ArticlePDF Available

Hydrogen gas: from clinical medicine to an emerging ergogenic molecule for sports athletes

Canadian Science Publishing
Canadian Journal of Physiology and Pharmacology
Authors:

Abstract and Figures

H2 has been clinically demonstrated to provide antioxidant and anti-inflammatory effects, which makes it an attractive agent in exercise medicine. Although exercise provides a multiplicity of benefits including decreased risk of disease, it can also have detrimental effects. For example, chronic high-intensity exercise in elite athletes, or sporadic bouts of exercise (i.e., noxious exercise) in untrained individuals, result in similar pathological factors such as inflammation, oxidation, and cellular damage that arise from and result in disease. Paradoxically, exercise-induced pro-inflammatory cytokines and reactive oxygen species largely mediate the benefits of exercise. Ingestion of conventional antioxidants and anti-inflammatories often impairs exercise-induced training adaptations. Disease and noxious forms of exercise promote redox dysregulation and chronic inflammation, changes that are mitigated by H2 administration. Beneficial exercise and H2 administration promote cytoprotective hormesis, mitochondrial biogenesis, ATP production, increased NAD⁺/NADH ratio, cytoprotective phase II enzymes, heat-shock proteins, sirtuins, etc. We review the biomedical effects of exercise and those of H2, and we propose that hydrogen may act as an exercise mimetic and redox adaptogen, potentiate the benefits from beneficial exercise, and reduce the harm from noxious exercise. However, more research is warranted to elucidate the potential ergogenic and therapeutic effects of H2 in exercise medicine.
This content is subject to copyright. Terms and conditions apply.
REVIEW
Hydrogen gas: from clinical medicine to an emerging
ergogenic molecule for sports athletes
1
Tyler W. LeBaron, Ismail Laher, Branislav Kura, and Jan Slezak
Abstract: H
2
has been clinically demonstrated to provide antioxidant and anti-inflammatory effects, which makes it an attrac-
tive agent in exercise medicine. Although exercise provides a multiplicity of benefits including decreased risk of disease, it can
also have detrimental effects. For example, chronic high-intensity exercise in elite athletes, or sporadic bouts of exercise (i.e.,
noxious exercise) in untrained individuals, result in similar pathological factors such as inflammation, oxidation, and cellular
damage that arise from and result in disease. Paradoxically, exercise-induced pro-inflammatory cytokines and reactive oxygen
species largely mediate the benefits of exercise. Ingestion of conventional antioxidants and anti-inflammatories often impairs
exercise-induced training adaptations. Disease and noxious forms of exercise promote redox dysregulation and chronic inflam-
mation, changes that are mitigated by H
2
administration. Beneficial exercise and H
2
administration promote cytoprotective
hormesis, mitochondrial biogenesis, ATP production, increased NAD
+
/NADH ratio, cytoprotective phase II enzymes, heat-shock
proteins, sirtuins, etc. We review the biomedical effects of exercise and those of H
2
, and we propose that hydrogen may act as an
exercise mimetic and redox adaptogen, potentiate the benefits from beneficial exercise, and reduce the harm from noxious exercise.
However, more research is warranted to elucidate the potential ergogenic and therapeutic effects of H
2
in exercise medicine.
Key words: reactive oxygen species, antioxidants, molecular hydrogen, free radicals, exercise, inflammation, redox dysregulation,
anti-inflammatory, hormesis, mitochondria.
Résumé : Des données cliniques ont montré que l’H
2
a des effets antioxydants et anti-inflammatoires, ce qui en fait un agent
attrayant en médecine de l’exercice physique. Bien que l’exercice physique offre une multitude de bienfaits, y compris une
diminution du risque de maladie, il peut aussi avoir des effets délétères. Par exemple, les effets de l’exercice physique de haute
intensité de manière prolongée chez les athlètes d’élite, ou de pointes sporadiques d’exercice physique c’est-à-dire de l’exercice
physique nocif chez des personnes non entraînées, se résument à des facteurs pathologiques similaires comme l’inflammation,
l’oxydation et des dommages cellulaires, lesquels entraînent des maladies ou en sont le résultat. Paradoxalement, les cytokines
pro-inflammatoires et les dérivés réactifs de l’oxygène produits pendant l’exercice physique contribuent largement à la média-
tion des bienfaits de l’exercice physique. En outre, l’ingestion d’antioxydants et d’anti-inflammatoires classiques nuit souvent
aux modes d’action d’adaptation à l’entraînement mis en jeu pendant l’exercice physique. De fait, la maladie et les formes
nocives d’exercice physique favorisent le dérèglement redox et l’inflammation chronique, changements pourtant atténués par
l’administration d’H
2
. Effectivement, l’exercice physique bénéfique et l’administration d’H
2
favorisent l’hormèse cytoprotec-
trice, la biogenèse mitochondriale, la production d’ATP, l’augmentation du ratio NAD
+
/NADH, les enzymes cytoprotecteurs de
phase II, les protéines de choc thermique, les sirtuines, etc. En résumé, nous offrons une synthèse des effets biomédicaux de
l’exercice physique et de ceux de l’H
2
, et nous proposons que l’hydrogène puisse agir en tant que mimétique de l’exercice
physique et d’adaptogène redox, potentialiser les bienfaits de l’exercice physique bénéfique et atténuer les dommages causés par
l’exercice physique nocif. Il serait cependant justifié de procéder à plus de recherches en vue d’élucider les effets ergogènes et
thérapeutiques de l’H
2
en médecine de l’exercice physique. [Traduit par la Rédaction]
Mots-clés : dérivés réactifs de l’oxygène, antioxydants, hydrogène moléculaire, radicaux libres, exercice physique, inflammation,
dérèglement redox, anti-inflammatoire, hormèse, mitochondrie.
Introduction
Hydrogen gas may have potential as an ergogenic and therapeutic
molecule for sports athletes. The biomedical interest in molecular
hydrogen has grown exponentially since the publication of a pa-
per by Ohsawa et al. (2007). They reported that inhalation of only
2%–4% H
2
gas significantly reduced the cerebral infarct volumes in
a rat model of ischemia–reperfusion injury induced by middle
cerebral artery occlusion. The authors further demonstrated that
dissolved hydrogen in the media of cultured cells, at biologically
feasible concentrations, selectively reduced levels of toxic hy-
Received 1 February 2019. Accepted 27 March 2019.
T.W. LeBaron. Molecular Hydrogen Institute, Utah, USA; Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences,
Bratislava, Slovak Republic.
I. Laher. Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, The University of British Columbia, 217 - 2176 Health
Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
B. Kura and J. Slezak.* Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
Corresponding author: Tyler W. LeBaron (email: LeBaronT@molecularhydrogeninstitute.com).
*Jan Slezak served as a Guest Editor; peer review and editorial decisions regarding this manuscript were handled by Tatiana Ravingerova.
1
This paper is part of a Special Issue of selected papers from the 5th European Section Meeting of the International Academy of Cardiovascular Sciences held
in Smolenice, Slovakia, on 23–26 May 2018.
Copyright remains with the author(s) or their institution(s). Permission for reuse (free in most cases) can be obtained from RightsLink.
797
Can. J. Physiol. Pharmacol. 97: 797–807 (2019) dx.doi.org/10.1139/cjpp-2019-0067 Published at www.nrcresearchpress.com/cjpp on 10 April 2019.
droxyl radicals (
OH), but did not decrease other physiologically
important reactive oxygen species (ROS) (e.g., superoxide, nitric
oxide, hydrogen peroxide) (Ohsawa et al. 2007). This is a critical
benefit for athletes because ROS play an active role not only in
injury and overtraining, but also in mediating the benefits of
exercise-induced training adaptations (Merry and Ristow 2016).
Although the research on molecular hydrogen is still in its in-
fancy, with only ≈1200 scientific publications on the subject, these
preclinical and clinical studies suggest that H
2
may have thera-
peutic potential, with no toxicity, in over 170 different human and
animal disease models (Ichihara et al. 2015;LeBaron et al. 2019).
Erratic and prolonged intense exercise may be considered a
potentially useful disease model due to the accompanying oxida-
tive stress, inflammation, muscle damage, and abnormal metab-
olites, which may strengthen the applications for H
2
in exercise
and sports performance. Not only does conventional antioxidant
supplementation fail to prevent these exercise-induced patholog-
ical changes, it may also negate exercise benefits. In contrast, we
hypothesize that, just as H
2
mitigates those pathological factors in
diseases, it may similarly attenuate those same factors when in-
duced by exercise, and may actually promote beneficial exercise
adaptations. Thus, we propose that due to these potential bene-
fits, combined with the current exercise studies on H
2
, hydrogen
is uniquely qualified as an ergogenic and therapeutic molecule for
exercise performance and sports medicine.
Exercise benefits
Exercise provides many benefits, including decreased risk of
diabetes, cardiovascular disease, cancer, and even premature
mortality (Levinger et al. 2015). On the molecular level, exercise
provides benefits by modulating signal transduction and via alter-
ing DNA acetylation and methylation patterns, which in turn
influences gene expression. Consequently, exercise increases en-
dogenous antioxidant levels (e.g., glutathione (GSH), superoxide
dismutase (SOD), catalase (CAT), etc.), neurotropic factors (e.g.,
brain-derived neurotropic factor), circulating adiponectin levels,
enhances the expression of glucose transporter type 4 transport-
ers, induces mitochondrial biogenesis, promotes DNA repair
mechanisms, and provides a host of other physiological benefits
(Tang et al. 2005;Gomez-Pinilla et al. 2011;Gomes et al. 2012).
Although the benefits of exercise are well documented, the exact
underlying molecular mechanisms of how exercise affords these
therapeutic effects have remained unclear. Recent research suggests
that the exercise-induced increase of endogenously produced reac-
tive molecules may be a primary mediator for these beneficial
effects (Gomes et al. 2012). This appears to be at odds with an
earlier proposed model of aging and disease known as the “Free
Radical Theory of Aging”. This theory of aging was proposed in the
1950s by Denham Harman, and was largely based on the strong
correlations between oxidative stress and diseases (Harman 2009).
Indeed, some have suggested that oxidative stress plays a caus-
ative role in the pathogenesis of virtually every disease (Liochev
2013).
Oxidative stress
Oxidative stress exists when the number of oxidants and reac-
tive molecules overcomes the body’s endogenous enzymatic and
nonenzymatic antioxidant self-defense systems (Bentley et al.
2015). Reactive molecules include ROS and reactive nitrogen spe-
cies. ROS is a category that includes radicals and nonradical chem-
ical species. Free radicals are chemical species that contain an
unpaired electron; for example, hydroxyl radicals (
OH), superox-
ide anion radicals (
O
2
), alkoxyl radicals (R-O
), peroxyl radicals
(R-OO
), carbon radicals (R-C
), and various reactive nitrogen spe-
cies (e.g., nitric oxide, NO
), and thiyl radicals (RS
). Nonradical
reactive molecules include hydrogen peroxide (H
2
O
2
), singlet
oxygen (
1
O
2
), and various reactive nitrogen species (e.g., peroxyni-
trite (ONOO
), nitrogen dioxide (NO
2
), dinitrogen trioxide (N
2
O
3
))
(Palmieri and Sblendorio 2006;Jones 2008). Most ROS are pro-
duced in the electron transport chain of the mitochondria, pri-
marily at complex 1, and not 3, as was commonly believed (Liu
et al. 2002). Other sites of ROS generation include NADPH oxidase,
nitric oxide synthase, xanthine oxidase, cytochrome p450, alde-
hyde oxidase, and heme proteins, etc. (Halliwell and Gutteridge
2015). The increased metabolism and the exercise-induced activa-
tion of these systems result in a significant increase in ROS pro-
duction.
ROS regulation
Importantly, these reactive molecules can either have noxious
or beneficial effects, depending on their (i) identity, (ii) concentra-
tion, (iii) location, and (iv) duration. For example, under normal
metabolic conditions, superoxide radicals are constantly being
formed by single-electron reduction of molecular oxygen in the
electron transport chain of the mitochondria, in nuclear and
plasma membranes via NADPH oxidases, in the endoplasmic re-
ticulum for protein folding, and in macrophages (Finkel 2011).
Superoxide production is increased during immune responses, as
it is essential for killing pathogens, and increasing levels of in-
flammatory cytokines, and for the formation of NOD-like receptor
prin domain-containing 3 inflammasome (Finkel 2011). Similarly,
superoxide formation is increased during bouts of exercise due to
the increased oxygen intake. Exercise-induced superoxide acts as an
important signalling molecule that activates various transcription
factors, resulting in improved exercise capacity (Gomes et al. 2012).
The concentration of superoxide is regulated within a narrow
range by controlling its production and its clearance (Halliwell
and Gutteridge 2015). Superoxide can be dismutated by the body’s
antioxidant enzyme SOD to form H
2
O
2
. The produced H
2
O
2
,as
another important signalling molecule, can further regulate gene
expression and induce favourable cellular changes before being
converted to water and O
2
by CAT (Jones 2008). If the concentra-
tion of superoxide or H
2
O
2
exceeds regulatory systems, then oxi-
dative stress occurs. The higher levels may also result in an
increased production of toxic hydroxyl radicals (
OH) via the Haber–
Weiss and Fenton reactions (eqs. 1 and 2). Ferrous iron and super-
oxide can then be regenerated (eq. 3) and participate in eqs. 1 and 2,
respectively, which further propagates the production of hy-
droxyl radicals (Halliwell and Gutteridge 2015).
(1) Fe3O2¡Fe2O2
(2) Fe2H2O2¡Fe3OHOH
(3) Fe3H2O2¡Fe2O22H
Due to their high reactivity, hydroxyl radicals (
OH) react with essen-
tially any biomolecule at a diffusion-controlled rate (≈10
10
·M
−1
·s
−1
),
which makes them so damaging. Therefore, the regulation of the
signalling molecules superoxide and hydrogen peroxide is essen-
tial to reduce the production of hydroxyl radicals.
The nitric oxide radical (NO
) is also an essential signalling mol-
ecule, and is needed for endothelial function (e.g., vasodilation),
gene expression, and cell proliferation (Nathan and Xie 1994), and
is produced near its targets (Clements et al. 2014). However, simi-
lar to superoxide, when its concentration is too high, such as from
hyperactivation of inducible nitric oxide synthase, then excess NO
can combine nearly instantly with superoxide to form pernicious
peroxynitrite molecules (Brown and Neher 2010). Peroxynitrite is
a stronger oxidant than either nitric oxide or superoxide, and
besides being able to oxidize proteins, lipids, and DNA, is also a
non-Fenton source of toxic hydroxyl radicals (Pacher et al. 2007).
These 2 oxidants (i.e., ONOO
and
OH) have no known physiolog-
798 Can. J. Physiol. Pharmacol. Vol. 97, 2019
Published by NRC Research Press
ical benefit, and easily oxidize cellular biomolecules leading to
disease, injury, and impaired athletic performance.
Disproportionate levels of ROS generation either from disease
or from vigorous exercise can rapidly deplete the body’s antioxi-
dant system (e.g., SOD, GSH, vitamin C, etc.) leading to oxidative
stress and its subsequent toxic consequences (Finkel 2011). The
location of ROS formation is also critical in determining whether
they will have a beneficial or a noxious effect. Deliberate super-
oxide production via NO
x
systems or within the mitochondria
often occurs near redox-sensitive transcription factors and phos-
phatases (D’Autréaux and Toledano 2007). For example, there are
specifically located aquaporins through which the produced H
2
O
2
can transverse, permitting interaction with various biomolecules
(e.g., phosphatases for growth factor signalling (Juarez et al. 2008))
before being converted to water and oxygen by glutathione per-
oxidase or CAT (Finkel 2011). The colocalization of the oxidant-
producing system with the intended target affords specificity of
the oxidant, thus ensuring a higher probability for beneficial ef-
fects (D’Autréaux and Toledano 2007;Dickinson and Chang 2011).
Redox dysregulation
Reactive molecules have both deleterious disease-causing ef-
fects, and essential and therapeutic disease-preventing effects
(Levinger et al. 2015). This paradoxical nature of oxidant signalling
may be considered a form of hormesis, in which low doses of
potentially harmful stress result in cytoprotective cellular adap-
tations. Thus, the 2 opposing effects of oxidants on health are not
simply paradoxical, but are homeostatically interconnected and
interdependent. Redox homeostasis is required for normal cellu-
lar and bodily function. The key issue is not the presence or ab-
sence of oxidants and antioxidants in inducing detrimental
consequences, but the harmful disturbance to the delicate ho-
meostasis between oxidation and reduction (Gomes et al. 2012).
This disturbance or dyshomeostasis is referred to as redox dys-
regulation.
Paradoxically, it is possible to have too much and too little
oxidation at the same time within the same cell. For example,
aging is associated with excessive oxidation in the cytosol, but a
loss of oxidizing potential in the endoplasmic reticulum, which is
essential for the proper folding of proteins (Feleciano and Kirstein
2016). In contrast to focusing exclusively on free radicals as being
responsible for aging and disease, redox dysregulation appears to
be a more accurate proposition.
ROS-mediated exercise benefits
Exercise naturally elicits the hormesis effect due to the tran-
sient spikes in ROS production, and thus improves optimal redox
homeostasis and mitigates against redox dysregulation (Ristow
and Zarse 2010). This helps explain why many of the benefits of
exercise are thought to be mediated by exercise-induced ROS pro-
duction, which activates a plethora of transcription factors and
modulates various signal transduction pathways (Finkel 2011). Ex-
ercise greatly increases oxygen demand and thus also subsequent
ROS production. It has been estimated that 0.2%–2% of the O
2
we
breathe is converted into various ROS (Balaban et al. 2005). ROS is
also produced under other types of hypoxia–ischemia type condi-
tions and in sarcomeres of exercising muscles (Powers et al. 2011).
Parabolic nature of exercise
Similar to disease remediation, wound healing, and injury re-
covery, the relationship between exercise-induced ROS benefits
and exercise-induced ROS damage follows a parabolic type func-
tion as shown in Fig. 1. An increasing level of ROS facilitates injury
healing and beneficial exercise adaptations, but excessive ROS has
the opposite effect. This cytotoxic effect can occur with prolonged
high-intensity exercise (Fisher-Wellman and Bloomer 2009;Merry
and Ristow 2016). Elite athletes frequently exercise chronically at
these high intensities, which consequently increases ROS produc-
tion leading to cytotoxic injuries and a decrease in endogenous
antioxidant status (Fisher-Wellman and Bloomer 2009). The
chronic assault of ROS can result in potentially detrimental cellu-
lar adaptations for the athlete. For example, during exercise,
mediation of vasodilation switches from NO
to mitochondrial-
derived H
2
O
2
(Durand and Gutterman 2014). Excessive ROS pro-
duction and subsequent cellular damage also occurs in the
elderly (due to loss of endogenous antioxidant levels) (Evans
2000), and in “weekend warriors”, where their body has not made
necessary adaptations to cope with the stresses of erratic and (or)
overly intense exercise bouts (Davies et al. 1982;Evans 2000;Urso
and Clarkson 2003).
Antioxidant supplementation
Exercise-induced excessive ROS production provides a rationale
for the ingestion of exogenous antioxidant supplements by many
athletes. However, several clinical trials on antioxidant supple-
mentation report that their use is not only ineffective, but can, in
fact, be deleterious. Several studies suggest that antioxidants can
negate training benefits, such as vascularization and mitochon-
drial biogenesis (Merry and Ristow 2016). Studies of human ath-
letes are equivocal, showing that antioxidant supplementation
can have ergogenic, ergolytic, or neutral effects on exercise per-
formance (Fisher-Wellman and Bloomer 2009). However, the re-
sults in those studies showing potential ergogenic effects of
antioxidant supplementation (Lafay et al. 2009;Braakhuis et al.
2013) are obfuscated by their potential nonradical scavenging ac-
tions (e.g., effectors of signal transduction). For example, the poly-
phenolic compounds in tea, grapes, and other plants often have
pharmacological effects in cells. They do not act simply as radical
scavengers, but exist in the cell as important sensors and effectors
of key redox-regulated pathways (Finkel 2011).
Several well-designed human studies show that daily antioxidant
supplementation (e.g., alpha tocopherol) can impair exercise perfor-
mance (Sharman et al. 1971;Olesen et al. 2014). An 8-week study
found that vitamin C blunted the exercise-induced increase in max-
imal oxygen consumption (V
˙O
2
max) by 50% (Gomez-Cabrera et al.
2008). Another study showed V
˙O
2
max improved only in the placebo
group (Morrison et al. 2015), and biochemical analysis of muscle
biopsy revealed that the exercise-induced upregulation in GSH
and PGC-1, a marker of mitochondrial biogenesis, was abolished
by antioxidant ingestion (Morrison et al. 2015). Similarly, an 11-
Fig. 1. Hypothetical relation between increasing levels of reactive
oxygen species (ROS) production and various health benefits.
Horizontal axis follows ROS production from sedentary to elite
athletes and (or) various disease conditions (e.g., ischemia–reperfusion).
LeBaron et al. 799
Published by NRC Research Press
week study with 54 subjects found that compared with placebo,
ingestion of 1000 mg of vitamin C and 235 mg of vitamin E during
training blunted the endurance-training-induced increases of im-
portant biomolecules, such as cytochrome c oxidase (COX4), per-
oxisome proliferator-activated receptor gamma coactivator 1-alpha
(PGC-1), cell division control protein 42, and mitogen-activated
protein kinase 1 (Paulsen et al. 2014). These data are in agreement
with another study showing that several antioxidant enzymes (i.e.,
SOD1, SOD2, glutathione peroxidase (GPx1), and CAT), and mitochon-
drial biogenesis (i.e., PGC-1), were only increased in untreated athletes,
i.e., in the absence of antioxidant supplementation (Ristow et al. 2009).
Similar negative effects have been reported by others (Wray et al. 2009;
Gliemann et al. 2013,2014).
Inflammation and injury
Chronic exercise also increases levels of inflammation by up-
regulating various pro-inflammatory mediators (Pedersen et al.
2007). Many of these increases are also associated with disease
conditions in which excessive levels of inflammation further dam-
age tissue and delay healing (Hotamisligil 2006). Similar to the
effects of acute and chronic physical activity on oxidative stress,
regular exercise reduces systemic inflammation, whereas chronic
high-intensity exercise training increases inflammation (Gleeson
2007). Athletes engaged in chronic and prolonged exercise train-
ing at high intensities can exhibit exercise-induced immunode-
pression, thus also increasing their risk for infection (Gleeson
et al. 2004).
The biphasic relationship between the beneficial and noxious
effects of myokines (cytokines secreted from skeletal muscle) fol-
lows a similar paradoxical pattern, as does exercise-induced ROS
generation (Handschin and Spiegelman 2008) (see Fig. 1). For ex-
ample, the myokines interleukin (IL)-6, IL-1, IL-8, IL-15, etc., se-
creted from exercising muscle fibers mediate some of the benefits
of exercise (Pedersen et al. 2007). These myokines not only medi-
ate the benefits (e.g., increased expression of p38-AMPK, ERK1/2,
PGC-1) to the exercising muscles themselves, but they also pro-
vide systemic beneficial effects on nonskeletal muscle tissue (e.g.,
improved glucose homeostasis) (Handschin and Spiegelman 2008).
However, prolonged elevation of these pro-inflammatory mediators
(i.e., cytokines, tumor necrosis factor-, chemokines, etc.) increases
the risk of diseases such as cancer, diabetes, neurodegeneration,
sarcopenia, and others (Hotamisligil 2006), as well as injury and im-
paired athletic performance (Handschin and Spiegelman 2008).
Chronic ingestion of anti-inflammatory agents is prevalent
among both those with debilitating diseases, and with athletes
trying to train or compete with various injuries. Estimated daily
use of nonsteroidal anti-inflammatories in the general population
is around 4%, and in elite athletes as high as 35% (Paoloni et al.
2009). However, in addition to the potential toxic side effects of
prolonged use of steroidal and nonsteroidal anti-inflammatory
agents, there is also evidence suggesting that their chronic use
may also blunt exercise training benefits similar to antioxidants
(Schoenfeld 2012;Urso 2013).
Effects of molecular hydrogen
In contrast to these conventional antioxidants and anti-
inflammatories, the physicochemical properties and biomedical
studies of molecular hydrogen suggest that H
2
may be useful in
mitigating the effects of excessive ROS and inflammation (Ohta
2015;Slezák et al. 2016;Nogueira et al. 2018), without abolishing
the desired exercise training adaptations and benefits. This is be-
cause H
2
is a stable molecule incapable of reacting with important
reactive signalling molecules under biological conditions without
a catalyst. It hasbeen proposed that, although H
2
cannot neutralize
important ROS, it can selectively scavenge cytotoxic hydroxyl rad-
icals, and to a lesser extent peroxynitrite (Ohsawa et al. 2007),
which is exactly what is most needed for athletes. Although the
2
nd
-order reaction rate constant between hydroxyl radicals and H
2
(4.2 × 10
7
·M
−1
·s
−1
) is about 3 orders of magnitude lower than that
between other more abundant nucleophilic cellular components,
it is clear that H
2
reduces the markers (e.g., 3=-p-(aminophenyl)
fluorescein, 3=-p-(hydroxyphenyl) fluorescein) for these oxidants
(Ohta 2015). Molecular hydrogen has become an attractive agent
in the biomedical field by acting as a gaseous-signalling modula-
tor that effectively decreases oxidative stress and inflammation
(Dixon et al. 2013). Molecular hydrogen has been shown to have
therapeutic potential in over 170 different animal and human
disease models, and in essentially every organ of the human body.
Several animal studies have shown that H
2
is effective at increas-
ing resilience and mitigating the negative effects of acute and
chronic stress such as inflammation, elevated ROS, and anxiety
and depressive-like behaviours (Nagata et al. 2009;Slezak et al.
2015;Zhang et al. 2016;Gao et al. 2017).
Erratic or prolonged intense exercise may be considered a
model for disease due to the many parallels and common patho-
logical factors (i.e., ROS, inflammation, metabolic changes, etc.)
between them. For example, in a rat study comparing early aerobic
exercise with and without H
2
administration as preconditioning to
protect against myocardial injury induced by acute myocardial
infarction, it was demonstrated that H
2
was as effective as, and
sometimes more effective than, early aerobic exercise, and the
greatest benefit was often the combination (e.g., infarct size,
troponin 1, SOD, CAT, GSH, total antioxidant status, malondialde-
hyde (MDA), creatine kinase, and mitochondrial protein translo-
case of outer membrane 20, translocase of inner membrane 23)
(Feng et al. 2018). Therefore, H
2
may be beneficial for elite and
nonelite athletes because (i) administration of H
2
elicits many of
the same benefits provided by exercise, thus acting as an “exercise
mimetic”, and (ii) just as H
2
restores redox homeostasis, prevents
pathological changes, and mitigates excessive inflammation aris-
ing from and induced by disease, it may similarly have those
protective and therapeutic effects against noxious forms of exer-
cise (Ostojic 2014;Nogueira et al. 2018). Table 1 summarizes some
of the biomolecular changes present in disease, beneficial or nox-
ious forms of exercise, and by H
2
administration. The changes in
disease conditions are pathological in nature, and do not repre-
sent any specific disease, but represent common findings of dis-
eases in general (Hotamisligil 2006). The reader is referred to
several reviews on diseases for more details on these detrimental
changes (Hotamisligil 2006;Gleeson 2007;Pedersen et al. 2007).
For exercise, the changes are either negative or positive, which
would depend on the frequency, intensity, duration, and recov-
ery. For example, it is not uncommon for athletes to push them-
selves to extreme levels of chronic prolonged intense training, or
for even untrained individuals to complete a marathon or other
physically demanding events (e.g., weekend basketball). These
chronic or erratic intense activities can directly damage muscle
tissue, resulting in cell death and decreased mitochondrial func-
tion or number, and (or) other pathological changes. However,
regular optimal exercise induces beneficial changes to these bio-
markers (i.e., increases or decreases) compared with the biomark-
ers of sedentary individuals, and thus provides protection against
their pathological changes in maintaining and improving optimal
homeostatic levels (Gleeson et al. 2004).
The arrows in Table 1 that represent the changes by H
2
admin-
istration indicate the respective attenuation of the pathologically
induced changes compared with control. These changes of H
2
administration were gathered from data involving humans and
animals (Dixon et al. 2013;LeBaron et al. 2019;Nicolson et al. 2016),
and are not directly quantified or compared with respect to each
other or exercise. Thus, caution should be used when interpreting
the table. However, the improved homeostasis and cellular viabil-
ity from H
2
administration has been reported in different animal
species (e.g., rodent, dog, horse, pig, etc.) and in many different
800 Can. J. Physiol. Pharmacol. Vol. 97, 2019
Published by NRC Research Press
animal disease models, as well as in clinical human studies (Zhai
et al. 2014;Ichihara et al. 2015).
H
2
acts as a hormetic effector to improve redox
status
Intriguingly, hydrogen does not always reduce markers of oxi-
dation, but it appears that it only reduces excessive levels. The fact
that H
2
does not scavenge ROS, and only decreases excessive levels
of ROS, makes it a safe and effective medical gas for use in clinical
management and also to preserve the benefits and reduce the
harm of sporadic or chronic intense exercise. Not only does H
2
reduce excessive ROS production, but it may also have beneficial
mild pro-oxidant properties, by exerting hormetic benefits simi-
lar to those produced by exercise. Some studies show that the
therapeutic and neuroprotective benefits of H
2
are also correlated
with slightly increased levels of MDA, a marker of lipid peroxida-
tion (Eckermann et al. 2011), even in the sham group (Matchett
et al. 2009). We reported similar changes in rats, where H
2
pre-
vented irradiation-induced increases in tumor necrosis factor-
and MDA. But when H
2
was administered alone, tumor necrosis
factor-initially increased above control, and then decreased and
remained below both the irradiated group and the nonirradiated
control, whereas MDA tended to initially decrease then increase
(Kura et al. 2019); thus, demonstrating that, although H
2
primarily
reduces MDA, sometimes its therapeutic effects are associated
with transiently increased levels of MDA. Similarly, some of the
benefits of H
2
in plants are also mediated by increases in ROS
production (Xie et al. 2014). Furthermore, pretreatment of
SH-SY5Y cells with H
2
protected them from subsequent oxidative
stress induced by H
2
O
2
(Murakami et al. 2017). In this case, H
2
acted as a mitohormetic effector by transiently increasing mito-
chondrial superoxide production, followed by an upregulation of
Nrf2 transcription and induction of other cytoprotective phase II
proteins (Murakami et al. 2017;Kura et al. 2018). We recently
demonstrated that short-term inhalation of H
2
mildly increased
urinary 8-hydroxy-2=-deoxyguanine (8-OHdG) in patients with
Parkinson’s disease by 16% (p= 0.02) (Hirayama et al. 2018). This is
significantly less than the several hundred percent increase seen
in various diseases, and comparable with a mild bout of exercise
training (Hirayama et al. 2018). In contrast, a 4-week open label
cross-over study in patients with rheumatoid arthritis, found that
ingestion of hydrogen-rich water (HRW) significantly decreased
8-OHdG by 14.3% (p< 0.01), and remained below baseline for an
additional 4 weeks during the washout period (Ishibashi et al.
2012). This illustrates the emerging pattern that H
2
has dual ef-
fects depending on the situation. In addition to the ROS-induced
hormesis, H
2
has paradoxically been reported to provide thera-
peutic effects via transiently activating the NF-B/Bcl-xL pathway
in the early phase (Zhuang et al. 2013), which may also be a form
of hormesis (Hirayama et al. 2018). Lastly, it has been reported that
H
2
can induce heat-shock response (Nishiwaki et al. 2018) and the
mitochondrial unfolded protein response (Sobue et al. 2017).
These may also be considered forms of hormesis that are also
induced by exercise.
These 2 opposing effects of hydrogen (e.g., increased and de-
creased ROS production) are not mutually exclusive. Depending
on the challenge, timing, and need, hydrogen treatment appears
to perform either function to help maintain optimal redox ho-
meostasis. Hydrogen seems to act as a redox adaptogen at main-
taining redox homeostasis either by acting hormetically and (or)
via modulating redox-sensitive processes.
Methods of H
2
administration
There are several methods for hydrogen gas administration in-
cluding inhalation of H
2
gas (Hayashida et al. 2008), tube feeding
of H
2
-rich solution (Li et al. 2013), intravenous injection of H
2
-rich
saline (Cui et al. 2014), H
2
-rich dialysis solution for hemodialysis
(Nakayama et al. 2010), hyperbaric H
2
chamber (Dole et al. 1975),
bathing in H
2
-rich water (Kato et al. 2012), increasing H
2
produc-
tion by intestinal bacteria (Chen et al. 2013), topical application
(Ostojic et al. 2014), oral ingestion of hydrogen-producing tablets
(LeBaron et al. 2019;Ostojic et al. 2018), and simply drinking HRW
(Nakao et al. 2010). Regardless of the mode of administration, the
cellular bioavailability of molecular hydrogen is extremely high
due to its unique physicochemical properties. Its small size, low
mass, neutral charge and nonpolar nature, coupled with its high
rate of diffusion, allow it to easily penetrate cellular biomem-
branes and diffuse into the mitochondria and nucleus (Nicolson
et al. 2016;Ohta 2015).
Hydrogen can be dissolved in water up to 0.8 mM (1.6 mg·L
−1
)at
standard ambient temperature and pressure. A concentration of
1.6 mg·L
−1
may not seem significant, but because H
2
is the lightest
and smallest molecule, it should be compared using moles rather
than mass. Ingestion of1LofH
2
-saturated water provides more
Table 1. Biological markers in disease, by exercise (negative and positive), and by H
2
treatment.
Biological markers Disease
Exercise
H
2
Negative Positive
Antioxidant status: BAP, SOD, GSH, CAT, GPx, GST, Nrf2, HO-1 ↓↓ ↑ ↑
Vascular function: eNOS, DDAH2 ↓↓ ↑ ↑
Brain effects: CREB, BDNF ↓↓ ↑ ↑
Mitochondrial function: ATP, membrane potential, complexes 1–5, PGC1-↓↓ ↑ ↑
Miscellaneous effects: GLUT4, TST, SIRT3, AMPK, NAD
+
/NADH ↓↓ ↑ ↑
Inflammatory response: TNF-, ILs1–20, NFATC1, COX-2, NF-B, NLRP3 ↑↑ ↓ ↓
Oxidative stress: MDA, d-ROM, CRP, 8OHdG, 4HNE, TBARS, NTY ↑↑ ↓ ↓
Tissue damage: Caspase-3,-8,-9,-12, AST, ALT, BUN, Cr, LDH ↑↑ ↓ ↓
Vascular: nNOS, iNOS ↑↑ ↓ ↓
Note: When interpreting the above data, note that the effects of H
2
treatment were gathered from data involving humans and
animals and are not directly quantified or compared with respect to each other or exercise. BAP, biological antioxidant potential; SOD,
superoxide dismutase; GSH, glutathione; CAT, catalase; GPx, glutathione peroxidase; GST, glutathione-S-transferase; Nrf2, nuclear
factor erythroid 2–related factor 2; HO-1, heme oxygenase-1; eNOS, endothelial nitric oxide synthase; DDAH2, dimethylarginine
dimethylaminohydrolase; CREB, cAMP response element binding protein; BDNF, brain-derived neurotropic factor; PGC1-, peroxi-
some proliferator-activated receptor-coactivator-1; GLUT4, glucose transporter type 4; TST, testosterone; SIRT3, sirtuin 3; AMPK,
AMP-activated protein kinase; TNF-, tumor necrosis factor-; ILs1–20, interleukins 1–20; NFATC1, Nuclear Factor Of Activated T-Cells
1; COX-2, cyclooxygenase-2; NF-B, nuclear factor kappa light chain enhancer of activated B cells; NLRP3, NOD-like receptor prin
domain-containing 3; MDA, malondialdehyde; d-ROM, diacron reactive oxygen metabolite; CRP, C-reactive protein; 8OHdG,
8-hydroxy-2=-deoxyguanine; 4HNE, 4-hydroxynonenal; TBARS, thiobarbituric acid reactive substances; NTY, nitrotyrosine; AST, aspar-
tate aminotransferase; ALT, alanine aminotransferase; BUN, blood urea nitrogen; Cr, creatinine; LDH, lactate dehydrogenase; nNOS,
neural nitric oxide synthase; iNOS, inducible nitric oxide synthase.
LeBaron et al. 801
Published by NRC Research Press
“therapeutic moles” than would ingestion of a 100 mg dose of
vitamin C (0.79 millimoles H
2
vs. 0.57 millimoles vitamin C). Al-
though, the amount of H
2
ingested from inhalation of H
2
gas can
be many times higher than from ingestion of HRW, drinking
HRW is often as effective as, and in some cases more effective
than, inhalation (Ito et al. 2012;Ohno et al. 2012). This is likely
attributed to hydrogen’s activity as a signal modulator (Ichihara
et al. 2015). HRW can be prepared by bubbling H
2
gas into water
under pressure, electrolysis of water (2H
2
O¡2H
2
+O
2
), and also
by reaction with metallic magnesium (Mg + 2H
2
O¡H
2
+ Mg(OH)
2
)
or other metals. Several products from ready-to-drink beverages
in aluminum pouches and cans and electrolytic devices to H
2
-
producing tablets and inhalation machines, are readily available
to consumers. However, not all products may produce or contain
concentrations of H
2
equivalent to or with the stability of those
used in human studies.
Human studies with H
2
administration
There are a limited number of clinical human studies to con-
firm the promising effects observed in laboratory animals. How-
ever, the approximately 60 studies in humans published so far
strengthen hydrogen’s potential as a feasible therapeutic agent.
These clinical studies have demonstrated beneficial effects in a
wide range of diseases including metabolic syndrome (Nakao et al.
2010), diabetes (Kajiyama et al. 2008), hyperlipidemia (Song et al.
2013), Parkinson’s disease (Yoritaka et al. 2013), cognitive impair-
ments (Nishimaki et al. 2018), rheumatoid arthritis (Ishibashi et al.
2012), chronic hepatitis B (Xia et al. 2013), vascular function (Sakai
et al. 2014), exercise performance (Ostojic´ et al. 2011), and others
(Ichihara et al. 2015). Currently, inhalation of hydrogen gas is
being clinically investigated in post-cardiac arrest patients in a
large 360-patient multicentered study with promising prelimi-
nary results (Ono et al. 2017). Animal studies suggest that H
2
inha-
lation may be more effective than conventional hypothermia at
mitigating the ischemia–reperfusion injury following cardiac re-
suscitation (Katsumata et al. 2017).
Table 2 summarizes the known studies on hydrogen admin-
istration and exercise in humans and animals, illustrating
hydrogen’s potential benefit in exercise medicine and sports per-
formance. In a double-blinded placebo-controlled cross-over trial
of 10 elite soccer players (20.9 ± 1.3 years), subjects ingested 1.5 L of
HRW (≈1 mM) prior to exercise (Aoki et al. 2012). Athletes under-
took ergometer cycling at 75% of V
˙O
2
max for 30 min, followed by
performing 100 repetitions of maximal isokinetic knee extensions
to evaluate peak torque and muscle activity as indicators of fa-
tigue. Peak torque was significantly decreased by 20%–25% after
40–60 contractions in the placebo group, but was not statistically
reduced in the HRW group, suggesting that H
2
attenuated the
exercise-induced decline of muscle function. Additionally, HRW
reduced the increase in blood lactate levels that occurred post-
cycling (reduction of 1 mM, p< 0.05). Lactate production, although
not directly responsible for acidosis or fatigue, is correlated with
both events as it represents a shift from ATP derived from aerobic
respiration (in the mitochondrial electron transport chain) to
substrate-level phosphorylation (in glycolytic anaerobic metabo-
lism). The ability of the athletes to maintain a similar exercise
output with reduced lactate levels may suggest improved mito-
chondrial function, increased rates of NADH/NAD
+
recycling, or
lactate metabolism. The decreased lactate levels were also re-
ported in a clinical study in patients with mitochondrial myopa-
thies (Ito et al. 2011). Of interest is that the slightly elevated levels
of oxidative markers post-exercise in the elite soccer players were
not attenuated, and were even mildly elevated by HRW (no signif-
icant changes in diacron reactive oxygen metabolites (d-ROMs) or
biological antioxidant potential (BAP)), suggesting that, unlike the
case of conventional antioxidant use, hydrogen should not abol-
ish, put perhaps increase, beneficial training adaptations. The
authors concluded that replacement of regular water with HRW
as a hydration strategy may prevent the adverse effects associated
with heavy exercise (Aoki et al. 2012).
In another double-blinded randomized cross-over design,
18 athletes consumed 1 L·day
−1
of HRW; there were decreased maxi-
mal rates of perceived exertion and reduced lactate production at
the critical running speed (8.1 miles·hour
−1
; 1 mile = 1.609 344 km)
during maximal exercise, without increases in serum antioxidant
capacity (Ostojic´ et al. 2011;Ostojic 2014). Another double-blinded
placebo-controlled cross-over study with 9 subjects reported that
bathing in HRW for 20 min significantly decreased delayed onset
muscle soreness after downhill running (p< 0.033), without sig-
nificantly reducing the markers of oxidation (i.e., MDA, d-ROMs)
(Kawamura et al. 2016).
The lack of difference in oxidative markers were also seen in a
study of 5 thoroughbred horses undergoing maximum levels of
treadmill exercises to exhaustion (Tsubone et al. 2013). Compared
with the placebo group, the pre-exercise oxygen metabolites
(d-ROMs) tended to be lower in the HRW group, but then signifi-
cantly increased immediately post-exercise (p< 0.001 vs. p< 0.05).
The BAP increased similarly in both groups. However, after
30 min, the BAP/d-ROMs ratio was still elevated in the placebo
group (p< 0.05), but normalized in the HRW group (Tsubone et al.
2013). This earlier recovery of the BAP/d-ROMs ratio in the HRW
group suggests less radical damage and faster recovery after an
intense exercise session. However, a similar follow-up study with
13 thoroughbred horses (Yamazaki et al. 2015) reported that, al-
though H
2
similarly did not reduce d-ROMs, it did significantly
reduce 8-OHdG immediately after, and 1, 3, and 24 h post-exercise
(p< 0.01, p= 0.0196, p< 0.01, and p< 0.01, respectively). This
marker, 8-OHdG, was significantly increased above baseline post-
exercise, and remained elevated throughout the 24 h in the pla-
cebo group; whereas in the H
2
group, it did not significantly
change from its baseline level (Yamazaki et al. 2015).
It is important to note that these studies used trained athletes
(or horses), whose bodies and cells may have already adapted to
combat the exercise-induced ROS production. Several double-
blinded human studies involving untrained subjects show that H
2
supplementation increased antioxidant enzymes (e.g., GSH, SOD,
etc.) and decreased markers of oxidation. For example, ingestion
of HRW for 4 weeks in 16 young healthy men resulted in a 25%
increase in GSH (p< 0.003) and a 11% increase in SOD (p< 0.007)
along with an accompanying decrease in MDA levels compared
with placebo (−25.8% vs. 11.7%; p< 0.001) (Trivic et al. 2017). How-
ever, in another double-blinded, placebo-controlled, cross-over
study with 26 healthy subjects (13 females, 13 males; mean age
34.4 ± 9.9), ingestion of HRW for 4 weeks did improve mood,
reduce anxiety, and decrease sympathetic nerve activation, but it
did not significantly reduce levels of oxidative stress. However,
the levels were all within a normal healthy range (Mizuno et al.
2017). In contrast, in an 8-week study of patients with metabolic
syndrome, ingestion of HRW caused a 39% increase in SOD
(p< 0.05) and a 43% decrease in thiobarbituric acid reactive sub-
stances (Nakao et al. 2010). Comparable increases in endogenous an-
tioxidants from regular exercise have been reported in some studies
(Ortenblad et al. 1997), while other studies show no increased antioxi-
dant levels from exercise training (Tiidus et al. 1996).
H
2
may also be helpful in improving the rate of recovery in soft
tissue injuries. In a study of 36 professional athletes, hydrogen
treatment was effective in sports-related soft tissue injury by increas-
ing the rate of return-to-normal range of motion for the injured limb
(Ostojic et al. 2014). Additional randomized, placebo-controlled,
cross-over studies suggest that H
2
decreases the rate of perceived
exertion and lowers heart rate during submaximal exercise in young
healthy adults (n= 19) (LeBaron et al. 2019), as well as improves
V
˙O
2
max in mid-age overweight women (n= 12) (Ostojic et al. 2018),
and reduces exercise-induced psychometric fatigue (n= 159) (Mikami
et al. 2019). More research on the acute and chronic effects of molec-
802 Can. J. Physiol. Pharmacol. Vol. 97, 2019
Published by NRC Research Press
ular hydrogen administration and its varying methods of delivery in
exercise medicine and sports performance is needed to determine its
true efficacy, and from which types of exercises and in which popu-
lations the most benefit would occur.
Safety
One important advantage of hydrogen is its lack of toxicity,
giving it a high safety profile. Hydrogen has been used since the
1940s to prevent decompression sickness in deep-sea diving (Case
and Haldane 1941;Dougherty 1976). No toxic effects were observed
even at 98.87% H
2
and 1.13% O
2
at 19.1 atm (Friess et al. 1978).
Additionally, hydrogen gas is naturally found in our blood and
breath due to normal fermentation of nondigestible carbohy-
drates from intestinal bacteria (Strocchi and Levitt 1992). This
bacterially produced hydrogen gas has also been shown to be
therapeutic (Kajiya et al. 2009). Even though the amount of gas is
often more than what is ingested from drinking HRW, ingestion
of HRW is still effective, and at least in some cases more effective
(Ito et al. 2012). Perhaps hydrogen is mildly toxic as suggested by
its hormetic actions. Thus, the toxic effects are potent enough to
induce hormesis, yet mild enough to be obscured and essentially
be converted to beneficial effects. This hypothesis would also ex-
plain why constant exposure to molecular hydrogen provides no
biological effects (Ito et al. 2012).
Summary of hydrogen’s potential benefits
We suggest that hydrogen can benefit athletic performance
because it can (i) rapidly reach subcellular compartments via
Table 2. Selected studies on hydrogen administration on sports exercise.
Reference Type
Model
(quality*) Design
Sample
size, nPrimary outcome
Mikami et al. 2019 Aerobic, mental fatigue Humans (2) RT, DB, PC 159 Improved endurance via V
˙O
2
, and reduced
psychometric fatigue
LeBaron et al. 2019 Aerobic endurance (V
˙O
2
) Humans (3) RT, DB, PC, CO 19 Decreased exercising heart rate during
submaximal intensity
Ostojic et al. 2018 Aerobic endurance (V
˙O
2
) Humans (3) RT, DB, PC, CO 12 Improved V
˙O
2
max, increased exercise time, and
more work done
Kawamura et al. 2018 Neutrophil dynamics and
function
Humans (3) RT, DB, PC, CO 9 No statistically significant differences were
observed in function or IL-6
Shin et al. 2018 Run-induced oxidative
stress
Humans (2) RT, DB, PC, CO 15 Reduced oxidative markers (8-OHdG, MDA) and
faster return to baseline
Sha et al. 2018 Microbiome, ROS
inflammation
Humans (2) RT, DB, PC 20 Improved microbiome; decreased MDA, IL-1, IL-6,
and TNF-; increased SOD and TAC
Aoki et al. 2012 Muscle fatigue Humans (3) RT, DB, PC, CO 10 Reduced lactate and fatigue; no changes in d-ROMs
or BAP
Ostojic´ et al. 2011 Running performance Humans (3) RT, DB PC, CO 18 Decreased perceived exertion and lactate levels; no
change in TAC
Ostojic and Stojanovic
2014
Safety in athletes Humans (3) RT, DB, PC 52 No adverse effects from 14 days of oral ingestion of
2 L HRW per day
Ponte et al. 2017 Prolonged repetitive
sprints
Humans (2) RT, PC, SB, CO 8 Maintained peak-power output during repetitive
sprints to exhaustion
Sun and Sun 2017 Two hours of intense
swimming exercise
Humans (2) RT, PC 60 Significantly improved SOD and TAC; decreased
superoxide levels
Ostojic et al. 2014 Musculotendinous injury
in athletes
Humans (2) RT, SB 36 Decreased plasma viscosity; faster recovery from
soft tissue injury
Kawamura et al. 2016 Muscle soreness Humans (2) RT, DB, PC, CO 9 Reduced delayed onset muscle soreness; no change
in d-ROMs or BAP
Shibayama et al. 2017 Redox status in intense
exercise
Humans (2) RT, SB, PC, CO 8 Significantly suppressed the exercise-induced
reduction in BAP/d-ROMs ratio
Koyama et al. 2008 DNA damage from
exercise
Humans (1) RT, DB, PC Suppressed urinary excretion of exercise-induced
increases of 8-OHdG
Drid et al. 2016 Judo post exercise
recovery
Humans (1) RT, DB, PC, CO 5 Decreased blood lactate and a trend for reduced
post-exercise heart rate
Drid et al. 2013 Intense judo training Humans (1) RT, SB, PC 12 Significantly reduced lactate elevation induced
after 1 h of intense judo training
Tsubone et al. 2013 Oxidative stress in
maximal exercise
Horses (2) RT, PC 5 Attenuated oxidative stress induced from maximal
exercise
Yamazaki et al. 2015 Oxidative stress in
maximal exercise
Horses (2) RT, PC 13 Significantly reduced oxidative stress immediately,
3 h, and 24 h after exercise
Wang et al. 2015 Eccentric exercise Mice (1) RT, PC 40 Significantly increased sirtuin-3 and MnSOD;
decreased NLRP3 and Il-1
Ara et al. 2018 Forced swim test fatigue
and stress
Mice (2) RT, PC 21 Decreased lactate, BUN, TNF-, IL-6, IL-17, and IL-1;
increased GPx
Nogueira et al. 2018 Run-induced oxidative
stress
Mice (2) RT, PC 60 Increased SOD, decreased TBARS, mildly blunted
inflammation (i.e., TNF-, IL-6)
Note: RT, randomized trial; DB, double-blinded; PC, placebo-controlled; CO, cross-over; SB, single-blinded. 8OHdG, 8-hydroxy-2=-deoxyguanine; BAP, biological
antioxidant potential; BUN, blood urea nitrogen; d-ROM, diacron reactive oxygen metabolite; GPx, glutathione peroxidase; HRW, hydrogen-rich water; IL, interleukin;
MDA, malondialdehyde; NLRP3, NOD-like receptor prin domain-containing 3; SOD, superoxide dismutase; TAC, total antioxidant capacity; TBARS, thiobarbituric acid
reactive substances; TNF-, tumor necrosis factor-;V
˙O
2
, oxygen consumption; V
˙O
2
max, maximal oxygen consumption.
*Quality Rating 1–3, where 1 is of the lowest quality either because it was not controlled and (or) because the details are not open to the public (e.g., non-English
language), and 3 is of the highest reliability performed in hydrogen research.
LeBaron et al. 803
Published by NRC Research Press
passive diffusion and protect DNA, RNA, proteins, cell mem-
branes, and mitochondria from damage (Ohta 2015); (ii) selectively
decrease only the most cytotoxic radicals without eliminating
beneficial signalling (Ohsawa et al. 2007); (iii) maintain redox ho-
meostasis by decreasing the oxidant load via signal modulation
(e.g., downregulation of the NADPH oxidase system) (Sato et al.
2008); (iv) activate the Nrf2 pathway with subsequent upregula-
tion of endogenous antioxidants (e.g., GSH, CAT, GPx, and induc-
tion of heme oxygenase-1) (Zhai et al. 2014); and (v) decrease
excessive levels of pro-inflammatory mediators (e.g., cytokines,
COX2, NFAT, etc.) Additionally, in some cases, molecular hydrogen
also increases oxidant production (e.g., superoxide) (Murakami et al.
2017), and so potentially provides hormetic benefits similar to those
due to exercise. Lastly, hydrogen also increases sirtuin 3 expres-
sion (Wang et al. 2015), maintains mitochondrial membrane po-
tential (Cui et al. 2014), increases ATP production (Dohi et al. 2014),
and has other benefits as shown in Table 1, all of which can pro-
vide an ergogenic and cytoprotective benefit for elite and nonelite
athletes.
Conclusion
Exercise is associated with many effects that are either noxious
or beneficial, depending on its intensity, duration, and frequency.
In either case, these effects are at least partly mediated by
exercise-induced increases in ROS and inflammation that activate
various transcription factors, leading to their phenotypic expres-
sion. Similar to prolonged high-intensity exercise, various disease
conditions are also associated with excessive ROS and inflamma-
tion. Molecular hydrogen attenuates many of these pathological
disease conditions in animal and human studies, suggesting that
it may similarly mitigate the toxic effects of chronic, high-intensity
exercise training in elite athletes, or sporadic, high-intensity exer-
cise bouts in untrained individuals. Hydrogen may play an impor-
tant role as an exercise mimetic and redox adaptogen to regulate
the exercise-induced production of inflammation, and protect
against harmful cellular stress. These biomedical properties of
hydrogen and the human studies on exercise performance sug-
gest that it has ergogenic potential worthy of further explora-
tion. However, additional human studies with different doses,
durations, and methods of administration are required before
stronger recommendations or conclusions can be made.
Acknowledgement
This study was supported by grants APVV-15-0376, VEGA 2/0021/15,
and ITMS 26230120009.
References
Aoki, K., Nakao, A., Adachi, T., Matsui, Y., and Miyakawa, S. 2012. Pilot study:
Effects of drinking hydrogen-rich water on muscle fatigue caused by acute
exercise in elite athletes. Med. Gas Res. 2(1): 12. doi:10.1186/2045-9912-2-12.
PMID:22520831.
Ara, J., Fadriquela, A., Ahmed, M.F., Bajgai, J., Sajo, M.E.J., Lee, S.P., et al. 2018.
Hydrogen Water Drinking Exerts Antifatigue Effects in Chronic Forced
Swimming Mice via Antioxidative and Anti-Inflammatory Activities. BioMed
Res. Int. 2018: 2571269. doi:10.1155/2018/2571269. PMID:29850492.
Balaban, R.S., Nemoto, S., and Finkel, T. 2005. Mitochondria, oxidants, and
aging. Cell, 120(4): 483–495. doi:10.1016/j.cell.2005.02.001. PMID:15734681.
Bentley, D.J., Ackerman, J., Clifford, T., and Slattery, K.S. 2015. Acute and Chronic
Effects of Antioxidant Supplementation on Exercise Performance. In Antiox-
idants in Sport Nutrition. Edited by M. Lamprecht. Boca Raton (FL).
Braakhuis, A.J., Hopkins, W.G., and Lowe, T.E. 2013. Effects of dietary antioxi-
dants on training and performance in female runners. Eur. J. Sport Sci. 14(2):
160–168. doi:10.1080/17461391.2013.785597. PMID:23600891.
Brown, G.C., and Neher, J.J. 2010. Inflammatory neurodegeneration and mecha-
nisms of microglial killing of neurons. Mol. Neurobiol. 41(2–3): 242–247.
doi:10.1007/s12035-010-8105-9. PMID:20195798.
Case, E.M., and Haldane, J.B. 1941. Human physiology under high pressure: I.
Effects of Nitrogen, Carbon Dioxide, and Cold. J. Hyg. 41(3): 225–249. doi:10.
1017/S0022172400012432.
Chen, X., Zhai, X., Shi, J., Liu, W.W., Tao, H., Sun, X., and Kang, Z. 2013. Lactulose
Mediates Suppression of Dextran Sodium Sulfate-Induced Colon Inflamma-
tion by Increasing Hydrogen Production. Dig. Dis. Sci. 58: 1560–1568. doi:10.
1007/s10620-013-2563-7.
Clements, W.T., Lee, S.R., and Bloomer, R.J. 2014. Nitrate ingestion: a review of
the health and physical performance effects. Nutrients, 6(11): 5224–5264.
doi:10.3390/nu6115224. PMID:25412154.
Cui, Y., Zhang, H., Ji, M., Jia, M., Chen, H., Yang, J., and Duan, M. 2014. Hydrogen-
rich saline attenuates neuronal ischemia-reperfusion injury by protecting
mitochondrial function in rats. J. Surg. Res. 192: 564–572. doi:10.1016/j.jss.
2014.05.060.
D’Autréaux, B., and Toledano, M.B. 2007. ROS as signalling molecules: mecha-
nisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol.
8(10): 813–824. doi:10.1038/nrm2256.
Davies, K.J., Quintanilha, A.T., Brooks, G.A., and Packer, L. 1982. Free radicals and
tissue damage produced by exercise. Biochem. Biophys. Res. Commun.
107(4): 1198–1205. doi:10.1016/S0006-291X(82)80124-1. PMID:6291524.
Dickinson, B.C., and Chang, C.J. 2011. Chemistry and biology of reactive oxygen
species in signaling or stress responses. Nat. Chem. Biol. 7(8): 504–511. doi:
10.1038/nchembio.607. PMID:21769097.
Dixon, B.J., Tang, J., and Zhang, J.H. 2013. The evolution of molecular hydrogen:
a noteworthy potential therapy with clinical significance. Med. Gas Res. 3(1): 10.
doi:10.1186/2045-9912-3-10. PMID:23680032.
Dohi, K., Kraemer, B.C., Erickson, M.A., McMillan, P.J., Kovac, A.,
Flachbartova, Z., et al. 2014. Molecular Hydrogen in Drinking Water Protects
against Neurodegenerative Changes Induced by Traumatic Brain Injury. PLoS
One, 9(9): e108034. doi:10.1371/journal.pone.0108034. PMID:25251220.
Dole, M., Wilson, F.R., and Fife, W.P. 1975. Hyperbaric hydrogen therapy: a
possible treatment for cancer. Science, 190(4210): 152–154. doi:10.1126/science.
1166304. PMID:1166304.
Dougherty, J.H., Jr. 1976. Use of H2 as an inert gas during diving: pulmonary
function during H2-O2 breathing at 7.06 ATA. Aviat. Space Environ. Med.
47(6): 618–626. PMID:938397.
Drid, P., Ostojic, S.M., Stojanovic, M., and Trivic, T. 2013. Hydrogen-Rich Water in
Judo Training. Psycho-Physiol. Spiritual Ethical Aspects, 4: 129.
Drid, P., Stojanovic, M.D., Trivic, T., and Ostojic, S.M. 2016. Molecular Hydrogen
Affected Post-Exercise Recovery in Judo Athletes: 3820 Board# 259 June 4,
9:30 AM–11:00 AM. Med. Sci. Sports Exerc. 48(5): 1071.
Durand, M.J., and Gutterman, D.D. 2014. Exercise and vascular function: how
much is too much? Can. J. Physiol. Pharmacol. 92(7): 551–557. doi:10.1139/cjpp-
2013-0486. PMID:24873760.
Eckermann, J.M., Chen, W., Jadhav, V., Hsu, F.P., Colohan, A.R., Tang, J., and
Zhang, J.H. 2011. Hydrogen is neuroprotective against surgically induced
brain injury. Med. Gas Res. 1(1): 7. doi:10.1186/2045-9912-1-7. PMID:22146427.
Evans, W.J. 2000. Vitamin E, vitamin C, and exercise. Am. J. Clin. Nutr. 72(S2):
647S–652S. doi:10.1093/ajcn/72.2.647S. PMID:10919971.
Feleciano, D.R., and Kirstein, J. 2016. Collapse of redox homeostasis during aging
and stress. Mol. Cell. Oncol. 3(2): e1091060. doi:10.1080/23723556.2015.1091060.
PMID:27308612.
Feng, R., Cai, M., Wang, X., Zhang, J., and Tian, Z. 2018. Early Aerobic Exercise
Combined with Hydrogen-Rich Saline as Preconditioning Protects Myocar-
dial Injury Induced by Acute Myocardial Infarction in Rats. Appl. Biochem.
Biotechnol. 187: 663–676. doi:10.1007/s12010-018-2841-0.
Finkel, T. 2011. Signal transduction by reactive oxygen species. J. Cell Biol. 194(1):
7–15. doi:10.1083/Jcb.201102095. PMID:21746850.
Fisher-Wellman, K., and Bloomer, R.J. 2009. Acute exercise and oxidative stress:
a 30 year history. Dyn. Med. 8: 1. doi:10.1186/1476-5918-8-1. PMID:19144121.
Friess, S.L., Hudak, W.V., and Boyer, R.D. 1978. Toxicology of hydrogen-
containing diving environments. I. Antagonism of acute CO2 effects in the
rat by elevated partial pressures of H2 gas. Toxicol. Appl. Pharmacol. 46(3):
717–725. doi:10.1016/0041-008X(78)90317-4. PMID:746557.
Gao, Q., Song, H., Wang, X.T., Liang, Y., Xi, Y.J., Gao, Y., et al. 2017. Molecular
hydrogen increases resilience to stress in mice. Sci. Rep. 7(1): 9625. doi:10.
1038/s41598-017-10362-6. PMID:28852144.
Gleeson, M. 2007. Immune function in sport and exercise. J. Appl. Physiol. 103(2):
693–699. doi:10.1152/japplphysiol.00008.2007. PMID:17303714.
Gleeson, M., Nieman, D.C., and Pedersen, B.K. 2004. Exercise, nutrition and immune
function. J. Sports Sci. 22(1): 115–125. doi:10.1080/0264041031000140590. PMID:
14971437.
Gliemann, L., Schmidt, J.F., Olesen, J., Biensø, R.S., Peronard, S.L., Grandjean, S.U.,
et al. 2013. Resveratrol blunts the positive effects of exercise training on
cardiovascular health in aged men. J. Physiol. 591(20): 5047–5059. doi:10.1113/
jphysiol.2013.258061. PMID:23878368.
Gliemann, L., Olesen, J., Biensø, R.S., Schmidt, J.F., Akerstrom, T., Nyberg, M.,
et al. 2014. Resveratrol modulates the angiogenic response to exercise train-
ing in skeletal muscles of aged men. Am. J. Physiol. Heart Circ. Physiol.
307(8): H1111–H1119. doi:10.1152/ajpheart.00168.2014. PMID:25128170.
Gomes, E.C., Silva, A.N., and de Oliveira, M.R. 2012. Oxidants, antioxidants, and
the beneficial roles of exercise-induced production of reactive species. Oxid.
Med. Cell Longev. 2012: 756132. doi:10.1155/2012/756132. PMID:22701757.
Gomez-Cabrera, M.C., Domenech, E., Romagnoli, M., Arduini, A., Borras, C.,
Pallardo, F.V., et al. 2008. Oral administration of vitamin C decreases muscle
mitochondrial biogenesis and hampers training-induced adaptations in en-
durance performance. Am. J. Clin. Nutr. 87(1): 142–149. doi:10.1093/ajcn/87.1.
142. PMID:18175748.
804 Can. J. Physiol. Pharmacol. Vol. 97, 2019
Published by NRC Research Press
Gomez-Pinilla, F., Zhuang, Y., Feng, J., Ying, Z., and Fan, G. 2011. Exercise impacts
brain-derived neurotrophic factor plasticity by engaging mechanisms of
epigenetic regulation. Eur. J. Neurosci. 33(3): 383–390. doi:10.1111/j.1460-9568.
2010.07508.x. PMID:21198979.
Halliwell, B., and Gutteridge, J. 2015. Free radicals in biology and medicine.
5
th
edition, Oxford University Press, U.S.A.
Handschin, C., and Spiegelman, B.M. 2008. The role of exercise and PGC1alpha in
inflammation and chronic disease. Nature, 454(7203): 463–469. doi:10.1038/
nature07206. PMID:18650917.
Harman, D. 2009. About “Origin and evolution of the free radical theory of
aging: a brief personal history, 1954-2009”. Biogerontology, 10(6): 783. doi:10.
1007/s10522-009-9253-z. PMID:19856210.
Hayashida, K., Sano, M., Ohsawa, I., Shinmura, K., Tamaki, K., Kimura, K., et al.
2008. Inhalation of hydrogen gas reduces infarct size in the rat model of
myocardial ischemia-reperfusion injury. J. Card. Fail. 14(7): S168–S168. doi:10.
1016/J.Cardfail.2008.07.178.
Hirayama, M., Ito, M., Minato, T., Yoritaka, A., LeBaron, T.W., and Ohno, K. 2018.
Inhalation of hydrogen gas elevates urinary 8-hydroxy-2=-deoxyguanine
in Parkinson’s disease. Med. Gas Res. 8(4): 144–149. doi:10.4103/2045-9912.
248264. PMID:30713666.
Hotamisligil, G.S. 2006. Inflammation and metabolic disorders. Nature, 444(7121):
860–867. doi:10.1038/nature05485. PMID:17167474.
Ichihara, M., Sobue, S., Ito, M., Ito, M., Hirayama, M., and Ohno, K. 2015. Benefi-
cial biological effects and the underlying mechanisms of molecular hydro-
gen - comprehensive review of 321 original articles. Med. Gas Res. 5: 12.
doi:10.1186/s13618-015-0035-1. PMID:26483953.
Ishibashi, T., Sato, B., Rikitake, M., Seo, T., Kurokawa, R., Hara, Y., et al. 2012.
Consumption of water containing a high concentration of molecular hydro-
gen reduces oxidative stress and disease activity in patients with rheumatoid
arthritis: an open-label pilot study. Med. Gas Res. 2(1): 27. doi:10.1186/2045-
9912-2-27. PMID:23031079.
Ito, M., Ibi, T., Sahashi, K., Ichihara, M., and Ohno, K. 2011. Open-label trial and
randomized, double-blind, placebo-controlled, crossover trial of hydrogen-
enriched water for mitochondrial and inflammatory myopathies. Med. Gas
Res. 1(1): 24. doi:10.1186/2045-9912-1-24. PMID:22146674.
Ito, M., Hirayama, M., Yamai, K., Goto, S., Ichihara, M., and Ohno, K. 2012.
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. 2(1): 15. doi:10.1186/2045-
9912-2-15. PMID:22608009.
Jones, D.P. 2008. Radical-free biology of oxidative stress. Am. J. Physiol.-Cell
Physiol. 295(4): C849–C868. doi:10.1152/ajpcell.00283.2008. PMID:18684987.
Juarez, J.C., Manuia, M., Burnett, M.E., Betancourt, O., Boivin, B., Shaw, D.E.,
et al. 2008. Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated
oxidation and inactivation of phosphatases in growth factor signaling. Proc.
Natl. Acad. Sci. U. S. A. 105(20): 7147–7152. doi:10.1073/pnas.0709451105. PMID:
18480265.
Kajiya, M., Sato, K., Silva, M.J., Ouhara, K., Do, P.M., Shanmugam, K.T., and
Kawai, T. 2009. Hydrogen from intestinal bacteria is protective for Conca-
navalin A-induced hepatitis. Biochem. Biophys. Res. Commun. 386(2): 316–
321. doi:10.1016/j.bbrc.2009.06.024. PMID:19523450.
Kajiyama, S., Hasegawa, G., Asano, M., Hosoda, H., Fukui, M., Nakamura, N., et al.
2008. Supplementation of hydrogen-rich water improves lipid and glucose
metabolism in patients with type 2 diabetes or impaired glucose tolerance.
Nutr. Res. 28: 137–143. doi:10.1016/j.nutres.2008.01.008. PMID:19083400.
Kato, S., Saitoh, Y., Iwai, K., and Miwa, N. 2012. Hydrogen-rich electrolyzed warm
water represses wrinkle formation against UVA ray together with type-I col-
lagen production and oxidative-stress diminishment in fibroblasts and cell-
injury prevention in keratinocytes. J. Photochem. Photobiol. B: Biol. 106:
24–33. doi:10.1016/j.jphotobiol.2011.09.006. PMID:22070900.
Katsumata, Y., Sano, F., Abe, T., Tamura, T., Fujisawa, T., Shiraishi, Y., et al. 2017.
The Effects of Hydrogen Gas Inhalation on Adverse Left Ventricular Remod-
eling After Percutaneous Coronary Intervention for ST-Elevated Myocardial
Infarction-First Pilot Study in Humans. Circ J. doi:10.1253/circj.CJ-17-0105.
Kawamura, T., Gando, Y., Takahashi, M., Hara, R., Suzuki, K., and Muraoka, I.
2016. Effects of hydrogen bathing on exercise-induced oxidative stress and
delayed-onset muscle soreness. Jpn J. Phys. Fitness Sports Med. 65(3): 297–
305. doi:10.7600/jspfsm.65.297.
Kawamura, T., Suzuki, K., Takahashi, M., Tomari, M., Hara, R., Gando, Y., et al.
2018. Involvement of Neutrophil Dynamics and Function in Exercise-Induced
Muscle Damage and Delayed-Onset Muscle Soreness: Effect of Hydrogen
Bath. Antioxidants, 7(10): 127. doi:10.3390/antiox7100127.
Koyama, K., Tanaka, Y., Saihara, Y., Ando, D., Goto, Y., and Katayama, A. 2008.
Effect of hydrogen saturated alkaline electrolyzed water on urinary oxidative
stress markers after an acute exercise: A randomized controlled trial. Anti-
aging Med. 4: 117–122.
Kura, B., Bagchi, A.K., Singal, P.K., Barancik, M., LeBaron, T.W., Valachova, K.,
et al. 2018. Molecular hydrogen (H2): Potential in mitigating oxidative stress-
induced cardiotoxicity. Can. J. Physiol. Pharmacol. 97: 287–292. doi:10.1139/
cjpp-2018-0604.
Kura, B., Kalocayova, B., LeBaron, T.W., Frimmel, K., Buday, J., Surovy, J., and
Slezak, J. 2019. Regulation of microRNAs by molecular hydrogen contributes
to the prevention of radiation-induced damage in the rat myocardium. Mol.
Cell. Biochem. 1–12. doi:10.1007/s11010-019-03512-z.
Lafay, S., Jan, C., Nardon, K., Lemaire, B., Ibarra, A., Roller, M., et al. 2009. Grape
extract improves antioxidant status and physical performance in elite male
athletes. J. Sports Sci. Med. 8(3): 468–480. PMID:24150013.
LeBaron, T.W., Kura, B., Kalocayova, B., Tribulova, N., and Slezak, J. 2019. A new
approach for the prevention and treatment of cardiovascular disorders. Mo-
lecular hydrogen significantly reduces the effects of oxidative stress. Mole-
cules. 24(11): 2076. doi:10.3390/molecules24112076.
LeBaron, T.W., Larson, A., Ohta, S., Mikami, T., Barlow, J., Bulloch, J., and
DeBeliso, M. 2019. Acute Supplementation with Molecular Hydrogen Benefits
Submaximal Exercise Indices. Randomized, Double-blinded, Placebo-controlled
Crossover Pilot Study. J. Lifestyle Med. 9(1): 36–43. doi:10.15280/jlm.2019.9.1.36.
PMID:30918832.
Levinger, I., Shaw, C.S., Stepto, N.K., Cassar, S., McAinch, A.J., Cheetham, C., and
Maiorana, A.J. 2015. What Doesn’t Kill You Makes You Fitter: A Systematic
Review of High-Intensity Interval Exercise for Patients with Cardiovascular
and Metabolic Diseases. Clin. Med. Insights Cardiol. 9: 53–63. doi:10.4137/
CMC.S26230. PMID:26157337.
Li, Q., Kato, S., Matsuoka, D., Tanaka, H., and Miwa, N. 2013. Hydrogen water
intake via tube-feeding for patients with pressure ulcer and its reconstructive
effects on normal human skin cells in vitro. Med. Gas Res. 3(1): 20. doi:10.
1186/2045-9912-3-20. PMID:24020833.
Liochev, S.I. 2013. Reactive oxygen species and the free radical theory of aging.
Free Radic. Biol. Med. 60: 1–4. doi:10.1016/j.freeradbiomed.2013.02.011. PMID:
23434764.
Liu, Y., Fiskum, G., and Schubert, D. 2002. Generation of reactive oxygen species
by the mitochondrial electron transport chain. J. Neurochem. 80(5): 780–787.
doi:10.1046/j.0022-3042.2002.00744.x. PMID:11948241.
Matchett, G.A., Fathali, N., Hasegawa, Y., Jadhav, V., Ostrowski, R.P., Martin, R.D.,
et al. 2009. Hydrogen gas is ineffective in moderate and severe neonatal
hypoxia-ischemia rat models. Brain Res. 1259: 90–97. doi:10.1016/j.brainres.
2008.12.066. PMID:19168038.
Merry, T.L., and Ristow, M. 2016. Do antioxidant supplements interfere with
skeletal muscle adaptation to exercise training? J. Physiol. 594(18): 5135–
5147. doi:10.1113/JP270654. PMID:26638792.
Mikami, T., Tano, K., Hosung, L., Lee, H., Park, J., Ohta, F., LeBaron, T.W., and
Ohta, S. 2019. Drinking hydrogen water enhances endurance and relieves
psychometric fatigue: randomized, double-blind, placebo-controlled study.
Can. J. Physiol. Pharmacol. In press. doi:10.1139/cjpp-2019-0059.
Mizuno, K., Sasaki, A.T., Ebisu, K., Tajima, K., Kajimoto, O., Nojima, J., et al. 2017.
Hydrogen-rich water for improvements of mood, anxiety, and autonomic
nerve function in daily life. Med. Gas Res. 7(4): 247–255. doi:10.4103/2045-9912.
222448. PMID:29497485.
Morrison, D., Hughes, J., Della Gatta, P.A., Mason, S., Lamon, S., Russell, A.P., and
Wadley, G.D. 2015. Vitamin C and E supplementation prevents some of the
cellular adaptations to endurance-training in humans. Free Radic. Biol. Med.
89: 852–862. doi:10.1016/j.freeradbiomed.2015.10.412. PMID:26482865.
Murakami, Y., Ito, M., and Ohsawa, I. 2017. Molecular hydrogen protects against
oxidative stress-induced SH-SY5Y neuroblastoma cell death through the pro-
cess of mitohormesis. PLoS One, 12(5): e0176992. doi:10.1371/journal.pone.
0176992. PMID:28467497.
Nagata, K., Nakashima-Kamimura, N., Mikami, T., Ohsawa, I., and Ohta, S. 2009.
Consumption of Molecular Hydrogen Prevents the Stress-Induced Impair-
ments in Hippocampus-Dependent Learning Tasks during Chronic Physical
Restraint in Mice. Neuropsychopharmacology, 34(2): 501–508. doi:10.1038/
npp.2008.95. PMID:18563058.
Nakao, A., Toyoda, Y., Sharma, P., Evans, M., and Guthrie, N. 2010. Effectiveness
of Hydrogen Rich Water on Antioxidant Status of Subjects with Potential
Metabolic Syndrome-An Open Label Pilot Study. J. Clin. Biochem. Nutr. 46(2):
140–149. doi:10.3164/jcbn.09-100. PMID:20216947.
Nakayama, M., Nakano, H., Hamada, H., Itami, N., Nakazawa, R., and Ito, S. 2010.
A novel bioactive haemodialysis system using dissolved dihydrogen (H-2)
produced by water electrolysis: a clinical trial. Nephrol. Dial. Transplant.
25(9): 3026–3033. doi:10.1093/ndt/gfq196. PMID:20388631.
Nathan, C., and Xie, Q.W. 1994. Nitric oxide synthases: roles, tolls, and controls.
Cell, 78(6): 915–918. doi:10.1016/0092-8674(94)90266-6. PMID:7522969.
Nicolson, G.L., de Mattos, G.F., Settineri, R., Costa, C., Ellithorpe, R., Rosenblatt, S.,
et al. 2016. Clinical Effects of Hydrogen Administration: From Animal and
Human Diseases to Exercise Medicine. Int. J. Clin. Med. 7(1): 32–76. doi:10.
4236/ijcm.2016.71005.
Nishimaki, K., Asada, T., Ohsawa, I., Nakajima, E., Ikejima, C., Yokota, T., et al.
2018. Effects of Molecular Hydrogen Assessed by an Animal Model and a
Randomized Clinical Study on Mild Cognitive Impairment. Curr. Alzheimer
Res. 15(5): 482–492. doi:10.2174/1567205014666171106145017. PMID:29110615.
Nishiwaki, H., Ito, M., Negishi, S., Sobue, S., Ichihara, M., and Ohno, K. 2018.
Molecular hydrogen upregulates heat shock response and collagen biosyn-
thesis, and downregulates cell cycles - Meta-analyses of gene expression pro-
files. Free Radic. Res. 52: 434–445. doi:10.1080/10715762.2018.1439166.
Nogueira, J.E., Passaglia, P., Mota, C.M.D., Santos, B.M., Batalhão, M.E.,
Carnio, E.C., and Branco, L.G.S. 2018. Molecular hydrogen reduces acute
exercise-induced inflammatory and oxidative stress status. Free Radic. Biol.
Med. 129: 186–193. doi:10.1016/j.freeradbiomed.2018.09.028. PMID:30243702.
LeBaron et al. 805
Published by NRC Research Press
Ohno, K., Ito, M., and Ichihara, M. 2012. Molecular hydrogen as an emerging
therapeutic medical gas for neurodegenerative and other diseases. Oxid.
Med. Cell. Longevity, 2012: 353152. doi:10.1155/2012/353152.
Ohsawa, I., Ishikawa, M., Takahashi, K., Watanabe, M., Nishimaki, K., Yamagata, K.,
et al. 2007. Hydrogen acts as a therapeutic antioxidant by selectively reducing
cytotoxic oxygen radicals. Nat. Med. 13(6): 688–694. doi:10.1038/nm1577.
PMID:17486089.
Ohta, S. 2015. Molecular hydrogen as a novel antioxidant: overview of the ad-
vantages of hydrogen for medical applications. Methods Enzymol. 555: 289–
317. doi:10.1016/bs.mie.2014.11.038. PMID:25747486.
Olesen, J., Gliemann, L., Biensø, R., Schmidt, J., Hellsten, Y., and Pilegaard, H.
2014. Exercise training, but not resveratrol, improves metabolic and inflam-
matory status in skeletal muscle of aged men. J. Physiol. 592(8): 1873–1886.
doi:10.1113/jphysiol.2013.270256. PMID:24514907.
Ono, H., Nishijima, Y., Ohta, S., Sakamoto, M., Kinone, K., Horikosi, T., et al.
2017. Hydrogen Gas Inhalation Treatment in Acute Cerebral Infarction: A
Randomized Controlled Clinical Study on Safety and Neuroprotection. J.
Stroke Cerebrovasc. Dis. 26: 2587–2594. doi:10.1016/j.jstrokecerebrovasdis.
2017.06.012.
Ortenblad, N., Madsen, K., and Djurhuus, M.S. 1997. Antioxidant status and lipid
peroxidation after short-term maximal exercise in trained and untrained
humans. Am. J. Physiol.: Regul. Integr. Comp. Physiol. 272(4.2): R1258–R1263.
doi:10.1152/ajpregu.1997.272.4.R1258.
Ostojic, S.M. 2014. Molecular Hydrogen in Sports Medicine: New Therapeutic
Perspectives. Int. J. Sports Med. 36: 273–279. doi:10.1055/s-0034-1395509.
PMID:25525953.
Ostojic, S.M., and Stojanovic, M.D. 2014. Hydrogen-rich water affected blood
alkalinity in physically active men. Res. Sports Med. 22(1): 49– 60. doi:10.1080/
15438627.2013.852092. PMID:24392771.
Ostojic´ , S.M., Stojanovic´, M.D., Calleja-Gonzalez, J., Obrenovic´, M.D., Veljovic´, D.,
Međedovic´ , B., et al. 2011. Drinks with alkaline negative oxidative reduction
potential improve exercise performance in physically active men and women:
Double-blind, randomized, placebo-controlled, cross-over trial of efficacy
and safety. Serbian J. Sports Sci. 5(1–4): 83–89.
Ostojic, S.M., Vukomanovic, B., Calleja-Gonzalez, J., and Hoffman, J.R. 2014.
Effectiveness of oral and topical hydrogen for sports-related soft tissue inju-
ries. Postgrad. Med. 126(5): 187–195. doi:10.3810/pgm.2014.09.2813. PMID:
25295663.
Ostojic, S.M., Korovljev, D., Stajer, V., and Javorac, D. 2018. 28-days Hydrogen-
rich Water Supplementation Affects Exercise Capacity in Mid-age Over-
weight Women: 2942 Board# 225. Med. Sci. Sports Exerc. 50(S5): 728–
729.
Pacher, P., Beckman, J.S., and Liaudet, L. 2007. Nitric oxide and peroxynitrite in
health and disease. Physiol. Rev. 87(1): 315–424. doi:10.1152/physrev.00029.
2006. PMID:17237348.
Palmieri, B., and Sblendorio, V. 2006. Oxidative stress detection: what for? Part I.
Eur. Rev. Med. Pharmacol. Sci. 10(6): 291–317. PMID:17274534.
Paoloni, J.A., Milne, C., Orchard, J., and Hamilton, B. 2009. Non-steroidal anti-
inflammatory drugs in sports medicine: guidelines for practical but sensible
use. Br. J. Sports Med. 43(11): 863–865. doi:10.1136/bjsm.2009.059980. PMID:
19546098.
Paulsen, G., Cumming, K.T., Holden, G., Hallén, J., Rønnestad, B.R., Sveen, O.,
et al. 2014. Vitamin C and E supplementation hampers cellular adaptation
to endurance training in humans: a double-blind, randomised, controlled trial.
J. Physiol. 592(8): 1887–1901. doi:10.1113/jphysiol.2013.267419. PMID:24492839.
Pedersen, B.K., Åkerström, T.C., Nielsen, A.R., and Fischer, C.P. 2007. Role of
myokines in exercise and metabolism. J. Appl. Physiol. 103(3): 1093–1098.
doi:10.1152/japplphysiol.00080.2007. PMID:17347387.
Ponte, D.A., Giovanelli, N., Nigris, D., and Lazzer, S. 2017. Effects of hydrogen
rich water on prolonged intermittent exercise. J. Sports Med. Phys. Fitness.
58: 612–621. doi:10.23736/S0022-4707.17.06883-9.
Powers, S.K., Ji, L.L., Kavazis, A.N., and Jackson, M.J. 2011. Reactive oxygen spe-
cies: impact on skeletal muscle. Compr. Physiol. 1(2): 941–969. doi:10.1002/
cphy.c100054. PMID:23737208.
Ristow, M., and Zarse, K. 2010. How increased oxidative stress promotes longev-
ity and metabolic health: The concept of mitochondrial hormesis (mito-
hormesis). Exp. Gerontol. 45(6): 410–418. doi:10.1016/j.exger.2010.03.014. PMID:
20350594.
Ristow, M., Zarse, K., Oberbach, A., Kloting, N., Birringer, M., Kiehntopf, M., et al.
2009. Antioxidants prevent health-promoting effects of physical exercise in
humans. Proc. Natl. Acad. Sci. U. S. A. 106(21): 8665–8670. doi:10.1073/pnas.
0903485106. PMID:19433800.
Sakai, T., Sato, B., Hara, K., Hara, Y., Naritomi, Y., Koyanagi, S., et al. 2014.
Consumption of water containing over 3.5 mg of dissolved hydrogen could
improve vascular endothelial function. Vasc. Health Risk Manage. 10: 591–
597. doi:10.2147/VHRM.S68844.
Sato, Y., Kajiyama, S., Amano, A., Kondo, Y., Sasaki, T., Handa, S., et al. 2008.
Hydrogen-rich pure water prevents superoxide formation in brain slices of
vitamin C-depleted SMP30/GNL knockout mice. Biochem. Biophys. Res. Com-
mun. 375(3): 346–350. doi:10.1016/j.bbrc.2008.08.020. PMID:18706888.
Schoenfeld, B.J. 2012. The use of nonsteroidal anti-inflammatory drugs for
exercise-induced muscle damage: implications for skeletal muscle devel-
opment. Sports Med. 42(12): 1017–1028. doi:10.1007/BF03262309. PMID:
23013520.
Sha, J.B., Zhang, S.S., Lu, Y.M., Gong, W.J., Jiang, X.P., Wang, J.J., et al. 2018.
Effects of the long-term consumption of hydrogen-rich water on the anti-
oxidant activity and the gut flora in female juvenile soccer players from
Suzhou, China. Med. Gas Res. 8(4): 135–143. doi:10.4103/2045-9912.248263.
PMID:30713665.
Sharman, I.M., Down, M.G., and Sen, R.N. 1971. The effects of vitamin E and training on
physiological function and athletic performance in adolescent swimmers. Br. J.
Nutr. 26(2): 265–276. doi:10.1079/BJN19710033. PMID:5571788.
Shibayama, Y., Takeuchi, K., Dobashi, S., and Koyama, K. 2017. Hydrogen-rich
Water Modulates Redox Status Repeated Three Consecutive Days Of Strenu-
ous Exercise (3298 Board# 203 June 2). Med. Sci. Sports Exerc. 49(S5): 941.
doi:10.1249/01.mss.0000519562.97355.a8.
Shin, D.S., Jung, S.H., Hong, E.Y., Shin, Y.H., Park, J.Y., and Chung, M.H. 2018.
Removal Effect of Hydrogen Water Drinking on Exercise-induced Production
of Reactive Oxygen Species in Adult Men and Women. Exerc. Sci. 27(4): 289–
295. doi:10.15857/ksep.2018.27.4.289.
Slezak, J., Kura, B., Ravingerová, T., Tribulova, N., Okruhlicova, L., and
Barancik, M. 2015. Mechanisms of cardiac radiation injury and potential
preventive approaches. Can. J. Physiol. Pharmacol. 93(9): 737–753. doi:10.1139/
cjpp-2015-0006. PMID:26030720.
Slezák, J., Kura, B., Frimmel, K., Zálešák, M., Ravingerová, T., Viczenczová, C.,
et al. 2016. Preventive and Therapeutic Application of Molecular Hydrogen in
Situations With Excessive Production of Free Radicals. Physiol. Res. 65(S1):
S11–S28. PMID:27643933.
Sobue, S., Inoue, C., Hori, F., Qiao, S., Murate, T., and Ichihara, M. 2017. Molecular
hydrogen modulates gene expression via histone modification and induces
the mitochondrial unfolded protein response. Biochem. Biophys. Res. Com-
mun. 493(1): 318–324. doi:10.1016/j.bbrc.2017.09.024. PMID:28890349.
Song, G., Li, M., Sang, H., Zhang, L., Li, X., Yao, S., et al. 2013. Hydrogen-rich water
decreases serum LDL-cholesterol levels and improves HDL function in pa-
tients with potential metabolic syndrome. J. Lipid Res. 54(7): 1884–1893. doi:
10.1194/jlr.M036640. PMID:23610159.
Strocchi, A., and Levitt, M.D. 1992. Maintaining intestinal H2 balance: credit the
colonic bacteria. Gastroenterology, 102(4.1): 1424–1426. doi:10.1016/0016-5085(92)
90790-6.
Sun, Y.P., and Sun, L. 2017. Selective protective effect of hydrogen water on free
radical injury of athletes after high-intensity exercise. Biomed. Res. Trace
Elements, 28(10): 4558–4561.
Tang, Z., Yuan, L., Gu, C., Liu, Y., and Zhu, L. 2005. Effect of exercise on the
expression of adiponectin mRNA and GLUT4 mRNA in type 2 diabetic rats.
J. Huazhong Univ. Sci. Technol. Med. Sci. 25(2): 191–193, 201. doi:10.1007/
BF02873574. PMID:16116970.
Tiidus, P.M., Pushkarenko, J., and Houston, M.E. 1996. Lack of antioxidant adap-
tation to short-term aerobic training in human muscle. Am. J. Physiol.: Regul.
Integr. Comp. Physiol. 271(4.2): R832–R836. doi:10.1152/ajpregu.1996.271.4.
R832.
Trivic, T., Vojnovic, M., Drid, P., and Ostojic, S.M. 2017. Drinking hydrogen-rich
water for 4 weeks positively affects serum antioxidant enzymes in healthy
men: a pilot study. Curr. Topics Nutraceut. Res. 15(1): 45–48.
Tsubone, H., Hanafusa, M., Endo, M., Manabe, N., Hiraga, A., Ohmura, H., and
Aida, H. 2013. Effect of Treadmill Exercise and Hydrogen-rich Water In-
take on Serum Oxidative and Anti-oxidative Metabolites in Serum of Thor-
oughbred Horses. J. Equine Sci. 24(1): 1–8. doi:10.1294/jes.24.1. PMID:
24833996.
Urso, M.L. 2013. Anti-inflammatory interventions and skeletal muscle injury:
benefit or detriment? J. Appl. Physiol. 115(6): 920–928. doi: 10.1152/japplphysiol.
00036.2013.PMID:23539314.
Urso, M.L., and Clarkson, P.M. 2003. Oxidative stress, exercise, and antioxidant
supplementation. Toxicology, 189(1–2): 41–54. doi:10.1016/S0300-483X(03)00151-3.
PMID:12821281.
Wang, L., Liu, Z., Hou, Y., and Ge, Y. 2015. Hydrogen-rich water inhibits mito-
chondrial oxidative stress and inflammation in the skeletal muscle after
eccentric exercise. Chin. J. Tissue Eng. Res. 19(29): 4682–4687.
Wray, D.W., Uberoi, A., Lawrenson, L., Bailey, D.M., and Richardson, R.S. 2009.
Oral antioxidants and cardiovascular health in the exercise-trained and un-
trained elderly: a radically different outcome. Clin. Sci. (Lond) 116(5): 433–
441. doi:10.1042/CS20080337.
Xia, C., Liu, W., Zeng, D., Zhu, L., Sun, X., and Sun, X. 2013. Effect of hydrogen-
rich water on oxidative stress, liver function, and viral load in patients
with chronic hepatitis B. Clin. Translation. Sci. 6(5): 372–375. doi:10.1111/
cts.12076.
Xie, Y., Mao, Y., Zhang, W., Lai, D., Wang, Q., and Shen, W. 2014. Reactive Oxygen
Species-Dependent Nitric Oxide Production Contributes to Hydrogen-Promoted
Stomatal Closure in Arabidopsis. Plant Physiol. 165(2): 759–773. doi:10.1104/pp.
114.237925. PMID:24733882.
Yamazaki, M., Kusano, K., Ishibashi, T., Kiuchi, M., and Koyama, K. 2015. Intra-
venous infusion of H2-saline suppresses oxidative stress and elevates antiox-
idant potential in Thoroughbred horses after racing exercise. Sci. Rep. 5:
15514. doi:10.1038/srep15514. PMID:26493164.
Yoritaka, A., Takanashi, M., Hirayama, M., Nakahara, T., Ohta, S., and Hattori, N.
806 Can. J. Physiol. Pharmacol. Vol. 97, 2019
Published by NRC Research Press
2013. Pilot study of H(2) therapy in Parkinson’s disease: A randomized double-
blind placebo-controlled trial. Mov. Disord. doi:10.1002/mds.25375.
Zhai, X., Chen, X., Ohta, S., and Sun, X. 2014. Review and prospect of the biomed-
ical effects of hydrogen. Med. Gas Res. 4(1): 19. doi:10.1186/s13618-014-0019-6.
PMID:25485090.
Zhang, Y., Su, W.J., Chen, Y., Wu, T.Y., Gong, H., Shen, X.L., et al. 2016. Effects of
hydrogen-rich water on depressive-like behavior in mice. Sci. Rep. 6: 23742.
doi:10.1038/srep23742. PMID:27026206.
Zhuang, Z., Sun, X.J., Zhang, X., Liu, H.D., You, W.C., Ma, C.Y., et al. 2013. Nuclear
factor-kappaB/Bcl-XL pathway is involved in the protective effect of hydrogen-
rich saline on the brain following experimental subarachnoid hemorrhage in
rabbits. J. Neurosci. Res. 91(12): 1599–1608. doi:10.1002/jnr.23281. PMID:24105634.
LeBaron et al. 807
Published by NRC Research Press
... Different conventional antioxidant supplementations (e.g., vitamin C, vitamin E, resveratrol) have been implemented to reduce the ROS for athletes and other populations (8)(9)(10)(11). However, it has been shown that supplementing these types of antioxidants may induce some side effects, such as the reduction in muscle contractile strength and exercise adaptation (12)(13)(14)(15), which may potentially because these antioxidants may exacerbate redox dysregulation and induce over-removal of ROS, of which an appropriate level is actually helpful for the maintenance of physiological function, activation of signaling pathways and initiation of multiple biological processes (16,17). Given the limitations of conventional antioxidants, a novel antioxidant that can effectively combat oxidative stress without interfering with other important functions is highly demanded. ...
... Molecular hydrogen (H 2 ) is a potential antioxidant to alleviate exercise-induced oxidative stress (16,18,19). H 2 can selectively reduce ·OH and ·ONOO without reacting to other important signaling oxidants (e.g., H 2 O 2 ) (20,21). ...
... The experiments were evaluated for potential bias and categorized as low, moderate, or high risk. Specifically, one study (17) was deemed to have a high risk of bias, four studies (16,17,28,39) were classified as having a moderate risk, and the remainder were considered to have a low risk of bias. ...
Article
Full-text available
Objective Exercise-induced oxidative stress affects multiple neurophysiological processes, diminishing the exercise performance. Hydrogen (H2) can selectively reduce excessive free radicals, but studies observed its “dual effects” on exercise-induced oxidative stress, that is, increasing or decreasing the oxidative stress. Therefore, we here conducted a systematic review and meta-analysis to quantitatively assess the influence of H2 on exercise-induced oxidative stress in healthy adults. Methods We conducted a systematic review of publications across five databases. The following keywords were used for search strategy: [“hydrogen”[Mesh] or “molecular hydrogen” or “hydrogen rich water” or “hydrogen-rich water” or “hydrogen rich saline”] and [“Oxidative Stress”[Mesh] or “Antioxidative Stress” or “Oxidative Damage” or “Oxidative Injury” or “Oxidative Cleavage”] and [“randomized controlled trial”[Mesh] or “randomized” or “RCT”]. We included trials reporting the effects of H2 on exercise-induced oxidative stress and potential antioxidant capacity post-exercise in healthy adults. Additionally, subgroup analyses were conducted to explore how various elements of the intervention design affected those outcomes. Results Six studies, encompassing seven experiments with a total of 76 participants, were included in our analysis. Among these studies, hydrogen-rich water, hydrogen bathing, and hydrogen-rich gas were three forms used in H2 administration. The H2 was applied in different timing, including before, during, or after exercise only, both before and after exercise, and repeatedly over days. Single-dose, multi-dose within 1 day and/or multiple-dose over days were implemented. It was observed that compared to placebo, the effects of H2 on oxidative stress (diacron-reactive oxygen metabolites, d-ROMs) was not significant (SMD = −0.01, 95%CI-0.42 to 0.39, p = 0.94). However, H2 induced greater improvement in antioxidant potential capacity (Biological Antioxidant Potential, BAP) (SMD = 0.29, 95% CI 0.04 to 0.54, p = 0.03) as compared to placebo. Subgroup analyses revealed that H2 supplementation showed greater improvement (SMD = 0.52, 95%CI 0.16 to 0.87, p = 0.02) in the antioxidant potential capacity of intermittent exercises than continuous exercise. Conclusion H2 supplementation can help enhance antioxidant potential capacity in healthy adults, especially in intermittent exercise, but not directly diminish the levels of exercise-induced oxidative stress. Future studies with more rigorous design are needed to examine and confirm these findings. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=364123, Identifier CRD42022364123.
... In a further trial conducted in 2019, the efficacy of ingesting 2 tablets of HRW was evaluated on physical performance. It was noted that there was a reduction in heart rate and respiratory rate without any change in VO2 max [36]. Mikami et al. were able to show the efficacy of taking 500 mL of HRW 30 min before physical effort on an ergometric cycle on VO2max and the Borg scale [37]. ...
Article
Full-text available
Background. Due to its antioxidant, anti-inflammatory, anti-apoptosis, and anti-fatigue properties, molecular hydrogen (H2) is potentially a novel therapeutic nutrient for patients with coronavirus acute disease 2019 (COVID-19). We determined the efficacy and safety profile of hydrogen-rich water (HRW) to reduce the risk of COVID-19 progression. Methods: We also conducted a phase 3, triple-blind, randomised, placebo-controlled trial to evaluate treatment with HRW initiated within 5 days after the onset of signs or symptoms in primary care patients with mild-to-moderate, laboratory-confirmed COVID-19. Participants were randomised to receive HRW or placebo twice daily for 21 days. The incidence of clinical worsening and adverse events were the primary endpoints. Results: A total of 675 participants were followed up to day 30. HRW was not superior to placebo in preventing clinical worsening at day 14: in H2 group, 46.1% in the H2 group, 43.5% in the placebo group, hazard ratio 1.09, 90% confidence interval [0.90–1.31]. One death was reported at day 30 in the H2 group and two in the placebo group at day 30. Adverse events were reported in 91 (27%) and 89 (26.2%) participants, respectively. Conclusions: HRW taken twice daily from the onset of COVID-19 symptoms for 21 days did not reduce clinical worsening.
... 38 Hydrogen can promote cell protection, mitochondrial biogenesis, adenosine triphosphate (ATP) generation, an increased NAD(+)/NADH ratio, Phase II enzymes for cell protection, heat shock proteins, and sirtuins, among other effects. 39 Dohi et al reported that treatment with hydrogen-rich culture medium for 24 hours significantly enhanced mitochondrial basal respiration, reserve respiration, and nonmitochondrial respiration while increasing ATP production. 40 Nonmitochondrial respiration refers to the oxygen consumed by enzymes in the cell after the mitochondrial electron transfer chain is closed. ...
Article
Full-text available
Hydrogen, which is a novel biomedical molecule, is currently the subject of extensive research involving animal experiments and in vitro cell experiments, and it is gradually being applied in clinical settings. Hydrogen has been proven to possess anti-inflammatory, selective antioxidant, and antiapoptotic effects, thus exhibiting considerable protective effects in various diseases. In recent years, several studies have provided preliminary evidence for the protective effects of hydrogen on spinal cord injury (SCI). This paper provides a comprehensive review of the potential molecular biology mechanisms of hydrogen therapy and its application in treating SCI, with an aim to better explore the medical value of hydrogen and provide new avenues for the adjuvant treatment of SCI.
... In addition to its antioxidant properties, H 2 has been found to have antiinflammatory properties (Gharib et al., 2001), antiapoptotic properties (Nicolson et al., 2016), and properties that modulate signal transduction and gene expression (Ohta, 2014;Slezak et al., 2021). Due to its health benefits (Ohta, 2014;Ichihara et al., 2015;Botek et al., 2022b;Johnsen et al., 2023), supplementation with H 2 has become popular among athletes to enhance performance and reduce fatigue (LeBaron et al., 2019;Botek et al., 2020;Kawamura et al., 2020;Shibayama et al., 2020;Botek et al., 2021;Timón et al., 2021;Botek et al., 2022a;Jebabli et al., 2023;Zhou et al., 2023). Several recent studies demonstrated that H 2 reduces exerciseinduced pro-inflammatory response and oxidative stress (Ara et al., 2018;Nogueira and Branco, 2021), blood lactate concentrations and improves muscle function (Aoki et al., 2012;Botek et al., 2021). ...
Article
Full-text available
Purpose: Molecular hydrogen has been shown to possess antioxidant, anti-inflammatory, ergogenic, and recovery-enhancing effects. This study aimed to assess the effect of molecular hydrogen administration on muscle performance, damage, and perception of soreness up to 24 h of recovery after two strenuous training sessions performed on the same day in elite fin swimmers. Methods: Eight females (mean ± SD; age 21.5 ± 5.0 years, maximal oxygen consumption 45.0 ± 2.5 mL.kg⁻¹.min⁻¹) and four males (age 18.9 ± 1.3 years, maximal oxygen consumption 52.2 ± 1.7 mL.kg⁻¹.min⁻¹) performed 12 × 50 m sprints in the morning session and a 400 m competitive performance in the afternoon session. Participants consumed hydrogen-rich water (HRW) or placebo 3 days before the sessions (1,260 mL/day) and 2,520 mL on the experimental day. Muscle performance (countermovement jump), muscle damage (creatine kinase), and muscle soreness (100 mm visual analogue scale) were measured during the experimental day and at 12 and 24 h after the afternoon session. Results: HRW compared to placebo reduced blood activity of creatine kinase (156 ± 63 vs. 190 ± 64 U.L⁻¹, p = 0.043), muscle soreness perception (34 ± 12 vs. 42 ± 12 mm, p = 0.045), and improved countermovement jump height (30.7 ± 5.5 cm vs. 29.8 ± 5.8 cm, p = 0.014) at 12 h after the afternoon session. Conclusion: Four days of HRW supplementation is a promising hydration strategy for promoting muscle recovery after two strenuous training sessions performed on the same day in elite fin swimmers. Clinical Trial Registration: clinicaltrials.gov, identifier NCT05799911
Article
Full-text available
Aim To evaluate the real-life effectiveness and safety of hydrogen inhalation (HI) therapy as an additional treatment in Chinese adults with hypertension. Methods This observational, retrospective clinical study included hypertensive patients receiving routine antihypertensives with or without HI initiation from 2018 to 2023. Participants were assigned to the HI group or non-HI group (control group) after propensity score matching. The changes in mean systolic blood pressure (SBP) level during the 24-week follow-up period in different groups were examined primarily. The secondary outcome was the changes in diastolic blood pressure (DBP) and blood pressure (BP) control rate during the study. Several subgroup and sensitivity analyses were performed to confirm the robustness of our main findings. Adverse event (AE) was also assessed in patients of both groups. Results In total, we selected 2,364 patients into the analysis. Both mean SBP and DBP levels significantly decreased in the HI group compared to control group at each follow-up visit with the between group difference of −4.63 mm Hg (95% CI, −6.51 to −2.74) at week 8, −6.69 mm Hg (95% CI, −8.54 to −4.85) at week 16, −7.81 mm Hg (95% CI, −9.57 to −6.04) at week 24 for SBP, and −1.83 mm Hg (95% CI, −3.21 to −0.45) at week 8, −2.57 mm Hg (95% CI, −3.97 to −1.17) at week 16, −2.89 mm Hg (95% CI, −4.24 to −1.54) at week 24 for DBP. Patients in the HI group were more likely to attain controlled BP at the follow-up period with odds ratio of 1.44 (95% CI, 1.21–1.72) at week 8, 1.90 (95% CI, 1.59–2.27) at week 16, and 2.24 (95% CI, 1.87–2.68) at the end. The trends of subgroup and sensitivity analyses were mostly consistent with the main analysis. The incidences of AEs were similar between the HI group and control group with all p-value >0.05. Conclusion The HI therapy is related to significant amelioration in BP levels with acceptable safety profile in Chinese hypertensive adults after 24 weeks of treatment, building a clinical ground for further research to evaluate the antihypertensive effect of HI therapy.
Article
Hydrogen, as an antioxidant, may have the potential to mitigate fatigue and improve selected oxidative stress markers induced by strenuous exercise. This study focused on previously unexplored approach of pre-exercise inhalation of hydrogen-rich gas (HRG). Twenty-four healthy adult men first completed prelaboratories to determine maximum cycling power (Wmax) and maximum cycling time (Tmax). Then they were subjected to ride Tmax at 80% Wmax on cycle ergometers after inhaled HRG or placebo gas (air) for 60-minute in a double-blind, counterbalanced, randomized, and crossover design. The cycling frequency in the fatigue modelling process and the rating of perceived exertion (RPE) at the beginning and end of the ride were recorded. Before gas inhalation and after fatigue modeling, visual analog scale (VAS) for fatigue and counter-movement jump (CMJ) were tested, and blood samples were obtained. The results showed that compared to placebo, HRG inhalation induced significant improvement in VAS, RPE, the cycling frequency in the last 30 seconds, the ability to inhibit hydroxyl radicals, and serum lactate after exercise (p < 0.028), but not in CMJ height and glutathione peroxidase activit. In conclusions, HRG inhalation prior to acute exercise can alleviate exercise-induced fatigue, maintain functional performance, and improve hydroxyl radical and lactate levels.
Chapter
Hydrogen gas has garnered significant attention in recent years due to its remarkable antioxidant and anti-inflammatory properties. However, the extensive research on hydrogen’s applications in the energy sector often overshadows its potential as a medically and biologically active gas. Surprisingly, investigations into the biomedical aspects of H2 trace back to as early as 1793. Hydrogen exhibits exceptional pharmacokinetics, swiftly traversing cellular biomembranes, including the blood–brain and testes barriers, to access subcellular organelles. Following ingestion, hydrogen follows the path of least resistance through the circulatory system and is primarily eliminated through exhalation. Despite the intricate molecular mechanisms and precise targets remaining elusive, the antioxidant effects of hydrogen involve the upregulation of endogenous antioxidants via the activation of the Nrf2/keap1 pathway. Recent research highlights the potential role of Fe-porphyrin as a redox-related biosensor, facilitating hydrogen’s reactions with hydroxyl radicals and triggering additional signal transduction processes. Moreover, this review delves into the physicochemical properties of hydrogen, particularly emphasizing its molar solubility, considerations regarding the term saturation, and other unique characteristics of H2 are discussed. The expanding knowledge and research surrounding the history of H2 underscore its transformative potential in biomedical applications and pave the way for future advancements in harnessing its therapeutic properties.
Chapter
Molecular hydrogen (H2) is well known as a colorless gas, and water enriched with H2 (HRW) is an innovative, beneficial beverage for human health that improves gut microbiota management and the viability of the intestinal. Drinking water enriched with hydrogen has been found to have therapeutic effects on inflammatory bowel diseases (IBDs). The low molecular weight of H2 enables it to easily diffuse and permeate cell membranes to exert various biological effects. In addition, H2 may control the immune system, antioxidant and anti-inflammatory activities (metabolism of mitochondrial energy), and cell death processes (apoptosis, autophagy, and pyroptosis) by reducing excessive reactive oxygen species generation and altering nuclear transcription factors. The fundamental mechanism of H2 is still not fully understood. Given its safety and possible usefulness, H2 has a promising future as a treatment for a variety of illnesses, including IBD. This review aimed to comprehensively highlight the current knowledge in the fields of H2 function in antioxidative, anti-inflammatory, and anti-apoptotic effects as well as its underlying mechanism, with a focus on IBD, and also offer recommendations for using H2 medically to treat IBD.
Chapter
Molecular hydrogen (H2), supplied either as a gas or in a solution, has been gaining popularity as a treatment for a variety of conditions and diseases. For example, it has been suggested to be beneficial for neurodegenerative diseases, to ease the injuries caused by restoration of blood flow to previously ischaemic tissues, and even to alleviate the symptoms of COVID-19. It has also been suggested as an ergogenic sports supplement. However, the exact mode of action of H2 has yet to be definitively unravelled. It has been suggested that H2 acts as an antioxidant and, in particular, as a scavenger of hydroxyl radicals (·OH). This might be the case, but it is unlikely that this is the only mode of action of H2 in biological systems. Here we discuss some of the possible mechanisms by which H2 may have an effect, which may explain how it is acting in a medical context.
Article
Full-text available
Acute physical exercise increases reactive oxygen species in skeletal muscle, leading to tissue damage and fatigue. Molecular hydrogen (H2) acts as a therapeutic antioxidant directly or indirectly by inducing antioxidative enzymes. Here, we examined the effects of drinking H2 water (H2-infused water) on psychometric fatigue and endurance capacity in a randomized, double-blind, placebo-controlled fashion. In Experiment 1, all participants drank only placebo water in the first cycle ergometer exercise session, and for comparison they drank either H2 water or placebo water 30 min before exercise in the second examination. In these healthy non-trained participants (n = 99), psychometric fatigue judged by visual analogue scales was significantly decreased in the H2 group after mild exercise. When each group was divided into 2 subgroups, the subgroup with higher visual analogue scale values was more sensitive to the effect of H2. In Experiment 2, trained participants (n = 60) were subjected to moderate exercise by cycle ergometer in a similar way as in Experiment 1, but exercise was performed 10 min after drinking H2 water. Endurance and fatigue were significantly improved in the H2 group as judged by maximal oxygen consumption and Borg’s scale, respectively. Taken together, drinking H2 water just before exercise exhibited anti-fatigue and endurance effects.
Article
Full-text available
Cardiovascular diseases are the most common causes of morbidity and mortality worldwide. Redox dysregulation and a dyshomeostasis of inflammation arise from, and result in, cellular aberrations and pathological conditions, which lead to cardiovascular diseases. Despite years of intensive research, there is still no safe and effective method for their prevention and treatment. Recently, molecular hydrogen has been investigated in preclinical and clinical studies on various diseases associated with oxidative and inflammatory stress such as radiation-induced heart disease, ischemia-reperfusion injury, myocardial and brain infarction, storage of the heart, heart transplantation, etc. Hydrogen is primarily administered via inhalation, drinking hydrogen-rich water, or injection of hydrogen-rich saline. It favorably modulates signal transduction and gene expression resulting in suppression of proinflammatory cytokines, excess ROS production, and in the activation of the Nrf2 antioxidant transcription factor. Although H 2 appears to be an important biological molecule with anti-oxidant, anti-inflammatory, and anti-apoptotic effects, the exact mechanisms of action remain elusive. There is no reported clinical toxicity; however, some data suggests that H 2 has a mild hormetic-like effect, which likely mediate some of its benefits. The mechanistic data, coupled with the pre-clinical and clinical studies, suggest that H 2 may be useful for ROS/inflammation-induced cardiotoxicity and other conditions.
Article
Full-text available
Background Clinical studies have reported hydrogen-rich water (HRW) to have therapeutic and ergogenic effects. The aim of this study was to determine the effect of acute supplementation with HRW on exercise performance as measured by VO2, respiratory exchange ratio (RER), heart rate (HR), and respiratory rate (RR). Methods Baseline levels of all exercise indices were determined in nineteen (4 female, 23.4 ± 9.1 yr; 15 male, 30.5 ± 6.8 yr) healthy subjects using a graded treadmill exercise test to exhaustion. Each subject was examined two additional times in a randomized double-blinded, placebo-controlled crossover fashion. Subjects received either HRW or placebo, which was consumed the day before and the day of the testing. HRW was delivered using the hydrogen-producing tablets, DrinkHRW (5 mg of H2). All data was analyzed with SPSS using pairwise comparisons with Bonferroni adjustment. Results HRW supplementation did not influence maximal or minimal indices of exercise performance (VO2, RER, HR and RR) (p < 0.05). However, HRW significantly decreased average exercising RR and HR (p < 0.05). HRW decreased exercising HR during minutes 1–9 of the graded exercise test (121 ± 26 bpm) compared to placebo (126 ± 26 bpm) and baseline (124 ± 27 bpm) (p < 0.001) without substantially influencing VO2. Conclusion Acute supplementation of DrinkHRW tablets may benefit submaximal aerobic exercise performance by lowering exercising HR. Further studies are needed to determine the influence and practical significance of HRW on varying exercise intensities as well as optimal dosing protocols and the effects of chronic use.
Article
Full-text available
microRNAs (miRNAs) constitute a large class of post-transcriptional regulators of gene expression. It has been estimated that miRNAs regulate up to 30% of the protein-coding genes in humans. They are implicated in many physiological and pathological processes, including those involved in radiation-induced heart damage. Biomedical studies indicate that molecular hydrogen has potential as a radioprotective agent due to its antioxidant, anti-inflammatory, and signal-modulating effects. However, the impact of molecular hydrogen on the expression of miRNAs in the heart after irradiation has not been investigated. This study aimed to explore the involvement of miRNA-1, -15b, and -21 in the protective action of molecular hydrogen on rat myocardium damaged by irradiation. The results showed that the levels of malondialdehyde (MDA) and tumor necrosis factor alpha (TNF-α) increased in the rat myocardium after irradiation. Treatment with molecular hydrogen-rich water (HRW) reduced these values to the level of non-irradiated controls. miRNA-1 is known to be involved in cardiac hypertrophy, and was significantly decreased in the rat myocardium after irradiation. Application of HRW attenuated this decrease in all evaluated time periods. miRNA-15b is considered to be anti-fibrotic, anti-hypertrophic, and anti-oxidative. Irradiation downregulated miRNA-15b, whereas administration of HRW restored these values. miRNA-21 is connected with cardiac fibrosis. We observed significant increase in miRNA-21 expression in the irradiated rat hearts. Molecular hydrogen lowered myocardial miRNA-21 levels after irradiation. This study revealed for the first time that the protective effects of molecular hydrogen on irradiation-induced heart damage may be mediated by regulating miRNA-1, -15b, and -21.
Article
Full-text available
Hyposmia is one of the earliest and the most common symptoms in Parkinson’s disease (PD). The benefits of hydrogen water on motor deficits have been reported in animal PD models and PD patients, but the effects of hydrogen gas on PD patients have not been studied. We evaluated the effect of inhalation of hydrogen gas on olfactory function, non-motor symptoms, activities of daily living, and urinary 8-hydroxy-2′-deoxyguanine (8-OHdG) levels by a randomized, double-blinded, placebo-controlled, crossover trial with an 8-week washout period in 20 patients with PD. Patients inhaled either ~1.2–1.4% hydrogen-air mixture or placebo for 10 minutes twice a day for 4 weeks. Inhalation of low dose hydrogen did not significantly influence the PD clinical parameters, but it did increase urinary 8-OHdG levels by 16%. This increase in 8-OHdG is markedly less than the over 300% increase in diabetes, and is more comparable to the increase after a bout of strenuous exercise. Although increased reactive oxygen species is often associated with toxicity and disease, they also play essential roles in mediating cytoprotective cellular adaptations in a process known as hormesis. Increases of oxidative stress by hydrogen have been previously reported, along with its ability to activate the Nrf2, NF-κB pathways, and heat shock responses. Although we did not observe any beneficial effect of hydrogen in our short trial, we propose that the increased 8-OHdG and other reported stress responses from hydrogen may indicate that its beneficial effects are partly or largely mediated by hormetic mechanisms. The study was approved by the ethics review committee of Nagoya University Graduate School of Medicine (approval number 2015-0295). The clinical trial was registered at the University Hospital Medical Information Network (identifier UMIN000019082).
Article
Full-text available
Expending a considerable amount of physical energy inevitably leads to fatigue during both training and competition in football. An increasing number of experimental findings have confirmed the relationship between the generation and clearance of free radicals, fatigue, and exercise injury. Recently, hydrogen was identified as a new selective antioxidant with potential beneficial applications in sports. The present study evaluated the effect of 2-month consumption of hydrogen-rich water on the gut flora in juvenile female soccer players from Suzhou. As demonstrated by enzyme linked immunosorbent assay and 16S rDNA sequence analysis of stool samples, the consumption of hydrogen-rich water for two months significantly reduced serum malondialdehyde, interleukin-1, interleukin-6, tumour necrosis factor-α levels; then significantly increased serum superoxide dismutase, total antioxidant capacity levels and haemoglobin levels of whole blood. Furthermore, the consumption of hydrogen-rich water improved the diversity and abundance of the gut flora in athletes. All examined indices, including the shannon, sobs, ace, and chao indices, were higher in the control group than those proposed to result from hydrogen-rich water consumption prior to the trial, but these indices were all reversed and were higher than those in the controls after the 2-month intervention. Nevertheless, there were some differences in the gut flora components of these two groups before the trial, whereas there were no significant changes in the gut flora composition during the trial period. Thus, the consumption of hydrogen-rich water for two months might play a role modulating in the gut flora of athletes based on its selective antioxidant and anti-inflammatory activities. The study protocol was approved by the ethics committee of the Suzhou Sports School (approved number: SSS-EC150903).
Article
Full-text available
Uncontrolled production of oxygen and nitrogen radicals results in oxidative and nitrosative stresses that impair cellular functions and have been regarded as causative common denominators of many pathological processes. In this review, we report on the beneficial effects of molecular hydrogen in scavenging radicals in an artificial system of •OH formation. As a proof of principle, we also demonstrate that in rat hearts in vivo, administration of molecular hydrogen led to a significant increase in superoxide dismutase as well as pAKT, a cell survival signaling molecule. Irradiation of the rats caused a significant increase in lipid peroxidation, which was mitigated by pre-treatment of the animals with molecular hydrogen. The nuclear factor erythroid 2-related factor 2 is regarded as an important regulator of oxyradical homeostasis, as well as it supports the functional integrity of cells, particularly under conditions of oxidative stress. We suggest that the beneficial effects of molecular hydrogen may be through the activation of nuclear factor erythroid 2-related factor 2 pathway that promotes innate antioxidants and reduction of apoptosis, as well as inflammation.
Article
Full-text available
PURPOSE This study aimed to investigate whether hydrogen (H2) water drinking reduced oxidative damage products (8-hydroxy-2-deoxyguanosine [8-oxo-dG], malondialdehyde [MDA], L-lactate) produced in blood after exercise-induced oxidative stress. METHODS Men (n=7) and women (n=8) (systolic blood pressure [BP]: 90-150 mmHg, diastolic BP: 60-100 mmHg) were recruited. Exercise was performed for 20 minutes (8 km/hr) using treadmill. Placebo or test group drunk water and hydrogen water (1.2-1.5 ppm, 1,000 mL), respectively, right after exercise. Three days after, the same experiment was performed in the cross-over manner. Blood samples were obtained 30 minutes before exercise, right after exercise, and 30, 60, 120 minutes after exercise. 8-oxo-dG, MDA and L-lactate in serum were determined using ELISA kit. RESULTS The blood levels of 8-oxo-dG were significantly increased after exercise (from 381±9.4 to 524±14.9 ng/mL for male; from 367±13.5 to 509±10.7 ng/mL for female). In the placebo group, the increased 8-oxo-dG at each time was not significantly different from the peak level induced by the exercise except that at 120 minutes in female. In the test group, its levels at each time were significantly lower than that in the peak level. The effect of hydrogen water was more pronounced in female than in male. The results obtained with MDA were quite similar to those with 8-oxo-dG. The L-lactate levels were not significant statistically in both groups. CONCLUSIONS The data suggest that the one time drinking of hydrogen water may suppress the exercise-induced oxidative stress.
Article
Full-text available
The purpose of this study was to investigate the involvement of neutrophil dynamics and function in exercise-induced muscle damage (EIMD) and delayed-onset muscle soreness (DOMS), and the effect of molecular hydrogen (H2) intake on these parameters. Nine healthy and active young men performed H2 and placebo bath trial in a crossover design. They carried out downhill running (−8% slope) for 30 min at a speed corresponding to 75~85% of peak oxygen uptake (VO2peak). Subsequently, they repeated bathing for 20 min per day for one week. Degree of muscle soreness (visual analogue scale: VAS), peripheral leukocyte counts, neutrophil dynamics and function, muscle damage, and inflammation markers were measured. Plasma interleukin (IL)-6 concentration was significantly correlated with peripheral neutrophil count, VAS, and serum creatine kinase activity, respectively, after downhill running. Peripheral neutrophil count and serum myoglobin concentration were also significantly correlated. Conversely, there were no effects of H2 bath. These results suggest that IL-6 may be involved in the mobilization of neutrophils into the peripheral blood and subsequent EIMD and DOMS after downhill running; however, it is not likely that H2 bath is effective for the inflammatory process that was centered on neutrophils after downhill running.
Article
Physical exercise induces inflammatory and oxidative markers production in the skeletal muscle and this process is under the control of both endogenous and exogenous modulators. Recently, molecular hydrogen (H 2 ) has been described as a therapeutic gas able to reduced oxidative stress in a number of conditions. However, nothing is known about its putative role in the inflammatory and oxidative status during a session of acute physical exercise in sedentary rats. Therefore, we tested the hypothesis that H 2 attenuates both inflammation and oxidative stress induced by acute physical exercise. Rats ran at 80% of their maximum running velocity on a closed treadmill inhaling either the H 2 gas (2% H 2 , 21% O 2 , balanced with N 2 ) or the control gas (0% H 2 , 21% O 2 , balanced with N 2 ) and were euthanized immediately or 3h after exercise. We assessed plasma levels of inflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6] and oxidative markers [superoxide dismutase (SOD), thiobarbituric acid reactive species (TBARS) and nitrite/nitrate (NOx)]. In addition, we evaluated the phosphorylation status of intracellular signaling proteins [glycogen synthase kinase type 3 (GSK3α/β) and the cAMP responsive element binding protein (CREB)] that modulate several processes in the skeletal muscle during exercise, including changes in exercise-induced reactive oxygen species (ROS) production. As expected, physical exercise increased virtually all the analyzed parameters. In the running rats, H 2 blunted exercise-induced plasma inflammatory cytokines (TNF-α and IL-6) surges. Regarding the oxidative stress markers, H 2 caused further increases in exercise-induced SOD activity and attenuated the exercise-induced increases in TBARS 3h after exercise. Moreover, GSK3α/β phosphorylation was not affected by exercise or H 2 inhalation. Otherwise, exercise caused an increased CREB phosphorylation which was attenuated by H 2 . These data are consistent with the notion that H 2 plays a key role in decreasing exercise-induced inflammation, oxidative stress, and cellular stress.