Next Article in Journal
Trajectory Planning of the Exit Point for a Cable-Driven Parallel Mechanism by Considering the Homogeneity of Tension Variation
Previous Article in Journal
Temperature-Programmed Reduction of NiO/Al2O3 by Biochar In Situ Generated from Citric Acid
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effects of Alkaline-Reduced Water on Exercise-Induced Oxidative Stress and Fatigue in Young Male Healthy Adults

1
Department of Medical Sciences, Convergence Research Center for Medical Sciences, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si 55069, Jeollabuk-do, Korea
2
Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
3
Department of Laboratory Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
4
Department of Biology, University of the Philippines, Baguio 2600, Philippines
5
Department of Physical Education, Sungkyul University, Anyang 14097, Gyeonggi-do, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Processes 2022, 10(8), 1543; https://doi.org/10.3390/pr10081543
Submission received: 28 June 2022 / Revised: 22 July 2022 / Accepted: 1 August 2022 / Published: 6 August 2022
(This article belongs to the Section Biological Processes and Systems)

Abstract

:
Regular physical activity confers health benefits and improves the general quality of life. Recently, alkaline-reduced water (ARW) consumption has garnered increasing attention in the field of sports. ARW effectively inhibits the oxidative stress generated in cells during high-intensity exercises; however, whether it exerts similar effects during exhaustive exercises remains unknown. This study was designed as a randomized, controlled, crossover, double-blind clinical trial with a single intervention of ARW intake (pH 9.5, 10 mL/kg body weight) after intense exercise. The participants were divided into two groups, wherein they consumed either purified water (PW group) or ARW (ARW group). Blood samples were collected before exercise, immediately after exercise, and 15 min after drinking water. The serum levels of oxidative stress markers and fatigue markers were determined. The results showed that ROS (p < 0.01) and NO levels (p < 0.001) were significantly decreased after ARW intake, and the reduction was more pronounced than that in the PW group. Interestingly, the increase in GPx and MDA levels was mediated by ARW intake (both p < 0.05) after exercise. The levels of fatigue markers, such as lactate (p < 0.001), lactate dehydrogenase (p < 0.001), and phosphate (p < 0.001), were significantly reduced in both groups, with ARW intervention showing more decreased markers. The correlation analysis results showed that ARW may help maintain homeostatic conditions for ROS, antioxidant systems, and fatigue markers. These findings indicate that ARW consumption is effective in reducing oxidative stress and fatigue following exhaustive exercise and that ARW could be used as an antioxidant and anti-fatigue supplement after exhaustive physical exercise.

1. Introduction

It is well known that regular physical activity confers health benefits, improves functional ability, and elevates the general quality of life. Exercise induces a variety of physiological and metabolic changes in the body, depending on the type of exercise or training undertaken. However, exhaustive physical exercise can lead to deleterious health effects, such as muscle damage, inflammation, and oxidative stress [1,2]. Specifically, intense exercise, such as anaerobic exercise, results in repetitive muscle contraction, which increases oxygen consumption in cells and may cause an imbalance of pro-oxidant and antioxidant levels. The resulting oxidative stress leads to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) [2]. Furthermore, increased ROS and RNS levels during intense exercise can overwhelm the endogenous antioxidants, such as catalase and glutathione peroxidase (GPx), thereby causing damage to tissues, cells, and their components, including lipids, proteins, and nucleic acids, and possibly contributing to a rise in the levels of protein carbonyls and lipid peroxidation products, such as malondialdehyde (MDA) [3,4,5]. Intense physical exercise elevates the levels of MDA in blood and other tissues and reduces antioxidant levels [6,7,8]. Besides MDA, other biomarkers of muscle metabolism include lactate, inorganic phosphate, and calcium, which play important roles in muscular fatigue during exercise [9]. During intense exercise, an increase in the blood concentration of lactate can be detected [10,11]. Under similar conditions, the levels of intracellular enzymes, such as lactate dehydrogenase (LDH), are also known to increase, which is attributed to the damage sustained by muscle fibers during excessive exercising [12].
Exogenous antioxidants, which can detoxify certain peroxides by scavenging the ROS, may prevent oxidative damage caused by the ROS produced during intense exercise [13,14]. In this regard, studies have been undertaken to explore the therapeutic effects of alkaline-reduced water (ARW) on various diseases, including gastrointestinal [15], metabolic, and lifestyle diseases [16,17]. ARW is a type of functional water that has an alkaline pH (pH 8–10), active molecular hydrogen (H2), a negative oxidation-reduction potential (ORP), and an ability to protect deoxyribonucleic acid from oxidative damage by scavenging ROS [18]. These properties of ARW, especially as an effective antioxidant and free-radical scavenger, have been indicated in several studies [17,19]. In general, alkaline agents can modify the blood buffering system in the body by enhancing bicarbonate concentration in the blood (thus increasing the pH of the blood), which in turn increases the utilization of lactate in muscles during intense exercise [20,21]. Based on studies conducted in the sports science field, ARW consumption has been demonstrated to enhance athletes’ hydration and anaerobic exercise performance, as well as increase the fasting and post-exercise pH levels in the arteries [22,23,24]. However, the effects of ARW on the oxidative stress and fatigue induced by exhaustive exercise have not been elucidated. Therefore, this clinical study was conducted to evaluate the effect of ARW consumption on the oxidative stress and fatigue markers induced by exhaustive exercise in healthy young participants.

2. Materials and Methods

2.1. Participants

Twenty-four young and healthy male volunteers were recruited for this clinical study according to the following inclusion criteria: (i) healthy male participants aged 19–25 years; (ii) physically active with a normal body mass index (BMI); and (iii) had not smoked for 3 months or consumed alcohol for more than a week prior to the start of the study. Participants were excluded if they had any kind of visible or known disease, hypertension, musculoskeletal injuries in the previous three months, breathing difficulties, metabolic diseases, autoimmune disease, rash, or urticaria. In addition, all participants were assessed for height, weight, the BMI (kg/m2), and heart rate before the start of the study. G*Power analysis [25] was performed to estimate the appropriate sample size (G*Power 3.1.9.7, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany). A sample size of 26 subjects was calculated using the following standard assumptions from a previous study: α = 0.05, power = 0.80, dropout rate = 20%, partial η2 = 0.07. Two participants dropped out in the crossover week and were not included in the study.

2.2. Study Design

This study was designed as a randomized, controlled, crossover, double-blind clinical trial with a single intervention of ARW intake (pH 9.5, 10 mL/kg body weight) after intense exercise. The participants were randomly divided into two groups (n = 12) by a principal investigator, according to the sequence of random numbers obtained through an online number generator. The subjects were randomized into the purified water drinking group (PW group) and ARW drinking group (ARW group). A wash-out period of one week was applied to prevent carry-over before crossing over. All participants fasted for 4 h before experimental testing to standardize the blood biochemistry (Figure 1). The maximal graded exercise test using the Bruce treadmill protocol was used in this study [26]. All participants signed informed consent forms prior to the commencement of the study, and ethical approval was granted by the Institutional Review Board, Yonsei University, Wonju Severance Christian Hospital (IRBN CR222022).
After obtaining the baseline information and the randomization of participants into two groups, blood was collected from each volunteer/participant before and after a 15-min intense exercise session on an indoor treadmill (treadmill, Cosmed T170DE, h/p Cosmos, Nussdorf-Traunstein, Germany) and 15 min after the intake of ARW or PW (10 mL/kg body weight). Participants were instructed to drink their test water according to their grouping within 10 min of intense exercise. Blood samples were centrifuged at 4 °C at 2500 rpm for 15 min, and the serum was separated and stored at −80 °C until needed for the biochemical analysis of oxidative stress and fatigue markers.

2.3. Preparation of Test Water

Both sets of experimental water were generated from an alkaline ionized water generator (CGM MWPI-2101, CERAGEM Co., Ltd., Cheonan, Korea), which is approved as a grade 2 medical device by the Korean Food and Drug Administration. The participants of the ARW group were administered water of pH 9.5 according to their body weights (10 mL/kg).

2.4. Measurement of ROS

Blood serum ROS levels were analyzed using the 2′,7′-dichlorofluorescein diacetate (DCFH-DA) [27] assay kit (Abcam, Cambridge, MA, USA), according to the manufacturer’s instructions. The serum samples (10 μL) were incubated with 20 µM of DCFH-DA (100 µL) for 30 min at 37 °C in the dark. Absorbance was read and analyzed using a DTX-880 multimode microplate reader (Beckman Coulter Inc., Fullerton, CA, USA) at excitation and emission wavelengths of 488 and 525 nm, respectively.

2.5. Measurement of NO Activity

The blood nitrite (NO2) concentration was analyzed using the Griess reaction [28], according to the manufacturer’s instructions (Promega Corp., Madison, WI, USA). The serum samples (50 μL) were mixed with equal volumes of Griess reagent in a 96-well microplate and incubated at room temperature for 15 min. Absorbance was measured at 540 nm using an ABP-00627 absorbance microplate reader (SpectraMax ABS Plus, San Jose, CA, USA).

2.6. Measurement of GPx

Serum GPx concentration was analyzed using a GPx assay kit (Biovision, Milpitas, CA, USA). The serum samples (10 μL) were directly tested in a 96-well pate following the manufacturer’s protocol. The optical density (OD) was measured at 340 nm using an absorbance microplate reader (SpectraMax ABS Plus, San Jose, CA, USA) at T1. The plate was then incubated at 25 °C for 10 min and read again at T2. GPx activity was computed based on the amount of NADPH generated between T1 and T2. GPx activity was quantified using the method described by Flohé and Günzler [29].

2.7. Measurement of MDA

The serum MDA levels were measured using a thiobarbituric acid reactive substances assay kit (Biovision, Milpitas, CA, USA). The assay was performed according to the manufacturer’s instructions. The reaction product was measured calorimetrically at 532 nm using an ABP-00627 absorbance microplate reader (SpectraMax ABS Plus, San Jose, CA, USA). MDA levels were measured using the procedure described by Buege and Aust [30].

2.8. Measurement of Lactate Levels and LDH Assay

Blood lactate assay was performed using a commercial kit according to the manufacturer’s protocol (Biomax, Seoul, Korea). Lactate standards were prepared at the following concentrations: 0, 2, 4, 6, 8, and 10 nmol/well. Samples and standards were added to a 96-well plate. The reaction mix (50 µL) was prepared and added to each sample and the standards. The plate was then incubated in the dark for 30 min at room temperature. The OD was measured at 570 nm using an ABP-00627 absorbance microplate reader (SpectraMax ABS Plus, San Jose, CA, USA). Results were calculated using a standard curve.
Blood LDH activity was measured using an LDH assay kit (Biovision, Milpitas, CA, USA), according to the manufacturer’s protocol. NADH standards were prepared at the following concentrations: 0, 2.5, 5.0, 7.5, 10.0, and 12.5 mM. The samples and standards were added to a 96-well plate. A reaction mix (50 µL) was prepared and added to each sample and standard. The OD was measured at 450 nm using an ABP-00627 absorbance microplate reader (SpectraMax ABS Plus, San Jose, CA, USA) at T1. The plate was incubated at 37 °C for 30 min and was read again at T2. The LDH activity was computed based on the amount of NADH generated between T1 and T2. Lactate and LDH measurements were measured following the methods of Ara et al. [13].

2.9. Phosphate Assay

Blood phosphate activity, as described by Bonet et al. [31], was measured using a phosphate assay kit (Biovision, Milpitas, CA, USA) according to the manufacturer’s instructions. Phosphate standards were prepared at the following concentrations: 0, 200, 400, 600, 800, and 1000 nmol/well. The samples and standards were added to a 96-well plate. The reaction mix (50 µL) was added to each sample and standard and mixed thoroughly. The plates were then incubated in the dark for 1 h at room temperature. The absorbance of the plate was measured at excitation/emission wavelengths of 535/587 nm. Results were calculated using a standard curve.

2.10. Data Management and Statistical Analysis

Data standards are presented as mean ± standard error of the mean (SEM). All data for each marker were normalized, and fold changes were computed according to normal controls and analyzed and compared using a one-way analysis of variance (ANOVA), followed by a multiple comparison test with the GraphPad Prism 8.0 software package (GraphPad, La Jolla, CA, USA). Correlation data were obtained using Pearson’s correlation, and a heatmap was used to obtain the linear relationship between oxidative stress and fatigue markers. Differences were considered statistically significant at p < 0.05.

3. Results

3.1. Characteristics of Study Participants

Table 1 shows the baseline characteristics of the participants. Male participants aged 18–25 years were recruited for the study. Height, weight, BMI, lean mass, and body fat percentage were recorded. Heart rates were monitored during rest and exercise.

3.2. Effect of Drinking ARW after Exercise on Oxidative Stress

ROS and NO levels are considered as markers of oxidative stress. After intense exercise, an increase in ROS and NO levels was observed. The increased serum ROS levels decreased more significantly after water consumption in the ARW group (p < 0.01) than in the PW group (p < 0.05) (Figure 2A). Similarly, the increased NO levels caused by intense exercise were significantly reduced after drinking PW (p < 0.01) and ARW (p < 0.001). Notably, a significant difference in NO levels was observed between the PW and ARW groups (p < 0.05) (Figure 2B).

3.3. Effect of Drinking ARW after Exercise on Antioxidant Enzymes

Serum GPx and MDA levels were measured as markers of antioxidant enzymes. As expected, the antioxidant levels increased post-exercise. The increase in serum GPx levels observed post-exercise was significantly reduced after ARW consumption (p < 0.05) (Figure 3A). This reduction was not observed in the PW group. Similarly, serum MDA levels significantly decreased after water intake in the ARW group (p < 0.05), but not in the PW group (Figure 3B).

3.4. Effects of Drinking ARW after Exercise on Lactate, LDH, and Phosphate Levels

Lactate and LDH levels were markedly increased post-exercise. A significant reduction in these parameters was observed in both the PW (p < 0.001) and ARW groups (p < 0.001) (Figure 4). However, the reduction was more pronounced in the ARW group than in the PW group, especially in LDH levels. Nevertheless, the difference was not statistically significant.

3.5. Correlation of ARW Consumption with Oxidative Stress and Fatigue Markers

The relationship between oxidative stress and fatigue markers was further subjected to correlation analysis through the integration of linear regression models (Figure 5). The heatmap revealed differences in the correlation patterns of oxidative stress and fatigue markers in the pre-exercise, post-exercise, PW, and ARW groups. Interestingly, the correlation pattern in the PW group deviated from that in the rest of the groups. Among the parameters tested, GPx activity showed a weak but statistically significant positive association with lactate (r = 0.381, p < 0.05) and LDH (r = 0.375, p < 0.05) levels in the pre-exercise group. This mild positive correlation between GPx and LDH levels (r = 0.367, p < 0.05) was maintained in the post-exercise group. Surprisingly, PW and ARW treatments altered the interaction of GPx with lactate and LDH. Administration of PW following exercise eliminated the relationship between GPx and three variables. In contrast, ARW consumption significantly strengthened the relationship between GPx and LDH (r = 0.527, p < 0.05). Additionally, ARW consumption strengthened the relationship between GPx and MDA (r = 0.478, p < 0.05).

4. Discussion

In this study, we investigated the effects of ARW consumption on exercise-induced oxidative stress and fatigue markers in healthy young adults. We found that the administration of ARW reduced free radical generation, mediated antioxidant enzyme activity, and improved the levels of fatigue markers after exhaustive exercise in healthy adults.
In recent years, the popularity of ARW in sports has grown rapidly. Consumption of ARW, which has a high mineral content, alkaline pH, and antioxidants with negative ORP, has been proposed as an alternative sports drink by various researchers [22,23,24]. A few studies have observed a positive effect of ARW consumption on exercise performance, which stems from improving the acid balance, hydration, and utilization of lactate [22,23]. Intense exercises increase the demand for oxygen in the skeletal muscle and damage the muscles by excessive production of free radicals, due to oxidation of biomolecules [33,34]. Muscular damage due to free radicals is associated with oxidative stress, which refers to an imbalance between the production of ROS and RNS, and the potential of the antioxidant defense system and the antioxidant enzymes to scavenge the ROS and RNS being produced. Exercise generally increases NO formation in the vasculature via endothelial nitric oxide synthase. Oxidative stress plays important roles in both fatigue- and chronic fatigue-related conditions [33,35]. Under normal conditions, regular physical exercise improves the body function, but during intense exercise, excessive production of ROS causes the peroxidation of lipids in the cell membrane, resulting in structural damage to the cells of skeletal muscles [36]. Therefore, we examined the serum levels of biomarkers associated with oxidative stress and antioxidant biomarkers before and after exhaustive exercise and post administration of ARW. The results showed that the serum levels of both ROS and NO were markedly increased after intense exercise, and ARW administration inhibited the increase in ROS and NO levels in the blood. In fact, NO levels were significantly reduced in the ARW group than in the PW group. Previous research has demonstrated that ARW scavenges ROS and inhibits ROS-induced DNA damage [37], thus functioning as an antioxidant.
Antioxidant enzymes, such as superoxide dismutase (SOD) and GPx, are regarded as the principal components of enzymatic antioxidant defense systems that fight fatigue. They play a protective role in cells by reducing the generation of active oxygen radicals. Glutathione, an important intracellular antioxidant, is converted to its oxidized form glutathione disulfide by GPx, when used as a scavenger against free radicals [38]. Studies have reported that the levels of antioxidant enzymes, such as GPx, SOD, and catalase, increase during high-intensity exercise [39,40]; our results were similar to those findings. Intense exercise increased GPx levels in the blood in both the groups. However, following ARW administration, GPx levels markedly decreased post-exercise. This result is consistent with the results of the studies that reported that ARW consumption may exert positive effects by increasing antioxidant defense and reducing oxidant production after exercise [41,42]. Although SOD and catalase assays might be needed to confirm this antioxidant capacity, our results showed that GPx activity was mediated by ARW an antioxidant defense. Moreover, ARW exerts additional beneficial effects by scavenging the free radicals produced during exercise, thereby facilitating the maintenance of muscle performance and redox homeostasis during consecutive days of exercising [43]. Exhaustive exercises also increase the concentrations of lipid peroxides in the skeletal muscles and blood [44], thereby causing free radical-induced damage to membrane integrity. Administration of ARW could stabilize the lipid structure of the membrane disrupted by the assault of free radicals. This stabilizing effect was observed in the present study, as reflected by the reduction in MDA levels following the administration of ARW after exercise. These findings indicate that ARW exhibits antioxidant activity that prevents lipid peroxidation and protects the body from oxidative stress after exhaustive exercise.
Furthermore, we evaluated fatigue-related markers in the blood samples of all participants in both the groups. Intense exercise leads to the switching of aerobic muscular activity to anaerobic metabolism, which results in the conversion of pyruvate to lactic acid catalyzed by LDH [44]. Lactic acid or lactate is a common marker of fatigue. An excessive production of lactate inhibits the contractile activity of muscles and glycolysis. A similar mechanism underlying the increase in serum levels of LDH, a key enzyme marker required for lactate production, has been observed. Increased LDH levels are known to cause cellular necrosis and tissue damage [45,46,47]. Therefore, the accumulation of serum lactate and LDH could be an indication of the speed and degree of fatigue developed in participants after exhaustive exercise. As expected, exhaustive exercise on a treadmill for 15 min markedly elevated the serum levels of LA and LDH, whereas this increase was remarkably attenuated after the post-exercise intake of both PW and ARW. These results indicated that drinking either PW or ARW could enhance exercise endurance by reducing the levels of fatigue markers in the blood, wherein ARW could have mediated the reduction in the levels of these markers far more effectively than PW. Furthermore, we analyzed inorganic phosphate levels in the blood after intense exercise and after administration of ARW, as inorganic phosphate is also an important marker of fatigue [48]. In fact, impairment/inhibition of calcium production from the sarcoplasmic reticulum has been identified as a contributor to fatigue in skeletal muscle fibers, owing to the involvement of inorganic phosphate [49]. Similarly, our results showed that inorganic phosphate levels in the blood after intense exercise markedly increased, whereas the administration of ARW gradually reduced phosphate levels. These findings indicate that both PW and ARW may be effective in reducing fatigue, but ARW administration shows more promising results. Further study may be needed to explore the mechanisms underlying this effect. In addition, as intense exercise disturbs the acid-base balance in the body [50,51], more biochemical assays in this study might be needed to check if the ARW mediates metabolic acidosis, as shown by Chycki and colleagues [22]. Understanding the interaction between oxidative and fatigue markers may provide insights into the cumulative effect of ARW on exercise-induced oxidative stress.
With this, we anticipate that evaluating the interactions among ROS, antioxidant systems, and fatigue markers will clearly demonstrate the impact of experimental water on intensive exercise. Consequently, when the correlation coefficients of the variables in the present study were calculated, we observed that the association of GPx with LDH and MDA changed considerably. Remarkably, a positive correlation was observed between GPx and LDH levels in the pre-exercise group. This correlation persisted even after exercise. We hypothesized that the increase in GPx levels during resistance training was an initial response to the increase in ROS levels. However, excessive ROS production destabilized the antioxidant machinery. Numerous studies have demonstrated that antioxidant supplementation is necessary to restore redox equilibrium [14,44]. We propose that the administration of antioxidants after intense exercise is critical to maintaining a favorable relationship between GPx and other fatigue markers, such as LDH and MDA. Surprisingly, this relationship was preserved in the ARW group but not in the PW group. Although the effect of antioxidant administration after exercise remains controversial, this study showed that ARW intake may be a suitable alternative for maintaining homeostatic conditions of ROS, antioxidant systems, and fatigue markers.
However, this study had some limitations. First, the sample size was relatively small; a bigger study group might be warranted to draw conclusive results. Second, this study focused only on male participants of 19 to 25 years of age. As results might be affected by gender and age, including a female cohort and different age groups in further studies may warrant conclusive results. Third, the duration of the wait period after water intake and before testing was limited to 15 min. A better understanding of the efficacy of ARW consumption could be achieved by evaluating the prolonged effect of ARW on the study parameters to confirm the reversal effects and implications of drinking ARW and implications of drinking ARW, as reported by Weidman et al., wherein alkaline water showed rehydration and recovery at 120 min [41].

5. Conclusions

Our findings demonstrated that ARW administration is effective in preventing oxidative stress and fatigue following exhaustive exercise. Although the effect of antioxidant administration after exercise remains controversial, this study showed that ARW intake may be a suitable alternative for maintaining homeostatic conditions of ROS, antioxidant systems, and fatigue markers. Therefore, ARW may be used as an antioxidant and anti-fatigue supplement by individuals who participate in exhaustive endurance physical exercise. Further study may be needed to explore the mechanisms underlying this effect.

Author Contributions

Conceptualization, K.-J.L., M.L. and J.B.; writing—original draft preparation, J.B., A.F. and J.M.A.; writing—review and editing, M.L., K.-J.L., I.-Y.C. and C.-S.K.; data curation, J.B., A.F., J.M.A., M.L. and K.-E.K.; investigation, I.-Y.C. W.-S.A. and H.-Y.J.; supervision, C.-S.K., M.L. and K.-J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Institutional Review Board Statement

This study was performed after obtaining approval from the institutional review board (IRB) of the Wonju Severance Christian Hospital in the Republic of Korea (IRB number: CR222022).

Informed Consent Statement

Written informed consent has been obtained from the participants to publish this paper.

Data Availability Statement

The data presented in this study are available in the article (tables and figures).

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Knez, W.L.; Coombes, J.S.; Jenkins, D.G. Ultra-endurance exercise and oxidative damage. Sports Med. 2006, 36, 429–441. [Google Scholar] [CrossRef] [PubMed]
  2. Zuo, L.; Nogueira, L.; Hogan, M.C. Reactive oxygen species formation during tetanic contractions in single isolated xenopus myofibers. J. Appl. Physiol. 2011, 111, 898–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Neubauer, O.; Reichhold, S.; Nersesyan, A.; König, D.; Wagner, K.-H. Exercise-induced DNA damage: Is there a relationship with inflammatory responses? Exerc. Immunol. Rev. 2008, 14, 51–72. [Google Scholar] [PubMed]
  4. Powers, S.K.; Nelson, W.B.; Hudson, M.B. Exercise-induced oxidative stress in humans: Cause and consequences. Free Radic. Biol. Med. 2011, 51, 942–950. [Google Scholar] [CrossRef] [PubMed]
  5. Vollaard, N.B.; Shearman, J.P.; Cooper, C.E. Exercise-induced oxidative stress. Sports Med. 2005, 35, 1045–1062. [Google Scholar] [CrossRef]
  6. Davies, K.J.; Quintanilha, A.T.; Brooks, G.A.; Packer, L. Free radicals and tissue damage produced by exercise. Biochem. Biophys. Res. Commun. 1982, 107, 1198–1205. [Google Scholar] [CrossRef]
  7. Duthie, G.G.; Robertson, J.D.; Maughan, R.J.; Morrice, P.C. Blood antioxidant status and erythrocyte lipid peroxidation following distance running. Arch. Biochem. Biophys. 1990, 282, 78–83. [Google Scholar] [CrossRef]
  8. Marzatico, F.; Pansarasa, O.; Bertorelli, L.; Somenzini, L.; Della Valle, G. Blood free radical antioxidant enzymes and lipid peroxides following long-distance and lactacidemic performances in highly trained aerobic and sprint athletes. J. Sports Med. Phys. Fit. 1997, 37, 235–239. [Google Scholar]
  9. Todd, J. Lactate: Valuable for physical performance and maintenance of brain function during exercise. Biosci. Horiz. 2014, 7, hzu001. [Google Scholar] [CrossRef]
  10. Garnacho-Castaño, M.V.; Domínguez, R.; Ruiz-Solano, P.; Maté-Muñoz, J.L. Acute physiological and mechanical responses during resistance exercise at the lactate threshold intensity. J. Strength Cond. Res. 2015, 29, 2867–2873. [Google Scholar] [CrossRef]
  11. Nikseresht, A.; Yabande, I.; Rahmanian, K.; Jahromi, A.S. Blood lactate level in elite boy swimmers after lactate tolerance exercise test. Biomed. Res. Ther. 2017, 4, 1318–1326. [Google Scholar] [CrossRef] [Green Version]
  12. Lippi, G.; Schena, F.; Salvagno, G.; Montagnana, M.; Gelati, M.; Tarperi, C.; Banfi, G.; Guidi, G. Acute variation of biochemical markers of muscle damage following a 21-km, half-marathon run. Scand. J. Clin. Lab. Investig. 2008, 68, 667–672. [Google Scholar] [CrossRef] [PubMed]
  13. Ara, J.; Fadriquela, A.; Ahmed, M.F.; Bajgai, J.; Sajo, M.E.J.; Lee, S.P.; Kim, T.S.; Jung, J.Y.; Kim, C.S.; Kim, S.-K. Hydrogen water drinking exerts antifatigue effects in chronic forced swimming mice via antioxidative and anti-inflammatory activities. BioMed. Res. Int. 2018, 2018, 2571269. [Google Scholar] [CrossRef]
  14. Morillas-Ruiz, J.; García, J.V.; López, F.; Vidal-Guevara, M.; Zafrilla, P. Effects of polyphenolic antioxidants on exercise-induced oxidative stress. Clin. Nutr. 2006, 25, 444–453. [Google Scholar] [CrossRef] [PubMed]
  15. Bajgai, J.; Kim, C.-S.; Rahman, M.; Jeong, E.-S.; Jang, H.-Y.; Kim, K.-E.; Choi, J.; Cho, I.-Y.; Lee, K.-J.; Lee, M. Effects of alkaline-reduced water on gastrointestinal diseases. Processes 2022, 10, 87. [Google Scholar] [CrossRef]
  16. Delos Reyes, F.S.L.G.; Mamaril, A.C.C.; Matias, T.J.P.; Tronco, M.K.V.; Samson, G.R.; Javier, N.D.; Fadriquela, A.; Antonio, J.M.; Sajo, M.E. The search for the elixir of life: On the therapeutic potential of alkaline reduced water in metabolic syndromes. Processes 2021, 9, 1876. [Google Scholar] [CrossRef]
  17. Ignacio, R.M.C.; Joo, K.-B.; Lee, K.-J. Clinical effect and mechanism of alkaline reduced water. J. Food Drug Anal. 2012, 20, 394–397. [Google Scholar] [CrossRef]
  18. Hamasaki, T.; Harada, G.; Nakamichi, N.; Kabayama, S.; Teruya, K.; Fugetsu, B.; Gong, W.; Sakata, I.; Shirahata, S. Electrochemically reduced water exerts superior reactive oxygen species scavenging activity in ht1080 cells than the equivalent level of hydrogen-dissolved water. PLoS ONE 2017, 12, e0171192. [Google Scholar] [CrossRef]
  19. Shirahata, S.; Hamasaki, T.; Teruya, K. Advanced research on the health benefit of reduced water. Trends Food Sci. Technol. 2012, 23, 124–131. [Google Scholar] [CrossRef] [Green Version]
  20. Sahlin, K. Intracellular ph and energy metabolism in skeletal muscle of man with special reference to exercise. Acta Physiol. Scand. Suppl. 1978, 455, 1–56. [Google Scholar]
  21. Zajac, A.; Cholewa, J.; Poprzecki, S.; Waskiewicz, Z.; Langfort, J. Effects of sodium bicarbonate ingestion on swim performance in youth athletes. J. Sports Sci. Med. 2009, 8, 45–50. [Google Scholar]
  22. Chycki, J.; Kurylas, A.; Maszczyk, A.; Golas, A.; Zajac, A. Alkaline water improves exercise-induced metabolic acidosis and enhances anaerobic exercise performance in combat sport athletes. PLoS ONE 2018, 13, e0205708. [Google Scholar] [CrossRef]
  23. Kurylas, A.; Zajac, T.; Chycki, J.; Maszczyk, A.Z.A.; Zajac, A. naerobic performance and acid-base balance in basketball players after the consumption of highly alkaline water. Trends Food Sci. Technol. 2018, 5, 134–139. [Google Scholar]
  24. Ostojic, S.M.; Stojanovic, M.D. Hydrogen-rich water affected blood alkalinity in physically active men. Res. Sports Med. 2014, 22, 49–60. [Google Scholar] [CrossRef]
  25. Faul, F.; Erdfelder, E.; Lang, A.-G.; Buchner, A. G * power 3: A flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav. Res. Methods 2007, 39, 175–191. [Google Scholar] [CrossRef]
  26. Riebe, D.; Ehrman, J.K.; Liguori, G.; Magal, M. ACSM’s guidelines for exercise testing and prescription. In American College of Sports Medicine; Wolters Kluwer: Alphen aan den Rijn, The Netherlands, 2018. [Google Scholar]
  27. Eruslanov, E.; Kusmartsev, S. Identification of ros using oxidized dcfda and flow-cytometry. In Advanced Protocols in Oxidative Stress II; Springer: Berlin/Heidelberg, Germany, 2010; pp. 57–72. [Google Scholar]
  28. Poveda, J.; Riestra, A.; Salas, E.; Cagigas, M.; López-Somoza, C.; Amado, J.; Berrazueta, J. Contribution of nitric oxide to exercise-induced changes in healthy volunteers: Effects of acute exercise and long-term physical training. Eur. J. Clin. Investig. 1997, 27, 967–971. [Google Scholar] [CrossRef]
  29. Flohé, L.; Günzler, W.A. [12] assays of glutathione peroxidase. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 1984; Volume 105, pp. 114–120. [Google Scholar]
  30. Buege, J.A.; Aust, S.D. [30] Microsomal lipid peroxidation. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 1978; Volume 52, pp. 302–310. [Google Scholar]
  31. Bonet, J.B.; Javierre, C.; Guimarães, J.T.; Martins, S.; Rizo-Roca, D.; Beleza, J.; Viscor, G.; Pagès, T.; Magalhães, J.; Torrella, J.R. Benefits on hematological and biochemical parameters of a high-intensity interval training program for a half-marathon in recreational middle-aged women runners. Int. J. Environ. Health Res. 2022, 19, 498. [Google Scholar] [CrossRef]
  32. Bruce, R.A.; Kusumi, F.; Hosmer, D. Maximal oxygen intake and nomographic assessment of functional aerobic impairment in cardiovascular disease. Am. Heart J. 1973, 85, 546–562. [Google Scholar] [CrossRef]
  33. Chen, Q.; Wei, P. Biotechnology. Icariin supplementation protects mice from exercise-induced oxidant stress in liver. Food Sci. Biotechnol. 2013, 22, 1–5. [Google Scholar]
  34. Dillard, C.; Litov, R.; Savin, W.; Dumelin, E.; Tappel, A.L. Effects of exercise, vitamin e, and ozone on pulmonary function and lipid peroxidation. J. Appl. Physiol. 1978, 45, 927–932. [Google Scholar] [CrossRef]
  35. Yan, F.; Wang, B.; Zhang, Y. Polysaccharides from cordyceps sinensis mycelium ameliorate exhaustive swimming exercise-induced oxidative stress. Pharm. Biol. 2014, 52, 157–161. [Google Scholar] [CrossRef]
  36. Zhonghui, Z.; Xiaowei, Z.; Fang, F. Ganoderma lucidum polysaccharides supplementation attenuates exercise-induced oxidative stress in skeletal muscle of mice. Saudi J. Biol. Sci. 2014, 21, 119–123. [Google Scholar] [CrossRef] [Green Version]
  37. 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. [Google Scholar] [CrossRef]
  38. Powers, S.K.; Criswell, D.; Lawler Martin, D.; Herb, R.A.; Dudley, G. Influence of exercise and fiber type on antioxidant enzyme activity in rat skeletal muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 1994, 266, R375–R380. [Google Scholar] [CrossRef]
  39. Ji, L.L.; Fu, R.; Mitchell, E.W. Glutathione and antioxidant enzymes in skeletal muscle: Effects of fiber type and exercise intensity. J. Appl. Physiol. 1992, 73, 1854–1859. [Google Scholar] [CrossRef]
  40. Ji, L.L.; Fu, R. Responses of glutathione system and antioxidant enzymes to exhaustive exercise and hydroperoxide. J. Appl. Physiol. 1992, 72, 549–554. [Google Scholar] [CrossRef]
  41. Weidman, J.; Holsworth, R.E.; Brossman, B.; Cho, D.J.; Cyr, J.S.; Fridman, G.J. Effect of electrolyzed high-pH alkaline water on blood viscosity in healthy adults. J. Int. Soc. Sports Nutr. 2016, 13, 45. [Google Scholar] [CrossRef] [Green Version]
  42. Rias, Y.A.; Kurniawan, A.L.; Chang, C.W.; Gordon, C.J.; Tsai, H.T.J.A. Synergistic effects of regular walking and alkaline electrolyzed water on decreasing inflammation and oxidative stress, and increasing quality of life in individuals with type 2 diabetes: A community based randomized controlled trial. Antioxidants 2020, 9, 946. [Google Scholar] [CrossRef]
  43. Dobashi, S.; Takeuchi, K.; Koyama, K. Hydrogen-rich water suppresses the reduction in blood total antioxidant capacity induced by 3 consecutive days of severe exercise in physically active males. Med. Gas Res. 2020, 10, 21–26. [Google Scholar]
  44. Kayatekin, B.; Gönenç, S.; Açikgöz, O.; Uysal, N.; Dayi, A. Effects of sprint exercise on oxidative stress in skeletal muscle and liver. Eur. J. Appl. Physiol. 2002, 87, 141–144. [Google Scholar] [CrossRef]
  45. Vetharaniam, I.; Thomson, R.A.; Devine, C.; Daly, C. Modelling muscle energy-metabolism in anaerobic muscle. Meat Sci. 2010, 85, 134–148. [Google Scholar] [CrossRef]
  46. Duan, F.-F.; Guo, Y.; Li, J.-W.; Yuan, K. Antifatigue effect of luteolin-6-c-neohesperidoside on oxidative stress injury induced by forced swimming of rats through modulation of nrf2/are signaling pathways. Oxidative Med. Cell. Longev. 2017, 2017, 3159358. [Google Scholar] [CrossRef]
  47. Liu, J.; Du, C.; Wang, Y.; Yu, Z. Anti-fatigue activities of polysaccharides extracted from hericium erinaceus. Exp. Ther. Med. 2015, 9, 483–487. [Google Scholar] [CrossRef] [Green Version]
  48. Wan, J.-J.; Qin, Z.; Wang, P.-Y.; Sun, Y.; Liu, X. Muscle fatigue: General understanding and treatment. Exp. Mol. Med. 2017, 49, e384. [Google Scholar] [CrossRef] [Green Version]
  49. Allen, D.G.; Lamb, G.D.; Westerblad, H. Impaired calcium release during fatigue. J. Appl. Physiol. 2008, 104, 296–305. [Google Scholar] [CrossRef]
  50. Ratel, S.; Duche, P.; Hennegrave, A.; Van Praagh, E.; Bedu, M. Acid-base balance during repeated cycling sprints in boys and men. J. Appl. Physiol. 2002, 92, 479–485. [Google Scholar] [CrossRef] [Green Version]
  51. Russell, M.; Kingsley, M.I.C. Changes in acid-base balance during simulated soccer match play. J. Strength Cond. Res. 2012, 26, 2593–2599. [Google Scholar] [CrossRef]
Figure 1. Experimental design of the cross-over study.
Figure 1. Experimental design of the cross-over study.
Processes 10 01543 g001
Figure 2. Effects of ARW consumption on ROS and NO levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of ROS (A) and NO (B) were calculated pre- to post-exercise and post-exercise to post-drinking. The statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). * p < 0.05, ** p < 0.01, *** p < 0.001. Abbreviations: ROS, reactive oxygen species; NO, nitric oxide; PW, purified water-drinking group; ARW, alkaline reduced water-drinking group.
Figure 2. Effects of ARW consumption on ROS and NO levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of ROS (A) and NO (B) were calculated pre- to post-exercise and post-exercise to post-drinking. The statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). * p < 0.05, ** p < 0.01, *** p < 0.001. Abbreviations: ROS, reactive oxygen species; NO, nitric oxide; PW, purified water-drinking group; ARW, alkaline reduced water-drinking group.
Processes 10 01543 g002
Figure 3. Effects of ARW consumption on GPx and MDA levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of GPx (A) and MDA (B) were calculated pre- to post-exercise and post-exercise to post-drinking. Statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). * p < 0.05. Abbreviations: GPx, glutathione peroxidase; MDA, malondialdehyde; ns, not significant; PW, purified water-drinking group; ARW, alkaline reduced water-drinking group.
Figure 3. Effects of ARW consumption on GPx and MDA levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of GPx (A) and MDA (B) were calculated pre- to post-exercise and post-exercise to post-drinking. Statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). * p < 0.05. Abbreviations: GPx, glutathione peroxidase; MDA, malondialdehyde; ns, not significant; PW, purified water-drinking group; ARW, alkaline reduced water-drinking group.
Processes 10 01543 g003
Figure 4. Effects of ARW consumption on lactate and LDH levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of lactate (A), LDH (B), and phosphate (C) were calculated pre- to post-exercise and post-exercise to post-drinking. Statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). *** p < 0.001. Abbreviations: LDH, lactate dehydrogenase; PW, purified water; ARW, alkaline reduced water.
Figure 4. Effects of ARW consumption on lactate and LDH levels of healthy individuals after intense exercise. Blood was collected from the participants pre- and post-exercise. Participants consumed PW or ARW after exercise and blood was collected. Fold changes in the serum levels of lactate (A), LDH (B), and phosphate (C) were calculated pre- to post-exercise and post-exercise to post-drinking. Statistical significance was analyzed via a one-way ANOVA (Mann–Whitney U test). *** p < 0.001. Abbreviations: LDH, lactate dehydrogenase; PW, purified water; ARW, alkaline reduced water.
Processes 10 01543 g004
Figure 5. Pearson correlation between oxidative stress and fatigue markers. (A) Heatmap depicting the linear relationship between oxidative stress and fatigue markers. The color is directly proportional to the size of the circle. The deeper the hue, the larger the circle size. The circle is rated based on the absolute value of the correlation coefficient (R). The greater the size of the circle, the greater the correlation. (B) Pearson correlation of GPx with other oxidative stress and fatigue markers in the pre- and post-exercise groups after the consumption of PW and ARW. Abbreviations: PW, purified water; ARW, alkaline reduced water; ROS, reactive oxygen species; NO, nitric oxide; GPx, glutathione peroxidase; MDA, malondialdehyde; LDH, lactate dehydrogenase.
Figure 5. Pearson correlation between oxidative stress and fatigue markers. (A) Heatmap depicting the linear relationship between oxidative stress and fatigue markers. The color is directly proportional to the size of the circle. The deeper the hue, the larger the circle size. The circle is rated based on the absolute value of the correlation coefficient (R). The greater the size of the circle, the greater the correlation. (B) Pearson correlation of GPx with other oxidative stress and fatigue markers in the pre- and post-exercise groups after the consumption of PW and ARW. Abbreviations: PW, purified water; ARW, alkaline reduced water; ROS, reactive oxygen species; NO, nitric oxide; GPx, glutathione peroxidase; MDA, malondialdehyde; LDH, lactate dehydrogenase.
Processes 10 01543 g005
Table 1. Baseline characteristics of the participants in the cross-over study.
Table 1. Baseline characteristics of the participants in the cross-over study.
VariablesUnitMedianMinimumMaximum
Ageyear22.018.025.0
Heightcm175.1164.4185.6
Weightkg70.561.583.9
BMIkg/m223.419.527.1
Lean masskg33.129.142.2
Body fat%16.412.221.8
RPE max 19.017.020.0
HR maxbeats·min−1200.0186.0214.0
Exercise timeseconds826.0717.01037.0
VO2 max *mL·kg·min−150.144.062.0
*: VO2 max predictive formula [32] = 6.70–2.82 (gender: male = 1, female = 2) + 0.056 (Ts). Abbreviations: BMI, body mass index; RPE, rate of perceived exertion; HR, heart rate; VO2 max, maximal oxygen consumption.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, M.; Fadriquela, A.; Antonio, J.M.; Kim, C.-S.; Cho, I.-Y.; Kim, K.-E.; An, W.-S.; Jang, H.-Y.; Bajgai, J.; Lee, K.-J. Effects of Alkaline-Reduced Water on Exercise-Induced Oxidative Stress and Fatigue in Young Male Healthy Adults. Processes 2022, 10, 1543. https://0-doi-org.brum.beds.ac.uk/10.3390/pr10081543

AMA Style

Lee M, Fadriquela A, Antonio JM, Kim C-S, Cho I-Y, Kim K-E, An W-S, Jang H-Y, Bajgai J, Lee K-J. Effects of Alkaline-Reduced Water on Exercise-Induced Oxidative Stress and Fatigue in Young Male Healthy Adults. Processes. 2022; 10(8):1543. https://0-doi-org.brum.beds.ac.uk/10.3390/pr10081543

Chicago/Turabian Style

Lee, Mihyun, Ailyn Fadriquela, Jayson M. Antonio, Cheol-Su Kim, Il-Young Cho, Ka-Eun Kim, Wan-Sik An, Hong-Young Jang, Johny Bajgai, and Kyu-Jae Lee. 2022. "Effects of Alkaline-Reduced Water on Exercise-Induced Oxidative Stress and Fatigue in Young Male Healthy Adults" Processes 10, no. 8: 1543. https://0-doi-org.brum.beds.ac.uk/10.3390/pr10081543

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