Next Article in Journal
Distinct Roles of NANOS1 and NANOS3 in the Cell Cycle and NANOS3-PUM1-FOXM1 Axis to Control G2/M Phase in a Human Primordial Germ Cell Model
Next Article in Special Issue
Acetylated Tau Protein: A New Piece in the Puzzle between Brain Ischemia and Alzheimer’s Disease
Previous Article in Journal
Large Benefit from Simple Things: High-Dose Vitamin A Improves RBP4-Related Retinal Dystrophy
Previous Article in Special Issue
Relationship between Neuronal Damage/Death and Astrogliosis in the Cerebral Motor Cortex of Gerbil Models of Mild and Severe Ischemia and Reperfusion Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Hydrogen Neuroprotection in Post-Ischemic Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy: Underlying Mechanisms and Potential for Clinical Implementation—Fantasy or Reality?

by
Ryszard Pluta
1,*,
Sławomir Januszewski
1 and
Stanisław J. Czuczwar
2
1
Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
2
Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(12), 6591; https://doi.org/10.3390/ijms23126591
Submission received: 12 May 2022 / Revised: 10 June 2022 / Accepted: 11 June 2022 / Published: 13 June 2022
(This article belongs to the Special Issue Ischemic Brain Neurodegeneration 2.0)

Abstract

:
Currently, there is a lot of public interest in naturally occurring substances with medicinal properties that are minimally toxic, readily available and have an impact on health. Over the past decade, molecular hydrogen has gained the attention of both preclinical and clinical researchers. The death of pyramidal neurons in especially the CA1 area of the hippocampus, increased permeability of the blood-brain barrier, neuroinflammation, amyloid accumulation, tau protein dysfunction, brain atrophy, cognitive deficits and dementia are considered an integral part of the phenomena occurring during brain neurodegeneration after ischemia. This review focuses on assessing the current state of knowledge about the neuroprotective effects of molecular hydrogen following ischemic brain injury. Recent studies in animal models of focal or global cerebral ischemia and cerebral ischemia in humans suggest that hydrogen has pleiotropic neuroprotective properties. One potential mechanism explaining some of the general health benefits of using hydrogen is that it may prevent aging-related changes in cellular proteins such as amyloid and tau protein. We also present evidence that, following ischemia, hydrogen improves cognitive and neurological deficits and prevents or delays the onset of neurodegenerative changes in the brain. The available evidence suggests that molecular hydrogen has neuroprotective properties and may be a new therapeutic agent in the treatment of neurodegenerative diseases such as neurodegeneration following cerebral ischemia with progressive dementia. We also present the experimental and clinical evidence for the efficacy and safety of hydrogen use after cerebral ischemia. The therapeutic benefits of gas therapy open up new promising directions in breaking the translational barrier in the treatment of ischemic stroke.

1. Introduction

Molecular hydrogen is an odorless, colorless gas that is physiologically inert. Hydrogen is the lightest and most abundant element in the Earth’s atmosphere. Furthermore, hydrogen is a biological gas that is produced in mammals by intestinal bacteria [1,2,3]. It is believed to be a new type of natural antioxidant with a low ability to react with most biomolecules, which is potentially therapeutic. The first use of hydrogen in humans was hydreliox, a breathing gas mixture of hydrogen, helium and oxygen that is used to prevent decompression sickness and nitrogen narcosis during very deep technical dives [4]. The therapeutic use of hydrogen was first demonstrated in 1975, showing that hyperbaric hydrogen caused marked regression of skin squamous carcinoma in mice [5]. In 2001, hydrogen was documented to have anti-inflammatory properties in experimental parasitic hepatitis [6]. Six years later, it was found that hydrogen was supposed to act as an antioxidant by selectively removing the neurotoxic hydroxyl radical and peroxynitrite from the parenchyma of the rat brain as a result of oxidative stress caused by ischemic brain injury [7]. Hydrogen has been shown to have many advantages as a neuroprotective gas. First, it can penetrate biomembranes, diffuse into the cytosol and organelles and cross the blood-brain barrier [7,8,9]. Secondly, the repeated administration of hydrogen does not cause tolerance [10]. Thirdly, different easy and convenient approaches to administer it are available [11,12]. Fourthly, hydrogen has a protective effect against many diseases, including peripheral and central nervous system diseases such as neuropathic pain, Alzheimer’s disease, stroke, animal cerebral ischemia and neonatal hypoxic-ischemic brain injury [11,13,14,15,16,17,18,19,20,21,22,23]. Fifth, there are no documented serious side effects [13,24]. Sixth, in Japan, 2% molecular hydrogen inhalation has been approved for the clinical treatment of cerebral ischemia due to cardiac arrest [24,25,26,27,28]. The Chinese National Health and Medical Commission in 2020 recommended the use of inhaled hydrogen in addition to oxygen therapy for anti-cancer, anti-inflammatory and anti-oxidant treatments [29]. Hydrogen has been suggested as a new complementary therapy against stroke, which, e.g., reduces oxidative stress, neuroinflammation and apoptosis [22,30,31,32]. Despite many inaccuracies, the selective ability to scavenge free radicals and heal inflammation are still widely accepted mechanisms of the action of hydrogen [32]. Clinical trials have shown that hydrogen treatment is safe and effective in patients with asthma and chronic obstructive pulmonary disease [33,34].
Recently, it has been proposed to prevent and treat coronavirus disease by inhaling oxygen mixed with hydrogen (33.4% oxygen and 66.6% hydrogen) due to the important role of hydrogen in alleviating the worsening of lung function, emphysema, and acute or chronic inflammation [29,35,36]. Thus, molecular hydrogen is an interesting potential therapeutic gas for the prevention and treatment of various diseases, including neurological disorders. In this way, hydrogen contributes to the further development of the healthcare industry by using its healing properties. In this review, we summarized the current understanding of the potential neuroprotective properties of molecular hydrogen towards global and focal cerebral ischemia and the possible molecular mechanisms associated with its beneficial activity.

2. Search Criteria and Data Collection

Published scientific papers on the use of molecular hydrogen have been screened for in vitro, in vivo, animal and clinical investigations and side effects. Searches were performed digitally using databases including PubMed, MEDLINE, SCOPUS, Google Scholar and Science Direct to identify peer-reviewed original articles and reviews over the past twenty years (1 January 2001–31 December 2021). The search strategy was carried out using the following key words: “Molecular hydrogen therapy and brain ischemia”, “brain ischemia and molecular hydrogen therapy”, “molecular hydrogen therapy and ischemic stroke”, “ischemic stroke and molecular hydrogen therapy”, “molecular hydrogen therapy and cardiac arrest”, “cardiac arrest and molecular hydrogen therapy”, “molecular hydrogen neuroprotection and brain ischemia”, “molecular hydrogen neuroprotection and ischemic stroke”, “ischemic stroke and molecular hydrogen neuroprotection”, “molecular hydrogen therapy and amyloid”, “molecular hydrogen therapy and tau protein”, “tau protein and molecular hydrogen therapy“, “molecular hydrogen and bioavailability”, “molecular hydrogen and side effects”. The work covered by the study had to be related to the search terms and be as up-to-date as possible. The excluded works did not include hydrogen compounds, including hydrogen sulfide and numerous works by one author or from the same laboratory, in this case the latest works were taken. A total of 575 original papers and reviews were found, and 109 publications closely related to the subject of the review were used.

3. Molecular Hydrogen Neuroprotection in Post-Ischemic Brain Injury

It is now known that neurodegeneration following cerebral ischemia is caused by numerous proteomic and genomic changes that lead to neuronal death by necrosis and apoptosis, with progressive brain neuroinflammation and atrophy, ultimately leading to full-blown dementia [37,38,39,40,41,42,43,44,45,46,47,48,49]. Research indicates that, after cerebral ischemia, neurodegeneration of the Alzheimer’s disease type develops with selective neuronal death in the hippocampus with its complete atrophy [50,51,52,53,54,55]. The development of neuroinflammatory lesions has been shown to play a key role in the progression of post-ischemic brain neurodegeneration [39,44,48]. Amyloid processing, tau protein modification, autophagy and mitophagy genes are involved in post-ischemic neurodegeneration in the same way as in Alzheimer’s disease [40,42,49,56,57,58,59,60,61,62,63,64,65,66,67].
Restoration of blood flow in the brain following cerebral ischemia and ischemic stroke triggers an outbreak of reactive oxygen species, triggering a neuroinflammatory response and oxidative damage [39,44,48,65]. Reactive oxygen species destroy the membranes of neuronal and neuroglial cells, inducing lipid peroxidation, so antioxidants come into play as a therapeutic option [65]. Molecular hydrogen has been recognized as an antioxidant that can buffer the destructive effects of oxidative stress in the brain following ischemia by selectively reducing cytotoxic reactive oxygen species [7,22,30].

3.1. In Animals

In mice with focal cerebral ischemia with reperfusion, molecular hydrogen significantly increases SOD and GSH-Px activity, reduces malondialdehyde levels and infarct volume, relieves cerebral edema, improves neurological outcomes and alleviates cognitive deficits (Table 1) [19,68,69]. In global cerebral ischemia caused by cardiac arrest in rats, hydrogen inhalation improves neurological outcomes, cognitive deficits and survival [20,70]. Hydrogen injection or inhalation after global cerebral ischemia due to cardiac arrest effectively controls neuronal death and microglia activation in the hippocampus and lowers serum levels of S100b protein (Table 1) [8,31,71]. Inhalation of hydrogen or in combination with hypothermia has been shown to be superior to hypothermia alone in global cerebral ischemia from cardiac arrest in rats [31,71,72]. Molecular hydrogen has been shown to protect the permeability of the blood-brain barrier after focal and global cerebral ischemia (Table 1) [8,9]. Hydrogen has been shown to protect against oxidative stress, neuroinflammation, and prevents the ischemic site from turning into a hemorrhagic focus in rats with local cerebral ischemia (Table 1) [73]. Additionally, it has been shown that the intraperitoneal injection of hydrogen-rich saline has healing properties after transient global cerebral ischemia in rats [74]. Since most of the damage in the above model occurs between 6 and 24 h after ischemia, the effective hydrogen protection period was much less than the 6 h of recirculation, so the protective effect of hydrogen-rich saline in this situation is quite limited [74]. Hydrogen treatment of mice after bilateral closure of the common carotid artery improves cognitive abilities and induces anti-apoptotic and antioxidant effects [75]. In rats, up to 7 days after middle cerebral artery occlusion with reperfusion and administration of hydrogen, a reduction in infarct volume, ischemic penumbra hyperperfusion, neurological and behavioral disorders and white matter damage were observed (Table 1) [76]. It was documented that hydrogen therapy significantly improved the 7-day survival rate of mice after global brain ischemia, from 8.3 to 50% [77]. Histopathological analysis revealed that hydrogen therapy significantly attenuated neuronal injury and autophagy in the hippocampal CA1 area and also brain edema, after 24 h of reperfusion [77]. The beneficial effects of hydrogen therapy on post-ischemic brain injury were associated with significantly lower levels of oxidative stress markers: malondialdehyde and 8-hydroxy-2′-deoxyguanosine in the brain parenchyma [77]. Hydrogen inhalation following 10-min transient global cerebral ischemia in rats that survived 3 days attenuated cognitive impairment [78]. This neuroprotective effect was associated with decreased pyramidal neuronal death in the CA1 region of the hippocampus and inhibition of oxidative stress [78]. Hydrogen inhalation improved survival and neurological deficit after global ischemia caused by cardiac arrest in rats [31]. It also prevented the increase in left ventricular end-diastolic pressure and the increase in serum IL-6 levels, and reduced mortality [31,79,80]. Hydrogen treatment increased the level of interleukin-10, vascular endothelial growth factor and leptin [81]. In addition, a reduction in mortality in rats after cardiac arrest and an effect on the restoration of the bioelectrical activity of the brain was noted [81]. The survival rate at 4 h was 78% in the hydrogen group and 22% in the placebo group [81]. In another global model of cerebral ischemia due to cardiac arrest in rats, increased survival and inhibition of autophagy were observed [82]. Hydrogen inhalation for 4 days improves neurological outcomes and survival after global cerebral ischemia due to cardiac arrest in systemic hypertension rats and is superior to treatment with mild hypothermia [71]. The intraperitoneal injection of hydrogen into rabbits in cardiac arrest improved 3-day survival and neurological deficits, reduced neuronal damage, and inhibited neuronal apoptosis [83]. The intraperitoneal injection of hydrogen decreased the indicators of oxidative stress in the blood and the parenchyma of the hippocampus and increased the activity of the antioxidant enzyme [83]. Rats given hydrogen-rich water before and after occlusion of the middle cerebral artery, surviving up to 14 days after focal ischemia, showed reduced infarct volume and improved neurological outcomes [84]. In addition, hydrogen prevented ischemia-induced decreases in parvalbumin and hypocalcin, and also reduced neuronal cell death induced by toxic glutamate [84]. In addition, hydrogen lowered the increased levels of intracellular Ca2+ caused by glutamate toxicity [84].
Subsequent studies found that rats that underwent global cerebral ischemia and were treated with hydrogen-rich saline had milder neuronal injury and a limited number of irreversibly damaged neurons in the brain [15]. Expression of miR-210, miR21 and NF-κB in the ischemic hippocampus at 6, 24 and 96 h was significantly reduced in the hydrogen-treated group [15]. Moreover, the number of Tregs cells after cerebral ischemia treated with hydrogen increased on days one and four after reperfusion [15]. These results indicate that the recovery from global cerebral ischemia in rats dosed with hydrogen-rich saline is most likely associated with an upregulation of Treg cell numbers [15]. In this study, hydrogen significantly increased the number of surviving ischemic pyramidal neurons in the CA1 region of the hippocampus [15]. In addition, the neurobehavioral test confirmed that hydrogen reduced damage to the brain after ischemia [15]. This neuroprotective effect may be due to the extensive spread of hydrogen throughout the brain. Hydrogen has a good diffusion rate, can easily penetrate the blood-brain barrier and reach deep brain structures, and is also able to reach the site of injury before revascularization to remove toxic oxygen free radicals [85].
In behavioral studies of rats after focal cerebral ischemia, administration of lactulose, which induces endogenous hydrogen production in the intestine, resulted in higher neurological scores and shorter escape latency in the Morris test [86]. Morphological studies using 2,3,5-triphenyltetrazolium chloride showed a smaller infarct volume, Nissel staining showed relatively distinct and intact neuronal cells and TUNEL staining showed fewer apoptotic neurons [86]. In biochemical studies, lactulose decreased the content of malondialdehyde in the brain, the activity of caspase-3, the concentration of 3-nitrotyrosine and 8-hydroxy-2-deoxyguanosine and increased the activity of superoxide dismutase [86]. Orally administered lactulose activated expression of NF-E2 related factor 2 (Nrf2) in the brain [86]. The antibiotics suppressed the neuroprotective effects of lactulose by reducing hydrogen generation [86]. Lactulose administered intragastrically had a neuroprotective effect in post-ischemic brain injury in rats, which is attributed to the production of hydrogen via fermentation of lactulose by intestinal bacteria and activation of Nrf2 [86].
Studies that assessed the most effective timing of hydrogen administration after local cerebral ischemia in rats showed a post-ischemic time interval of up to 6 h during which significant reductions in infarct volume and brain edema were observed, and neurological outcomes improved [87]. At that time, after local cerebral ischemia, hydrogen decreased 8-hydroxyl-2′-deoxyguanosine (8-OHdG), reduced the content of malondialdehyde, interleukin-1β, tumor necrosis factor-α and suppressed caspase 3 activity [87]. These results indicate that hydrogen has a neuroprotective effect when administered during 6 h post-ischemia [87].
One study used hydrogen-saturated saline for focal cerebral ischemia in rats to test whether hydrogen-saturated saline reduces apoptosis of neuronal cells through the p38 MAPK-caspase-3 signaling pathway [88]. The obtained data showed that hydrogen reduced apoptotic neuronal cell death and infiltration of inflammatory cells in the brain cortex of rats post-ischemia [88]. In the hydrogen-treated group, there was a significant decrease in p38 MAPK protein expression compared to the untreated group [88]. It was concluded that hydrogen-rich saline could exert anti-apoptotic neuroprotective effects via the p38 MAPK signaling pathway [88]. It has also been found that, in acute post-ischemic brain injury in mice, hydrogen reduces the levels of Bax and TNFα and induces an anti-inflammatory response by regulating IL-2 and IL-10 [75].

3.2. In Humans

Studies in patients with acute focal cerebral ischemia have shown that administration of molecular hydrogen by inhalation or intravenous infusion is safe, hydrogen has been found in the blood, and there was no effect of hydrogen on physiological parameters [13,25,89]. Patients with local cerebral infarction who received hydrogen inhalation as part of their treatment had a reduced infarct size, improved neurological outcomes and the ability to perform daily activities compared to untreated patients (Table 2) [16]. In a human study of global cerebral ischemia due to cardiac arrest, inhalation of low concentration molecular hydrogen positively affected brain function without adverse events (Table 2) [25]. In Japan, a phase II clinical trial was conducted in patients with cerebral ischemia after cardiac arrest, finding a positive effect on neurological functioning and the safety of molecular hydrogen inhalation (Table 2) [25,27]. While no side effects of hydrogen have been detected in animal studies, potential side effects should still be investigated due to diarrhea reported by a small number of patients after receiving hydrogen [89].

4. Molecular Hydrogen versus Amyloid and Tau Protein Modification

It is now well known that ischemic neurodegeneration of the brain is caused by a set of proteomic and genetic changes that lead to the death of neuronal cells in an amyloid and tau protein dependent manner [40,46,47,56,57,58,59,63,64]. Amyloid causes oxidative stress with progressive neuroinflammation leading to brain atrophy and dementia [38,39,42,43,44,46,48,49,50,52,53,65]. Hard data show that cerebral ischemia in animals and humans leads to the production and accumulation of amyloid in the form of diffuse and senile plaques [37,53,95,96,97,98,99]. Direct evidence suggests that tau protein hyperphosphorylation leading to the development of neurofibrillary tangles also plays a role in the development of ischemic neurodegeneration of the brain, such as in Alzheimer’s disease [41,47,100,101]. The signaling mechanisms generated by amyloid and the tau protein following cerebral ischemia are believed to play a critical role in the development of irreversible neurodegeneration and dementia [37,41,47,53,57,58,59,63,100,101].
The amyloid protein precursor was found to be significantly lowered in the brain in a transgenic mouse model of Alzheimer’s disease after treatment with hydrogen-rich water [102]. In these animals, the level of β-secretase was also significantly lowered in the brain after administration of hydrogen-rich water [102]. The result of the above changes was a reduction in amyloid deposition in the CA3 area of the hippocampus [102]. Additionally, the decreased level of the soluble amyloid protein precursor α in this model was partially recovered by the administration of hydrogen-rich water, clearly confirming the neuroprotective effect of hydrogen [102]. Taken together, these results indicate that treatment with hydrogen-rich water prevents proteolysis of the amyloid protein precursor towards amyloid [102].
In the transgenic model of Alzheimer’s disease, intracerebral administration of Pd hydride nanoparticles (a highly charged hydrogen carrier) effectively removes hydroxyl radicals, reduces amyloid production and aggregation, alleviates mitochondrial dysfunction, reverses synaptic deficits and inhibits neuronal death [22]. In vitro, hydrogen treatment enhances the antioxidant system in human SK-N-MC neuroblastoma cells under the influence of amyloid-stimulated oxidative stress through the induction of AMPK and upregulation of the Sirt1-FoxO3a axis, which prevents mitochondrial dysfunction and the generation of reactive oxygen species, thereby ultimately maintaining cells survival [103].
The administration of hydrogen-rich water significantly inhibited the phosphorylation of the tau protein in Ser404 and Ser422 in the transgenic model of Alzheimer’s disease [102]. This resulted in a significant reduction in the number of neurofibrillary tangles in the CA3 region of the mouse hippocampus [102]. These results indicate that treatment with hydrogen-rich water can reduce pathological modifications of the tau protein, which prevents damage to neurons and synapses and memory deficits in the transgenic model of Alzheimer’s disease [102].

5. Molecular Hydrogen Bioavailability

Currently, there are several methods of molecular hydrogen administration, i.e., hydrogen inhalation, infusion of hydrogen-rich saline, hydrogen-rich water provided by various types of molecular hydrogen donors/suppliers, as well as functional micro/nanomaterials that increase the concentration of hydrogen administered and thus the effectiveness of treatment [12,104,105]. In the clinic, the most common methods of administering hydrogen are inhalation, infusion of hydrogen-rich saline and drinking hydrogen-rich water [28]. In clinical conditions, breathing hydrogen gas in concentrations of 1–4% is a hassle-free, direct and easy-to-use method [28]. It should be added that drinking hydrogen-rich water is safer and more convenient to use [28]. In contrast, hydrogen-rich saline is commonly used by intravenous infusion or intraperitoneal injection [28] Drinking hydrogen-rich water causes 59% of the consumed hydrogen to be expelled through the breath, almost 40% is used up in the human body, and about 1% is released through the skin [106]. It has been proven that after inhalation of 3% or 4% hydrogen, its level in venous and arterial blood rises rapidly and reaches a plateau within 20 min [13,24]. After stopping hydrogen inhalation, its level in arterial blood drops below 10% of the plateau level after around 6 min, and in venous blood after around 18 min [13,24]. However, after a 30-min intravenous infusion of hydrogen-rich saline in two volunteers, the levels of hydrogen in the venous and arterial blood rose to a plateau within 15 min and fell sharply after the infusion was stopped [13,24]. Data from clinical and experimental studies have shown that inhaling hydrogen produces higher concentrations of hydrogen in the brain tissue than other ways of its administration [13,24,107]. In order to select the most effective hydrogen treatment for a given disease entity, additional studies of the pharmacokinetics and medical benefits of combining hydrogen with water or other hydrogen carriers are necessary [28].

6. Conclusions

Molecular hydrogen exerts a significant neuroprotective effect on almost all animal models of cerebral ischemia studied. The neuroprotective properties of hydrogen are often documented by assessing neuronal survival and improved neurocognitive function (Figure 1). In addition, its protective properties against mitochondrial damage and the permeability of the blood-brain barrier were noted (Figure 1). Furthermore, studies showed that hydrogen-rich water prevented neuronal death and synaptic loss, inhibited the development of senile amyloid plaques and reduced tau protein hyperphosphorylation and the growth of neurofibrillary tangles in the transgenic model of Alzheimer’s disease (Figure 1). Few side effects indicate that the therapeutic potential of hydrogen is not limited and its neuroprotective effects are pleiotropic. Molecular mechanisms of hydrogen-initiated neuroprotection have been studied in animal focal and global cerebral ischemia demonstrating a reduction in oxidative stress, inhibition of pro-apoptotic effects with simultaneous activation of anti-apoptotic pathways and inhibition of the neuroinflammatory process with a simultaneous increase in the neurotrophic effect of microglia. It is well known that oxidative stress affects the expression of a large number of genes that stimulate many biological phenomena, such as increased production of amyloid, modification of the tau protein, autophagy, apoptosis and neuroinflammation, hence the antioxidant effect of hydrogen may be its most important neuroprotective property (Figure 1). Currently, two proposals for the molecular mechanisms underlying the antioxidant properties of hydrogen are put forward. A widely accepted theory is that hydrogen directly reacts with hydroxyl radicals and peroxynitrite, the traditional scavenger theory [28]. The second theory suggests that hydrogen controls the production of reactive oxygen species by acting as a rectifier for the electron flow in the mitochondria [28]. Although the neuroprotective effects of hydrogen have been demonstrated in the last decade in numerous experimental studies and after preliminary clinical trials, further work is needed in the clinical use of hydrogen. Research has shown that hydrogen is non-toxic but has minor side effects including heartburn, diarrhea and headache, which have been reported in a few cases [89,108]. The distribution of hydrogen in organs and tissues varies with the mode of administration, which impacts the medical benefit. It seems that the limited therapeutic effect of hydrogen may be related to the duration, concentration and method of application, beyond the phase of a specific disease entity. For these reasons, additional studies are required to study the pharmacokinetics and effect of hydrogen dosage and then develop more effective delivery regimens/procedures. Carefully completed translational studies in various models of cerebral ischemia in animals are needed to demonstrate whether the neuroprotective effects of hydrogen on post-experimental cerebral ischemia fully exist, thus paving the way for a design for clinical use.

7. Outlook

Currently, ischemic stroke is one of the most important causes of morbidity and mortality worldwide. At present, tissue plasminogen activator is administered to induce thrombolysis and restore cerebral blood flow following focal brain ischemia, but therapeutic benefit is only achieved in a small percentage of patients qualified for fibrinolysis. Despite the evident preclinical confirmation of the neuroprotective hydrogen treatment of local post-ischemic brain injury, it has not been successfully translated into clinical use. So, the treatment that will please most acute stroke patients remains a mystery. Currently, despite the lack of effective treatment of the consequences of ischemic stroke, research is being carried out on new methods of treating this disease, however, the use of medicinal gases, especially hydrogen, is not very popular. Nevertheless, it should be stated that gases such as normobaric and hyperbaric oxygen and hydrogen exert medical effects in preclinical studies of cerebral ischemia. There are significant advantages in using gases in terms of low cost, high quantity and ease of administration, all of these properties making them ideal candidates for the translational treatment of ischemic stroke. In conclusion, the influence of cellular gaseous mediators such as carbon monoxide, nitric oxide and hydrogen could be an interesting alternative for treating ischemic stroke [1,2,3]. Breathing with these gas mediators may additionally generate neuroprotection, but this plan must be validated as an effective treatment for stroke. This review reveals the neuroprotective potential of hydrogen treatment, supporting the possibility of modulating cellular gas mediators following cerebral ischemia. The benefits of hydrogen therapy open up new promising directions in breaking the translational barrier for brain ischemia in humans.
The evidence introduced in this review shows a promising neuroprotective effect of hydrogen after cerebral ischemia with recirculation (Figure 1). However, the limited number of experimental and clinical studies demonstrating the neuroprotective effects of hydrogen following cerebral ischemia does not provide sufficient scientific support. In addition, recirculation times following cerebral ischemia were short, and therefore the effects of hydrogen treatment related to long-term follow-up following ischemic injury are currently unknown. Therefore, future randomized clinical investigations are needed to confirm the effectiveness of hydrogen therapy and to provide data on some of the currently unresolved issues, such as the timing of hydrogen use. Despite very scarce evidence, the current research on hydrogen in the treatment of cerebral ischemia with reperfusion seems interesting and promising for the future as a neuroprotective molecule and preventing the deposition of various amyloid plaques and dysfunctional tau protein in the form of neurofibrillary tangles (Figure 1). In the last decade, the interest in hydrogen and its reputation for its numerous pharmacological effects has been steadily increasing. Due to the fact that hydrogen, like many other natural substances, has more than one drug target (Figure 1), it indicates its versatile use and low risk of treatment resistance. There is no doubt that, due to preclinical data, the next step in investigating the therapeutic properties of hydrogen must be well-designed and controlled randomized clinical trials. Double-blind research is imperative to finally elucidate the medical properties of hydrogen. A definitive elucidation of the therapeutic benefits of hydrogen may offer hope for the long-term effect of a therapy that is currently a feverish subject of research. Hydrogen is currently not approved for clinical use except in Japan and China [13,24,25,26,27,29]. High bioavailability and negligible side effects of hydrogen administration are an invaluable positive element of its usefulness in the clinic. We hope that future clinical research will help us better understand the medical potential of hydrogen and place this fascinating gas at the forefront of new neuroprotective therapies in neurodegeneration.

Author Contributions

Conceptualization, R.P.; methodology, R.P. and S.J.; software, R.P. and S.J.; validation, R.P. and S.J.C.; formal analysis, R.P. and S.J.C.; investigation, R.P. and S.J.; resources, R.P. and S.J.; data curation, R.P. and S.J.C.; writing—original draft preparation, R.P. and S.J.C.; writing—review and editing, R.P. and S.J.C.; visualization, R.P. and S.J.; supervision, R.P.; project administration, R.P.; funding acquisition, R.P. and S.J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data is available from the author for correspondence.

Acknowledgments

Authors acknowledge support by the Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland (T3-R.P.) and Medical University of Lublin (DS 475/22-S.J.C.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ohta, S. Molecular hydrogen is a novel antioxidant to efficiently reduce oxidative stress with potential for the improvement of mitochondrial diseases. Biochim. Biophys. Acta 2012, 1820, 586–594. [Google Scholar] [CrossRef] [PubMed]
  2. Zhou, J.; Wu, P.-F.; Wang, F.; Chen, J.-G. Targeting gaseous molecules to protect against cerebral ischaemic injury: Mechanisms and prospects. Clin. Exp. Pharmacol. Physiol. 2012, 39, 566–576. [Google Scholar] [CrossRef] [PubMed]
  3. Htun, Y.; Nakamura, S.; Kusaka, T. Hydrogen and therapeutic gases for neonatal hypoxic-ischemic encephalopathy: Potential neuroprotective adjuncts in translational research. Pediatr. Res. 2021, 89, 753–759. [Google Scholar] [CrossRef] [PubMed]
  4. Lanphier, E.H. Human respiration under increased pressures. Symp. Soc. Exp. Biol. 1972, 26, 379–394. [Google Scholar] [PubMed]
  5. Dole, M.; Wilson, F.R.; Fife, W.P. Hyperbaric hydrogen therapy: A possible treatment for cancer. Science 1975, 190, 152–154. [Google Scholar] [CrossRef]
  6. Gharib, B.; Hanna, S.; Abdallahi, O.M.; Lepidi, H.; Gardette, B.; De Reggi, M. Anti-inflammatory properties of molecular hydrogen: Investigation on parasite-induced liver inflammation. Comptes Rendus L’académie Sci.-Ser. III-Sci. Vie 2001, 324, 719–724. [Google Scholar] [CrossRef]
  7. Ohsawa, I.; Ishikawa, M.; Takahashi, K.; Watanabe, M.; Nishimaki, K.; Yamagata, K.; Katsura, K.; Katayama, Y.; Asoh, S.; Ohta, S. Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat. Med. 2007, 13, 688–694. [Google Scholar] [CrossRef]
  8. Huo, T.-T.; Zeng, Y.; Liu, X.-N.; Sun, L.; Han, H.-Z.; Chen, H.-G.; Lu, Z.-H.; Huang, Y.; Nie, H.; Dong, H.-L.; et al. Hydrogen rich saline improves survival and neurological outcome after cardiac arrest and cardiopulmonary resuscitation in rats. Anesth. Analg. 2014, 119, 368–380. [Google Scholar] [CrossRef]
  9. Takeuchi, S.; Nagatani, K.; Otani, N.; Nawashiro, H.; Sugawara, T.; Wada, K.; Mori, K. Hydrogen improves neurological function through attenuation of blood-brain barrier disruption in spontaneously hypertensive stroke-prone rats. BMC Neurosci. 2015, 16, 22. [Google Scholar] [CrossRef] [Green Version]
  10. Ge, Y.; Wu, F.; Sun, X.; Xiang, Z.; Yang, L.; Huang, S.; Lu, Z.; Sun, Y.; Yu, W.-F. Intrathecal infusion of hydrogen-rich normal saline attenuates neuropathic pain via inhibition of activation of spinal astrocytes and microglia in rats. PLoS ONE 2014, 9, e97436. [Google Scholar] [CrossRef]
  11. Iketani, M.; Ohsawa, I. Molecular hydrogen as a neuroprotective agent. Curr. Neuropharmacol. 2017, 15, 324–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Liu, B.; Qin, S. Different types of molecular hydrogen donors and their pharmacokinetics in vivo. Sheng Li Xue Bao 2019, 71, 371–377. [Google Scholar] [PubMed]
  13. Ono, H.; Nishijima, Y.; Adachi, N.; Sakamoto, M.; Kudo, Y.; Nakazawa, J.; Nakao, A. Hydrogen (H2) treatment for acute erythymatous skin diseases. A report of 4 patients with safety data and a non-controlled feasibility study with H2 concentration measurement on two volunteers. Med. Gas Res. 2012, 2, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kawaguchi, M.; Satoh, Y.; Otsubo, Y.; Kazama, T. Molecular hydrogen attenuates neuropathic pain in mice. PLoS ONE 2014, 9, e100352. [Google Scholar]
  15. Li, Q.; Yu, P.; Zeng, Q.; Luo, B.; Cai, S.; Hui, K.; Yu, G.; Zhu, C.; Chen, X.; Duan, M.; et al. Neuroprotective effect of hydrogen-rich saline in global cerebral ischemia/reperfusion rats: Up-regulated tregs and down-regulated miR-21, miR-210 and NF-jB expression. Neurochem. Res. 2016, 41, 2655–2665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ono, H.; Nishijima, Y.; Ohta, S.; Sakamoto, M.; Kinone, K.; Horikosi, T.; Tamaki, M.; Takeshita, H.; Futatuki, T.; Ohishi, W.; et al. Hydrogen gas inhalation treatment in acute cerebral infarction: A randomized controlled clinical study on safety and neuroprotection. J. Stroke Cerebrovasc. Dis. 2017, 26, 2587–2594. [Google Scholar] [CrossRef] [Green Version]
  17. Hou, C.; Peng, Y.; Qin, C.; Fan, F.; Liu, J.; Long, J. Hydrogen-rich water improves cognitive impairment gender-dependently in APP/PS1 mice without affecting Ab clearance. Free. Radic. Res. 2018, 52, 1311–1322. [Google Scholar] [CrossRef]
  18. Wang, H.; Huo, X.; Chen, H.; Li, B.; Liu, J.; Ma, W.; Wang, X.; Xie, K.; Yu, Y.; Shi, K. Hydrogen rich saline activated autophagy via HIF-1a pathways in neuropathic pain model. BioMed Res. Int. 2018, 2018, 4670834. [Google Scholar]
  19. Huang, J.; Liu, W.; Manaenko, A.; Sun, X.; Mei, Q.; Hu, Q. Hydrogen inhibits microglial activation and regulates microglial phenotype in a mouse middle cerebral artery occlusion model. Med. Gas Res. 2019, 9, 127–132. [Google Scholar]
  20. Huang, L.; Applegate, R.L., II; Applegate, P.M.; Gong, L.; Ocak, U.; Boling, W.; Zhang, J.H. Inhalation of high-concentration hydrogen gas attenuates cognitive deficits in a rat model of asphyxia induced cardiac arrest. Med. Gas Res. 2019, 9, 122–126. [Google Scholar] [CrossRef]
  21. Wang, Y.-Z.; Li, T.-T.; Cao, H.-L.; Yang, W.-C. Recent advances in the neuroprotective effects of medical gases. Med. Gas Res. 2019, 9, 80–87. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, Z.-Y.; Fang, Y.-J.; Luo, Y.-J.; Lenahan, C.; Zhang, J.-M.; Chen, S. The role of medical gas in stroke: An updated review. Med. Gas Res. 2019, 9, 221–228. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, L.; Zhao, P.; Yue, C.; Jin, Z.; Liu, Q.; Du, X.; He, Q. Sustained release of bioactive hydrogen by Pd hydride nanoparticles overcomes Alzheimer’s disease. Biomaterials 2019, 197, 393–404. [Google Scholar] [CrossRef] [PubMed]
  24. Ono, H.; Nishijima, Y.; Adachi, N.; Sakamoto, M.; Kudo, Y.; Kaneko, K.; Nakao, A.; Imaoka, T. A basic study on molecular hydrogen (H2) inhalation in acute cerebral ischemia patients for safety check with physiological parameters and measurement of blood H2 level. Med. Gas Res. 2012, 2, 21. [Google Scholar] [CrossRef] [Green Version]
  25. Tamura, T.; Hayashida, K.; Sano, M.; Suzuki, M.; Shibusawa, T.; Yoshizawa, J.; Kobayashi, Y.; Suzuki, T.; Ohta, S.; Morisaki, H.; et al. Feasibility and safety of hydrogen gas inhalation for post-cardiac arrest syndrome—First in human pilot study. Circ. J. 2016, 80, 1870–1873. [Google Scholar] [CrossRef] [Green Version]
  26. Tamura, T.; Hayashida, K.; Sano, M.; Onuki, S.; Suzuki, M. Efficacy of inhaled hydrogen on neurological outcome following brain ischemia during post-cardiac arrest care (HYBRID II trial): Study protocol for a randomized controlled trial. Trials 2017, 18, 488. [Google Scholar] [CrossRef] [Green Version]
  27. Tamura, T.; Suzuki, M.; Hayashida, K.; Kobayashi, Y.; Yoshizawa, J.; Shibusawa, T. Hydrogen gas inhalation alleviates oxidative stress in patients with post-cardiac arrest syndrome. J. Clin. Biochem. Nutr. 2020, 67, 214–221. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, W.; Zhang, H.-T.; Qin, S.-C. Neuroprotective effects of molecular hydrogen: A critical review. Neurosci. Bull. 2021, 37, 389–404. [Google Scholar] [CrossRef]
  29. Sano, M.; Tamura, T. Hydrogen gas therapy: From preclinical studies to clinical trials. Curr. Pharm. Des. 2021, 27, 650–665. [Google Scholar] [CrossRef]
  30. Li, H.; Luo, Y.; Yang, P.; Liu, J. Hydrogen as a complementary therapy against ischemic stroke: A review of the evidence. J. Neurol. Sci. 2019, 396, 240–246. [Google Scholar] [CrossRef]
  31. Hayashida, K.; Sano, M.; Kamimura, N.; Yokota, T.; Suzuki, M.; Ohta, S.; Fukuda, K.; Hori, S. Hydrogen inhalation during normoxic resuscitation improves neurological outcome in a rat model of cardiac arrest independently of targeted temperature management. Circulation 2014, 130, 2173–2180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tian, Y.; Zhang, Y.; Wang, Y.; Chen, Y.; Fan, W.; Zhou, J.; Qiao, J.; Wei, Y. Hydrogen, a novel therapeutic molecule, regulates oxidative stress, inflammation, and apoptosis. Front. Physiol. 2021, 12, 789507. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, S.-T.; Bao, C.; He, Y.; Tian, X.; Yang, Y.; Zhang, T. Hydrogen gas (XEN) inhalation ameliorates airway inflammation in asthma and COPD patients. QJM Int. J. Med. 2020, 113, 870–875. [Google Scholar] [CrossRef] [PubMed]
  34. Zheng, Z.-G.; Sun, W.-Z.; Hu, J.-Y.; Jie, Z.-J.; Xu, J.-F.; Cao, J.; Song, Y.-L.; Wang, C.-H.; Wang, J.; Zhao, H.; et al. Hydrogen/oxygen therapy for the treatment of an acute exacerbation of chronic obstructive pulmonary disease: Results of a multicenter, randomized, double-blind, parallel-group controlled trial. Respir. Res. 2021, 22, 149. [Google Scholar] [CrossRef]
  35. Guan, W.-J.; Chen, R.-C.; Zhong, N.-S. Strategies for the prevention and management of coronavirus disease 2019. Eur. Respir. J. 2020, 55, 2000597. [Google Scholar] [CrossRef] [Green Version]
  36. Guan, W.-J.; Wei, C.-H.; Chen, A.-L.; Sun, X.-C.; Guo, G.-Y.; Zou, X.; Shi, J.-D.; Lai, P.-Z.; Zheng, Z.-G.; Zhong, N.-S. Hydrogen/oxygen mixed gas inhalation improves disease severity and dyspnea in patients with coronavirus disease 2019 in a recent multicenter, open-label clinical trial. J. Thorac. Dis. 2020, 12, 3448–3452. [Google Scholar] [CrossRef]
  37. Pluta, R.; Kida, E.; Lossinsky, A.S.; Golabek, A.A.; Mossakowski, M.J.; Wisniewski, H.M. Complete cerebral ischemia with short-term survival in rats induced by cardiac arrest. I. Extracellular accumulation of Alzheimer’s β-amyloid protein precursor in the brain. Brain Res. 1994, 649, 323–328. [Google Scholar] [CrossRef]
  38. Kiryk, A.; Pluta, R.; Figiel, I.; Mikosz, M.; Ułamek, M.; Niewiadomska, G.; Jabłoński, M.; Kaczmarek, L. Transient brain ischemia due to cardiac arrest causes irreversible long-lasting cognitive injury. Behav. Brain Res. 2011, 219, 1–7. [Google Scholar] [CrossRef]
  39. Sekeljic, V.; Bataveljic, D.; Stamenkovic, S.; Ułamek, M.; Jabłoński, M.; Radenovic, L.; Pluta, R.; Andjus, P.R. Cellular markers of neuroinflammation and neurogenesis after ischemic brain injury in the long-term survival rat model. Brain Struct. Funct. 2012, 217, 411–420. [Google Scholar] [CrossRef]
  40. Pluta, R.; Bogucka-Kocka, A.; Ułamek-Kozioł, M.; Bogucki, J.; Czuczwar, S.J. Ischemic tau protein gene induction as an additional key factor driving development of Alzheimer’s phenotype changes in CA1 area of hippocampus in an ischemic model of Alzheimer’s disease. Pharmacol. Rep. 2018, 70, 881–884. [Google Scholar] [CrossRef]
  41. Pluta, R.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J. Tau protein dysfunction after brain ischemia. J. Alzheimer’s Dis. 2018, 66, 429–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pluta, R. Brain Ischemia: Alzheimer’s Disease Mechanisms; Nova Science Publishers, Inc.: New York, NY, USA, 2019; p. 311. [Google Scholar]
  43. Pluta, R.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J. Amyloid pathology in the brain after ischemia. Folia Neuropathol. 2019, 57, 220–226. [Google Scholar] [CrossRef] [PubMed]
  44. Radenovic, L.; Nenadic, M.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J.; Andjus, P.R.; Pluta, R. Heterogeneity in brain distribution of activated microglia and astrocytes in a rat ischemic model of Alzheimer’s disease after 2 years of survival. Aging 2020, 12, 12251–12267. [Google Scholar] [CrossRef] [PubMed]
  45. Pluta, R.; Januszewski, S.; Czuczwar, S.J. The role of gut microbiota in an ischemic stroke. Int. J. Mol. Sci. 2021, 22, 915. [Google Scholar] [CrossRef] [PubMed]
  46. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Brain ischemia as a prelude to Alzheimer’s disease. Front. Aging Neurosci. 2021, 13, 636653. [Google Scholar] [CrossRef] [PubMed]
  47. Pluta, R.; Czuczwar, S.J.; Januszewski, S.; Jabłoński, M. The many faces of post-ischemic tau protein in brain neurodegeneration of the Alzheimer’s disease type. Cells 2021, 10, 2213. [Google Scholar] [CrossRef] [PubMed]
  48. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Neuroinflammation in post-ischemic neurodegeneration of the brain: Friend, foe, or both? Int. J. Mol. Sci. 2021, 22, 4405. [Google Scholar] [CrossRef]
  49. Pluta, R. Brain ischemia as a bridge to Alzheimer’s disease. Neural Regen. Res. 2022, 17, 791–792. [Google Scholar] [CrossRef]
  50. Hossmann, K.A.; Schmidt-Kastner, R.; Ophoff, B.G. Recovery of integrative central nervous function after one hour global cerebro-circulatory arrest in normothermic cat. J. Neurol. Sci. 1987, 77, 305–320. [Google Scholar] [CrossRef]
  51. Snowdon, D.A.; Greiner, L.H.; Mortimer, J.A.; Riley, K.P.; Greiner, P.A.; Markesbery, W.R. Brain infarction and the clinical expression of Alzheimer disease: The Nun Study. JAMA 1997, 277, 813–817. [Google Scholar] [CrossRef]
  52. Pluta, R. The role of apolipoprotein E in the deposition of β-amyloid peptide during ischemia–reperfusion brain injury. A model of early Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2000, 903, 324–334. [Google Scholar] [CrossRef] [PubMed]
  53. Pluta, R.; Ułamek, M.; Jabłoński, M. Alzheimer’s mechanisms in ischemic brain degeneration. Anat. Rec. 2009, 292, 1863–1881. [Google Scholar] [CrossRef]
  54. Pluta, R.; Januszewski, S.; Jabłoński, M.; Ułamek, M. Factors in creepy delayed neuronal death in hippocampus following brain ischemia-reperfusion injury with long-term survival. Acta Neurochir. 2010, 106, 37–41. [Google Scholar]
  55. Pluta, R.; Ułamek, M.; Jabłoński, M. Consideration of the ischaemic basis and treatment of Alzheimer’s disease. Folia Neuropathol. 2010, 48, 11–26. [Google Scholar] [PubMed]
  56. Pluta, R.; Barcikowska, M.; Debicki, G.; Ryba, M.; Januszewski, S. Changes in amyloid precursor protein and apolipoprotein E immunoreactivity following ischemic brain injury in rat with long-term survival: Influence of idebenone treatment. Neurosci. Lett. 1997, 232, 95–98. [Google Scholar] [CrossRef]
  57. Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Januszewski, S.; Jabłonski, M.; Gil-Kulik, P.; Brzozowska, J.; Petniak, A.; Furmaga-Jabłonska, W.; Bogucki, J.; et al. Dysregulation of amyloid precursor protein, β-secretase, presenilin 1 and 2 genes in the rat selectively vulnerable CA1 subfield of hippocampus following transient global brain ischemia. J. Alzheimer’s Dis. 2015, 47, 1047–1056. [Google Scholar] [CrossRef] [Green Version]
  58. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Petniak, A.; Gil-Kulik, P.; Januszewski, S.; Bogucki, J.; Jabłoński, M.; Brzozowska, J.; Furmaga-Jabłońska, W.; et al. Discrepancy in expression of β-secretase and amyloid-β protein precursor in Alzheimer-related genes in the rat medial temporal lobe cortex following transient global brain ischemia. J. Alzheimer’s Dis. 2016, 51, 1023–1031. [Google Scholar] [CrossRef] [Green Version]
  59. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Gil-Kulik, P.; Januszewski, S.; Jabłoński, M.; Petniak, A.; Brzozowska, J.; Bogucki, J.; et al. Alzheimer-associated presenilin 2 gene is dysregulated in rat medial temporal lobe cortex after complete brain ischemia due to cardiac arrest. Pharmacol. Rep. 2016, 68, 155–161. [Google Scholar] [CrossRef]
  60. Ułamek-Kozioł, M.; Kocki, J.; Bogucka-Kocka, A.; Petniak, A.; Gil-Kulik, P.; Januszewski, S.; Bogucki, J.; Jabłoński, M.; Furmaga-Jabłońska, W.; Brzozowska, J.; et al. Dysregulation of autophagy, mitophagy and apoptotic genes in the medial temporal lobe cortex in an ischemic model of Alzheimer’s disease. J. Alzheimer’s Dis. 2016, 54, 113–121. [Google Scholar] [CrossRef] [Green Version]
  61. Ułamek-Kozioł, M.; Kocki, J.; Bogucka-Kocka, A.; Januszewski, S.; Bogucki, J.; Czuczwar, S.J.; Pluta, R. Autophagy, mitophagy and apoptotic gene changes in the hippocampal CA1 area in a rat ischemic model of Alzheimer’s disease. Pharmacol. Rep. 2017, 69, 1289–1294. [Google Scholar] [CrossRef]
  62. Ułamek-Kozioł, M.; Czuczwar, S.J.; Kocki, J.; Januszewski, S.; Bogucki, J.; Bogucka-Kocka, A.; Pluta, R. Dysregulation of autophagy, mitophagy, and apoptosis genes in the CA3 region of the hippocampus in the ischemic model of Alzheimer’s disease in the rat. J. Alzheimer’s Dis. 2019, 72, 1279–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Pluta, R.; Ułamek-Kozioł, M.; Kocki, J.; Bogucki, J.; Januszewski, S.; Bogucka-Kocka, A.; Czuczwar, S.J. Expression of the tau protein and amyloid protein precursor processing genes in the CA3 area of the hippocampus in the ischemic model of Alzheimer’s disease in the rat. Mol. Neurobiol. 2020, 57, 1281–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Pluta, R.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J. Participation of amyloid and tau protein in neuronal death and neurodegeneration after brain ischemia. Int. J. Mol. Sci. 2020, 21, 4599. [Google Scholar] [CrossRef] [PubMed]
  65. Pluta, R.; Kiś, J.; Januszewski, S.; Jabłoński, M.; Czuczwar, S.J. Cross-talk between amyloid, tau protein and free radicals in post-ischemic brain neurodegeneration in the form of Alzheimer’s disease proteinopathy. Antioxidants 2022, 11, 146. [Google Scholar] [CrossRef]
  66. Pluta, R.; Furmaga-Jabłońska, W.; Januszewski, S.; Czuczwar, S.J. Post-ischemic brain neurodegeneration in the form of Alzheimer’s disease proteinopathy: Possible therapeutic role of curcumin. Nutrients 2022, 14, 248. [Google Scholar] [CrossRef]
  67. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Post-ischemic neurodegeneration of the hippocampus resembling Alzheimer’s disease proteinopathy. Int. J. Mol. Sci. 2022, 23, 306. [Google Scholar] [CrossRef]
  68. Huang, J.; Liu, W.; Sun, X. Hydrogen inhalation improves mouse neurological outcomes after cerebral ischemia/reperfusion independent of anti-necroptosis. Med. Gas Res. 2018, 8, 1–5. [Google Scholar]
  69. Chen, L.; Chao, Y.; Cheng, P.; Li, N.; Zheng, H.; Yang, Y. UPLCQTOF/MS-based metabolomics reveals the protective mechanism of hydrogen on mice with ischemic stroke. Neurochem. Res. 2019, 44, 1950–1963. [Google Scholar] [CrossRef]
  70. Huang, L.; Applegate, R.L., II; Applegate, P.M.; Boling, W.; Zhang, J.H. Inhalation of high concentration hydrogen gas improves shortterm outcomes in a rat model of asphyxia induced-cardiac arrest. Med. Gas Res. 2018, 8, 73–78. [Google Scholar] [CrossRef]
  71. Chen, G.; Chen, B.; Dai, C.; Wang, J.; Wang, J.; Huang, Y.; Li, Y. Hydrogen inhalation is superior to mild hypothermia for improving neurological outcome and survival in a cardiac arrest model of spontaneously hypertensive rat. Shock 2018, 50, 689–695. [Google Scholar] [CrossRef]
  72. Wang, P.; Jia, L.; Chen, B.; Zhang, L.; Liu, J.; Long, J.; Li, Y. Hydrogen inhalation is superior to mild hypothermia in improving cardiac function and neurological outcome in an asphyxial cardiac arrest model of rats. Shock 2016, 46, 312–318. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, C.H.; Manaenko, A.; Zhan, Y.; Liu, W.W.; Ostrowki, R.P.; Tang, J.; Zhang, J.H. Hydrogen gas reduced acute hyperglycemia-enhanced hemorrhagic transformation in a focal ischaemia rat model. Neuroscience 2010, 169, 402–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ji, Q.; Hui, K.; Zhang, L.; Sun, X.; Li, W.; Duan, M. The effect of hydrogen-rich saline on the brain of rats with transient ischemia. J. Surg. Res. 2011, 168, e95–e01. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, D.; Choi, J.-I. Hydrogen-rich water improves cognitive ability and induces antioxidative, antiapoptotic, and anti-inflammatory effects in an acute ischemia-reperfusion injury mouse model. BioMed Res. Int. 2021, 9956938, 12p. [Google Scholar] [CrossRef]
  76. Jiang, Z.; Alamuri, T.T.; Muir, E.R.; Choi, D.W.; Duong, T.Q. Longitudinal multiparametric MRI study of hydrogen-enriched water with minocycline combination therapy in experimental ischemic stroke in rats. Brain Res. 2020, 1748, 147122. [Google Scholar] [CrossRef]
  77. Nagatani, K.; Wada, K.; Takeuchi, S.; Kobayashi, H.; Uozumi, Y.; Otani, N.; Fujita, M.; Tachibana, S.; Nawashiro, H. Effect of hydrogen gas on the survival rate of mice following global cerebral ischemia. Shock 2012, 37, 645–652. [Google Scholar] [CrossRef]
  78. Ge, P.; Zhao, J.; Li, S.; Ding, Y.; Yang, F.; Luo, Y. Inhalation of hydrogen gas attenuates cognitive impairment in transient cerebral ischemia via inhibition of oxidative stress. Neurol. Res. 2012, 34, 187–194. [Google Scholar] [CrossRef]
  79. Hayashida, K.; Sano, M.; Kamimura, N.; Yokota, T.; Suzuki, M.; Maekawa, Y.; Kawamura, A.; Abe, T.; Ohta, S.; Fukuda, K.; et al. H2 gas improves functional outcome after cardiac arrest to an extent comparable to therapeutic hypothermia in a rat model. J. Am. Heart Assoc. 2012, 1, e003459. [Google Scholar] [CrossRef] [Green Version]
  80. Hayashida, K.; Miyara, S.J.; Shinozaki, K.; Takegawa, R.; Yin, T.; Rolston, D.M.; Choudhary, R.C.; Guevara, S.; Molmenti, E.P.; Becker, L.B. Inhaled gases as therapies for post-cardiac arrest syndrome: A narrative review of recent developments. Front. Med. 2021, 7, 586229. [Google Scholar] [CrossRef]
  81. Yin, T.; Becker, L.B.; Choudhary, R.C.; Takegawa, R.; Shoaib, M.; Shinozaki, K.; Endo, Y.; Homma, K.; Rolston, D.M.; Eguchi, S.; et al. Hydrogen gas with extracorporeal cardiopulmonary resuscitation improves survival after prolonged cardiac arrest in rats. J. Transl. Med. 2021, 19, 462. [Google Scholar] [CrossRef]
  82. Gong, X.; Fan, X.; Yin, X.; Xu, T.; Li, J.; Guo, J.; Zhao, X.; Wei, S.; Yuan, Q.; Wang, J.; et al. Hydrogen therapy after resuscitation improves myocardial injury involving inhibition of autophagy in an asphyxial rat model of cardiac arrest. Exp. Ther. Med. 2022, 23, 376. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, G.; Zhou, J.; Zhan, W.; Xiong, Y.; Hu, C.; Li, X.; Li, X.; Li, Y.; Liao, X. The neuroprotective effects of intraperitoneal injection of hydrogen in rabbits with cardiac arrest. Resuscitation 2013, 84, 690–695. [Google Scholar] [CrossRef] [PubMed]
  84. Han, L.; Tian, R.; Yan, H.; Pei, L.; Hou, Z.; Hao, S.; Li, Y.V.; Tian, Q.; Liu, B.; Zhang, Q. Hydrogen-rich water protects against ischemic brain injury in rats by regulating calcium buffering proteins. Brain Res. 2015, 1615, 129–138. [Google Scholar] [CrossRef] [PubMed]
  85. Jiang, Z.; Li, C.; Manuel, M.L.; Yuan, S.; Kevil, C.G.; McCarter, K.D.; Lu, W.; Sun, H. Role of hydrogen sulfide in early blood-brain barrier disruption following transient focal cerebral ischemia. PLoS ONE 2015, 10, e0117982. [Google Scholar] [CrossRef] [PubMed]
  86. Zhai, X.; Chen, X.; Shi, J.; Shi, D.; Ye, Z.; Liu, W.; Li, M.; Wang, Q.; Kang, Z.; Bi, H.; et al. Lactulose ameliorates cerebral ischemia–reperfusion injury in rats by inducing hydrogen by activating Nrf2 expression. Free Radic. Biol. Med. 2013, 65, 731–741. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, Y.; Liu, W.; Sun, X.; Li, R.; Sun, Q.; Cai, J.; Kang, Z.; Lv, S.; Zhang, J.H.; Zhang, W. Hydrogen saline offers neuroprotection by reducing oxidative stress in a focal cerebral ischemia-reperfusion rat model. Med. Gas Res. 2011, 1, 15. [Google Scholar] [CrossRef] [Green Version]
  88. Li, D.; Ai, Y. Hydrogen saline suppresses neuronal cell apoptosis and inhibits the p38 mitogen-activated protein kinase-caspase-3 signaling pathway following cerebral ischemia-reperfusion injury. Mol. Med. Rep. 2017, 16, 5321–5325. [Google Scholar] [CrossRef] [Green Version]
  89. Nagatani, K.; Nawashiro, H.; Takeuchi, S.; Tomura, S.; Otani, N.; Osada, H.; Wada, K.; Katoh, H.; Tsuzuki, N.; Mori, K. Safety of intravenous administration of hydrogen-enriched fluid in patients with acute cerebral ischemia: Initial clinical studies. Med. Gas Res. 2013, 3, 13. [Google Scholar] [CrossRef] [Green Version]
  90. Hugyecz, M.; Mracskó, E.; Hertelendy, P.; Farkas, E.; Domoki, F.; Bari, F. Hydrogen supplemented air inhalation reduces changes of prooxidant enzyme and gap junction protein levels after transient global cerebral ischemia in the rat hippocampus. Brain Res. 2011, 1404, 31–38. [Google Scholar] [CrossRef]
  91. Cui, Y.; Zhang, H.; Ji, M.; Jia, M.; Chen, H.; Yang, J.; Duan, M. Hydrogen-rich saline attenuates neuronal ischemia-reperfusion injury by protecting mitochondrial function in rats. J. Surg. Res. 2014, 192, 564–572. [Google Scholar] [CrossRef]
  92. Chen, K.; Wang, N.; Diao, Y.; Dong, W.; Sun, Y.; Liu, L.; Wu, X. Hydrogen-rich saline attenuates brain injury induced by cardiopulmonary bypass and inhibits microvascular endothelial cell apoptosis via the PI3K/Akt/GSK3b signaling pathway in rats. Cell. Physiol. Biochem. 2017, 43, 1634–1647. [Google Scholar] [CrossRef] [PubMed]
  93. Gao, Y.; Gui, Q.; Jin, L.; Yu, P.; Wu, L.; Cao, L.; Wang, Q.; Duan, M. Hydrogen-rich saline attenuates hippocampus endoplasmic reticulum stress after cardiac arrest in rats. Neurosci. Lett. 2017, 640, 29–36. [Google Scholar] [CrossRef] [PubMed]
  94. Cole, A.R.; Perry, D.A.; Raza, A.; Nedder, A.P.; Pollack, E.; Regan, W.L.; van den Bosch, S.J.; Polizzotti, B.D.; Yang, E.; Davila, D.; et al. Perioperatively inhaled hydrogen gas diminishes neurologic injury following experimental circulatory arrest in swine. JACC Basic Transl. Sci. 2019, 4, 176–187. [Google Scholar] [CrossRef] [PubMed]
  95. Jendroska, K.; Poewe, W.; Daniel, S.E.; Pluess, J.; Iwerssen-Schmidt, H.; Paulsen, J.; Barthel, S.; Schelosky, L.; Cervos-Navarro, J.; DeArmond, S.J. Ischemic stress induces deposition of amyloid beta immunoreactivity in human brain. Acta Neuropathol. 1995, 90, 461–466. [Google Scholar] [CrossRef]
  96. Wiśniewski, H.M.; Maślińska, D. Beta-protein immunoreactivity in the human brain after cardiac arrest. Folia Neuropathol. 1996, 34, 65–71. [Google Scholar]
  97. Jendroska, K.; Hoffmann, O.M.; Patt, S. Amyloid β peptide and precursor protein (APP) in mild and severe brain ischemia. Ann. N. Y. Acad. Sci. 1997, 826, 401–405. [Google Scholar] [CrossRef]
  98. Van Groen, T.; Puurunen, K.; Maki, H.M.; Sivenius, J.; Jolkkonen, J. Transformation of diffuse beta-amyloid precursor protein and beta-amyloid deposits to plaques in the thalamus after transient occlusion of the middle cerebral artery in rats. Stroke 2005, 36, 1551–1556. [Google Scholar] [CrossRef] [Green Version]
  99. Qi, J.; Wu, H.; Yang, Y.; Wand, D.; Chen, Y.; Gu, Y.; Liu, T. Cerebral ischemia and Alzheimer’s disease: The expression of amyloid-β and apolipoprotein E in human hippocampus. J. Alzheimer’s Dis. 2007, 12, 335–341. [Google Scholar] [CrossRef]
  100. Kato, T.; Hirano, A.; Katagiri, T.; Sasaki, H.; Yamada, S. Neurofibrillary tangle formation in the nucleus basalis of Meynert ipsilateral to a massive cerebral infarct. Ann. Neurol. 1988, 23, 620–623. [Google Scholar] [CrossRef]
  101. Hatsuta, H.; Takao, M.; Nogami, A.; Uchino, A.; Sumikura, H.; Takata, T.; Morimoto, S.; Kanemaru, K.; Adachi, T.; Arai, T.; et al. Tau and TDP-43 accumulation of the basal nucleus of Meynert in individuals with cerebral lobar infarcts or hemorrhage. Acta Neuropathol. Commun. 2019, 7, 49. [Google Scholar] [CrossRef]
  102. Lin, Y.-T.; Shi, Q.-Q.; Zhang, L.; Yue, C.-P.; He, Z.-J.; Li, X.-X.; He, Q.-J.; Liu, Q.; Du, X.-B. Hydrogen-rich water ameliorates neuropathological impairments in a mouse model of Alzheimer’s disease through reducing neuroinflammation and modulating intestinal microbiota. Neural Regen. Res. 2022, 17, 409–417. [Google Scholar] [PubMed]
  103. Lin, C.-L.; Huang, W.-N.; Li, H.-H.; Huang, C.-N.; Hsieh, S.; Lai, C.; Lu, F.-J. Hydrogen-rich water attenuates amyloid-induced cytotoxicity through upregulation of Sirt1-FoxO3a by stimulation of AMP-activated protein kinase in SK-N-MC cells. Chem. Biol. Interact. 2015, 240, 12–21. [Google Scholar] [CrossRef] [PubMed]
  104. Zhao, L.; Wang, Y.; Zhang, G.; Zhang, T.; Lou, J.; Liu, J. L-arabinose elicits gut-derived hydrogen production and ameliorates metabolic syndrome in C57BL/6J mice on high-fat-diet. Nutrients 2019, 11, 3054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zhou, G.; Goshi, E.; He, Q. Micro/nanomaterials-augmented hydrogen therapy. Adv. Healthc. Mater. 2019, 8, e1900463. [Google Scholar] [CrossRef]
  106. Shimouchi, A.; Nose, K.; Shirai, M.; Kondo, T. Estimation of molecular hydrogen consumption in the human whole body after the ingestion of hydrogen-rich water. Adv. Exp. Med. Biol. 2012, 737, 245–250. [Google Scholar]
  107. Liu, C.; Kurokawa, R.; Fujino, M.; Hirano, S.; Sato, B.; Li, X. Estimation of the hydrogen concentration in rat tissue using an airtight tube following the administration of hydrogen via various routes. Sci. Rep. 2014, 4, 5485. [Google Scholar] [CrossRef] [Green Version]
  108. Nakao, A.; Toyoda, Y.; Sharma, P.; Evans, M.; Guthrie, N. Effectiveness of hydrogen rich water on antioxidant status of subjects with potential metabolic syndrome-an open label pilot study. J. Clin. Biochem. Nutr. 2010, 46, 140–149. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Neuroprotective mechanisms of molecular hydrogen on post-ischemic brain neurodegeneration phenomena. BBB-blood-brain barrier. ↓—reduction, ↑—increase.
Figure 1. Neuroprotective mechanisms of molecular hydrogen on post-ischemic brain neurodegeneration phenomena. BBB-blood-brain barrier. ↓—reduction, ↑—increase.
Ijms 23 06591 g001
Table 1. Neuroprotective effects of molecular hydrogen in experimental post-ischemic brain injury.
Table 1. Neuroprotective effects of molecular hydrogen in experimental post-ischemic brain injury.
IschemiaAnimalStrainTreatmentBenefitsReferences
FocalMiceC57B/LInhalation of 66.7% hydrogen/33.3% oxygen for 90 min post-ischemia.Inhibition of microglial activity and regulation of microglial phenotype. Improvement of neurological outcome.[19,68]
GlobalMiceC57BL/6JInhalation hydrogen (1.3%), oxygen (30%), and nitrogen (68.7%).
45 min of ischemia and 180 min of reperfusion, and 3 h/d, from 1 to 3 days post-ischemia.
Improved survival. Attenuation of neuronal injury, autophagy and brain edema.[77]
GlobalRatWistar2.1% hydrogen supplemented by room air ventilation for 4 h after ischemia.Reduction changes of prooxidant enzyme and gap junction protein levels.[90]
GlobalRatSprague-DawleyHydrogen-rich saline (5 mL/kg) was injected immediately post-ischemia.Significant improvement of surviving cells. Reduction tissue damage, the degree of mitochondrial swelling, and the loss of mitochondrial membrane potential but also preservation the mitochondrial cytochrome c content.[91]
GlobalRatSprague-DawleyI.V. hydrogen-rich saline (1 mL/kg, 4 mL/kg, or 6 mL/kg),
HRS was given before hypoxia and during reoxygenation.
Inhibition of hippocampus endoplasmic reticulum stress and microvascular endothelial cells apoptosis via PI3K/Akt/GSK3β signaling pathway.[92]
GlobalRatSprague-DawleyHydrogen-rich saline 5 mL/kg was intraperitoneally injected immediately and 6 h post-ischemia.Significant improvement survival rate and neurological function. The beneficial effects associated with decreased levels of oxidative products, as well as the increased levels of antioxidant enzymes and accompanied by the increased activity of glucose-regulated protein 78, the decreased activity of cysteinyl aspartate specific proteinase-12 (caspase-12).[93]
GlobalRatWistarInhalation of 2% hydrogen started immediately at the end of ischemia and lasted for 3 h.Attenuation of cognitive impairment. Decreased pyramidal neuronal death in CA1 region of hippocampus.[78]
GlobalRatSprague-DawleyHydrogen-rich saline was administered i.v. at 1 min before end of ischemia, followed by injections at 6 and 12 h post-ischemia.Improves survival and neurological outcome.[8]
FocalRatSprague-Dawley6 mL/kg i.p. per rat before and after ischemia.Reduction brain infarct volume and improvement of neurological function. Prevention the ischemia-induced reduction of parvalbumin and hippocalcin levels and also reduced the glutamate toxicity-induced death of neurons. Attenuation the glutamate toxicity-induced by elevate in intracellular calcium.[84]
FocalRatSprague-Dawley0.5 mL/kg/day saturated hydrogen saline (0.6 mmol/L) i.p. 3 days prior to ischemia and immediately during 24 h of reperfusion.Significantly reduction the number of apoptotic cells, and the protein expression of p38 MAPK and caspase-3. These effects may be associated with the p38MAPK signaling pathway.[88]
FocalRatSprague-DawleyHydrogen saline was injected i.p. (1 mL/100 g body weight) at designed time points 0, 3 or 6 h after reperfusion onset.Reduction 8-hydroxyl-2′-deoxyguanosine, malondidehyde, interleukin-1β, tumor necrosis factor-α, and suppressed caspase 3 activity in ischemic brain.[87]
GlobalRabbitWhiteBefore ischemia i.p. injection of hydrogen low dose (10 mL/kg) or high dose (20 mL/kg).Improvement survival and neurological outcomes, reduction of neuronal damage and inhibition of neuronal apoptosis. Reduction indicators of oxidative stress in the blood and the hippocampus and increased activity of antioxidant enzyme.[83]
GlobalSwineYorkshireInhalation of hydrogen (2.40%) for a 24-h period during and after the ischemic injury.Reduced neurological injury.[94]
Table 2. Neuroprotective effects of molecular hydrogen in clinical post-ischemic brain injury.
Table 2. Neuroprotective effects of molecular hydrogen in clinical post-ischemic brain injury.
IschemiaNumber of ParticipantsTreatmentBenefitsStudyReferences
Focal50 patientsInhalation 3% hydrogen gas (1 h twice a day) for initial 7 days.Reduced infarct size, improved neurological outcome and daily living activity.Randomized[16]
Global5 patients2% hydrogen with oxygen was supplied via a respirator
after admission to the intensive care unit for 18 h.
4 patients survived 90 days with a favorable neurological outcome.Pilot study[25]
Global360 patients2% hydrogen with 24 to 50% oxygen was supplied via mechanical ventilation
after admission for 18 h.
The first multicenter randomized trial is underway to confirm the efficacy of hydrogen on neurological outcomes in comatose out-of-hospital cardiac arrest survivors.Randomized, double-blind, placebo-controlled trial.[26]
Global5 patientsInhalation 2% hydrogen with titrated oxygen was initiated upon admission for 18 h.Oxidative stress markers were reduced in cardiogenic post-cardiac arrest patients but were slightly elevated in the patient with sepsis. Inflammatory cytokine levels remained unchanged in cardiogenic post-cardiac arrest patients, whereas a dramatic reduction was observed in one patient with sepsis.Pilot study[27]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pluta, R.; Januszewski, S.; Czuczwar, S.J. Molecular Hydrogen Neuroprotection in Post-Ischemic Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy: Underlying Mechanisms and Potential for Clinical Implementation—Fantasy or Reality? Int. J. Mol. Sci. 2022, 23, 6591. https://doi.org/10.3390/ijms23126591

AMA Style

Pluta R, Januszewski S, Czuczwar SJ. Molecular Hydrogen Neuroprotection in Post-Ischemic Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy: Underlying Mechanisms and Potential for Clinical Implementation—Fantasy or Reality? International Journal of Molecular Sciences. 2022; 23(12):6591. https://doi.org/10.3390/ijms23126591

Chicago/Turabian Style

Pluta, Ryszard, Sławomir Januszewski, and Stanisław J. Czuczwar. 2022. "Molecular Hydrogen Neuroprotection in Post-Ischemic Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy: Underlying Mechanisms and Potential for Clinical Implementation—Fantasy or Reality?" International Journal of Molecular Sciences 23, no. 12: 6591. https://doi.org/10.3390/ijms23126591

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

Article Metrics

Back to TopTop