Next Article in Journal
Intrinsic Antibacterial Urushiol-Based Benzoxazine Polymer Coating for Marine Antifouling Applications
Previous Article in Journal
Dissect Gender-Dependent Susceptibility SNPs in Progressive Osteoarthritis Using Regulator Motif Candidate of Genetic Association Strategy (RMCGA)
Previous Article in Special Issue
The Role of Mast Cells in the Remodeling Effects of Molecular Hydrogen on the Lung Local Tissue Microenvironment under Simulated Pulmonary Hypertension
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Hydrogen in the Treatment of Respiratory Diseases

Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(9), 4116; https://doi.org/10.3390/ijms26094116
Submission received: 18 February 2025 / Revised: 22 April 2025 / Accepted: 25 April 2025 / Published: 26 April 2025
(This article belongs to the Special Issue Physiology and Molecular Medicine of Molecular Hydrogen)

Abstract

:
Molecular hydrogen is gaining increasing attention as an antioxidant, anti-inflammatory, and antiapoptotic agent. Once considered an inert gas, it reveals current therapeutic potential among others in inflammatory diseases, cancer, and sports medicine, among others. The present review aims to provide a consistent summary of the findings of the last twenty years on the use of molecular hydrogen in major respiratory diseases, including allergies, asthma, COPD, pulmonary fibrosis, lung injury of various origins, as well as cancer and infections of the respiratory tract. In addition, the basic mechanisms through which molecular hydrogen exercises its biological activity on the respiratory system are described.

Graphical Abstract

1. Introduction

Molecular hydrogen is gaining increasing attention in modern medicine for its potential in the treatment of various diseases. It possesses unique antioxidative properties [1] not only as a highly selective free radical scavenger [2,3], but also as an inducer of endogenous detoxifying enzymes operating at the level of key transcription factors. These broad antioxidant properties contribute to its anti-inflammatory and antiapoptotic activities, which may alleviate many symptoms associated with various diseases in animal models and human therapy [4,5,6]. Molecular hydrogen, a theoretically inert gas, has emerged as a promising therapeutic agent with a wide range of potential applications.
Hydrogen (H2) is a colorless, odorless, and tasteless gas with poor water solubility. It reaches a maximum level of about 0.78 mM (≈1.6 mg/L) at room temperature with a loss of about 2–5% per 3 min [7]. Under normal conditions, its reactivity is negligible due to its high dissociation energy, which classifies it as an inert gas. Consequently, it is approved as a food additive in the European Union [8]. It is produced (and consumed) by bacteria of the gut microbiota [9,10] thought the degradation of oligosaccharides and mammals lack of specific enzymes (hydrogenases) required for this process. The most prominent bacterial phyla involved in this process are the Firmicutes and Bacteroidetes phyla, which include the anaerobic Clostridium species [9,10,11,12].
Following its discovery by Antoine Lavoisier in 1783, hydrogen has long been considered an inert gas without significant biological activity. This perception changed in 1888, when H2 was utilized for the first time as a diagnostic tool for visceral pain for the first time [13]. It was not until 1944 that hydrogen mixed with oxygen at a ratio of 96%-to-4%, known as the Hydrox gas mixture, was used by deep-sea divers to prevent decompression sickness and allow diving to depths of up to 500 m [14].
Later, in 1975, the initial observations on the possible use of hyperbaric hydrogen in the treatment of cancer in animals were conducted [15]. Despite these observations, molecular hydrogen remained neglected until the beginning of the 21st century, when the first publications on the anti-inflammatory [16] and antioxidant [3] properties of H2 were released, and a new era of the application of molecular hydrogen in medicine began.
Since that time, several hundred publications have been published exploring the use of molecular hydrogen in various fields of medicine, veterinary sciences, and human wellness. These studies cover a wide range of topics, including adjunctive therapies for inflammatory diseases, cancer treatment, and even preventing muscle fatigue in athletes, improving skin quality, or curing common hangovers. During the recent COVID-19 pandemic, H2 has received considerable attention as a potential therapeutic agent.
Over the past two decades, many excellent reviews have been published that focus on the potential mechanisms of H2 action [1,3,5,6] and the potential application of molecular hydrogen in various diseases [4,17,18,19,20,21]. Most reviews on respiratory diseases include studies on the application of molecular hydrogen in one specific disease: cancer [22,23,24,25,26,27,28], sepsis-related lung injury [29,30] or COVID-19 [31,32]. There is only one review that focuses on several respiratory diseases [33]. Our article, on the other hand, collects and describes research on the use of molecular hydrogen not only in the most common lung diseases—asthma, chronic obstructive pulmonary disease (COPD), lung cancer, and lung injury—but also on allergies and respiratory infections (Figure 1). In addition, this review has been updated with studies not included in the mentioned article [33] and also recent research.It also provides a brief summary of the mechanisms of the action of H2 related to the respiratory system. The present review covers animal studies, including preliminary studies, reviews, research papers performed solely on cell cultures, case reports, observational studies, and clinical trials. A number of studies lack conclusive results or statistical power. Therefore, they should be treated as preliminary results or observations that inspire other researchers to conduct further research.

2. Mechanisms of Action of Molecular Hydrogen

2.1. Anti-Oxidant Activity of Molecular Hydrogen

The antioxidant activity of H2 is based on two processes: a direct scavenging of the most toxic reactive oxygen and nitrogen species (ROS/RNS), the hydroxyl radical (•OH) and peroxynitrite (ONOO), and an indirect modulation of detoxifying enzymes and pathways (Figure 2).
The hydroxyl radical, a particularly potent ROS, exhibits a reactivity approximately 100 times greater than that of superoxide (O2) [25]. It has been demonstrated to oxidize DNA and to induce lipid peroxidation processes, which result in the production of oxidative stress markers such as 4-hydroxyl-2-nonenal (4HNE), malondialdehyde (MDA) [34], and 8-Hydroxydeoxyguanosine (8-OHdG), a marker of oxidative DNA damage [35].
Peroxynitrite is one of the most potent RNS involved in nitrosative stress processes, including nitration of tyrosine residues within proteins. This process leads to the subsequent deterioration of protein function, resulting in cell necrosis or apoptosis [36].
Molecular hydrogen has been shown to selectively neutralize both •OH and ONOO without any reactivity towards other biologically active radicals such as the superoxide anion (O2) and hydrogen peroxide (H2O2) [1,3]. Consequently, H2 does not disrupt physiological metabolic oxidoreduction reactions.
A significant domain of molecular hydrogen’s activity relates to its role in the maintenance of mitochondrial membrane potential, in part by reducing oxidoreductase activity, thereby preventing its dissipation. This, in turn, leads to the preservation of mitochondrial cytochrome C content [21,37]. In addition, as H2 diminishes the levels of mitochondrial ROS, it impedes electron leakage in the electron transport chain, modulates the mitochondrial respiratory chain complex I (though not complex II) [38,39], and increases the expression of mitofusin-2 (a protein essential for mitochondrial fusion) [40] while decreasing the levels of the dynamin-related protein 1 (Drp1 protein), a regulator of mitochondrial fission) [41,42]. Consequently, this results in the restoration and subsequent regulation of mitochondrial dynamics and function [41,43]. In parallel, hydrogen has been shown to inhibit mitochondrial stress and modulate mitophagy [44].
In addition to the direct neutralization of •OH and ONOO, which results in the protection of the genome from oxidative damage, H2 also activates a variety of detoxifying enzymes. These include the glutathione/thioredoxin system [8], superoxide dismutase (SOD), catalase (CAT), and hemoxygenase-1 (HO-1), which are key antioxidant enzymes [5,8,21,45,46,47,48,49].
Furthermore, additionally, H2 activates the Nrf2 (nuclear factor erythroid 2-related factor 2) pathway, a key transcription factor involved in oxidative stress-related responses, including cytoprotective, antioxidant, and detoxifying enzymes such as HO-1 [50]. Concurrently, the Nrf2 pathway plays a pivotal role in respiratory diseases and links oxidative stress and inflammation [51]. It is noteworthy that HO-1, which is activated via the Nrf2 pathway, plays a pivotal role in maintaining the balance between pro- and anti-oxidative, pro- and anti-inflammatory, and pro- and antiapoptotic processes within the lung [52]. The protective effects of HO-1 upregulation have been observed in models of lung injury and its symptoms, including pulmonary edema, inflammation, and lung cell apoptosis [53]. These effects may be attributed to the removal of free heme and inhibition of the Fenton reaction [1,52]. The Fenton reaction, in turn, has been associated with the development of symptoms of lung injury through the enhancement of oxidative stress, as it results in the production of the most toxic ROS, namely, the hydroxyl radical [25]. Prolonged or excessive oxidative stress, which cannot be counterbalanced by the endogenous antioxidant system [54], leads to oxidative damage of alveolar macrophages and epithelial cells, their apoptosis, and, thus, to induced histological changes of the lungs such as pulmonary edema. Concurrently, it has been observed to induce overactivation of proinflammatory pathways such as the NFκB pathway and thereby contribute to the development of inflammation [55]. Disturbances in HO-1 expression and its levels have been reported in several lung diseases [56]. Notably, the activation of the Nrf2 pathway has been shown to inhibit the NLRP3 (NLR family pyrin domain containing 3) inflammasome, a protein complex that plays a pivotal role in the propagation of inflammatory responses and the regulation of cell death processes [57].
Concurrently, H2 suppresses the activation of the NADPH oxidase pathway and downregulates the expression of NOX2 and NOX4, two subunits of NADPH oxidase responsible for the generation of ROS [5,21,58,59]. Furthermore, H2 has been shown to reduce the overactivation of myeloperoxidase (MPO), one of the markers of neutrophil activation and degranulation, leading to the inhibition of neutrophilic inflammation, a hallmark of several chronic respiratory diseases [21,60,61].

2.2. Anti-Inflammatory Activity of Molecular Hydrogen

The anti-inflammatory properties of molecular hydrogen are primarily attributed to its antioxidant activity as a scavenger of ROS and RNS (Figure 3). A decrease in the production of ROS coupled with an increase in their elimination or scavenging by H2 would result in the downregulation of the overactivated NLRP3 inflammasome. This, in turn, would further suppress the NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells, the key transcription factor involved in oxidative stress and inflammation-related responses) pathway [4,62,63].
Concurrently, H2 exercises its inhibitory effect on proinflammatory pathways encompassing NFκB and MAPK (mitogen-activated protein kinase) cascades (such as p38, ERK, and JNK) [64]. Consequently, the ROS-dependent proinflammatory pathways like NLRP3 and TLR4 (Toll-like receptor 4), which play a role in chronic inflammation and responses to LPS (Lipopolysaccharide), respectively, are attenuated [43,63,65]. Furthermore, expression of anti-inflammatory cyto- and chemokines such as interleukin (IL)-4, IL-10, IL-13, is upregulated [5,17,43,66,67]. Conversely, proinflammatory cytokines such as IL-1β, IL-6, tumor necrosis factor alpha (TNF-α), interferon gamma (INF-γ), and high mobility group box 1 protein (HMGB1) [5,21,43,66] have been observed to be downregulated along with adhesion molecules (such as vascular cell adhesion molecule 1 (VCAM-1) or intercellular adhesion molecule 1 (ICAM-1)) [21,67,68,69,70]. In addition, H2 is directly involved in the regulation of signaling protein Sirtuin-1 (SIRT-1), a potential link between oxidative stress and inflammation [71]. SIRT-1 plays a pivotal role in the regulation of cell proliferation and differentiation, stress responses, and apoptosis. Additionally, it functions as a protective mechanism during acute lung injury (ALI) [72]. At the cellular level, the anti-inflammatory properties of H2 are manifested by decreased neutrophil and macrophage activity and migration into the injured tissue [43,68,73]. Additionally, H2 promotes the polarization of macrophages from the proinflammatory M1 type to the anti-inflammatory M2 type [43,74]. It also inhibits Th2 responses, restores regulatory T cells (Treg), and, thus, normalizes an overactivated immune system [21,43].

2.3. Antiapoptotic Activity of Molecular Hydrogen

Another category of properties that emerge from its antioxidant characteristics is the antiapoptotic one. As mentioned previously, molecular hydrogen affects a number of signaling pathways, including the PI3K/Akt/GSK3b, Ask1/JNK, JAK2/STAT3, Ras/ERK1,2/MEK1,2 pathways, all of which play a crucial role in maintaining the pro/antiapoptotic balance. Consequently, H2 has been shown to downregulate the expression of proapoptotic factors such as Bax and the ROS-p53 signaling pathway [29,43] and upregulate the antiapoptotic factors, including Bcl-2 and Bcl-xl. This phenomenon is likely facilitated by the activation of the PI3K/Akt and JAK2/STAT3 signaling pathways [21,43,72,75]. Concurrently, protective mechanisms at the mitochondrial level resulting from decreased ROS production and reduced oxidative stress also play a critical role in the protective effects of molecular hydrogen [5].
Furthermore, molecular hydrogen has been demonstrated to regulate endoplasmic reticulum (ER)-related stress, which is believed to be the link between ROS generation and cell death [72]. This process depends on the excessive accumulation of unfolded proteins in the ER. H2 has been shown to impede this process by interacting with the PERK-eIF2a-ATF4, IRE1-XBP1, and ATF6 [21]. Furthermore, it regulates autophagy, a process involved in the degradation and recycling of macromolecules. While autophagy plays a protective role under normal physiological conditions, its overactivation can lead to inflammation and tissue damage [6]. The activity of H2 relies mainly on the regulation of the expression of p-mTOR/mTOR and p62 proteins [17,21,65].

3. Methods of Administration

There are several methods to administer H2 [76], the most prominent being inhalations (2–4% in air or 66% in oxygen), the drinking of hydrogen-enriched water (HRW), and injections of hydrogen-enriched saline (HRS) [1,19] (see Figure 4). The generation of pure H2 typically occurs through dedicated devices that employ hydrolysis of water or solutions containing electrocatalysts such as potassium hydroxide [20,76].
HRW is intended for consumption (drinking) and can be obtained from magnesium- or calcium-containing sources [76,77]. In some Asian countries, some of them are approved as dietary supplements [78] while others, containing magnesium, citric acid, and ascorbic acid, comply with European regulations [79]. It has been found that a single calcium-containing tablet can produce up to 2.5 µg of H2 [80]. Additionally, another source of H2 includes commercially available hydrogen-generating water tanks or flasks, which are mostly based on electrolysis of filtered water [76,81].
The consumption of certain prebiotics, especially those rich in dietary fiber, indigestible starches, and sugars (lactulose), has been demonstrated to enhance intestinal H2 production through the activity of intestinal flora [10,28,82].
A number of alternative methods are currently under development, including the use of magnesium-hyaluronic acid complexes, palladium cells for the long-term storage of H2, nanobubbles, and nanotechnology-based magnesium alloys. Other methods include implantable magnesium galvanic cells for targeted delivery to tumors. Their initial testing has been performed in animal studies [49,76,83,84].
In all cases, safety issues, such as the risk of explosion at concentrations exceeding 4% in air, present concerns regarding biodegradation, and the general tolerability of the implants, as well as stability and limited storage time of the prepared H2-enriched solutions, should have to be taken into account.

4. Molecular Hydrogen and Allergies

The potential benefits of molecular hydrogen in the treatment or prevention of allergic reactions or their exacerbations remain to be fully elucidated. To date, the majority of studies on this topic have been conducted on allergy-related diseases, specifically allergic rhinitis (AR) and allergic asthma in the ovalbumin (OVA) animal models, along with observations in humans suffering from AR with and without nasal polyps. In all cases, molecular hydrogen was administered via inhalation, direct instillation into the nostrils, or as nasal lavage. All studies reported a decrease in the severity of nasal symptoms, such as itching, scratching, or a runny nose [85,86]. In addition, lower inflammatory markers, such as blood eosinophil count, levels of eosinophilic protein or mucosal eotaxin, IL-4, IL-5, IL-13, and monocyte chemoattractant protein-1 (MCP-1), were measured [86,87,88]. Concurrently, patients observed reduced tissue congestion, nasal edema, a general feeling of inflammation and sickness, and improved quality of life. At the same time, no adverse effects of H2 administration have been reported [89,90,91].
Specifically, molecular hydrogen has been demonstrated to restore the Th1/Th2 imbalance by downregulating Th2 cell responses, upregulating the levels of IL-10 and transforming growth factor (TGFβ) [85], and decreasing CD4+ T cell infiltration [6,92]. H2 increased the population of CD4+CD25+Foxp3+ Treg cells, which are often decreased in allergic rhinitis (AR) [87]. Consequently, H2 exerted a regulatory effect on the damaged nasal mucosa and thereby stabilized the impaired functions of macrophages [85,87].
The anti-inflammatory effects of H2 appear to be strongly associated with its antioxidant properties, as a parallel reduction in oxidative stress has been reported [86]. Oxidative stress, a key marker in AR and other nasal inflammatory processes, has been identified as a pivotal mediator of these pathologies. Inflammation has been shown to promote oxidative stress responses, which, in turn, exacerbate allergic inflammation [89]. Increased ROS was found in nasal polyps, and the blockade of the Nrf2 pathway in nasal epithelial cells has been shown to enhance susceptibility to sinusitis [89,93,94].
A study on type I hypersensitivity has been published by Itoh et al. [58]. In this study, H2 reduced mast cell degranulation via the inhibition of high-affinity immunoglobulin E receptor (FcεRI IgE) accumulation, which, in turn, led to a decreased release of histamine, leukotrienes, and cytokines, the reduced production of H2O2 by inhibiting NADPH oxidase activity, and its downstream signaling. The consumption of HRW by sensitized mice resulted in reduced vascular permeability and reduced histamine release.
A study by Choi et al. demonstrated the potential use of molecular hydrogen to prevent allergic reactions by shortening the duration of allergen exposure to the respiratory system. The study indicated that the administration of HRW led to an augmentation of elimination rates of carbon nanoparticles and a diminution of their proallergic properties, along with the pro-oxidant and proinflammatory potential of these particles [95].

5. Molecular Hydrogen and Asthma

Asthma is among the most prevalent inflammatory diseases of the airways. At present, approximately 300 million people worldwide are affected by this disorder [96]. The symptoms are predominantly associated with persistent airway inflammation and reversible airway obstruction, and include wheezing, dyspnea, cough, and chest tightness. At the cellular level, the symptoms are associated with an increased influx of cells into the airways, chronic oxidative stress, and an imbalance of Th1/Th2 responses, as well as pro- and anti-inflammatory cytokines, which, subsequently, result in airway hyperreactivity.
In experimental models of allergic (OVA) asthma, H2 administration through inhalations or drinking of HRW has been shown to ameliorate most asthma symptoms. The studies reported a decrease in airway hyperreactivity and lung resistance [33,97], airway inflammation measured as the inflammatory cell influx into the airways [74], the total number of these cells and the levels of their markers, such as MPO [33,97,98,99], and the levels of proinflammatory cytokines and chemokines (IL-4, IL-5, IL-6, IL-13, TNF-α, CXCL15) in blood and bronchoalveolar lavage fluid (BALF) [33,97,98]. In addition, goblet cell hyperplasia, mucus accumulation, and mucin-5AC (MUC5AC, the major glycoprotein of mucus [100]) expression was found to be reduced [33,74,97,98]. H2 administration attenuated oxidative stress expressed as lower MDA and other lipid peroxidation markers along with an enhancement in the expression and activity of endogenous antioxidant enzymes such as SOD or CAT [33,97]. Furthermore, airway remodeling, evidenced by extensive collagen I deposition and collagen III and VEGF expression, was prevented [33,98]. Epithelial barrier damage was prevented through the increased expression of E-cadherin and the tight junction protein zonula occludens-1 (ZO-1) [101], along with inhibition of proapoptotic processes like the caspase 3 and 9 pathways [99]. The phagocytic activity of macrophages was also restored [74]. Again, H2 inhalations or drinking had no effect on non-asthmatic, healthy mice [99].
Huang et al. described that H2 treatment reversed the phagocytic defect of macrophages [74]. The observed effects were attributed to the activation of the antioxidant and anti-inflammatory Nrf2 pathway, co-dominant over the inhibition of the proinflammatory NFκB pathway [74,98].
An interesting theory was proposed by Niu et al. [102]. One of the probable molecular mechanisms underlying the development and progression of asthma would be a metabolic switch from oxidative phosphorylation to anaerobic glycolysis. The study further postulates that monocytes derived from human asthmatics and lungs from OVA mice exhibit augmented lactate production and elevated glycolytic enzyme activity, consequently leading to diminished activity of mitochondrial respiratory complex I and II. This, in turn, results in impaired ATP production. The potential benefits of H2 administration would depend on the reversal and regulation of this metabolic switch by normalizing the increased activity of glycolytic enzymes and decreased mitochondrial enzyme activity, which would lead to the normalization of ATP and lactate levels, along with the normalization of SOD and glutathione peroxidase (GPx) activities.
There have been initial attempts to investigate the possible beneficial effects of molecular hydrogen administration in asthma. The study by Singh et al. [103] demonstrated that the drinking of HRW by patients with asthma and COPD leads to an increase in blood oxygen saturation, vitamin E levels, along with lower oxidative stress markers such as thiobarbituric acid reactive substances (TBARS), MDA, or diene conjugates. In addition, the study noted an enhancement in the patients’ exercise tolerance. Another study by Wang et al. [104] showed the beneficial effect of a 45-min inhalation of 2.4% H2 in air on inflammatory markers such as IL-4, IL-6 in exhaled breath condensate (EBC), and MCP-1 and IL-8 in blood.
Two other studies examined not asthma itself but concentrated rather on its common triggers of specifically occupational and air-pollution-related asthma. Choi et al. [95] demonstrated that drinking HRW and intraperitoneal (ip.) injections of HRS led to an augmentation in the elimination of carbon nanoparticles, accompanied by a reduction in lung inflammation and allergic responses following exposure. The mechanisms underlying H2’s activity are associated with the inhibition of lipid peroxidation via the suppression of Fenton reactions, consequently via the antioxidant properties of H2.
In a related study, Feng et al. [105] administered particulate matter with a diameter of less than 2.5 µm (PM2.5) to rats, followed by H2 inhalations. PM2.5 has been shown to react via the aryl hydrocarbon receptors, thereby increasing oxidative stress and promoting pro-oxidant and proinflammatory responses in the lungs. Consistent with the findings in other asthma studies, H2 inhalations were found to mitigate histopathological changes, mucus hypersecretion, and excessive mucin 5AC (MUC5AC) expression. Furthermore, H2 inhalations have been demonstrated to reduce oxidative stress and oxidative damage, as well as lung inflammation, as evidenced by lower levels of IL-1β, IL-8, and TNF-α. Consequently, H2 demonstrated a protective effect against pulmonary dysfunction and decreased lung mechanics. The beneficial effects of H2 can be further related to direct inhibition of aryl hydrocarbon receptors.
In summary, the findings suggest that hydrogen gas inhalations could serve as a valuable adjunct therapy during acute asthma exacerbations or in response to direct exposure to environmental triggers.

6. Molecular Hydrogen and COPD

Another prevalent lung disease is chronic obstructive pulmonary disease (COPD), which is characterized by progressive airway obstruction, shortness of breath, dyspnea, emphysema, and chronic bronchitis. No single cause is known, and risk factors include smoking, air pollution, and occupational exposure to dusts, chemicals, and fumes. At the cellular level, both inflammation and chronic oxidative stress play a pivotal role [106,107,108,109].
Animal models are based on the long-term exposure of rodents to cigarette smoke (CS), which is indeed the major exposure risk for the development of COPD. H2 has been administered by inhalations, ip. injections of HRS or drinking of HRW with similar results. In general, H2 administration has been found to lead to enhanced survival and reduced weight loss [110], improved lung function and static lung compliance, and decreased arterial blood pressure [110,111,112].
In addition, reduced lung damage was observed, along with ultrastructural changes such as alveolar disorders, alveolar wall thickening, ciliated cells degeneration, small airway remodeling, and emphysema, accompanied by reduced epithelial cell apoptosis [110,111,112,113]. Furthermore, a decrease in inflammation and inflammatory cell infiltration was observed [110], along with a reduction in goblet cell hyperplasia, and a CS-induced upregulation of MUC5AC expression was also documented.
Furthermore, the levels of proinflammatory cytokines (IL-6, IL-17, IL-23, Matrix metalloproteinase-12 (MMP-12)) and the expression of proapoptotic markers in lung tissue and BALF were reduced [110,111,113]. Furthermore, a tissue inhibitor of metalloproteinase-1 (TIMP-1, a pivotal factor in maintaining tissue integrity [114]) expression was elevated [110].
Observations indicated a decrease in oxidative stress, the primary pathology in COPD, accompanied by a reduction in levels of oxidative DNA damage markers [112].
At the molecular level, H2 decreased the CS-upregulated phosphorylation of the epidermal growth factor receptor (EGFR), but without changes in EGFR protein levels [113] along with decreased activation of the ERK1/2 and NFκB pathways [111].
In human studies, the drinking of HRW by COPD patients has been found to increase their blood oxygen saturation, exercise tolerance, and reduce oxidative stress markers and general hypoxia [103]. A double blind, parallel-controlled trial demonstrated that inhaled combination therapy comprising hydrogen (H2) and oxygen (O2) resulted in better outcomes during COPD exacerbations compared to inhaled pure oxygen alone [115]. Patients exhibited better breathless, cough, and sputum scale (BCSS) scores, suggesting a reduced severity of COPD exacerbations. However, no significant differences were observed in pulmonary function or blood gas parameters. In a separate study, COPD patients inhaled 45 min of XEN gas (2.4% H2 in air), and even such a brief inhalation period led to a reduction in inflammatory markers in EBC and blood [104].
In conclusion, the beneficial effects of molecular hydrogen are related to its antioxidant and anti-inflammatory properties, suggesting it to be a useful adjunctive therapy during COPD exacerbations.

7. Molecular Hydrogen and Pulmonary Fibrosis

Another lung disease in which molecular hydrogen has been shown to be beneficial is pulmonary fibrosis (PF), a type of interstitial lung disease characterized by fibrotic changes within the lungs and a progressive and irreversible decline in lung function. It has been associated with overactivation of myofibroblasts originating from the epithelium via the epithelial-to-mesenchymal transition (EMT), which, when prolonged, leads to the destruction of alveolar-capillary units and excessive extracellular matrix deposition. This process contributes to structural remodeling and pulmonary fibrosis. EMT is often initiated by excessive oxidative stress, which activates the transforming growth factor TGF1β via an ROS-dependent pathway [41].
Animal models using different fibrosis-inducing agents (paraquat, LPS, or bleomycin) have shown that the administration of H2 by inhalations or drinking (HRW) has been shown to reduce lung stiffness and preserve the lung’s ability to expand, along with an augmented lung capacity [116,117]. At the cellular level, decreased inflammation (IL-4, IL-6, IL-13), oxidative stress (ROS, MDA, hydroxyproline), and fibrosis (fibronectin, collagen deposition) were observed [116,117,118,119]. Furthermore, a decrease in M2 macrophages and EMT, along with reduced α-smooth muscle actin (α-SMA) and TGF1β expression in the lungs, has been described. The potential mechanisms underlying these processes involve the H2’s ability to reduce oxidative stress, diminish p38/MAPK and JNK pathway activation, and attenuate general inflammation, thereby modulating fibrotic responses [33].

8. Molecular Hydrogen and Other Pulmonary Diseases

Molecular hydrogen has been utilized in the treatment of other, less prevalent pulmonary diseases. Inhalations of a 2:1 ratio of hydrogen to oxygen (H2:O2) have been demonstrated to reduce airway resistance and inspiratory effort in patients with tracheal stenosis [120]. In a separate study, mice with obliterative airway disease exhibited reduced airway occlusion, increased CD4+/CD3+ ratio, decreased IL-6, and increased Treg activity after drinking HRW, with no significant impact on the Nrf2 pathway [121].
Muramatsu et al. [122] demonstrated that even prenatal exposure to H2 can influence the neonate’s lung function. In a mouse model of pulmonary dysplasia, LPS injections into the amniotic fluid during pregnancy resulted in an oxidative stress-dependent arrest of alveolar development in the offspring. The drinking of HRW by pregnant or feeding mice led to the normalization of the abnormally enlarged alveoli, the reduction in oxidative stress markers such as nitrotyrosine and 8OHdG, and the augmentation of LPS-suppressed expressions of HO-1, fibroblast growth factor receptor 4 (FGFR4), and vascular endothelial growth factor receptor 2 (VEGFR2), the latter being essential for proper maintenance of alveolar structures and is involved in proper lung development [123].
Similar observations have been documented by Hattori et al. [124]. Maternal infections frequently result in preterm birth and severe respiratory impairment in the neonate. In the present study, pregnant rats were injected ip. with LPS and received HRW orally. The maternal intake of HRW resulted in enhanced neonatal lung function, morphology, and developmental status, as evidenced by higher arterial blood pO2 and pH and lower pCO2. Furthermore, a reduction in the number of caspase-3-positive cells and IL-6 and 8OHdG levels was noticed. These observations suggest that maternal H2 consumption reduces apoptosis and oxidative stress-related damage of the lung and thereby enhances the lung function of the pup.
Another disease in which H2 may be beneficial is pulmonary hypertension, which is characterized, among others, by shortness of breath, elevated pulmonary arterial pressure, right ventricular hypertrophy, systemic inflammation, and increased oxidative and nitrative stress. The disease appears to be strongly associated with ROS, RNS, and chronic inflammation [69]. In experimental models in rats, H2 inhalations and HRW or HRS administration have been shown to reduce pulmonary arterial pressure, right ventricular weight and hypertrophy index, general and pulmonary inflammatory responses, macrophage infiltration, and markers of oxidative (measured as 8OHdG levels) and nitrative (measured as 3-nitrotyrosine levels) stress [69,125]. Again, these protective effects are based on the antioxidant and anti-inflammatory activities of H2 [33].

9. Molecular Hydrogen and Lung Injuries

9.1. Sepsis-Related Lung Injury

Sepsis, a serious condition marked by the organism’s excessive reaction to infection, can lead to various complications. Among the most severe are systemic inflammation, the so-called cytokine storm, acute lung injury (ALI), which can rapidly progress to acute respiratory distress syndrome (ARDS). This condition is characterized by excessive inflammatory responses, cytokine storm, and oxidative stress, along with dysregulated cell apoptosis and autophagy, as described by Zhang et al. [30]. Furthermore, the aforementioned processes are accompanied by a decline in lung function.
Research conducted on cecal-ligation-and-puncture (CLP)-induced murine sepsis models has demonstrated the efficacy of hydrogen inhalations and HRS injection during surgery [117]. In both cases, H2 increased survival rates, pulmonary function described as the PaO2/FiO2 coefficient, or blood gas parameters [126] together with a decreased severity of lung injury, pulmonary edema, and histological score, or lower neutrophil count [117,126]. At the molecular level, lower inflammatory markers such as proinflammatory cytokines (IL-1β, IL-6, TNF-α, HMGB1) and lower oxidative stress outcomes (lipid peroxidation, DNA oxidation, nitrotyrosine levels) were observed. The observed effects were mainly related to the antioxidant activities of H2 and its activation of the HO-1 and Nrf2 pathways and suppression of p38 and NFκB ones. [126]. Furthermore, this results in the upregulation of mitochondrial function and the activation of autophagy via the modulation of the mTOR/TFEB and PINK/Parkin phagocytic pathways in parallel with inhibition of the NLRP3 inflammasome, as has been demonstrated by Ren et al. [127]. In clinical settings, the co-administration of H2 has been observed to enhance the efficacy of standard treatment options [30,33].

9.2. General Lung Injury

One of the most important clinical problems in which H2 may be beneficial is lung injury and inflammation of any kind. These conditions can be classified according to their severity (measured as the PaO2/FiO2 index), duration (acute or chronic), or trigger or cause. Among these, bacterial and viral lung injury (LI), hypoxia/reoxygenation, hyperoxia, ventilator-, smoke-, burn, radiation-induced, and fetal LI are the most prominent. The main symptoms of acute lung injury (ALI) are histopathological changes within the lung with inflammatory cell infiltration, goblet cell hyperplasia, alveolar wall thickening, pulmonary and epithelial cell damage, edema, decreased lung function, and blood oxygenation (measured as PaO2/FiO2 index or blood gas levels). At the molecular level, ALI is characterized by increased levels of cytokines and chemokines, markers of oxidative stress and apoptosis, along with decreased levels of anti-inflammatory and antiapoptotic factors. Consequently, proinflammatory pathways, such as NFκB, are observed to be upregulated, whereas antioxidant pathways, such as the Nrf2 pathway, are downregulated. Consequently, ALI and its chronic form are a complex disorder of the pro- and antioxidant and pro- and anti-inflammatory balance.
There are numerous animal models of ALI, with the most prevalent being the LPS-instillation model. Lipopolysaccharide (LPS) constitutes a pivotal component of the bacterial cell wall of Gram-negative bacteria. Intratracheal (it.), intraperitoneal (ip.), or intravenous (iv.) administration of LPS in animals has been shown to reproduce the majority of ALI symptoms, including lung edema, leukocyte accumulation in the airways, severe lung inflammation, and others. The LPS-induced ALI exhibits a high degree of similarity with the human clinical picture of pneumonia- and sepsis-associated lung injury [128].
The H2 administration was carried out through inhalations at various concentrations (ranging from 2% to 66% in air or O2) or as injections of HRS at different time points.
The administration of H2 resulted in a near-complete abrogation of all symptoms associated with LPS-induced ALI, including the decreased lung function measured as blood gas parameters [29,65,129], PaO2/FiO2 index [67,130,131], ventilation parameters [129], and histological changes, including alveolar wall thickening [129], neutrophil influx [64,67,129,131], lung edema, increased wet-to-dry ratio, disturbed expression of aquaporin 1 and 5 [17,33,64,67,71,129,131,132], epithelial and alveolar cell damage [33,131,132], and increased protein contents in BALF [67,130]. Furthermore, H2 reduced elevated inflammatory markers, including IL-1β, IL-6, TNF-α, prostaglandin E2 (PGE2) [29,65,71,128,130], macrophage protein 1α 2 (MP1α), and monocyte chemoattractant protein-1 (MCP-1) [67], while increasing the levels of decreased anti-inflammatory markers such as IL-10 [133]. It also reduced the oxidative stress markers, including MDA and nitrotyrosine [64,132], as well as the production of ROS [29], and upregulated antioxidant enzymes such as HO-1 and SOD [64,130,132]. Moreover, the studies observed a decline in lung cell apoptosis attributed to the H2-induced decrease in the number of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) (an assay for cell apoptosis [134])-positive cells and the levels of caspases-3 and 8 [17,33,64,130]. It has been demonstrated to upregulate the antiapoptotic markers such as Bcl-2 [17,29,64,67]. These alterations were associated with the suppression of the NFκB pathway in a SIRT-1-dependent way [29,130] alongside a downregulation of the signaling pathways connected to MAPK and JNK kinases [129]. Autophagy was also regulated, with the activation of the AMPK/mTOR/TFEB pathways being decreased [17,132]. Furthermore, molecular hydrogen was found to regulate the disrupted alveolar barrier permeability [133].

9.3. Hypoxia-Reoxygenation Lung Injury

In addition to the well-known LPS-based model of ALI, there are several types of lung injury (LI) and inflammation. One such example is the model of LI induced by hypoxia-reoxygenation (H/R), analogous to injuries observed in lung transplants and following cardiac arrest. The pathomechanisms underlying this type of LI include extensive oxidative stress, increased migration of neutrophils and M1 macrophages into the airways, lung cell apoptosis, increased production of proinflammatory markers, and severe deterioration of lung function resulting in hypoxemia and hypercapnia.
Inhalations of molecular hydrogen in the hypoxia-reoxygenation model have been shown not only to ameliorate impaired lung function but also to significantly decrease oxidative stress and inflammation [68]. At the molecular level, these changes were mainly based on the H2-dependent reduction in hydroxyl radical production and induction of HO-1 and SOD, which led to the subsequent inhibition of inflammatory markers such as IL-1β and TNF-α. These changes also resulted in the inhibition of the pro-oxidant and proinflammatory p38 MAPK and NFκB pathways, which occurred in parallel to the inhibition of apoptosis and pyroptosis [30,33,68].
Pretreatment of the lung graft donor by H2 inhalation prevented lung inflammation after H/R, as evidenced by decreased levels of IL-1β, TNF-α, ICAM-1, and reduced inflammatory cells influx, along with improved lung function, as reflected in increased arterial pO2 and decreased pCO2 [135]. Reduced histological and apoptotic changes, such as alveolar septum thickening, were observed. Further, a reduction in oxidative stress markers such as lipid peroxidation and proapoptotic markers, including Bax and caspase-3, was observed. Concurrently, an increase in antiapoptotic proteins such as Bcl-2 and Bclx was noted. [136].
Consistent observations have been made by Haam et al. [137] in H2-inhaling pigs with cardiac arrest. The animals showed enhanced lung function, improved blood gas parameters and diminished lung injury severity score, and lung edema during both H2 inhalation and reperfusion. In addition, the study noted higher levels of antioxidant enzymes (HO-1, SOD) and anti-inflammatory cytokines (IL-10) along with a reduced number of apoptotic cells.

9.4. Hyperoxia-Related Lung Injury

Another type of lung injury frequently observed in clinical settings is hyperoxic (hyperoxia-related) LI. Unlike hypoxia and subsequent hypoxemia, which are characterized by insufficient levels of inspired oxygen, hyperoxia is mainly related to elevated oxygen levels resulting from the administration of high-concentration or pure oxygen. Hyperoxia has been demonstrated to be closely associated with oxygen toxicity and can result in elevated oxygen stress and its associated consequences. They include lung inflammation, the overactivation of neutrophils and macrophages, edema, and, ultimately, alveolar damage and pulmonary dysplasia [75,138,139]. There have been attempts to use molecular hydrogen, predominantly administered as HRS ip., as a therapeutic intervention in the case of hyperoxia-related lung injury. H2 administration resulted in a reduction in lung edema, inflammatory cells infiltration, pleural fluid volume, and hemorrhage. Further, it was observed that H2 administration led to a decrease in inflammation, indicated by reduced levels of IL-1β and TNF-α. At the molecular level, a study by Sun et al. reported a reduction in apoptosis (described as a lower number of TUNEL-positive cells) and its associated markers (p-Akt, p-FoxO3, cyclin D1, Bcl-2) [72]. The molecular mechanisms underlying this beneficial effect of H2 appear to encompass its antiapoptotic activity, which is associated with the activation of the PI3K/Akt/FoxO3+ pathway. Furthermore, H2 protected alveolar type II cells from hyperoxia [75], as well as increased the activation of the SIRT-1 proteins. As a result, it has been observed to decrease ER stress along with the activation of the antioxidant HO-1 and Nrf2 pathways [30,52].

9.5. Ventilator-Induced Lung Injury

Ventilator-induced lung injury (VILI) is a significant clinical problem that has been addressed in two murine studies [140,141]. In both studies, H2 was administered as inhalations, resulting in decreased lung edema, the infiltration of inflammatory cells, and decreased inflammatory mediators. Concurrently, lung function and gas exchange were enhanced [141]. An intriguing mechanism proposed by Huang et al. [140] suggests a biphasic response to H2 with the potential for phase-dependent alterations in downstream effects. In the initial phase, which commenced about one hour after the initiation of inhalation, the treatment resulted in NFκB pathway activation, augmented Bcl-2, and diminished Bax protein expression. In the subsequent phase, H2 would induce a decline in NFκB activation and NFκB DNA binding, leading to a reduction in lung cell apoptosis. As in the case of other ALIs, the antioxidant properties of H2 play a significant role.

9.6. Seawater-Induced Lung Injury

It is generally accepted that one of the most effective animal models of ALI is, in addition to the LPS-based model, the model in which seawater is instilled into the airways. Both models exhibit features such as lung edema, alveolar collapse, and surfactant disruption along with hypoxemia and increased oxidative stress. Seawater-induced ALI shows symptoms consistent with those observed in drowning victims, while LPS-induced ALI is more inflammatory in nature. Notably, osmotic stress has been identified as a contributing factor in the pathogenesis of seawater-induced ALI [61,128,142]. In a study in seawater-instilled rabbits, two-percent inhalations of H2 resulted in a reduction in lung epithelial permeability and histopathological changes. Furthermore, markers of both oxidative stress (such as MDA levels in lung tissue) and airway inflammation (including IL-1β, IL-6, TNF-α in BALF) were reduced, as was MPO activity as a marker of neutrophilic infiltration. In addition, proapoptotic markers such as caspase-3 and, consequently, apoptosis as a process were reduced. Other observations, regarding other types of ALI, HO-1, and Nrf2, along with their respective pathways, demonstrated increased expression, indicating their role in H2 activity and its impact on lung injury [61].

9.7. Other Forms of Lung Injury

A similar set of findings has been reported for other forms of ALI, induced by triggers such as smoke, burning, or radiation. In all cases, an improvement in alveolar structure, reduced alveolar wall thickening and lower levels of proinflammatory (IL-1β, IL-6, TNF-α), proapoptotic, and apoptotic index markers along with higher expression and activity of antioxidant enzymes such as SOD have been described. Consequently, the changes resulted in reduced ROS and MDA levels, attenuated inflammation, and lung cell apoptosis, regardless of the route of administration [30,143].

10. Molecular Hydrogen and Infections

Little is known about the potential application of molecular hydrogen in the treatment of infectious diseases other than COVID-19. There is a paucity of information regarding its use in the most common infections of the respiratory system, including those caused by pathogens such as Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, Streptococcus pyogenes, Mycoplasma pneumoniae, Bordetella pertussis, Burkholderia pseudomallei, Chlamydophila pneumoniae, Corynebacterium diphtheriae, Haemophilus influenzae, Mycoplasma pneumoniae, Streptococcus pneumoniae, Coxiella burnetii, and Legionella pneumophila. In addition, research papers addressing the use of molecular hydrogen in the treatment of viral infections of the respiratory tract (induced by rhinoviruses, adenoviruses, influenza and parainfluenza viruses, respiratory syncytial viruses, enteroviruses) are missing.
The only application of molecular hydrogen in respiratory infections is the treatment of the acute form of COVID-19 and post-COVID syndrome. The COVID-19 disease, caused by overactivation of the immune system by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus), leads to severe inflammatory pneumonia and direct alveolar injuries caused by the virus. These injuries result in decreased alveolar gas exchange, leading to hypoxemia and dyspnea [31,144]. Consequently, the development of secondary infections and the need for mechanical ventilation may occur [31,145]. At the cellular level, the SARS-CoV-2 virus activates the INF-γ pathway, leading to an overreaction of the immune system and the so-called cytokine storm, characterized by excessive secretion of proinflammatory mediators, including proinflammatory cytokines [145]. Concurrently, the inhibition of negative feedback and the amplification of feedback responses result in increased cytokine production and activation of the NADPH pathway in leukocytes. This, in turn, enhances oxidative stress via elevated levels of ROS [32,146]. COVID-19 infection is similar in its clinical presentation to sepsis, as both conditions are characterized by the rapid development of symptoms, including persistent neutrophilia [144,147].
Human studies during the recent pandemics have shown positive effects of the co-administration of hydrogen in conjunction with classical oxygen inhalatory therapy at various concentrations. In comparison with oxygen therapy alone, 4 to 66% H2 in O2 reduced the severity of the disease, dyspnea, chest distress, cough, fatigue, and the duration of hospitalization. Concurrently, the discharge rate, blood oxygen saturation, and general well-being of the patients were higher. The respiratory effects of oxygen–hydrogen co-administration are probably related to a deeper penetration of oxygen into the bronchial area, followed by decreased airway resistance, increased oxygenation, and the improvement of general respiratory function, together with a stimulation of mucus drainage [31,62,148].
At the cellular level, the effects of H2 inhalations are related to its antioxidant and anti-inflammatory properties [31]. Specifically, H2 reduced the infiltration of neutrophils and macrophages into the lung [149], thereby inhibiting their excessive activation and the subsequent release of proinflammatory mediators, including IL-1β, IL-6, TNF-α, HMGB-1, CCL, and MCP-1. In addition, H2 inhibited the activation of the NLRP3, NFκB, and MPO [30,31,32,150] and modulated the Nrf2 [30] pathways. Consequently, this led to the resolution of the cytokine storm, inflammation [149,151], and limitation of airway damage [32]. When administered at the onset of a mild infection, H2 has been observed to prevent the onset of the cytokine storm, likely through an early suppression of oxidative stress [6,31]. Notably, these effects are not attributable to direct interactions of H2 with the virus [31] and are independent of its variant (Omicron vs. other virus variants) [151].
In the context of the global pandemic, H2 inhalations have emerged as a promising therapy for the treatment of COVID-19. This approach, adapted mainly in China [149], has demonstrated significant potential in clinical trials. It is currently being explored as a safe and cost-effective treatment option. Furthermore, hydrogen inhalations have found application in post-COVID-19 respiratory rehabilitation. H2/O2 inhalations, varying in duration, have been observed to decrease symptom severity, anxiety (described as decreased fatigue assessment scale (FAS) scores). Additionally, such inhalations have been shown to improve respiratory functions (increased forced vital capacity (FVC), minute ventilation (VE), and forced expiratory volume in 1 s (FEV1)). Improved general fitness (measured as increased distance in the 6-min walking test (6-MWT)) and higher quality of life have also been documented [18,152,153].

11. Molecular Hydrogen and Cancer

Molecular hydrogen demonstrated efficacy in the treatment and as an adjuvant therapy for various cancers of the respiratory tract.
In vitro studies have demonstrated that an H2-rich medium reduced the colony size and formation of tongue cancer cells and decreased proliferation in human fibrosarcoma and esophageal cancer cells, as well as A549 cells [154,155]. Furthermore, the treatment of H1975 cells resulted in a decrease in cell viability, migration, and invasion, along with catalyzed cell apoptosis [155].
A reduction in tumor weight and size, as well as a lower number of cells of squamous cell carcinoma, was revealed by animal studies. Moreover, increased survival of mice with colorectal cancer was observed after H2 administration [25,156]. A549 xenografted mice exhibited diminished tumor growth accompanied by a suppression of ROS production and the progression of inflammation (measured as decreased IL-1β, IL-13, TNF-α levels), indicating the importance of the antioxidant and anti-inflammatory properties of H2 also in cancer treatment [25,155]. Notably, as previously reported, H2 administration exhibited no effect on healthy animals or non-cancerous cell lines [25,157].
However, the most compelling observations have been made in human subjects. H2 inhalations or the drinking of HRW have been shown to reduce tumor marker levels, tumor mass, and their formation. These interventions led to enhanced disease control, including longer progression-free periods and higher survival rates, with some cases of complete remission [22,23,25]. Patients reported improved quality of life with better physical status and fewer pulmonary symptoms [158]. In combination with conventional (such as cis-platin) and modern (including antibodies like nivolumab) therapeutics, H2 enhanced drug activity, resulting in enhanced outcomes and improved disease control and reduced side effects of the treatment, such as nephrotoxicity, weight loss, insomnia, pain, or hearing loss in the case of radiotherapy [23,159,160,161]. The aforementioned effects have been observed in patients diagnosed with a variety of cancers affecting the respiratory tract. The mechanisms of such activity involve a range of processes, including the inhibition of ROS production [159], the restoration of optimal levels and activity of immune cells, including NKT, NK, T cells, and the restoration of the proper immune system functionality [22,156,159]. These processes extend to the regulation of pro- and antiapoptotic pathways. As indicated by Yang et al., the latter include the activation of the ROS/NLRP3/caspase-3/gasdermin D-mediated pyroptotic pathways [162]. Additionally, the suppression of vascular endothelial growth factor (VEGF) expression and downregulation of regulators of chromosome condensation (such as the structural maintenance of chromosomes complex (SMC)-2, 5, 6) have been documented [25,155,163]. Furthermore, the effects of H2 result in the suppression of the overactivated Wnt/beta-catenin signaling pathways, which further leads to suppression of tumor progression [164].
Radiotherapy is a prevalent treatment option for cancer, yet it is associated with significant adverse effects. The efficacy of radiotherapy is mainly based on direct absorption of radiation energy at the level of nucleic acids, lipids, and proteins, as well as the subsequent generation of free radicals during water radiolysis [26,27]. Major adverse effects of radiotherapy include skin damage, bone marrow disturbances, immune system dysfunctions, gastrointestinal damage, and cognitive impairments, among others. In clinical settings, a range of radioprotective agents, including vitamins C, D, E, melatonin, α-lipolic acid, and N-acetylcysteine, are employed, predominantly functioning as free radical scavengers [24,165,166]. However, these agents are not without side effects. In this context, the administration of molecular hydrogen as a novel therapeutic option has been proposed as an adjunct therapy during radiotherapy.
In vitro studies have demonstrated reduced ROS production, improved oxidative stress, and apoptosis markers, together with higher survival rates of A549, HIEC, HaCaT, and BMSC cells exposed to radiotherapy in an H2-rich medium [167,168,169,170].
In animals, H2 administration in conjunction with radiotherapy has been shown to protect against immune dysfunction [92,171], acute and chronic lung [170], myocardium [172], and gastrointestinal [173] and mucosal [174] damage. Additionally, H2 administration has been associated with reduced incidence of dermatitis, nephrotoxicity, and weight loss [2,168]. Furthermore, it has been demonstrated to increase survival rates [175] and promote tissue recovery and wound healing [176]. Registered clinical trials and observational human studies have shown protection against bone marrow damage during modulated radiation therapy [177] and improved quality of life, including the improvement of taste disorders or anorexia when combined with H2 administration [178].
The radioprotective effects of H2 are primarily attributed to its hydroxyl radical scavenging activity [3], underscoring its antioxidant properties and antioxidant activity via decreased levels of oxidation of mitochondrial DNA and the NLRP3 cascades. The radioprotective effects of H2 are further enhanced by its antiapoptotic properties, which are a result of its antioxidant and anti-inflammatory properties. Furthermore, the regulation of pro- and antioxidant gene expression may play a significant role in radioprotection.

12. Future Perspectives and Limitations

Today, the popularity of treatments with molecular hydrogen is on the rise. This increase in popularity is observed not only in specialized medical centers but also as a component of wellness or beauty-related treatment options. The financial burden associated with acquiring professional equipment for the generation of molecular hydrogen is diminishing both in terms of purchase and rental costs. In several countries, including Japan, capsules containing magnesium for the generation of H2 have been introduced on the market, and H2 has been approved as a dietary supplement [179]. The broad range of applications and increasing popularity of hydrogen-related treatments have led to the perception of molecular hydrogen as a novel panacea. An important question is whether the administration of molecular hydrogen could serve as an in-house treatment, a form of add-on therapy for mild infections (e.g., the common cold), akin to brine inhalations [180]. Given the documented efficacy of H2 in the treatment of COVID-19, it is possible that it could also be effective in the treatment of other viral diseases, including influenza. Another research point is the possible bacteriostatic activity of H2. Some pathogens (such as Salmonella or Mycobacteria [181,182]) have been observed to utilize molecular hydrogen during the progression of infection; therefore, it cannot be used to reduce all bacterial infections and their progression. To date, there are no data on the successful use of H2 in the context of bacterial invasions within the respiratory tract. However, molecular hydrogen has been observed to alleviate symptoms of LPS- and sepsis-induced ALI, which can be of bacterial origin [181,182]. Also, the possible utility of H2 in bacterial-viral co-infection remains to be elucidated. The potential of H2 inhalation to prevent the exacerbations of allergies in atopic subjects, respiratory infections in prone subjects (e.g., school children during spring/autumn infection season), and its role as an immunomodulator due to its anti-inflammatory properties, remains, however, to be elucidated.
Despite the advantageous properties of molecular hydrogen, its utilization is limited both in clinical and domestic settings. The primary concern is safety since molecular hydrogen is explosive above a concentration of 4–5% in air; moreover, special equipment is required to assure safe administration above this limit, and, thus, this type of delivery is rather limited to specialized facilities. Second, the precise dosing of H2 is challenging, as all methods described in this review, with the exception of inhalations, fail to ensure the desired levels of H2. Thirdly, the stability of hydrogen-rich solutions poses significant challenges, as about 2–5% of dissolved hydrogen is lost from open containers every 3 min [8]. Consequently, there is an urgent need to develop cost-effective and efficient capture and storage systems to mitigate these losses. Another issue is the financial burden associated with the production of hydrogen, the costs of the hydrogen-generating and storage devices, and the maintenance of these systems.

13. Concluding Remarks

Molecular hydrogen has garnered increased attention as a relatively simple, cost-effective, and safe treatment option for acute and chronic respiratory diseases. Its multifaceted activity involving anti-inflammatory, antioxidant, and antiapoptotic mechanisms facilitates a more holistic approach to the treatment, addressing multiple facets of the disease rather than a single symptom or pathway. Consequently, there remains considerable potential for the utilization of molecular hydrogen in medical disciplines and associated domains of human and veterinary medicine.

Author Contributions

Conceptualization, D.Z., M.J. and P.W.; methodology, D.Z., M.J. and P.W.; formal analysis, D.Z., M.J. and P.W.; resources, D.Z., M.J. and P.W.; writing—original draft preparation, D.Z., M.J. and P.W.; writing—review and editing, D.Z., M.J. and P.W.; supervision, D.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Katarzyna Kaczynska for the critical reading and comments on the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
•OHhydroxyl radical
4HNE4-hydroxyl-2-nonenal
6-MWT6-min walking test
8-OHdG8-hydroxydeoxyguanosine
A549 cellsadenocarcinomic human alveolar basal epithelial cells
ALIacute lung injury
ARallergic rhinitis
ARDSacute respiratory distress syndrome
BALFbronchoalveolar lavage fluid
BCSSbreathless-cough-sputum scale
BMSCbone marrow-derived mesenchymal stem cells
CATcatalase
CLPcecal-ligation-and-puncture
COPDchronic obstructive pulmonary disease
COVID-19coronavirus disease 2019
CScigarette smoke
Drp1dynamin-related protein 1
EBCexhaled breath condensate
EGFRepidermal growth factor receptor
EMTepithelial-to-mesenchymal transition
ERendoplasmic reticulum
FASfatigue assessment scale
FcεRIhigh-affinity IgE receptor
FEV1 forced expiratory volume in 1 s
FGFR4fibroblast growth factor receptor 4
FVCforced vital capacity
GPxglutathione peroxidase
H/Rhypoxia-reoxygenation
H1975human non-small cell lung cancer cells
H2O2 hydrogen peroxide
HaCaTaneuploid immortal keratinocyte cell line from adult human skin
HIEChuman intestinal epithelial cells
HIF-1αhypoxia-inducible factor-1 α
HMGB1high mobility group box 1 protein
HO-1hemoxygenase-1
HRShydrogen-enriched saline
HRWhydrogen-enriched water
ICAM-1intercellular adhesion molecule 1
IgEimmunoglobulin E
ILinterleukin
INF-γinterferon gamma
ip.intraperitoneal (injection)
it.intratracheal (administration)
iv.intravenous (administration)
LIlung injury
LPSlipopolysaccharide
MAPKmitogen-activated protein kinase
MCP-1monocyte chemoattractant protein-1
MDAmalondialdehyde
MMP-12matrix metalloproteinase-12
MP1αmacrophage protein 1α
MPOmyeloperoxidase
MUC5ACmucin-5AC
NFκBnuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NLR family pyrin domain containing 3
Nrf2nuclear factor erythroid 2-related factor-2
O2-superoxide
ONOO⁻peroxynitrite
OVAovalbumin
pCO2partial pressure of carbon dioxide
PFpulmonary fibrosis
PGE2prostaglandin E2
PM2.5 particulate matter of a diameter of less than 2.5 µm
pO2partial pressure of oxygen
ROS/RNSreactive oxygen and nitrogen species
SARS-CoV-2severe acute respiratory syndrome coronavirus-2
SIRT-1Sirtuin-1
SMAsmooth muscle actin
SMCstructural maintenance of chromosomes complex
SODsuperoxide dismutase
TBARSthiobarbituric acid reactive substances
TGF1βtransforming growth factor
TIMP-1tissue inhibitor of metalloproteinase-1
TLR4toll-like receptor 4
TNF-αtumor necrosis factor-α
TUNELterminal deoxynucleotidyl transferase dUTP nick end labeling
VCAM-1vascular cell adhesion molecule-1
VEminute ventilation
VEGFvascular endothelial growth factor
VEGFR2vascular endothelial growth factor receptor 2
VILIventilatory-induced lung injury
XEN gas2.4% H2 in air
ZO-1tight junction protein ZO-1/zonula occludens-1

References

  1. Ohta, S. Molecular Hydrogen as a Novel Antioxidant: Overview of the Advantages of Hydrogen for Medical Applications. Methods Enzymol. 2015, 555, 289–317. [Google Scholar] [CrossRef]
  2. Hong, Y.; Chen, S.; Zhang, J.-M. Hydrogen as a Selective Antioxidant: A Review of Clinical and Experimental Studies. J. Int. Med. Res. 2010, 38, 1893–1903. [Google Scholar] [CrossRef]
  3. Ohsawa, I.; Ishikawa, M.; Takahashi, K.; Watanabe, M.; Nishimaki, K.; Yamagata, K.; Katsura, K.-I.; Katayama, Y.; Asoh, S.; Ohta, S. Hydrogen Acts as a Therapeutic Antioxidant by Selectively Reducing Cytotoxic Oxygen Radicals. Nat. Med. 2007, 13, 688–694. [Google Scholar] [CrossRef]
  4. Hirano, S.; Ichikawa, Y.; Sato, B.; Yamamoto, H.; Takefuji, Y.; Satoh, F. Potential Therapeutic Applications of Hydrogen in Chronic Inflammatory Diseases: Possible Inhibiting Role on Mitochondrial Stress. Int. J. Mol. Sci. 2021, 22, 2549. [Google Scholar] [CrossRef] [PubMed]
  5. Tao, G.; Song, G.; Qin, S. Molecular Hydrogen: Current Knowledge on Mechanism in Alleviating Free Radical Damage and Diseases. Acta Biochim. Biophys. Sin. 2019, 51, 1189–1197. [Google Scholar] [CrossRef]
  6. Tian, Y.; Zhang, Y.; Wang, Y.; Chen, Y.; Fan, W.; Zhou, J.; Qiao, J.; Wei, Y. Hydrogen, a Novel Therapeutic Molecule, Regulates Oxidative Stress, Inflammation, and Apoptosis. Front. Physiol. 2021, 12, 789507. [Google Scholar] [CrossRef] [PubMed]
  7. Shimouchi, A.; Nose, K.; Shirai, M.; Kondo, T. Estimation of Molecular Hydrogen Consumption in the Human Whole Body after the Ingestion of Hydrogen-Rich Water. Adv. Exp. Med. Biol. 2012, 737, 245–250. [Google Scholar] [CrossRef] [PubMed]
  8. Alwazeer, D.; Liu, F.F.-C.; Wu, X.Y.; LeBaron, T.W. Combating Oxidative Stress and Inflammation in COVID-19 by Molecular Hydrogen Therapy: Mechanisms and Perspectives. Oxid. Med. Cell. Longev. 2021, 2021, 5513868. [Google Scholar] [CrossRef]
  9. Hylemon, P.B.; Harris, S.C.; Ridlon, J.M. Metabolism of Hydrogen Gases and Bile Acids in the Gut Microbiome. FEBS Lett. 2018, 592, 2070–2082. [Google Scholar] [CrossRef]
  10. Smith, N.W.; Shorten, P.R.; Altermann, E.H.; Roy, N.C.; McNabb, W.C. Hydrogen Cross-Feeders of the Human Gastrointestinal Tract. Gut Microbes 2019, 10, 270–288. [Google Scholar] [CrossRef]
  11. Wang, J.; Yin, Y. Clostridium Species for Fermentative Hydrogen Production: An Overview. Int. J. Hydrogen Energy 2021, 46, 34599–34625. [Google Scholar] [CrossRef]
  12. Wolf, P.G.; Biswas, A.; Morales, S.E.; Greening, C.; Gaskins, H.R. H2 Metabolism Is Widespread and Diverse among Human Colonic Microbes. Gut Microbes 2016, 7, 235–245. [Google Scholar] [CrossRef] [PubMed]
  13. Pilcher, J.E. Senn on the diagnosis of gastro-intestinal perforation by the rectal insufflation of hydrogen gas. Ann. Surg. 1888, 8, 190–204. [Google Scholar] [CrossRef] [PubMed]
  14. Zetterstrom, A. Deep-Sea Diving with Synthetic Gas Mixtures. Mil. Surg. 1948, 103, 104–106. [Google Scholar] [CrossRef]
  15. Dole, M.; Wilson, F.R.; Fife, W.P. Hyperbaric Hydrogen Therapy: A Possible Treatment for Cancer. Science 1975, 190, 152–154. [Google Scholar] [CrossRef]
  16. Gharib, B.; Hanna, S.; Abdallahi, O.M.; Lepidi, H.; Gardette, B.; De Reggi, M. Anti-Inflammatory Properties of Molecular Hydrogen: Investigation on Parasite-Induced Liver Inflammation. C. R. Acad. Sci. III 2001, 324, 719–724. [Google Scholar] [CrossRef]
  17. Fu, Z.; Zhang, Z.; Wu, X.; Zhang, J. Hydrogen-Rich Saline Inhibits Lipopolysaccharide-Induced Acute Lung Injury and Endothelial Dysfunction by Regulating Autophagy through mTOR/TFEB Signaling Pathway. BioMed Res. Int. 2020, 2020, 9121894. [Google Scholar] [CrossRef]
  18. Johnsen, H.M.; Hiorth, M.; Klaveness, J. Molecular Hydrogen Therapy—A Review on Clinical Studies and Outcomes. Molecules 2023, 28, 7785. [Google Scholar] [CrossRef]
  19. Ohta, S. Molecular Hydrogen as a Preventive and Therapeutic Medical Gas: Initiation, Development and Potential of Hydrogen Medicine. Pharmacol. Ther. 2014, 144, 1–11. [Google Scholar] [CrossRef]
  20. Russell, G.; Nenov, A.; Hancock, J.T. Oxy-Hydrogen Gas: The Rationale Behind Its Use as a Novel and Sustainable Treatment for COVID-19 and Other Respiratory Diseases. Eur. Med. J. 2021, 10, 21-00027. [Google Scholar] [CrossRef]
  21. Yang, M.; Dong, Y.; He, Q.; Zhu, P.; Zhuang, Q.; Shen, J.; Zhang, X.; Zhao, M. Hydrogen: A Novel Option in Human Disease Treatment. Oxid. Med. Cell. Longev. 2020, 2020, e8384742. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, J.-B.; Kong, X.-F.; Qian, W.; Mu, F.; Lu, T.-Y.; Lu, Y.-Y.; Xu, K.-C. Two Weeks of Hydrogen Inhalation Can Significantly Reverse Adaptive and Innate Immune System Senescence Patients with Advanced Non-Small Cell Lung Cancer: A Self-Controlled Study. Med. Gas Res. 2020, 10, 149–154. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, J.-B.; Kong, X.-F.; Mu, F.; Lu, T.-Y.; Lu, Y.-Y.; Xu, K.-C. Hydrogen Therapy Can Be Used to Control Tumor Progression and Alleviate the Adverse Events of Medications in Patients with Advanced Non-Small Cell Lung Cancer. Med. Gas Res. 2020, 10, 75–80. [Google Scholar] [CrossRef]
  24. Hirano, S.; Ichikawa, Y.; Sato, B.; Yamamoto, H.; Takefuji, Y.; Satoh, F. Molecular Hydrogen as a Potential Clinically Applicable Radioprotective Agent. Int. J. Mol. Sci. 2021, 22, 4566. [Google Scholar] [CrossRef] [PubMed]
  25. Hirano, S.; Yamamoto, H.; Ichikawa, Y.; Sato, B.; Takefuji, Y.; Satoh, F. Molecular Hydrogen as a Novel Antitumor Agent: Possible Mechanisms Underlying Gene Expression. Int. J. Mol. Sci. 2021, 22, 8724. [Google Scholar] [CrossRef]
  26. Hu, Q.; Zhou, Y.; Wu, S.; Wu, W.; Deng, Y.; Shao, A. Molecular Hydrogen: A Potential Radioprotective Agent. Biomed. Pharmacother. 2020, 130, 110589. [Google Scholar] [CrossRef]
  27. Qian, L.; Shen, J.; Chuai, Y.; Cai, J. Hydrogen as a New Class of Radioprotective Agent. Int. J. Biol. Sci. 2013, 9, 887–894. [Google Scholar] [CrossRef]
  28. Zhou, W.; Zhang, J.; Chen, W.; Miao, C. Prospects of Molecular Hydrogen in Cancer Prevention and Treatment. J. Cancer Res. Clin. Oncol. 2024, 150, 170. [Google Scholar] [CrossRef]
  29. Qiu, P.; Liu, Y.; Zhang, J. Recent Advances in Studies of Molecular Hydrogen against Sepsis. Int. J. Biol. Sci. 2019, 15, 1261–1275. [Google Scholar] [CrossRef]
  30. Zhang, Y.; Zhang, J.; Fu, Z. Molecular Hydrogen Is a Potential Protective Agent in the Management of Acute Lung Injury. Mol. Med. 2022, 28, 27. [Google Scholar] [CrossRef]
  31. Li, Y.; Wang, Z.; Lian, N.; Wang, Y.; Zheng, W.; Xie, K. Molecular Hydrogen: A Promising Adjunctive Strategy for the Treatment of the COVID-19. Front. Med. 2021, 8, 671215. [Google Scholar] [CrossRef] [PubMed]
  32. Yang, F.; Yue, R.; Luo, X.; Liu, R.; Huang, X. Hydrogen: A Potential New Adjuvant Therapy for COVID-19 Patients. Front. Pharmacol. 2020, 11, 543718. [Google Scholar] [CrossRef]
  33. Fu, Z.; Zhang, J. Molecular Hydrogen Is a Promising Therapeutic Agent for Pulmonary Disease. J. Zhejiang Univ.-Sci. B 2022, 23, 102–122. [Google Scholar] [CrossRef] [PubMed]
  34. Niki, E. Biomarkers of Lipid Peroxidation in Clinical Material. Biochim. Biophys. Acta BBA-Gen. Subj. 2014, 1840, 809–817. [Google Scholar] [CrossRef]
  35. Valavanidis, A.; Vlachogianni, T.; Fiotakis, C. 8-Hydroxy-2′ -Deoxyguanosine (8-OHdG): A Critical Biomarker of Oxidative Stress and Carcinogenesis. J. Environ. Sci. Health Part C 2009, 27, 120–139. [Google Scholar] [CrossRef]
  36. Radi, R. Oxygen Radicals, Nitric Oxide, and Peroxynitrite: Redox Pathways in Molecular Medicine. Proc. Natl. Acad. Sci. USA 2018, 115, 5839–5848. [Google Scholar] [CrossRef] [PubMed]
  37. Cui, Y.; Zhang, H.; Ji, M.; Jia, M.; Chen, H.; Yang, J.; Duan, M. Hydrogen-Rich Saline Attenuates Neuronal Ischemia–Reperfusion Injury by Protecting Mitochondrial Function in Rats. J. Surg. Res. 2014, 192, 564–572. [Google Scholar] [CrossRef]
  38. Gvozdjáková, A.; Kucharská, J.; Kura, B.; Vančová, O.; Rausová, Z.; Sumbalová, Z.; Uličná, O.; Slezák, J. A New Insight into the Molecular Hydrogen Effect on Coenzyme Q and Mitochondrial Function of Rats. Can. J. Physiol. Pharmacol. 2020, 98, 29–34. [Google Scholar] [CrossRef]
  39. Ishihara, G.; Kawamoto, K.; Komori, N.; Ishibashi, T. Molecular Hydrogen Suppresses Superoxide Generation in the Mitochondrial Complex I and Reduced Mitochondrial Membrane Potential. Biochem. Biophys. Res. Commun. 2020, 522, 965–970. [Google Scholar] [CrossRef]
  40. Chandhok, G.; Lazarou, M.; Neumann, B. Structure, Function, and Regulation of Mitofusin-2 in Health and Disease. Biol. Rev. 2018, 93, 933–949. [Google Scholar] [CrossRef]
  41. Dong, A.; Yu, Y.; Wang, Y.; Li, C.; Chen, H.; Bian, Y.; Zhang, P.; Zhao, Y.; Yu, Y.; Xie, K. Protective Effects of Hydrogen Gas against Sepsis-Induced Acute Lung Injury via Regulation of Mitochondrial Function and Dynamics. Int. Immunopharmacol. 2018, 65, 366–372. [Google Scholar] [CrossRef] [PubMed]
  42. Li, D.; Li, Y.; Pan, W.; Yang, B.; Fu, C. Role of Dynamin-Related Protein 1-Dependent Mitochondrial Fission in Drug-Induced Toxicity. Pharmacol. Res. 2024, 206, 107250. [Google Scholar] [CrossRef] [PubMed]
  43. Fu, Z.; Zhang, J.; Zhang, Y. Role of Molecular Hydrogen in Ageing and Ageing-Related Diseases. Oxid. Med. Cell. Longev. 2022, 2022, 2249749. [Google Scholar] [CrossRef]
  44. Chen, H.; Mao, X.; Meng, X.; Li, Y.; Feng, J.; Zhang, L.; Zhang, Y.; Wang, Y.; Yu, Y.; Xie, K. Hydrogen Alleviates Mitochondrial Dysfunction and Organ Damage via Autophagy-mediated NLRP3 Inflammasome Inactivation in Sepsis. Int. J. Mol. Med. 2019, 44, 1309–1324. [Google Scholar] [CrossRef] [PubMed]
  45. Chelikani, P.; Fita, I.; Loewen, P.C. Diversity of Structures and Properties among Catalases. Cell. Mol. Life Sci. CMLS 2004, 61, 192–208. [Google Scholar] [CrossRef]
  46. Idriss, N.K.; Blann, A.D.; Lip, G.Y.H. Hemoxygenase-1 in Cardiovascular Disease. J. Am. Coll. Cardiol. 2008, 52, 971–978. [Google Scholar] [CrossRef]
  47. Liu, M.; Sun, X.; Chen, B.; Dai, R.; Xi, Z.; Xu, H. Insights into Manganese Superoxide Dismutase and Human Diseases. Int. J. Mol. Sci. 2022, 23, 15893. [Google Scholar] [CrossRef]
  48. Nguyen, N.H.; Tran, G.-B.; Nguyen, C.T. Anti-Oxidative Effects of Superoxide Dismutase 3 on Inflammatory Diseases. J. Mol. Med. 2020, 98, 59–69. [Google Scholar] [CrossRef]
  49. Zhai, X.; Chen, X.; Shi, J.; Shi, D.; Ye, Z.; Liu, W.; Li, M.; Wang, Q.; Kang, Z.; Bi, H.; et al. Lactulose Ameliorates Cerebral Ischemia–Reperfusion Injury in Ratsby Inducing Hydrogen by Activating Nrf2 Expression. Free Radic. Biol. Med. 2013, 65, 731–741. [Google Scholar] [CrossRef]
  50. He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
  51. Rangasamy, T.; Guo, J.; Mitzner, W.A.; Roman, J.; Singh, A.; Fryer, A.D.; Yamamoto, M.; Kensler, T.W.; Tuder, R.M.; Georas, S.N.; et al. Disruption of Nrf2 Enhances Susceptibility to Severe Airway Inflammation and Asthma in Mice. J. Exp. Med. 2005, 202, 47–59. [Google Scholar] [CrossRef] [PubMed]
  52. Kawamura, T.; Wakabayashi, N.; Shigemura, N.; Huang, C.-S.; Masutani, K.; Tanaka, Y.; Noda, K.; Peng, X.; Takahashi, T.; Billiar, T.R.; et al. Hydrogen Gas Reduces Hyperoxic Lung Injury via the Nrf2 Pathway in Vivo. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2013, 304, L646–L656. [Google Scholar] [CrossRef] [PubMed]
  53. Morse, D.; Lin, L.; Choi, A.M.K.; Ryter, S.W. Heme Oxygenase-1, a Critical Arbitrator of Cell Death Pathways in Lung Injury and Disease. Free Radic. Biol. Med. 2009, 47, 1–12. [Google Scholar] [CrossRef] [PubMed]
  54. Sies, H. Oxidative Stress: A Concept in Redox Biology and Medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef]
  55. Bezerra, F.S.; Lanzetti, M.; Nesi, R.T.; Nagato, A.C.; Silva, C.P.E.; Kennedy-Feitosa, E.; Melo, A.C.; Cattani-Cavalieri, I.; Porto, L.C.; Valenca, S.S. Oxidative Stress and Inflammation in Acute and Chronic Lung Injuries. Antioxidants 2023, 12, 548. [Google Scholar] [CrossRef]
  56. Fredenburgh, L.E.; Perrella, M.A.; Mitsialis, S.A. The Role of Heme Oxygenase-1 in Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2007, 36, 158–165. [Google Scholar] [CrossRef]
  57. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef]
  58. Itoh, T.; Fujita, Y.; Ito, M.; Masuda, A.; Ohno, K.; Ichihara, M.; Kojima, T.; Nozawa, Y.; Ito, M. Molecular Hydrogen Suppresses FcεRI-Mediated Signal Transduction and Prevents Degranulation of Mast Cells. Biochem. Biophys. Res. Commun. 2009, 389, 651–656. [Google Scholar] [CrossRef]
  59. Vermot, A.; Petit-Härtlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef]
  60. Aratani, Y. Myeloperoxidase: Its Role for Host Defense, Inflammation, and Neutrophil Function. Arch. Biochem. Biophys. 2018, 640, 47–52. [Google Scholar] [CrossRef]
  61. Diao, M.; Zhang, S.; Wu, L.; Huan, L.; Huang, F.; Cui, Y.; Lin, Z. Hydrogen Gas Inhalation Attenuates Seawater Instillation-Induced Acute Lung Injury via the Nrf2 Pathway in Rabbits. Inflammation 2016, 39, 2029–2039. [Google Scholar] [CrossRef] [PubMed]
  62. Guan, W.-J.; Wei, C.-H.; Chen, A.-L.; Sun, X.-C.; Guo, G.-Y.; Zou, X.; Shi, J.-D.; Lai, P.-Z.; Zheng, Z.-G.; Zhong, N.-S. Hydrogen/Oxygen Mixed Gas Inhalation Improves Disease Severity and Dyspnea in Patients with Coronavirus Disease 2019 in a Recent Multicenter, Open-Label Clinical Trial. J. Thorac. Dis. 2020, 12, 3448–3452. [Google Scholar] [CrossRef]
  63. Zou, R.; Wang, M.-H.; Chen, Y.; Fan, X.; Yang, B.; Du, J.; Wang, X.-B.; Liu, K.-X.; Zhou, J. Hydrogen-Rich Saline Attenuates Acute Lung Injury Induced by Limb Ischemia/Reperfusion via Down-Regulating Chemerin and NLRP3 in Rats. Shock 2019, 52, 134–141. [Google Scholar] [CrossRef] [PubMed]
  64. Qiu, X.; Li, H.; Tang, H.; Jin, Y.; Li, W.; YuSun; PingFeng; Sun, X.; Xia, Z. Hydrogen Inhalation Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Mice. Int. Immunopharmacol. 2011, 11, 2130–2137. [Google Scholar] [CrossRef]
  65. Zhang, Y.; Liu, Y.; Zhang, J. Saturated Hydrogen Saline Attenuates Endotoxin-Induced Lung Dysfunction. J. Surg. Res. 2015, 198, 41–49. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, H.; Xie, K.; Han, H.; Li, Y.; Liu, L.; Yang, T.; Yu, Y. Molecular Hydrogen Protects Mice against Polymicrobial Sepsis by Ameliorating Endothelial Dysfunction via an Nrf2/HO-1 Signaling Pathway. Int. Immunopharmacol. 2015, 28, 643–654. [Google Scholar] [CrossRef]
  67. Xie, K.; Yu, Y.; Huang, Y.; Zheng, L.; Li, J.; Chen, H.; Han, H.; Hou, L.; Gong, G.; Wang, G. Molecular Hydrogen Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Mice Through Reducing Inflammation and Apoptosis. Shock 2012, 37, 548–555. [Google Scholar] [CrossRef]
  68. Chen, M.; Zhang, J.; Chen, Y.; Qiu, Y.; Luo, Z.; Zhao, S.; Du, L.; Tian, D. Hydrogen Protects Lung from Hypoxia/Re-Oxygenation Injury by Reducing Hydroxyl Radical Production and Inhibiting Inflammatory Responses. Sci. Rep. 2018, 8, 8004. [Google Scholar] [CrossRef]
  69. He, B.; Zhang, Y.; Kang, B.; Xiao, J.; Xie, B.; Wang, Z. Protection of Oral Hydrogen Water as an Antioxidant on Pulmonary Hypertension. Mol. Biol. Rep. 2013, 40, 5513–5521. [Google Scholar] [CrossRef]
  70. Itoh, T.; Hamada, N.; Terazawa, R.; Ito, M.; Ohno, K.; Ichihara, M.; Nozawa, Y.; Ito, M. Molecular Hydrogen Inhibits Lipopolysaccharide/Interferon γ-Induced Nitric Oxide Production through Modulation of Signal Transduction in Macrophages. Biochem. Biophys. Res. Commun. 2011, 411, 143–149. [Google Scholar] [CrossRef]
  71. Du, J.; Li, J.; Li, R.; Yan, X. High Concentration of Hydrogen Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in a Sirt1-Dependent Manner. Respir. Physiol. Neurobiol. 2022, 296, 103808. [Google Scholar] [CrossRef] [PubMed]
  72. Sun, Q.; Han, W.; Hu, H.; Fan, D.; Li, Y.; Zhang, Y.; Lv, Y.; Li, M.; Pan, S. Hydrogen Alleviates Hyperoxic Acute Lung Injury Related Endoplasmic Reticulum Stress in Rats through Upregulation of SIRT1. Free Radic. Res. 2017, 51, 622–632. [Google Scholar] [CrossRef]
  73. Li, H.; Zhou, R.; Liu, J.; Li, Q.; Zhang, J.; Mu, J.; Sun, X. Hydrogen-Rich Saline Attenuates Lung Ischemia-Reperfusion Injury in Rabbits. J. Surg. Res. 2012, 174, e11–e16. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, P.; Wei, S.; Huang, W.; Wu, P.; Chen, S.; Tao, A.; Wang, H.; Liang, Z.; Chen, R.; Yan, J.; et al. Hydrogen Gas Inhalation Enhances Alveolar Macrophage Phagocytosis in an Ovalbumin-Induced Asthma Model. Int. Immunopharmacol. 2019, 74, 105646. [Google Scholar] [CrossRef]
  75. Wu, D.; Liang, M.; Dang, H.; Fang, F.; Xu, F.; Liu, C. Hydrogen Protects against Hyperoxia-Induced Apoptosis in Type II Alveolar Epithelial Cells via Activation of PI3K/Akt/Foxo3a Signaling Pathway. Biochem. Biophys. Res. Commun. 2018, 495, 1620–1627. [Google Scholar] [CrossRef] [PubMed]
  76. Russell, G.; Nenov, A.; Kisher, H.; Hancock, J.T. Molecular Hydrogen as Medicine: An Assessment of Administration Methods. Hydrogen 2021, 2, 444–460. [Google Scholar] [CrossRef]
  77. Korovljev, D.; Stajer, V.; Ostojic, J.; LeBaron, T.W.; Ostojic, S.M. Hydrogen-Rich Water Reduces Liver Fat Accumulation and Improves Liver Enzyme Profiles in Patients with Non-Alcoholic Fatty Liver Disease: A Randomized Controlled Pilot Trial. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 688–693. [Google Scholar] [CrossRef]
  78. LeBaron, T.W.; Laher, I.; Kura, B.; Slezak, J. Hydrogen Gas: From Clinical Medicine to an Emerging Ergogenic Molecule for Sports Athletes. Can. J. Physiol. Pharmacol. 2019, 97, 797–807. [Google Scholar] [CrossRef]
  79. Lucas, K.; Rosch, M.; Langguth, P. Molecular Hydrogen (H2) as a Potential Treatment for Acute and Chronic Fatigue. Arch. Pharm. 2021, 354, 2000378. [Google Scholar] [CrossRef]
  80. Alharbi, A.A.D.; Ebine, N.; Nakae, S.; Hojo, T.; Fukuoka, Y. Application of Molecular Hydrogen as an Antioxidant in Responses to Ventilatory and Ergogenic Adjustments during Incremental Exercise in Humans. Nutrients 2021, 13, 459. [Google Scholar] [CrossRef]
  81. Asada, R.; Kageyama, K.; Tanaka, H.; Matsui, H.; Kimura, M.; Saitoh, Y.; Miwa, N. Antitumor Effects of Nano-Bubble Hydrogen-Dissolved Water Are Enhanced by Coexistent Platinum Colloid and the Combined Hyperthermia with Apoptosis-like Cell Death. Oncol. Rep. 2010, 24, 1463–1470. [Google Scholar] [CrossRef] [PubMed]
  82. Ito, M.; Hirayama, M.; Yamai, K.; Goto, S.; Ito, M.; Ichihara, M.; Ohno, K. Drinking Hydrogen Water and Intermittent Hydrogen Gas Exposure, but Not Lactulose or Continuous Hydrogen Gas Exposure, Prevent 6-Hydorxydopamine-Induced Parkinson’s Disease in Rats. Med. Gas Res. 2012, 2, 15. [Google Scholar] [CrossRef] [PubMed]
  83. Yang, N.; Gong, F.; Liu, B.; Hao, Y.; Chao, Y.; Lei, H.; Yang, X.; Gong, Y.; Wang, X.; Liu, Z.; et al. Magnesium Galvanic Cells Produce Hydrogen and Modulate the Tumor Microenvironment to Inhibit Cancer Growth. Nat. Commun. 2022, 13, 2336. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, L.; Zhao, P.; Yue, C.; Jin, Z.; Liu, Q.; Du, X.; He, Q. Sustained Release of Bioactive Hydrogen by Pd Hydride Nanoparticles Overcomes Alzheimer’s Disease. Biomaterials 2019, 197, 393–404. [Google Scholar] [CrossRef]
  85. Xu, F.; Yu, S.; Qin, M.; Mao, Y.; Jin, L.; Che, N.; Liu, S.; Ge, R. Hydrogen-Rich Saline Ameliorates Allergic Rhinitis by Reversing the Imbalance of Th1/Th2 and Up-Regulation of CD4+CD25+Foxp3+Regulatory T Cells, Interleukin-10, and Membrane-Bound Transforming Growth Factor-β in Guinea Pigs. Inflammation 2018, 41, 81–92. [Google Scholar] [CrossRef]
  86. Yu, S.; Zhao, C.; Che, N.; Jing, L.; Ge, R. Hydrogen-Rich Saline Attenuates Eosinophil Activation in a Guinea Pig Model of Allergic Rhinitis via Reducing Oxidative Stress. J. Inflamm. 2017, 14, 1. [Google Scholar] [CrossRef]
  87. Fang, S.; Li, X.; Wei, X.; Zhang, Y.; Ma, Z.; Wei, Y.; Wang, W. Beneficial Effects of Hydrogen Gas Inhalation on a Murine Model of Allergic Rhinitis. Exp. Ther. Med. 2018, 16, 5178–5184. [Google Scholar] [CrossRef]
  88. Zhao, C.; Yu, S.; Li, J.; Xu, W.; Ge, R. Changes in IL-4 and IL-13 Expression in Allergic-Rhinitis Treated with Hydrogen-Rich Saline in Guinea-Pig Model. Allergol. Immunopathol. 2017, 45, 350–355. [Google Scholar] [CrossRef]
  89. Jin, L.; Tan, S.; Fan, K.; Wang, Y.; Yu, S. Research Progress of Hydrogen on Chronic Nasal Inflammation. J. Inflamm. Res. 2023, 16, 2149–2157. [Google Scholar] [CrossRef]
  90. Jin, L.; Fan, K.; Tan, S.; Liu, S.; Ge, Q.; Wang, Y.; Ai, Z.; Yu, S. The Beneficial Effects of Hydrogen-Rich Saline Irrigation on Chronic Rhinitis: A Randomized, Double-Blind Clinical Trial. J. Inflamm. Res. 2022, 15, 3983–3995. [Google Scholar] [CrossRef]
  91. Jin, L.; Yu, S.Q.; Zhang, X.; Ge, Q.; Zhang, X.L.; Wang, Y.; Qin, M.L. Clinical study of hydrogen-rich saline in the treatment of moderate to severe allergic rhinitis. Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi J. Clin. Otorhinolaryngol. Head Neck Surg. 2018, 32, 493–496. [Google Scholar] [CrossRef]
  92. Zhao, S.; Yang, Y.; Liu, W.; Xuan, Z.; Wu, S.; Yu, S.; Mei, K.; Huang, Y.; Zhang, P.; Cai, J.; et al. Protective Effect of Hydrogen-Rich Saline against Radiation-Induced Immune Dysfunction. J. Cell. Mol. Med. 2014, 18, 938–946. [Google Scholar] [CrossRef] [PubMed]
  93. Karlidaǧ, T.; Keles, E.; İlhan, N.; Yakclin, S.; Kaygusuz, İ.; Yildiz, M. Roles of Free Radicals, Nitric Oxide, and Scavenging Enzymes in Nasal Polyp Development. Ann. Otol. Rhinol. Laryngol. 2005, 114, 122–126. [Google Scholar] [CrossRef]
  94. Uneri, C.; Oztürk, O.; Polat, S.; Yüksel, M.; Haklar, G. Determination of Reactive Oxygen Species in Nasal Polyps. Rhinology 2005, 43, 185–189. [Google Scholar]
  95. Choi, J.; Suk An, E.; Ban, Y.-H.; Woom Seo, D.; Kim, T.-S.; Lee, S.-P.; Lin, Y.; Choi, E.-K.; Kim, Y.-B. Hydrogen-Enriched Water Eliminates Fine Particles from the Lungs and Blood by Enhancing Phagocytic Activity. J. Biomed. Res. 2017, 31, 503–511. [Google Scholar] [CrossRef]
  96. GINA 2022, Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention. Available online: https://ginasthma.org/gina-reports/ (accessed on 20 November 2024).
  97. Zhang, N.; Deng, C.; Zhang, X.; Zhang, J.; Bai, C. Inhalation of Hydrogen Gas Attenuates Airway Inflammation and Oxidative Stress in Allergic Asthmatic Mice. Asthma Res. Pract. 2018, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  98. Xiao, M.; Zhu, T.; Wang, T.; Wen, F.-Q. Hydrogen-Rich Saline Reduces Airway Remodeling via Inactivation of NF-κB in a Murine Model of Asthma. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 1033–1043. [Google Scholar]
  99. Zhang, J.; Feng, X.; Fan, Y.; Zhu, G.; Bai, C. Molecular Hydrogen Alleviates Asthma through Inhibiting IL-33/ILC2 Axis. Inflamm. Res. 2021, 70, 569–579. [Google Scholar] [CrossRef]
  100. Cho, H.-Y.; Park, S.; Miller, L.; Lee, H.-C.; Langenbach, R.; Kleeberger, S.R. Role for Mucin-5AC in Upper and Lower Airway Pathogenesis in Mice. Toxicol. Pathol. 2021, 49, 1077–1099. [Google Scholar] [CrossRef]
  101. Kuo, W.; Odenwald, M.A.; Turner, J.R.; Zuo, L. Tight Junction Proteins Occludin and ZO-1 as Regulators of Epithelial Proliferation and Survival. Ann. N. Y. Acad. Sci. 2022, 1514, 21–33. [Google Scholar] [CrossRef]
  102. Niu, Y.; Nie, Q.; Dong, L.; Zhang, J.; Liu, S.F.; Song, W.; Wang, X.; Wu, G.; Song, D. Hydrogen Attenuates Allergic Inflammation by Reversing Energy Metabolic Pathway Switch. Sci. Rep. 2020, 10, 1962. [Google Scholar] [CrossRef] [PubMed]
  103. Singh, R.B.; Tarnava, A.; Fatima, G.; Fedacko, J.; Mojto, V.; LeBaron, T.W. Can Hydrogen Water Enhance Oxygen Saturation in Patients with Chronic Lung Disease? A Non-Randomized, Observational Pilot Study. Diseases 2023, 11, 127. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, S.-T.; Bao, C.; He, Y.; Tian, X.; Yang, Y.; Zhang, T.; Xu, K.-F. Hydrogen Gas (XEN) Inhalation Ameliorates Airway Inflammation in Asthma and COPD Patients. QJM Int. J. Med. 2020, 113, 870–875. [Google Scholar] [CrossRef]
  105. Feng, S.; Duan, E.; Shi, X.; Zhang, H.; Li, H.; Zhao, Y.; Chao, L.; Zhong, X.; Zhang, W.; Li, R.; et al. Hydrogen Ameliorates Lung Injury in a Rat Model of Subacute Exposure to Concentrated Ambient PM2.5 via Aryl Hydrocarbon Receptor. Int. Immunopharmacol. 2019, 77, 105939. [Google Scholar] [CrossRef]
  106. 2025 GOLD Report. Available online: https://goldcopd.org/2025-gold-report/ (accessed on 11 February 2025).
  107. Barnes, P.J. Inflammatory Mechanisms in Patients with Chronic Obstructive Pulmonary Disease. J. Allergy Clin. Immunol. 2016, 138, 16–27. [Google Scholar] [CrossRef]
  108. Celli, B.; Fabbri, L.; Criner, G.; Martinez, F.J.; Mannino, D.; Vogelmeier, C.; Montes De Oca, M.; Papi, A.; Sin, D.D.; Han, M.K.; et al. Definition and Nomenclature of Chronic Obstructive Pulmonary Disease: Time for Its Revision. Am. J. Respir. Crit. Care Med. 2022, 206, 1317–1325. [Google Scholar] [CrossRef]
  109. Domej, W.; Oettl, K.; Renner, W. Oxidative Stress and Free Radicals in COPD--Implications and Relevance for Treatment. Int. J. Chron. Obstruct. Pulmon. Dis. 2014, 9, 1207–1224. [Google Scholar] [CrossRef]
  110. Liu, X.; Ma, C.; Wang, X.; Wang, W.; Li, Z.; Wang, X.; Wang, P.; Sun, W.; Xue, B. Hydrogen Coadministration Slows the Development of COPD-like Lung Disease in a Cigarette Smoke-Induced Rat Model. Int. J. Chron. Obstruct. Pulmon. Dis. 2017, 12, 1309–1324. [Google Scholar] [CrossRef] [PubMed]
  111. Lu, W.; Li, D.; Hu, J.; Mei, H.; Shu, J.; Long, Z.; Yuan, L.; Li, D.; Guan, R.; Li, Y.; et al. Hydrogen Gas Inhalation Protects against Cigarette Smoke-Induced COPD Development in Mice. J. Thorac. Dis. 2018, 10, 3232–3243. [Google Scholar] [CrossRef]
  112. Suzuki, Y.; Sato, T.; Sugimoto, M.; Baskoro, H.; Karasutani, K.; Mitsui, A.; Nurwidya, F.; Arano, N.; Kodama, Y.; Hirano, S.; et al. Hydrogen-Rich Pure Water Prevents Cigarette Smoke-Induced Pulmonary Emphysema in SMP30 Knockout Mice. Biochem. Biophys. Res. Commun. 2017, 492, 74–81. [Google Scholar] [CrossRef]
  113. Ning, Y.; Shang, Y.; Huang, H.; Zhang, J.; Dong, Y.; Xu, W.; Li, Q. Attenuation of Cigarette Smoke-Induced Airway Mucus Production by Hydrogen-Rich Saline in Rats. PLoS ONE 2013, 8, e83429. [Google Scholar] [CrossRef] [PubMed]
  114. Grünwald, B.; Schoeps, B.; Krüger, A. Recognizing the Molecular Multifunctionality and Interactome of TIMP-1. Trends Cell Biol. 2019, 29, 6–19. [Google Scholar] [CrossRef] [PubMed]
  115. Zheng, Z.-G.; Sun, W.-Z.; Hu, J.-Y.; Jie, Z.-J.; Xu, J.-F.; Cao, J.; Song, Y.-L.; Wang, C.-H.; Wang, J.; Zhao, H.; et al. Hydrogen/Oxygen Therapy for the Treatment of an Acute Exacerbation of Chronic Obstructive Pulmonary Disease: Results of a Multicenter, Randomized, Double-Blind, Parallel-Group Controlled Trial. Respir. Res. 2021, 22, 149. [Google Scholar] [CrossRef]
  116. Aokage, T.; Seya, M.; Hirayama, T.; Nojima, T.; Iketani, M.; Ishikawa, M.; Terasaki, Y.; Taniguchi, A.; Miyahara, N.; Nakao, A.; et al. The Effects of Inhaling Hydrogen Gas on Macrophage Polarization, Fibrosis, and Lung Function in Mice with Bleomycin-Induced Lung Injury. BMC Pulm. Med. 2021, 21, 339. [Google Scholar] [CrossRef] [PubMed]
  117. Dong, W.-W.; Zhang, Y.-Q.; Zhu, X.-Y.; Mao, Y.-F.; Sun, X.-J.; Liu, Y.-J.; Jiang, L. Protective Effects of Hydrogen-Rich Saline Against Lipopolysaccharide-Induced Alveolar Epithelial-to-Mesenchymal Transition and Pulmonary Fibrosis. Med. Sci. Monit. 2017, 23, 2357–2364. [Google Scholar] [CrossRef]
  118. Gao, L.; Jiang, D.; Geng, J.; Dong, R.; Dai, H. Hydrogen Inhalation Attenuated Bleomycin-induced Pulmonary Fibrosis by Inhibiting Transforming Growth Factor-β1 and Relevant Oxidative Stress and Epithelial-to-mesenchymal Transition. Exp. Physiol. 2019, 104, 1942–1951. [Google Scholar] [CrossRef]
  119. Liu, S.; Liu, K.; Sun, Q.; Liu, W.; Xu, W.; Denoble, P.; Tao, H.; Sun, X. Consumption of Hydrogen Water Reduces Paraquat-Induced Acute Lung Injury in Rats. BioMed Res. Int. 2011, 2011, 305086. [Google Scholar] [CrossRef]
  120. Zhou, Z.-Q.; Zhong, C.-H.; Su, Z.-Q.; Li, X.-Y.; Chen, Y.; Chen, X.-B.; Tang, C.-L.; Zhou, L.-Q.; Li, S.-Y. Breathing Hydrogen-Oxygen Mixture Decreases Inspiratory Effort in Patients with Tracheal Stenosis. Respiration 2019, 97, 42–51. [Google Scholar] [CrossRef]
  121. Ozeki, N.; Yamawaki-Ogata, A.; Narita, Y.; Mii, S.; Ushida, K.; Ito, M.; Hirano, S.; Kurokawa, R.; Ohno, K.; Usui, A. Hydrogen Water Alleviates Obliterative Airway Disease in Mice. Gen. Thorac. Cardiovasc. Surg. 2020, 68, 158–163. [Google Scholar] [CrossRef]
  122. Muramatsu, Y.; Ito, M.; Oshima, T.; Kojima, S.; Ohno, K. Hydrogen-Rich Water Ameliorates Bronchopulmonary Dysplasia (BPD) in Newborn Rats. Pediatr. Pulmonol. 2016, 51, 928–935. [Google Scholar] [CrossRef]
  123. Dao, D.T.; Nandivada, P.; Vuong, J.T.; Anez-Bustillos, L.; Pan, A.; Kishikawa, H.; Mitchell, P.D.; Baker, M.A.; Fell, G.L.; Martin, T.; et al. Vascular Endothelial Growth Factor Accelerates Compensatory Lung Growth by Increasing the Alveolar Units. Pediatr. Res. 2018, 83, 1182–1189. [Google Scholar] [CrossRef] [PubMed]
  124. Hattori, Y.; Kotani, T.; Tsuda, H.; Mano, Y.; Tu, L.; Li, H.; Hirako, S.; Ushida, T.; Imai, K.; Nakano, T.; et al. Maternal Molecular Hydrogen Treatment Attenuates Lipopolysaccharide-Induced Rat Fetal Lung Injury. Free Radic. Res. 2015, 49, 1026–1037. [Google Scholar] [CrossRef] [PubMed]
  125. Kishimoto, Y.; Kato, T.; Ito, M.; Azuma, Y.; Fukasawa, Y.; Ohno, K.; Kojima, S. Hydrogen Ameliorates Pulmonary Hypertension in Rats by Anti-Inflammatory and Antioxidant Effects. J. Thorac. Cardiovasc. Surg. 2015, 150, 645–654.e3. [Google Scholar] [CrossRef] [PubMed]
  126. Zhai, Y.; Zhou, X.; Dai, Q.; Fan, Y.; Huang, X. Hydrogen-Rich Saline Ameliorates Lung Injury Associated with Cecal Ligation and Puncture-Induced Sepsis in Rats. Exp. Mol. Pathol. 2015, 98, 268–276. [Google Scholar] [CrossRef]
  127. Ren, J.-D.; Wu, X.-B.; Jiang, R.; Hao, D.-P.; Liu, Y. Molecular Hydrogen Inhibits Lipopolysaccharide-Triggered NLRP3 Inflammasome Activation in Macrophages by Targeting the Mitochondrial Reactive Oxygen Species. Biochim. Biophys. Acta BBA-Mol. Cell Res. 2016, 1863, 50–55. [Google Scholar] [CrossRef]
  128. Chen, H.; Bai, C.; Wang, X. The Value of the Lipopolysaccharide-Induced Acute Lung Injury Model in Respiratory Medicine. Expert Rev. Respir. Med. 2010, 4, 773–783. [Google Scholar] [CrossRef]
  129. Tao, B.; Liu, L.; Wang, N.; Wang, W.; Jiang, J.; Zhang, J. Effects of Hydrogen-Rich Saline on Aquaporin 1, 5 in Septic Rat Lungs. J. Surg. Res. 2016, 202, 291–298. [Google Scholar] [CrossRef]
  130. Liu, H.; Liang, X.; Wang, D.; Zhang, H.; Liu, L.; Chen, H.; Li, Y.; Duan, Q.; Xie, K. Combination Therapy with Nitric Oxide and Molecular Hydrogen in a Murine Model of Acute Lung Injury. Shock 2015, 43, 504. [Google Scholar] [CrossRef]
  131. Liu, L.-D.; Wu, X.-Y.; Tao, B.-D.; Wang, N.; Zhang, J. Protective Effect and Mechanism of Hydrogen Treatment on Lung Epithelial Barrier Dysfunction in Rats with Sepsis. Genet. Mol. Res. 2016, 15, gmr.15016050. [Google Scholar] [CrossRef]
  132. Wang, Y.; Zhang, J.; Bo, J.; Wang, X.; Zhu, J. Hydrogen-Rich Saline Ameliorated LPS-Induced Acute Lung Injury via Autophagy Inhibition through the ROS/AMPK/mTOR Pathway in Mice. Exp. Biol. Med. 2019, 244, 721–727. [Google Scholar] [CrossRef]
  133. Saramago, E.A.; Borges, G.S.; Singolani, C.G., Jr.; Nogueira, J.E.; Soriano, R.N.; Cárnio, E.C.; Branco, L.G.S. Molecular Hydrogen Potentiates Hypothermia and Prevents Hypotension and Fever in LPS-Induced Systemic Inflammation. Brain. Behav. Immun. 2019, 75, 119–128. [Google Scholar] [CrossRef]
  134. Mirzayans, R.; Murray, D. Do TUNEL and Other Apoptosis Assays Detect Cell Death in Preclinical Studies? Int. J. Mol. Sci. 2020, 21, 9090. [Google Scholar] [CrossRef]
  135. Kawamura, T.; Huang, C.-S.; Peng, X.; Masutani, K.; Shigemura, N.; Billiar, T.R.; Okumura, M.; Toyoda, Y.; Nakao, A. The Effect of Donor Treatment with Hydrogen on Lung Allograft Function in Rats. Surgery 2011, 150, 240–249. [Google Scholar] [CrossRef] [PubMed]
  136. Kawamura, T.; Huang, C.-S.; Tochigi, N.; Lee, S.; Shigemura, N.; Billiar, T.R.; Okumura, M.; Nakao, A.; Toyoda, Y. Inhaled Hydrogen Gas Therapy for Prevention of Lung Transplant-Induced Ischemia/Reperfusion Injury in Rats. Transplantation 2010, 90, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
  137. Haam, S.; Lee, J.G.; Paik, H.C.; Park, M.S.; Lim, B.J. Hydrogen Gas Inhalation during Ex Vivo Lung Perfusion of Donor Lungs Recovered after Cardiac Death. J. Heart Lung Transpl. 2018, 37, 1271–1278. [Google Scholar] [CrossRef]
  138. Amarelle, L.; Quintela, L.; Hurtado, J.; Malacrida, L. Hyperoxia and Lungs: What We Have Learned From Animal Models. Front. Med. 2021, 8, 606678. [Google Scholar] [CrossRef] [PubMed]
  139. Kallet, R.H.; Matthay, M.A. Hyperoxic Acute Lung Injury. Respir. Care 2013, 58, 123–141. [Google Scholar] [CrossRef]
  140. Huang, C.-S.; Kawamura, T.; Peng, X.; Tochigi, N.; Shigemura, N.; Billiar, T.R.; Nakao, A.; Toyoda, Y. Hydrogen Inhalation Reduced Epithelial Apoptosis in Ventilator-Induced Lung Injury via a Mechanism Involving Nuclear Factor-Kappa B Activation. Biochem. Biophys. Res. Commun. 2011, 408, 253–258. [Google Scholar] [CrossRef]
  141. Huang, C.-S.; Kawamura, T.; Lee, S.; Tochigi, N.; Shigemura, N.; Buchholz, B.M.; Kloke, J.D.; Billiar, T.R.; Toyoda, Y.; Nakao, A. Hydrogen Inhalation Ameliorates Ventilator-Induced Lung Injury. Crit. Care 2010, 14, R234. [Google Scholar] [CrossRef]
  142. Ibsen, L.M.; Koch, T. Submersion and Asphyxial Injury. Crit. Care Med. 2002, 30, S402–S408. [Google Scholar] [CrossRef]
  143. Chen, X.; Liu, Q.; Wang, D.; Feng, S.; Zhao, Y.; Shi, Y.; Liu, Q. Protective Effects of Hydrogen-Rich Saline on Rats with Smoke Inhalation Injury. Oxid. Med. Cell. Longev. 2015, 2015, 106836. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, B.; Zhou, X.; Qiu, Y.; Song, Y.; Feng, F.; Feng, J.; Song, Q.; Jia, Q.; Wang, J. Clinical Characteristics of 82 Cases of Death from COVID-19. PLoS ONE 2020, 15, e0235458. [Google Scholar] [CrossRef]
  145. Tay, M.Z.; Poh, C.M.; Rénia, L.; MacAry, P.A.; Ng, L.F.P. The Trinity of COVID-19: Immunity, Inflammation and Intervention. Nat. Rev. Immunol. 2020, 20, 363–374. [Google Scholar] [CrossRef]
  146. Erlich, J.R.; To, E.E.; Liong, S.; Brooks, R.; Vlahos, R.; O’Leary, J.J.; Brooks, D.A.; Selemidis, S. Targeting Evolutionary Conserved Oxidative Stress and Immunometabolic Pathways for the Treatment of Respiratory Infectious Diseases. Antioxid. Redox Signal. 2020, 32, 993–1013. [Google Scholar] [CrossRef] [PubMed]
  147. Li, H.; Liu, L.; Zhang, D.; Xu, J.; Dai, H.; Tang, N.; Su, X.; Cao, B. SARS-CoV-2 and Viral Sepsis: Observations and Hypotheses. Lancet 2020, 395, 1517–1520. [Google Scholar] [CrossRef]
  148. Zeng, Y.; Guan, W.; Wang, K.; Jie, Z.; Zou, X.; Tan, X.; Li, X.; Chen, X.; Ren, X.; Jiang, J.; et al. Effect of Hydrogen/Oxygen Therapy for Ordinary COVID-19 Patients: A Propensity-Score Matched Case-Control Study. BMC Infect. Dis. 2023, 23, 440. [Google Scholar] [CrossRef]
  149. Luo, P.; Ding, Y.; He, Y.; Chen, D.; He, Q.; Huang, Z.; Huang, S.; Lei, W. Hydrogen-Oxygen Therapy Alleviates Clinical Symptoms in Twelve Patients Hospitalized with COVID-19: A Retrospective Study of Medical Records. Medicine 2022, 101, e27759. [Google Scholar] [CrossRef]
  150. Yu, Y.; Yang, Y.; Yang, M.; Wang, C.; Xie, K.; Yu, Y. Hydrogen Gas Reduces HMGB1 Release in Lung Tissues of Septic Mice in an Nrf2/HO-1-Dependent Pathway. Int. Immunopharmacol. 2019, 69, 11–18. [Google Scholar] [CrossRef] [PubMed]
  151. Shi, M.-M.; Chen, Y.-T.; Wang, X.-D.; Zhang, Y.-F.; Cheng, T.; Chen, H.; Sun, F.; Bao, H.; Chen, R.; Xiong, W.-N.; et al. The Efficacy of Hydrogen/Oxygen Therapy Favored the Recovery of Omicron SARS-CoV-2 Variant Infection: Results of a Multicenter, Randomized, Controlled Trial. J. Clin. Biochem. Nutr. 2023, 73, 228–233. [Google Scholar] [CrossRef]
  152. Botek, M.; Krejčí, J.; Valenta, M.; McKune, A.; Sládečková, B.; Konečný, P.; Klimešová, I.; Pastucha, D. Molecular Hydrogen Positively Affects Physical and Respiratory Function in Acute Post-COVID-19 Patients: A New Perspective in Rehabilitation. Int. J. Environ. Res. Public. Health 2022, 19, 1992. [Google Scholar] [CrossRef]
  153. Pozdnyakova, D.D.; Bakhareva, T.A.; Baranova, I.A.; Selemir, V.D.; Chuchalin, A.G. Rehabilitation Program of Post-COVID-19 Syndrome with the Use of Nitric Oxide and Molecular Hydrogen. Ter. Arkh. 2024, 96, 260–265. [Google Scholar] [CrossRef] [PubMed]
  154. Saitoh, Y.; Okayasu, H.; Xiao, L.; Harata, Y.; Miwa, N. Neutral pH Hydrogen-Enriched Electrolyzed Water Achieves Tumor-Preferential Clonal Growth Inhibition Over Normal Cells and Tumor Invasion Inhibition Concurrently with Intracellular Oxidant Repression. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2008, 17, 247–255. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, D.; Wang, L.; Zhang, Y.; Zhao, Y.; Chen, G. Hydrogen Gas Inhibits Lung Cancer Progression through Targeting SMC3. Biomed. Pharmacother. 2018, 104, 788–797. [Google Scholar] [CrossRef]
  156. Akagi, J.; Baba, H. Hydrogen Gas Restores Exhausted CD8+ T Cells in Patients with Advanced Colorectal Cancer to Improve Prognosis. Oncol. Rep. 2019, 41, 301–311. [Google Scholar] [CrossRef] [PubMed]
  157. Saitoh, Y.; Yoshimura, Y.; Nakano, K.; Miwa, N. Platinum Nanocolloid-Supplemented Hydrogendissolved Water Inhibits Growth of Human Tongue Carcinoma Cells Preferentially over Normal Cells. Exp. Oncol. 2009, 31, 156–162. [Google Scholar]
  158. Chen, J.-B.; Kong, X.-F.; Lv, Y.-Y.; Qin, S.-C.; Sun, X.-J.; Mu, F.; Lu, T.-Y.; Xu, K.-C. “Real World Survey” of Hydrogen-Controlled Cancer: A Follow-up Report of 82 Advanced Cancer Patients. Med. Gas Res. 2019, 9, 115–121. [Google Scholar] [CrossRef]
  159. Akagi, J.; Baba, H. Hydrogen Gas Activates Coenzyme Q10 to Restore Exhausted CD8+ T Cells, Especially PD-1+Tim3+terminal CD8+ T Cells, Leading to Better Nivolumab Outcomes in Patients with Lung Cancer. Oncol. Lett. 2020, 20, 258. [Google Scholar] [CrossRef]
  160. Kong, X.; Lu, T.; Lu, Y.-Y.; Yin, Z.; Xu, K. Effect of Hydrogen Inhalation Therapy on Hearing Loss of Patients with Nasopharyngeal Carcinoma After Radiotherapy. Front. Med. 2022, 9, 828370. [Google Scholar] [CrossRef]
  161. Nakashima-Kamimura, N.; Mori, T.; Ohsawa, I.; Asoh, S.; Ohta, S. Molecular Hydrogen Alleviates Nephrotoxicity Induced by an Anti-Cancer Drug Cisplatin without Compromising Anti-Tumor Activity in Mice. Cancer Chemother. Pharmacol. 2009, 64, 753–761. [Google Scholar] [CrossRef]
  162. Yang, Y.; Liu, P.Y.; Bao, W.; Chen, S.J.; Wu, F.S.; Zhu, P.Y. Hydrogen Inhibits Endometrial Cancer Growth via a ROS/NLRP3/Caspase-1/GSDMD-Mediated Pyroptotic Pathway. BMC Cancer 2020, 20, 28. [Google Scholar] [CrossRef]
  163. Meng, J.; Liu, L.; Wang, D.; Yan, Z.; Chen, G. Hydrogen Gas Represses the Progression of Lung Cancer via Down-Regulating CD47. Biosci. Rep. 2020, 40, BSR20192761. [Google Scholar] [CrossRef]
  164. Lin, Y.; Ohkawara, B.; Ito, M.; Misawa, N.; Miyamoto, K.; Takegami, Y.; Masuda, A.; Toyokuni, S.; Ohno, K. Molecular Hydrogen Suppresses Activated Wnt/β-Catenin Signaling. Sci. Rep. 2016, 6, 31986. [Google Scholar] [CrossRef]
  165. Mishra, K.; Alsbeih, G. Appraisal of Biochemical Classes of Radioprotectors: Evidence, Current Status and Guidelines for Future Development. 3 Biotech 2017, 7, 292. [Google Scholar] [CrossRef] [PubMed]
  166. Sato, T.; Kinoshita, M.; Yamamoto, T.; Ito, M.; Nishida, T.; Takeuchi, M.; Saitoh, D.; Seki, S.; Mukai, Y. Treatment of Irradiated Mice with High-Dose Ascorbic Acid Reduced Lethality. PLoS ONE 2015, 10, e0117020. [Google Scholar] [CrossRef] [PubMed]
  167. Chen, Y.; Zong, C.; Jia, J.; Liu, Y.; Zhang, Z.; Cai, B.; Tian, L. A Study on the Protective Effect of Molecular Hydrogen on Osteoradionecrosis of the Jaw in Rats. Int. J. Oral Maxillofac. Surg. 2020, 49, 1648–1654. [Google Scholar] [CrossRef] [PubMed]
  168. Mei, K.; Zhao, S.; Qian, L.; Li, B.; Ni, J.; Cai, J. Hydrogen Protects Rats from Dermatitis Caused by Local Radiation. J. Dermatol. Treat. 2014, 25, 182–188. [Google Scholar] [CrossRef]
  169. Qian, L.; Li, B.; Cao, F.; Huang, Y.; Liu, S.; Cai, J.; Gao, F. Hydrogen-Rich PBS Protects Cultured Human Cells from Ionizing Radiation-Induced Cellular Damage. Nucl. Technol. Radiat. Prot. 2010, 25, 23–29. [Google Scholar] [CrossRef]
  170. Terasaki, Y.; Ohsawa, I.; Terasaki, M.; Takahashi, M.; Kunugi, S.; Dedong, K.; Urushiyama, H.; Amenomori, S.; Kaneko-Togashi, M.; Kuwahara, N.; et al. Hydrogen Therapy Attenuates Irradiation-Induced Lung Damage by Reducing Oxidative Stress. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2011, 301, L415–L426. [Google Scholar] [CrossRef]
  171. Yang, Y.; Li, B.; Liu, C.; Chuai, Y.; Lei, J.; Gao, F.; Cui, J.; Sun, D.; Cheng, Y.; Zhou, C.; et al. Hydrogen-Rich Saline Protects Immunocytes from Radiation-Induced Apoptosis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2012, 18, BR144–BR148. [Google Scholar] [CrossRef]
  172. Qian, L.; Cao, F.; Cui, J.; Wang, Y.; Huang, Y.; Chuai, Y.; Zaho, L.; Jiang, H.; Cai, J. The Potential Cardioprotective Effects of Hydrogen in Irradiated Mice. J. Radiat. Res. 2010, 51, 741–747. [Google Scholar] [CrossRef]
  173. Qian, L.; Cao, F.; Cui, J.; Huang, Y.; Zhou, X.; Liu, S.; Cai, J. Radioprotective Effect of Hydrogen in Cultured Cells and Mice. Free Radic. Res. 2010, 44, 275–282. [Google Scholar] [CrossRef] [PubMed]
  174. Qiu, X.; Dong, K.; Guan, J.; He, J. Hydrogen Attenuates Radiation-Induced Intestinal Damage by Reducing Oxidative Stress and Inflammatory Response. Int. Immunopharmacol. 2020, 84, 106517. [Google Scholar] [CrossRef] [PubMed]
  175. Xiao, H.-W.; Li, Y.; Luo, D.; Dong, J.-L.; Zhou, L.-X.; Zhao, S.-Y.; Zheng, Q.-S.; Wang, H.-C.; Cui, M.; Fan, S.-J. Hydrogen-Water Ameliorates Radiation-Induced Gastrointestinal Toxicity via MyD88’s Effects on the Gut Microbiota. Exp. Mol. Med. 2018, 50, e433. [Google Scholar] [CrossRef]
  176. Zhou, P.; Lin, B.; Wang, P.; Pan, T.; Wang, S.; Chen, W.; Cheng, S.; Liu, S. The Healing Effect of Hydrogen-Rich Water on Acute Radiation-Induced Skin Injury in Rats. J. Radiat. Res. 2019, 60, 17–22. [Google Scholar] [CrossRef]
  177. Hirano, S.; Aoki, Y.; Li, X.-K.; Ichimaru, N.; Takahara, S.; Takefuji, Y. Protective Effects of Hydrogen Gas Inhalation on Radiation-Induced Bone Marrow Damage in Cancer Patients: A Retrospective Observational Study. Med. Gas Res. 2021, 11, 104–109. [Google Scholar] [CrossRef]
  178. Kang, K.-M.; Kang, Y.-N.; Choi, I.-B.; Gu, Y.; Kawamura, T.; Toyoda, Y.; Nakao, A. Effects of Drinking Hydrogen-Rich Water on the Quality of Life of Patients Treated with Radiotherapy for Liver Tumors. Med. Gas Res. 2011, 1, 11. [Google Scholar] [CrossRef]
  179. Alwazeer, D.; Tan, K.; Örs, B. Reducing Atmosphere Packaging as a Novel Alternative Technique for Extending Shelf Life of Fresh Cheese. J. Food Sci. Technol. 2020, 57, 3013–3023. [Google Scholar] [CrossRef] [PubMed]
  180. Zajac, D. Inhalations with Thermal Waters in Respiratory Diseases. J. Ethnopharmacol. 2021, 281, 114505. [Google Scholar] [CrossRef]
  181. Köster, S.; Upadhyay, S.; Chandra, P.; Papavinasasundaram, K.; Yang, G.; Hassan, A.; Grigsby, S.J.; Mittal, E.; Park, H.S.; Jones, V.; et al. Mycobacterium tuberculosis Is Protected from NADPH Oxidase and LC3-Associated Phagocytosis by the LCP Protein CpsA. Proc. Natl. Acad. Sci. USA 2017, 114, E8711–E8720. [Google Scholar] [CrossRef]
  182. Maier, R.J.; Olczak, A.; Maier, S.; Soni, S.; Gunn, J. Respiratory Hydrogen Use by Salmonella enterica Serovar Typhimurium Is Essential for Virulence. Infect. Immun. 2004, 72, 6294–6299. [Google Scholar] [CrossRef]
Figure 1. The most important respiratory disorders with proven activity of molecular hydrogen. Major respiratory disorders with proven therapeutic efficacy of molecular hydrogen (H2), including pulmonary fibrosis (PF), pulmonary hypertension (PH), sepsis, acute lung injury (ALI), and other conditions such as asthma, COPD, or cancer.
Figure 1. The most important respiratory disorders with proven activity of molecular hydrogen. Major respiratory disorders with proven therapeutic efficacy of molecular hydrogen (H2), including pulmonary fibrosis (PF), pulmonary hypertension (PH), sepsis, acute lung injury (ALI), and other conditions such as asthma, COPD, or cancer.
Ijms 26 04116 g001
Figure 2. Molecular hydrogen (H2) may exert cyto-protective effects through direct ROS scavenging, stabilization of mitochondrial activity, upregulation of protective enzymes (SOD, HO-1) and antioxidant pathways, as well as through the downregulation of enzymes generating ROS (NADPHox, iNOS). H2 presents a dual mechanism—direct scavenging and enzymatic regulation (Created in BioRender. Pan, I. (2025) https://BioRender.com/qbvn8pb accessed: 11 April 2025).
Figure 2. Molecular hydrogen (H2) may exert cyto-protective effects through direct ROS scavenging, stabilization of mitochondrial activity, upregulation of protective enzymes (SOD, HO-1) and antioxidant pathways, as well as through the downregulation of enzymes generating ROS (NADPHox, iNOS). H2 presents a dual mechanism—direct scavenging and enzymatic regulation (Created in BioRender. Pan, I. (2025) https://BioRender.com/qbvn8pb accessed: 11 April 2025).
Ijms 26 04116 g002
Figure 3. Anti-inflammatory activity of molecular hydrogen. Molecular hydrogen (H2) attenuates proinflammatory signaling by inhibiting NFκB nuclear translocation, blocking NLRP3 inflammasome activation, and modulating TLR4 signaling, thus limiting the release of inflammatory mediators (TNF-α, IL-6, IL-18, HMGB1). Concurrently, H2 promotes the production of anti-inflammatory cytokines (IL-4, IL-10, IL-13), which polarize M1 macrophages toward an M2-like phenotype. Together, these actions help restore the Th1/Th2 balance. Furthermore, molecular hydrogen mitigates macrophage and neutrophil infiltration into the bronchioles, reducing oxidative and tissue damage, preventing excessive mucus production, and ultimately preserving pulmonary function in inflammatory lung diseases. Notably, H2 achieves these effects without suppressing essential immune defenses, unlike conventional anti-inflammatory therapies (Created in BioRender. Pan, I. (2025) https://BioRender.com/mx9zdvq accessed: 11 April 2025).
Figure 3. Anti-inflammatory activity of molecular hydrogen. Molecular hydrogen (H2) attenuates proinflammatory signaling by inhibiting NFκB nuclear translocation, blocking NLRP3 inflammasome activation, and modulating TLR4 signaling, thus limiting the release of inflammatory mediators (TNF-α, IL-6, IL-18, HMGB1). Concurrently, H2 promotes the production of anti-inflammatory cytokines (IL-4, IL-10, IL-13), which polarize M1 macrophages toward an M2-like phenotype. Together, these actions help restore the Th1/Th2 balance. Furthermore, molecular hydrogen mitigates macrophage and neutrophil infiltration into the bronchioles, reducing oxidative and tissue damage, preventing excessive mucus production, and ultimately preserving pulmonary function in inflammatory lung diseases. Notably, H2 achieves these effects without suppressing essential immune defenses, unlike conventional anti-inflammatory therapies (Created in BioRender. Pan, I. (2025) https://BioRender.com/mx9zdvq accessed: 11 April 2025).
Ijms 26 04116 g003
Figure 4. Methods of administration of molecular hydrogen (*—methods relevant for lung diseases). Methods of molecular hydrogen (H2) administration for therapeutic applications, with inhalation, drinking hydrogen-rich water (HRW), or infusion of hydrogen-enriched saline (HRS) being the most relevant for the treatment of lung diseases. Other approaches include topical applications (gel/patches, baths), the use of probiotics to boost the endogenous H2 production by intestinal microbiota, or administration to the ocular surface.
Figure 4. Methods of administration of molecular hydrogen (*—methods relevant for lung diseases). Methods of molecular hydrogen (H2) administration for therapeutic applications, with inhalation, drinking hydrogen-rich water (HRW), or infusion of hydrogen-enriched saline (HRS) being the most relevant for the treatment of lung diseases. Other approaches include topical applications (gel/patches, baths), the use of probiotics to boost the endogenous H2 production by intestinal microbiota, or administration to the ocular surface.
Ijms 26 04116 g004
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

Zajac, D.; Jampolska, M.; Wojciechowski, P. Molecular Hydrogen in the Treatment of Respiratory Diseases. Int. J. Mol. Sci. 2025, 26, 4116. https://doi.org/10.3390/ijms26094116

AMA Style

Zajac D, Jampolska M, Wojciechowski P. Molecular Hydrogen in the Treatment of Respiratory Diseases. International Journal of Molecular Sciences. 2025; 26(9):4116. https://doi.org/10.3390/ijms26094116

Chicago/Turabian Style

Zajac, Dominika, Monika Jampolska, and Piotr Wojciechowski. 2025. "Molecular Hydrogen in the Treatment of Respiratory Diseases" International Journal of Molecular Sciences 26, no. 9: 4116. https://doi.org/10.3390/ijms26094116

APA Style

Zajac, D., Jampolska, M., & Wojciechowski, P. (2025). Molecular Hydrogen in the Treatment of Respiratory Diseases. International Journal of Molecular Sciences, 26(9), 4116. https://doi.org/10.3390/ijms26094116

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

Article Metrics

Back to TopTop