly, that prolonged stay of MCs within the inflammation focus leads to chronic activation accompanied by an increased expression of FcɛRI and MRGPRX2 receptors in MCs and, thus, an increased sensitivity to activating signals of a specific tissue microenvironment [
66,
84]. It goes without saying, this situation will increase secretion of cytokines, chemokines and enzymes with pro-inflammatory activity with each circle.
Thus, MCs under normal physiological conditions will differ from MCs in a pathological focus characterized by an inflammatory microenvironment. These data provide promising opportunities for H2 targeting to specific properties of MCs in tissues with an inflammatory process.
5. Reactive Oxygen Species in the Mechanisms of Activation of Mast Cell Secretory Pathways
The release of intracellular and extracellular ROS (ROS
in and ROS
ex respectively has an impact on immunocompetent cells, including macrophages, neutrophils and MCs [
31,
85,
86,
87]. It is known that, depending on ROS concentration, they can have a damaging effect on cell macromolecules and, along with this, act as important mediators involved in the regulation of cell growth and differentiation, including various types of cell death [
88,
89,
90,
91,
92,
93]. ROS can affect directly (as mutagens) and indirectly (as messengers and regulators) numerous structural and functional aspects of cell biology. An excess of ROS can result in genomic mutations, but, what is more, it can cause irreversible oxidative modification of proteins (oxidation and peroxidation of proteins), lipids and proteoglycans, disrupting their function and contributing to pathological changes as well [
41]. Conversely, local ROS at low concentrations are critical as redox signaling molecules in numerous signaling pathways involved in maintaining intracellular homeostasis (MAPK/ERK, PTK/PTP, PI3K-AKT-mTOR, etc.) and in regulation of key transcription factors (NFκB/IκB, Nrf2/KEAP1, AP-1, p53, HIF-1, etc.) [
94]. Therefore, ROS is able to regulate many cellular functions, including proliferation, differentiation, migration, and apoptosis. Fundamental studies of the molecular mechanisms of ROS biological effects and the potential of their power will be the basis for developing novel therapeutical approaches.
As repeatedly evidenced, MC degranulation attributable to chemical agents (salts of Hg and Au, substance 48/80, Ca
2+ ionophores, etc.) and physiological stimuli (antigens, neurotrophic growth factor, substance P, etc.) is accompanied by an increased content of ROS in the cytosol [
21,
23,
95,
96,
97]. Superoxide (O
2-) and hydrogen peroxide (H
2O
2) are the two main constituents of ROS in MCs [
98,
99,
100]. After MC activation, ROS
in is rapidly released, reaching a peak within a few minutes [
21,
22,
82]. Immune activation upon antigen identification by the IgE-FcɛRI complex, as well as non-immunological activation by thapsigargin, ionomycin and other compounds, is accompanied by an increase in ROS
in [
82,
101,
102]. Unlike ROS
in, there is no consensus regarding the release of ROS
ex. In addition to ROS
ex released exclusively from MCs, mast cells are also exposed to ROS
ex action due to close proximity to macrophages and other immunocompetent cells [
85,
103].
To date, multiple experimental data evidence crucial part of ROS in regulating MC degranulation regarding
in vitro and
in vivo models. ROS
in is actively involved in the activation of essential intracellular signaling pathways and can stimulate the production of a number of pro-inflammatory mediators of MC. There are multiple sources of ROS presented in MC mitochondria, which includes the electron transport chain, dehydrogenases in the matrix, intermembrane space proteins, monoamine oxidases in the outer membrane, etc. [
31,
104]. These enzymes form O
2–, which is further a source of H
2O
2 and hydroxyl radicals •OH. MCs produce ROS after stimulation of high-affinity IgE receptor (Fc epsilon RI). As known, there are ROS-regulated intracellular and/or plasma membrane Ca
2+ channels of MCs. In MCs, the activity of the store-operated Ca
2+ channel – (SOC) is regulated by O
2– and H
2O
2. H
2O
2 generation is dependent on Src family kinase and phosphatidylinositol-3-kinase activities. Concurrently, O
2– generation is dependent on Ca
2+ in the extracellular environment. Thus, generation of O
2– and H
2O
2 by separate signaling mechanisms reciprocally regulate SOC activity in MCs, which is presented in Ca
2+ signaling and mediator secretion activity [
23,
100]. Conditions that promote the formation of ROS in MCs can lead to MC activation by calcium signaling, including hypoxia, allergy, exposure to aryl hydrocarbon receptor ligands [
27,
105,
106].
ROS produced by NADPH oxidase regulate the pro-inflammatory response of MCs [
82]. Regular mitochondrial functions are necessary to provide physiological cellular dynamics, and their dysfunction triggers the development of numerous disorders, including those of the immune system. Impact of external factors on MCs can increase production of H
2O
2 with the participation of mitochondrial complex III, enhancing the secretion of histamine and serotonin by MCs [
107]. The use of H
2 as a substance with antioxidant properties is pathogenetically the most critical mechanism enabling reduction of MC degranulation activity, and, as a result, theoretically promising potential to decrease the inflammatory background in a specific tissue microenvironment (
Figure 2). The proton gradient across the inner mitochondrial membrane is a major driving force for mitochondrial ROS production, and it can be modified by representatives of the mitochondrial uncoupling protein (UCP) family. Of these, UCP2 uncouple oxidative phosphorylation, with concomitant decreases in ROS production, is an effective regulator of MC function [
108,
109,
110]. Recent evidence suggests that UCP2 and mitochondrial translocation regulate MC degranulation [
111]. UCP2 not only neutralize ROS, but also prevent their formation, influencing MC degranulation indirectly through an increased concentration of Ca
2+ [
112]. Therefore, UCP2 activators have the potential to reduce production of mitochondrial ROS [
113].
ROS can cause reversible post-translational changes in proteins involved in intracellular signaling. For example, particular proteins contain cysteine residues possessing the ability to be oxidized. Each of these modifications can change the activity of the protein, thereby affecting its function in the signal transduction pathway [
41,
114].
One of the central events mediated by the influence of ROS and changes in the redox status of the cell is an increased cytoplasmic concentration of Ca
2+, which are crucial in the mechanisms of MC degranulation [
115,
116,
117]. Concurrently, a change in the intracellular Ca
2+ concentration, in turn, also affects ROS generation [
87]. MC activation comes with ROS production; ROS regulate various signaling pathways, thus providing the release of inflammatory mediators and various cytokine production. Protein tyrosine phosphatases (PTPs)) are a superfamily of enzymes that are main targets for ROS due to an oxidation-susceptible nucleophilic cysteine at their active site [
118]. As reported, direct and indirect regulations of class I and II Cys-based protein tyrosine phosphatases (PTEN, LMW-PTP, SHP-2, PTP-PEST, PTP1B, DEP-1, TC45, LAR etc.) are possible. Notably, SHP-1, SHP-2, and PTEN phosphatases are known to be involved in MC activation [
119,
120,
121]. Phosphatase inhibition by H
2O
2 induces phosphorylation of tyrosine residues of β- and γ-subunits of FcεRI, Ca
2+ influx, and secretory activity of MC [
122].
Therefore, redox-regulated protein tyrosine phosphatases may be a target for the novel treatment options in allergies or inflammatory diseases using H
2 [
123]. Activation of protein kinase C dependent on ROS can be one of the mechanisms of MC regulation using H
2 [
124]. It should also be noted, that the adapter protein LAT (linker for activation of T cells) can serve as the target for ROS; interaction with this protein promotes induction of the FcεRI-dependent pathway of MC activation [
125]. Conversely, low-level local ROS are crucial both as redox-signaling molecules in a variety of pathways participating in cellular homeostasis (MAPK/ERK, PTK/PTP, PI3K-AKT-mTOR), and as regulators of key transcription factors (NFκB/IκB, Nrf2/KEAP1, AP-1, p53, HIF-1).
Thus, ROS may have a decisive role regulating the FcεRI signaling cascade for MC degranulation. As stated, there are many potential MC targets that are sensitive to the effects of ROS. Research investigating the ROS effect on diverse pathways of MC activation, in particular on the FcεRI-dependent pathway, provides great opportunities to further develop and implement into clinical practice drugs tailor-made on the basis of antioxidants and inhibitors resulted from ROS production [
16,
41]. H
2 can be used as such an agent, its antioxidant properties are widely discussed [
45,
52,
126]. H
2 can be used for effective therapeutic action on pathologies associated with MCs, primarily those of an atopic origin. H
2 applied using various approaches can act as blockers of MC secretory activity, limiting their potential to form a pro-inflammatory ground in a specific tissue microenvironment, and be used to treat multiple systemic inflammatory and allergic disorders [
41,
42]. Oral ingestion of water with an increased content of molecular hydrogen eliminated an immediate-type allergic reaction in mice [
41]. Namely, H
2 attenuates phosphorylation of the FcεRI-associated Lyn and its downstream signal transduction, thereby inhibiting the NADPH oxidase activity and decreasing H
2O
2 generation. The authors also demonstrated that, under an immediate allergic reaction, H
2 may develop its beneficial effect due to modulation of different unknown specific signaling pathways [
41].
Under induced intracerebral hemorrhage simulated in male mice, treatment with hydrogen reduced Lyn kinase phosphorylation and tryptase release, decreased MC accumulation and degranulation, which was ultimately accompanied by attenuated blood-brain barrier disruption, reduced cerebral edema, and better neurological status [
42]. In addition, administration of H
2-rich water was found to reduce skin MC infiltration in the treatment of atopic dermatitis and secretion of the pro-inflammatory cytokines such as interleukin IL-1β and IL-33 [
43]. Thus, ROS
in and ROS
ex formed under MC activation in a strict spatio-temporal dependence can be critical for the targeted action of molecular hydrogen.