Electrolyzed Hydrogen Water for CKD and Hemodialysis: Comparison
Please note this is a comparison between Version 2 by Sirius Huang and Version 1 by Masaaki Nakayama.

Chronic kidney disease (CKD), which is globally on the rise, has become an urgent challenge from the perspective of public health, given its risk factors such as end-stage renal failure, cardiovascular diseases, and infections. The pathophysiology of CKD, including dialysis patients, is deeply associated with enhanced oxidative stress in both the kidneys and the entire body. Therefore, the introduction of a safe and widely applicable antioxidant therapy is expected as a measure against CKD. Electrolyzed hydrogen water (EHW) generated through the electrolysis of water has been confirmed to possess chemical antioxidant capabilities. In Japan, devices producing this water have become popular for household drinking water. In CKD model experiments conducted to date, drinking EHW has been shown to suppress the progression of kidney damage related to hypertension. Furthermore, clinical studies have reported that systemic oxidative stress in patients undergoing dialysis treatment using EHW is suppressed, leading to a reduction in the incidence of cardiovascular complications.

  • electrolyzed hydrogen water
  • antioxidant
  • chronic kidney disease
  • hemodialysis

1. Introduction

1.1. Historical Background of Electrolyzed Hydrogen Water

“Electrolyzed hydrogen water” (EHW) refers to the water generated on the cathode side during the electrolysis of water. Chemically, it is characterized as weakly alkaline (with a pH of 9.0 or higher but lower than 10.0) and contains hydrogen molecules (H2). The concentration of H2 varies depending on the model of the generator, but it can be adjusted arbitrarily to 100–1300 ppb immediately after generation by adjusting the electrolysis intensity and water flow rate. The generator of this water is called an “electrolyzed water generator”, also known as a water ionizer. In Japan, it received medical device approval from the former Ministry of Health and Welfare in 1945 as a home drinking water generator. Furthermore, it has undergone double-blind trials, confirming its effectiveness in improving gastrointestinal symptoms such as abdominal discomfort, fullness, diarrhea, and constipation, i.e., significant global improvement of abdominal symptoms in the group with EHW as compared with the control [1]. Currently, approximately 200,000 units of this device are manufactured and sold annually in Japan, and the consumption of EHW is assumed to be incorporated into daily life at a certain level [2].

1.2. Cross over with Hydrogen Medicine

In the scientific exploration of EHW, Shirahata et al. first demonstrated its antioxidant capability in 1997 [3]. The chemical characteristics of this water include its ability to suppress the generation of superoxide anions and promote the decomposition of hydrogen peroxide. It has been shown to inhibit the production of reactive oxygen species and exhibits catalase-like activity and biological effects such as suppression of apoptosis via oxidative stress [4] and the extension of the lifespan of nematodes through its antioxidant action [5]. Regarding the mechanism of this antioxidative effect, Shirahata and others suggested that the mechanism of this antioxidative effect involves factors such as the influence of nano-sized platinum particles released from the electrodes used in electrolysis [5,6][5][6] as well as changes in water molecules due to the electrolysis of water. However, many details of the precise mechanism remain unclear.
In 2007, Ohsawa et al. reported that inhalation of H2 suppresses the expansion of cerebral infarcts caused by brain artery clamping, suggesting the involvement of direct hydroxyl radical elimination by H2 [7]. This triggered the development of hydrogen medicine, and various organ-protective effects from the antioxidant and anti-inflammatory effects of H2 have been confirmed in animal experiments [8]. In this context, the mechanism of the biological effects of EHW is now assumed to involve H2 [9].
Currently, research on the application of H2 for human disease prevention and treatment is underway [10]. In this sense, the situation in Japan, where electrolyzed hydrogen water generators are already in use among the general public, is intriguing. When considering real-world medical applications, researchers believe that the potential medical significance of EHW in preventing disease onset and suppressing exacerbation in the pre-symptomatic state, as well as in preventive healthcare, is significant. Cross-sectional comparative studies targeting healthy individuals have been conducted [11], reporting significantly lower oxidative stress values in the blood of daily EHW consumers compared to non-consumers, as well as significantly lower levels of blood urea nitrogen, a kidney function indicators.

2. Latest Insights into H2 Biology Research—Brief Summary

H2 has been demonstrated to possess anti-inflammatory, antioxidant, and anti-endoplasmic reticulum (ER) stress properties, and its involvement in the regulation of apoptosis, autophagy, and pyroptosis has been elucidated [12,13,14,15,16,17,18][12][13][14][15][16][17][18]. In this regard, H2 can be considered a unique molecule that influences fundamental biological responses. Although mechanisms for the multifaceted effects of H2 have been proposed, the fundamental processes still remain unclear. Ohsawa et al. suggested the possibility of H2 directly scavenging hydroxyl radicals [7]; however, subsequent studies revealed the effectiveness of H2 preconditioning in organ protection. As this phenomenon cannot be solely explained as due to chemical reactions of H2, it is speculated that H2 may impact the body’s inflammatory and antioxidant systems, activating the body’s defense mechanisms [11]. In this context, recent attention has been drawn to the relationship between H2, the redox system in the body, and the mitochondria [14,15,16][14][15][16]. Nuclear factor–erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (Keap1) serve as the master regulators of cellular redox in the body [19,20][19][20] (Figure 1a). Keap1 is present in the cytoplasm and functions as a stress sensor, serving as an enzyme that contributes to the degradation of Nrf2; i.e., in non-stressed cellular conditions, Nrf2 is sequestered by Keap1 and degraded by the ubiquitin–proteasome system. When cells are exposed to stimuli such as electrophilic substances, reactive oxygen species, or endoplasmic reticulum stress, Nrf2 is released from Keap1 inhibition and becomes activated as transcription factor to induce expression of antioxidant response element (ARE)/electrophile responsive element (EpRE) of genes, which include over 200 genes, including major antioxidant and anti-inflammatory molecules.
Figure 1. (a) Stress-sensing mechanisms of the Keap1-Nrf2 system. Abbreviations: Nrf2, nuclear factor–erythroid 2-related factor 2; Keap1, Kelch-like ECH-associated protein 1; Ub, ubiquitin; -SH, thiol; ARE, antioxidant response element; EpRE, electrophile responsive element. (b) Mechanisms underlying the manifestation of biological effects of electrolyzed hydrogen water (hypothesis). Abbreviations: H2O, water; OH, hydroxyl radical; H, hydrogen radical; -SH, thiol; -S-S-, disulfide.
Accumulated reports from animal experiments indicate that H2 administration enhances Nrf2 expression [21,22,23,24,25,26,27][21][22][23][24][25][26][27]. However, Nrf2 expression is supposed to be triggered by oxidative stress stimuli. From this perspective, there is a possibility that H2 induces a so-called hormesis phenomenon [28]. Indeed, the potential of H2 to induce mild oxidative stress has been reported [28[28][29][30],29,30], and activation of antioxidant systems via oxidative stimulation cannot be denied. This is analogous to the body’s response to exercise [31]. The hypothesis regarding the mechanism of H2 to activate Nrf2 is outlined in Figure 1b. The core of the matter lies in the fact demonstrated by Ohsawa et al. [7], who reported that H2 directly scavenges hydroxyl radicals in the form of electrophilic hydrogen radicals (H:H → H + H, H + OH → H2O). However, in general, a single electrophile could have both protective and toxic effects on cells. It is known that electrophiles react with nucleophiles, including protein thiols (-SH), such as those found in reduced glutathione (GSH) or guanine bases in DNA [32]. Each of them has an unshared pair of electrons, and the reaction of an electrophile with the -SH of a cysteine residue results in alkylation [33], leading to the decrease of the reductive capacity of cells, i.e., depletion of GSH. If we consider this phenomenon in terms of hydrogen radicals, theoretically it is possible that hydrogen radicals could react with GSH, resulting in a reduced reductive capacity of cells. Hydrogen radicals may also react with the -SH of cysteine residues in Keap1, leading to the generation of H2 and modification of the cysteine residue of Keap1 (disulfide reaction, -S-S-), which may trigger the activation of Nrf2 action. However, the responses of electrophiles are generally characterized by dose-response [34]; therefore, different doses of H2 and accompanying levels of hydrogen radicals may induce different responses. At present, the fate of hydrogen radicals in cells is completely unknown and requires further investigation. Mitochondria, the energy production mechanism, are the major source of reactive oxygen species in cells. It is assumed that the small molecule H2 is easily distributed within cells, and therefore, it is expected to be directly involved with mitochondria. It has been demonstrated that H2 supplementation is related to the preservation and maintenance of mitochondria [35,36][35][36]. The proposed mechanism suggests that H2 captures excess reactive oxygen species in mitochondria, preserving them from oxidative stress damag, and ultimately exhibiting organ protection effects [14,15,16][14][15][16]. Recently, a connection between the gut microbiota and mitochondrial function has been suggested [37,38,39,40][37][38][39][40]. H2 is involved in preserving mitochondrial function, while the gut microbiota serves as a source of H2 production in the body [41]. Future investigations are expected to explore whether H2 acts as a missing link between the gut microbiota and mitochondrial function. In recent reports, it has been revealed that the consumption of hydrogen-rich water can have an impact on the intestinal microbiota [42]. In this context, the intriguing point is whether externally adding H2 may enhance the interconnection between the gut microbiota and mitochondria within the body, potentially amplifying the anti-stress effects in the living organism. In summary, recent findings have been summarized, but many aspects of the fundamental mechanisms and starting points of H2’s actions on cells and the body remain unknown. However, considering that no adverse effects of H2 on the body have been confirmed, the clinical application of H2 has become a realistic challenge. Within this context, establishing methods of H2 administration that can demonstrate the effectiveness of H2, taking into account the characteristics of the disease, is considered a challenge.

3. H2 Intervention for CKD and Hemodialysis

Given the multifaceted involvement of oxidative stress in various pathologies, antioxidant therapy is considered extremely significant. However, the expected results are not always obtained in interventions employing antioxidants, including for CKD [61][43]. Reactive oxygen species are a double-edged sword, having both detrimental effects on the body and being crucial for the body’s defense. In this sense, excessive oxidative stress should be suppressed, but the degree of suppression should be at a level that does not compromise the benefits of reactive oxygen species to the body [15]. To date, while many preclinical studies using H2 have confirmed organ-protective effects and correction of metabolic abnormalities through its antioxidant and anti-inflammatory effects [12,13[12][13][14][15][16][17][18],14,15,16,17,18], no severe side effects of H2 loading have been observed. Therefore, the clinical application of antioxidant therapy with H2 is considered a realistic challenge. The following summarizes the research on CKD and dialysis-related topics.

3.1. Pre-Clinical Studies of H

2

in CKD Models

The reno-protective effects of H2 have been reported in various models of kidney diseases [62,63][44][45] via H2 administration through drinking water, intraperitoneal administration, and inhalation. These studies involve acute models such as acute kidney injury via ischemia–reperfusion of renal artery clamp [64,65,66[46][47][48][49][50][51][52][53][54][55][56],67,68,69,70,71,72,73,74], allograft rejection [75[57][58],76], drug-induced nephrotoxicity [77[59][60],78], renal calculi [79[61][62],80], and renal fibrosis via ureteral ligation [81,82,83][63][64][65]. However, reports on CKD are limited [21,84,85][21][66][67] (Table 1).
Table 1.
Effect of H
2
water ad libitum drinking for CKD model rats.

References

  1. Itokawa, Y. An overview on researches of portable alkaline water by electrolysis. Kinousuikenkyu 2004, 2, 59–64. (In Japanese)
  2. Alkaline Ionized Water Apparatus Market Trends. Available online: https://www.3aaa.gr.jp/english/markettrend.html (accessed on 1 December 2023).
  3. Shirahata, S.; Kabayama, S.; Nakano, M.; Miura, T.; Kusumoto, K.; Gotoh, M.; Hayashi, H.; Otsubo, K.; Morisawa, S.; Katakura, Y. Electrolyzed-reduced water scavenges active oxygen species and protects DNA from oxidative damage. Biochem. Biophys. Res. Commun. 1997, 234, 269–274.
  4. Li, Y.; Hamasaki, T.; Nakamichi, N.; Kashiwagi, T.; Komatsu, T.; Ye, J.; Teruya, K.; Abe, M.; Yan, H.; Kinjo, T.; et al. Suppressive effects of electrolyzed reduced water on alloxan-induced apoptosis and type 1 diabetes mellitus. Cytotechnology 2011, 63, 119–131.
  5. Yan, H.; Kinjo, T.; Tian, H.; Hamasaki, T.; Teruya, K.; Kabayama, S.; Shirahata, S. Mechanism of the lifespan extension of Caenorhabditis elegans by electrolyzed reduced water—participation of Pt nanoparticles. Biosci. Biotechnol. Biochem. 2011, 75, 1295–1299.
  6. Wang, Y.; Fugetsu, B.; Sakata, I.; Fujisue, C.; Kabayama, S.; Tahara, N.; Morisawa, S. Monolayered Platinum Nanoparticles as Efficient Electrocatalysts for the Mass Production of Electrolyzed Hydrogen Water. Sci. Rep. 2020, 10, 10126.
  7. Ohsawa, I.; Ishikawa, M.; Takahashi, K.; Watanabe, M.; Nishimaki, K.; Yamagata, K.; Katsura, K.-I.; Katayama, Y.; Asoh, S.; Ohta, S. Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat. Med. 2007, 13, 688–694.
  8. Ichihara, M.; Sobue, S.; Ito, M.; Ito, M.; Hirayama, M.; Ohno, K. Beneficial biological effects and the underlying mechanisms of molecular hydrogen—comprehensive review of 321 original articles. Med. Gas. Res. 2015, 5, 12.
  9. LeBaron, T.W.; Sharpe, R.; Ohno, K. Electrolyzed-Reduced Water: Review I. Molecular Hydrogen Is the Exclusive Agent Responsible for the Therapeutic Effects. Int. J. Mol. Sci. 2022, 23, 14750.
  10. Johnsen, H.M.; Hiorth, M.; Klaveness, J. Molecular Hydrogen Therapy-A Review on Clinical Studies and Outcomes. Molecules 2023, 28, 7785.
  11. Mizuno, K.; Watanabe, K.; Yamano, E.; Ebisu, K.; Tajima, K.; Nojima, J.; Ohsaki, Y.; Kabayama, S.; Watanabe, Y. Antioxidant effects of continuous intake of electrolyzed hydrogen water in healthy adults. Heliyon 2022, 8, e11853.
  12. Ohta, S. Molecular hydrogen as a preventive and therapeutic medical gas: Initiation, development and potential of hydrogen medicine. Pharmacol. Ther. 2014, 144, 1–11.
  13. Zhang, Y.; Tan, S.; Xu, J.; Wang, T. Hydrogen Therapy in Cardiovascular and Metabolic Diseases: From Bench to Bedside. Cell Physiol. Biochem. 2018, 47, 1–10.
  14. LeBaron, T.W.; Kura, B.; Kalocayova, B.; Tribulova, N.; Slezak, J. A New Approach for the Prevention and Treatment of Cardiovascular Disorders. Molecular Hydrogen Significantly Reduces the Effects of Oxidative Stress. Molecules 2019, 24, 2076.
  15. Yang, M.; Dong, Y.; He, Q.; Zhu, P.; Zhuang, Q.; Shen, J.; Zhang, X.; Zhao, M. Hydrogen: A Novel Option in Human Disease Treatment. Oxid. Med. Cell Longev. 2020, 2020, 8384742.
  16. 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.
  17. Rahman, M.H.; Jeong, E.-S.; You, H.S.; Kim, C.-S.; Lee, K.-J. Redox-Mechanisms of Molecular Hydrogen Promote Healthful Longevity. Antioxidants 2023, 12, 988.
  18. Saengsin, K.; Sittiwangkul, R.; Chattipakorn, S.C.; Chattipakorn, N. Hydrogen therapy as a potential therapeutic intervention in heart disease: From the past evidence to future application. Cell Mol. Life Sci. 2023, 80, 174.
  19. Suzuki, T.; Yamamoto, M. Stress-sensing mechanisms and the physiological roles of the Keap1-Nrf2 system during cellular stress. J. Biol. Chem. 2017, 292, 16817–16824.
  20. Suzuki, T.; Takahashi, J.; Yamamoto, M. Molecular Basis of the KEAP1-NRF2 Signaling Pathway. Mol. Cells. 2023, 46, 133–141.
  21. Zhu, W.J.; Nakayama, M.; Mori, T.; Hao, K.; Terawaki, H.; Katoh, J.; Kabayama, S.; Ito, S. Amelioration of cardio-renal injury with aging in dahl salt-sensitive rats by H2-enriched electrolyzed water. Med. Gas. Res. 2013, 3, 26.
  22. Yuan, J.; Wang, D.; Liu, Y.; Chen, X.; Zhang, H.; Shen, F.; Liu, X.; Fu, J. Hydrogen-rich water attenuates oxidative stress in rats with traumatic brain injury via Nrf2 pathway. J. Surg. Res. 2018, 228, 238–246.
  23. Kura, B.; Bagchi, A.K.; Singal, P.K.; Barancik, M.; LeBaron, T.W.; Valachova, K.; Šoltés, L.; Slezák, J. Molecular hydrogen: Potential in mitigating oxidative-stress-induced radiation injury. Can. J. Physiol. Pharmacol. 2019, 97, 287–292.
  24. Yu, Y.; Feng, J.; Lian, N.; Yang, M.; Xie, K.; Wang, G.; Wang, C.; Yu, Y. Hydrogen gas alleviates blood-brain barrier impairment and cognitive dysfunction of septic mice in an Nrf2-dependent pathway. Int. Immunopharmacol. 2020, 85, 106585.
  25. Lu, Y.; Li, C.F.; Ping, N.N.; Sun, Y.Y.; Wang, Z.; Zhao, G.X.; Yuan, S.H.; Zibrila, A.I.; Soong, L.; Liu, J.J. Hydrogen-rich water alleviates cyclosporine A-induced nephrotoxicity via the Keap1/Nrf2 signaling pathway. J. Biochem. Mol. Toxicol. 2020, 34, e22467.
  26. Hu, Y.; Wang, P.; Han, K. Hydrogen Attenuated Inflammation Response and Oxidative in Hypoxic Ischemic Encephalopathy via Nrf2 Mediated the Inhibition of NLRP3 and NF-κB. Neuroscience 2022, 485, 23–36.
  27. Peng, J.; He, Q.; Li, S.; Liu, T.; Zhang, J. Hydrogen-Rich Water Mitigates LPS-Induced Chronic Intestinal Inflammatory Response in Rats via Nrf-2 and NF-κB Signaling Pathways. Vet. Sci. 2022, 9, 621.
  28. Murakami, Y.; Ito, M.; Ohsawa, I. Molecular hydrogen protects against oxidative stress-induced SH-SY5Y neuroblastoma cell death through the process of mitohormesis. PLoS ONE 2017, 12, e0176992.
  29. Hirayama, M.; Ito, M.; Minato, T.; Yoritaka, A.; LeBaron, T.W.; Ohno, K. Inhalation of hydrogen gas elevates urinary 8-hydroxy-2’-deoxyguanine in Parkinson’s disease. Med. Gas. Res. 2019, 8, 144–149.
  30. Satta, H.; Iwamoto, T.; Kawai, Y.; Koguchi, N.; Shibata, K.; Kobayashi, N.; Yoshida, M.; Nakayama, M. Amelioration of hemodialysis-induced oxidative stress and fatigue with a hemodialysis system employing electrolyzed water containing molecular hydrogen. Ren. Replace. Ther. 2021, 7, 37.
  31. LeBaron, T.W.; Laher, I.; Kura, B.; Slezak, J. Hydrogen gas: From clinical medicine to an emerging ergogenic molecule for sports athletes 1. Can J. Physiol. Pharmacol. 2019, 97, 797–807.
  32. Satoh, T.; McKercher, S.R.; Lipton, S.A. Nrf2/ARE-mediated antioxidant actions of pro-electrophilic drugs. Free Radic. Biol. Med. 2013, 65, 645–657.
  33. Satoh, T.; Lipton, S.A. Redox regulation of neuronal survival mediated by electrophilic compounds. Trends Neurosci. 2007, 30, 37–45.
  34. Calabrese, V.; Cornelius, C.; Dinkova-Kostova, A.T.; Calabrese, E.J.; Mattson, M.P. Cellular stress responses, the hormesis paradigm, and vitagenes: Novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid. Redox Signal. 2010, 13, 1763–1811.
  35. Shingu, C.; Koga, H.; Hagiwara, S.; Matsumoto, S.; Goto, K.; Yokoi, I.; Noguchi, T. Hydrogen-rich saline solution attenuates renal ischemia-reperfusion injury. J. Anesth. 2010, 24, 569–574.
  36. Gvozdjáková, A.; Kucharská, J.; Kura, B.; Vančová, O.; Rausová, Z.; Sumbalová, Z.; Uličná, O.; Slezák, J. A new insight into the molecular hydrogen effect on coenzyme Q and mitochondrial function of rats. Can. J. Physiol. Pharmacol. 2020, 98, 29–34.
  37. Huang, Y.; Xin, W.; Xiong, J.; Yao, M.; Zhang, B.; Zhao, J. The Intestinal Microbiota and Metabolites in the Gut-Kidney-Heart Axis of Chronic Kidney Disease. Front. Pharmacol. 2022, 13, 837500.
  38. Han, B.; Sivaramakrishnan, P.; Lin, C.J.; Neve, I.A.A.; He, J.; Tay, L.W.R.; Sowa, J.N.; Sizovs, A.; Du, G.; Wang, J.; et al. Microbial Genetic Composition Tunes Host Longevity. Cell 2017, 169, 1249–1262.e13.
  39. Matsuhashi, T.; Sato, T.; Kanno, S.I.; Suzuki, T.; Matsuo, A.; Oba, Y.; Kikusato, M.; Ogasawara, E.; Kudo, T.; Suzuki, K.; et al. Mitochonic Acid 5 (MA-5) Facilitates ATP Synthase Oligomerization and Cell Survival in Various Mitochondrial Diseases. EBioMedicine 2017, 20, 27–38.
  40. Yardeni, T.; Tanes, C.E.; Bittinger, K.; Mattei, L.M.; Schaefer, P.M.; Singh, L.N.; Wu, G.D.; Murdock, D.G.; Wallace, D.C. Host mitochondria influence gut microbiome diversity: A role for ROS. Sci. Signal. 2019, 12, eaaw3159.
  41. Wolf, P.G.; Biswas, A.; Morales, S.E.; Greening, C.; Gaskins, H.R. H2 metabolism is widespread and diverse among human colonic microbes. Gut Microbes 2016, 7, 235–245.
  42. Xie, F.; Jiang, X.; Yi, Y.; Liu, Z.J.; Ma, C.; He, J.; Xun, Z.M.; Wang, M.; Liu, M.Y.; Mawulikplimi Adzavon, Y.; et al. Different effects of hydrogen-rich water intake and hydrogen gas inhalation on gut microbiome and plasma metabolites of rats in health status. Sci. Rep. 2022, 12, 7231.
  43. Colombijn, J.M.; Hooft, L.; Jun, M.; Webster, A.C.; Bots, M.L.; Verhaar, M.C.; Vernooij, R.W. Antioxidants for adults with chronic kidney disease. Cochrane Database Syst. Rev. 2023, 11, CD008176.
  44. Wang, B.; Li, Z.; Mao, L.; Zhao, M.; Yang, B.; Tao, X.; Li, Y.; Yin, G. Hydrogen: A Novel Treatment Strategy in Kidney Disease. Kidney Dis. 2022, 8, 126–136.
  45. Hirano, S.I.; Ichikawa, Y.; Sato, B.; Takefuji, Y.; Satoh, F. Clinical Use and Treatment Mechanism of Molecular Hydrogen in the Treatment of Various Kidney Diseases including Diabetic Kidney Disease. Biomedicines. 2023, 11, 2817.
  46. Yao, W.; Guo, A.; Han, X.; Wu, S.; Chen, C.; Luo, C.; Li, H.; Li, S.; Hei, Z. Aerosol inhalation of a hydrogen-rich solution restored septic renal function. Aging 2019, 11, 12097–12113.
  47. Kawamura, M.; Imamura, R.; Kobayashi, Y.; Taniguchi, A.; Nakazawa, S.; Kato, T.; Namba-Hamano, T.; Abe, T.; Uemura, M.; Kobayashi, H.; et al. Oral administration of Si-based agent attenuates oxidative stress and ischemia-reperfusion injury in a rat model: A novel hydrogen administration method. Front. Med. 2020, 7, 95.
  48. Li, J.; Hong, Z.; Liu, H.; Zhou, J.; Cui, L.; Yuan, S.; Chu, X.; Yu, P. Hydrogen-rich saline promotes the recovery of renal function after ischemia/reperfusion injury in rats via anti-apoptosis and anti-inflammation. Front. Pharmacol. 2016, 7, 106.
  49. Xu, X.; He, X.; Liu, J.; Qin, J.; Ye, J.; Fan, M. Protective effects of hydrogen-rich saline against renal ischemia-reperfusion injury by increased expression of heme oxygenase-1 in aged rats. Int. J. Clin. Exp. Pathol. 2019, 12, 1488–1496.
  50. Nishida, T.; Hayashi, T.; Inamoto, T.; Kato, R.; Ibuki, N.; Takahara, K.; Yoshikawa, Y.; Uchimoto, T.; Saito, K.; Tanda, N.; et al. Dual gas treatment with hydrogen and carbon monoxide attenuates oxidative stress and protects from renal ischemia-reperfusion injury. Transplant. Proc. 2018, 50, 250–258.
  51. Du, H.; Sheng, M.; Wu, L.; Zhang, Y.; Shi, D.; Weng, Y.; Xu, R.; Yu, W. Hydrogen-rich saline attenuates acute kidney injury after liver transplantation via activating p53-mediated autophagy. Transplantation. 2016, 100, 563–570.
  52. Wang, F.; Yu, G.; Liu, S.Y.; Li, J.B.; Wang, J.F.; Bo, L.L.; Qian, L.R.; Sun, X.J.; Deng, X.M. Hydrogen-rich saline protects against renal ischemia/reperfusion injury in rats. J. Surg. Res. 2011, 167, e339–e344.
  53. Chen, J.; Zhang, H.; Hu, J.; Gu, Y.; Shen, Z.; Xu, L.; Jia, X.; Zhang, X.; Ding, X. Hydrogen-rich saline alleviates kidney fibrosis following AKI and retains Klotho expression. Front. Pharmacol. 2017, 8, 499.
  54. Liu, W.; Dong, X.S.; Sun, Y.Q.; Liu, Z. A novel fluid resuscitation protocol: Provide more protection on acute kidney injury during septic shock in rats. Int. J. Clin. Exp. Med. 2014, 15, 919–926.
  55. Shi, Q.; Liao, K.S.; Zhao, K.L.; Wang, W.X.; Zuo, T.; Deng, W.H.; Chen, C.; Yu, J.; Guo, W.Y.; He, X.B.; et al. Hydrogen-rich saline attenuates acute renal injury in sodium taurocholate-induced severe acute pancreatitis by inhibiting ROS and NF-κB pathway. Mediators Inflamm. 2015, 2015, 685043.
  56. Guan, P.; Sun, Z.M.; Luo, L.F.; Zhou, J.; Yang, S.; Zhao, Y.S.; Yu, F.Y.; An, J.R.; Wang, N.; Ji, E.S. Hydrogen protects against chronic intermittent hypoxia induced renal dysfunction by promoting autophagy and alleviating apoptosis. Life Sci. 2019, 225, 46–54.
  57. Cardinal, J.S.; Zhan, J.; Wang, Y.; Sugimoto, R.; Tsung, A.; McCurry, K.R.; Billiar, T.R.; Nakao, A. Oral hydrogen water prevents chronic allograft nephropathy in rats. Kidney Int. 2010, 77, 101–109.
  58. Abe, T.; Li, X.K.; Yazawa, K.; Hatayama, N.; Xie, L.; Sato, B.; Kakuta, Y.; Tsutahara, K.; Okumi, M.; Tsuda, H.; et al. Hydrogen-rich University of Wisconsin solution attenuates renal cold ischemia-reperfusion injury. Transplantation 2012, 94, 14–21.
  59. Nakashima-Kamimura, N.; Mori, T.; Ohsawa, I.; Asoh, S.; Ohta, S. Molecular hydrogen alleviates nephrotoxicity induced by an anti-cancer drug cisplatin without compromising anti-tumor activity in mice. Cancer Chemother. Pharmacol. 2009, 64, 753–761.
  60. Li, F.Y.; Zhu, S.X.; Wang, Z.P.; Wang, H.; Zhao, Y.; Chen, G.P. Consumption of hydrogen-rich water protects against ferric nitrilotriacetate-induced nephrotoxicity and early tumor promotional events in rats. Food Chem. Toxicol. 2013, 61, 248–254.
  61. Peng, Z.; Chen, W.; Wang, L.; Ye, Z.; Gao, S.; Sun, X.; Guo, Z. Inhalation of hydrogen gas ameliorates glyoxylate-induced calcium oxalate deposition and renal oxidative stress in mice. Int. J. Clin. Exp. Pathol. 2015, 8, 2680–2689.
  62. Lu, H.; Ding, J.; Liu, W.; Peng, Z.; Chen, W.; Sun, X.; Guo, Z. UPLC/MS-based metabolomics investigation of the protective effect of hydrogen gas inhalation on mice with calcium oxalate-induced renal injury. Biol. Pharm. Bull. 2018, 41, 1652–1658.
  63. Xu, B.; Zhang, Y.B.; Li, Z.Z.; Yang, M.W.; Wang, S.; Jiang, D.P. Hydrogen-rich saline ameliorates renal injury induced by unilateral ureteral obstruction in rats. Int. Immunopharmacol. 2013, 17, 447–452.
  64. Xing, Z.; Pan, W.; Zhang, J.; Xu, X.; Zhang, X.; He, X.; Fan, M. Hydrogen rich water attenuates renal injury and fibrosis by regulation transforming growth factor-β induced Sirt1. Biol. Pharm. Bull. 2017, 40, 610–615.
  65. Mizutani, A.; Endo, A.A.; Saito, M.; Hara, T.; Nakagawa, M.; Sakuraya, K.; Murano, Y.; Nishizaki, N.; Hirano, D.; Fujinaga, S.; et al. Hydrogen-rich water reduced oxidative stress and renal fibrosis in rats with unilateral ureteral obstruction. Pediatr. Res. 2022, 91, 1695–1702.
  66. Xin, H.G.; Zhang, B.B.; Wu, Z.Q.; Hang, X.F.; Xu, W.S.; Ni, W.; Zhang, R.Q.; Miao, X.H. Consumption of hydrogen-rich water alleviates renal injury in spontaneous hypertensive rats. Mol. Cell Biochem. 2014, 392, 117–124.
  67. Zhu, W.J.; Nakayama, M.; Mori, T.; Nakayama, K.; Katoh, J.; Murata, Y.; Sato, T.; Kabayama, S.; Ito, S. Intake of water with high levels of dissolved hydrogen (H2) suppresses ischemia-induced cardio-renal injury in Dahl salt-sensitive rats. Nephrol. Dial. Transplant. 2011, 26, 2112–2118.
  68. Kelly, K.J. Distant effects of experimental renal ischemia/reperfusion injury. J. Am. Soc. Nephrol. 2003, 14, 1549–1558.
  69. Sugai, K.; Tamura, T.; Sano, M.; Uemura, S.; Fujisawa, M.; Katsumata, Y.; Endo, J.; Yoshizawa, J.; Homma, K.; Suzuki, M.; et al. Daily inhalation of hydrogen gas has a blood pressure-lowering effect in a rat model of hypertension. Sci. Rep. 2020, 10, 20173.
  70. Inoue, T. Neuroimmune system-mediated renal protection mechanisms. Clin. Exp. Nephrol. 2021, 25, 915–924.
  71. Xie, F.; Song, Y.; Yi, Y.; Jiang, X.; Ma, S.; Ma, C.; Li, J.; Zhanghuang, Z.; Liu, M.; Zhao, P.; et al. Therapeutic Potential of Molecular Hydrogen in Metabolic Diseases from Bench to Bedside. Pharmaceuticals 2023, 16, 541.
  72. Kajiyama, S.; Hasegawa, G.; Asano, M.; Hosoda, H.; Fukui, M.; Nakamura, N.; Kitawaki, J.; Imai, S.; Nakano, K.; Ohta, M.; et al. Supplementation of hydrogen-rich water improves lipid and glucose metabolism in patients with type 2 diabetes or impaired glucose tolerance. Nutr. Res. 2008, 28, 137–143.
  73. Ogawa, S.; Ohsaki, Y.; Shimizu, M.; Nako, K.; Okamura, M.; Kabayama, S.; Tabata, K.; Tanaka, Y.; Ito, S. Electrolyzed hydrogen-rich water for oxidative stress suppression and improvement of insulin resistance: A multicenter prospective double-blind randomized control trial. Diabetol. Int. 2021, 13, 209–219.
  74. 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.
  75. Liu, B.; Jiang, X.; Xie, Y.; Jia, X.; Zhang, J.; Xue, Y.; Qin, S. The effect of a low dose hydrogen-oxygen mixture inhalation in midlife/older adults with hypertension: A randomized, placebo-controlled trial. Front. Pharmacol. 2022, 13, 1025487.
  76. Sumida, K.; Yamagata, K.; Kovesdy, C.P. Constipation in CKD. Kidney Int. Rep. 2020, 5, 121–134.
  77. Cosola, C.; Rocchetti, M.T.; di Bari, I.; Acquaviva, P.M.; Maranzano, V.; Corciulo, S.; Di Ciaula, A.; Di Palo, D.M.; La Forgia, F.M.; Fontana, S.; et al. An Innovative Synbiotic Formulation Decreases Free Serum Indoxyl Sulfate, Small Intestine Permeability and Ameliorates Gastrointestinal Symptoms in a Randomized Pilot Trial in Stage IIIb-IV CKD Patients. Toxins 2021, 13, 334.
  78. Huang, K.C.; Yang, C.C.; Lee, K.T.; Chien, C.T. Reduced hemodialysis-induced oxidative stress in end-stage renal disease patients by electrolyzed reduced water. Kidney Int. 2003, 64, 704–714.
  79. Huang, K.C.; Yang, C.C.; Hsu, S.P.; Lee, K.T.; Liu, H.W.; Morisawa, S.; Otsubo, K.; Chien, C.T. Electrolyzed-reduced water reduced hemodialysis-induced erythrocyte impairment in end-stage renal disease patients. Kidney Int. 2006, 70, 391–398.
  80. Tange, Y.; Takesawa, S.; Yoshitake, S. Dialysate with high dissolved hydrogen facilitates dissociation of indoxyl sulfate from albumin. Nephrourol. Mon. 2015, 7, e26847.
  81. Nakayama, M.; Kabayama, S.; Ito, S. The hydrogen molecule as antioxidant therapy: Clinical application in hemodialysis and perspectives. Ren. Replace. Ther. 2016, 2, 23.
  82. Nakayama, M.; Kabayama, S.; Nakano, H.; Zhu, W.J.; Terawaki, H.; Nakayama, K.; Katoh, K.; Satoh, T.; Ito, S. Biological effects of electrolyzed water in hemodialysis. Nephron Clin. Pract. 2009, 112, c9–c15.
  83. Nakayama, M.; Nakano, H.; Hamada, H.; Itami, N.; Nakazawa, R.; Ito, S. A novel bioactive haemodialysis system using dissolved dihydrogen (H2) produced by water electrolysis: A clinical trial. Nephrol. Dial. Transplant. 2010, 25, 3026–3033.
  84. Maeda, K.; Yoshizaki, S.; Iida, T.; Terada, T.; Era, S.; Sakashita, K.; Arikawa, H. Improvement of the fraction of human mercaptalbumin on hemodialysis treatment using hydrogen-dissolved hemodialysis fluid: A prospective observational study. Ren. Replace. Ther. 2016, 2, 42.
  85. Nakayama, M.; Itami, N.; Suzuki, H.; Hamada, H.; Osaka, N.; Yamamoto, R.; Tsunoda, K.; Nakano, H.; Watanabe, K.; Zhu, W.J.; et al. in a 12 month observation. PLoS ONE 2017, 12, e0184535.
  86. Nakayama, M.; Itami, N.; Suzuki, H.; Hamada, H.; Yamamoto, R.; Tsunoda, K.; Osaka, N.; Nakano, H.; Maruyama, Y.; Kabayama, S.; et al. Novel haemodialysis (HD) treatment employing molecular hydrogen (H2)-enriched dialysis solution improves prognosis of chronic dialysis patients: A prospective observational study. Sci. Rep. 2018, 8, 254.
  87. Tsujimoto, Y.; Kuratsune, D.; Kabayama, S.; Miyazaki, M.; Watanabe, Y.; Nishizawa, Y.; Nakayama, M. Amelioration of fatigue in chronic dialysis patients with dialysis solution employing electrolyzed water containing molecular hydrogen (H2) and its association with autonomic function balance. Ren. Replace. Ther. 2021, 7, 58.
  88. Uemura, S.; Kegasa, Y.; Tada, K.; Tsukahara, T.; Kabayama, S.; Yamamoto, T.; Miyazaki, M.; Takada, J.; Nakayama, M. Impact of hemodialysis solutions containing different levels of molecular hydrogen (H2) on the patient-reported outcome of fatigue. Ren. Replace. Ther. 2022, 8, 32.
  89. Mouzakis, F.L.; Khadka, L.B.; da Silva, M.P.; Mottaghy, K. Quantification of dissolved H2 and continuous monitoring of hydrogen-rich water for haemodialysis applications: An experimental study. Int. J. Artif. Organs 2022, 45, 254–261.
More
Video Production Service