Open Access

Hydrogen gas alleviates acute alcohol‑induced liver injury by inhibiting JNK activation

  • Authors:
    • Yaxing Zhang
    • Mingmin Bi
    • Zifeng Chen
    • Min Dai
    • Ge  Zhou
    • Yuxuan Hu
    • Hongzhi Yang
    • Weibing Guan
  • View Affiliations

  • Published online on: March 1, 2021     https://doi.org/10.3892/etm.2021.9884
  • Article Number: 453
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Binge alcohol drinking is fast becoming a global health concern, with the liver among the first organ involved and the one afflicted with the greatest degree of injury. Oxidative stress, alterations in hepatic metabolism, immunity and inflammation have all been reported to contribute to the development of alcoholic liver disease (ALD). Hydrogen gas (H2) serves a key role in the modulation of hepatic redox, immune and inflammatory homeostasis. However, the effects of treatment using intraperitoneal injection of H2 on ALD remain unexplored. Therefore, the aim of the present study was to investigate the effects and underlying mechanism of intraperitoneal injection of H2 on acute alcohol‑induced liver injury in a mouse model. H2 was administered by daily intraperitoneal injections (1.0 ml/100 g) for 4 days. On day 4, the mice received H2 after fasting for 5.5 h. After 30 min, the mice were administered with 33% (v/v) ethanol at a cumulative dose of 4.5 g/kg body weight by four equally divided gavages at 20‑min intervals. Blood and liver tissues were collected at 16 h after the first ethanol gavage. Subsequently, serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), triglyceride and total cholesterol (TC) levels were analyzed using an Automatic Clinical Analyzer. Hepatic JNK activity and GAPDH levels were examined by western blotting. It was observed that acute ethanol gavage induced liver injury, as indicated by significantly increased serum ALT and AST levels, which were effectively decreased by H2 at 16 h after the first ethanol gavage. In addition, H2 treatment reduced serum TC levels in the Alcohol+H2 group when compared with those in Alcohol group. Mechanistically, H2 attenuated hepatic JNK phosphorylation induced by acute ethanol gavage. Therefore, the results of the present study demonstrated that treatment with exogenous H2 by intraperitoneal injection may alleviate acute alcohol‑induced liver injury by inhibiting hepatic JNK activation, which may represent a novel therapeutic strategy for ALD.

Introduction

Excessive alcohol intake, as result of binge drinking as well as chronic alcohol consumption of >40 g per day, is becoming a global healthcare concern (1,2). Globally, ~2 billion individuals consume alcohol regularly, where >75 million are diagnosed with disorders associated with alcohol abuse and are at risk of developing alcohol-associated liver diseases (3). The liver typically sustains the earliest and the greatest degree of tissue injury caused by excessive drinking, since it is the primary site of ethanol metabolism (1,4). Excessive alcohol consumption causes alcoholic liver disease (ALD), which is characterized by a wide spectrum of hepatic pathologies, from reversible fatty liver (simple steatosis) to acute alcoholic hepatitis, chronic fibrosis and cirrhosis and superimposed hepatocellular carcinoma (HCC) (2,5,6). Mechanistically, pathogenesis of ALD includes genetic susceptibility, oxidative stress, hepatic metabolism alteration and steatosis, acetaldehyde-mediated toxicity, cytokine- and chemokine-induced inflammation, alterations in immunity and dysbiosis, epigenetic changes and modifications in the regenerative process (2,6-8). Alcohol abstinence achieved by psychosomatic intervention is at present the best treatment for all stages of ALD (2). However, drinking discontinuation may be difficult, for example, the cheap price of hard liquor, easy accessibility to alcohol, and alcohol advertisement make it very difficult to prevent the increase in ALD (9). Therefore, new strategies for alleviating alcohol-induced liver injury remain to be in demand.

Hydrogen gas (H2) is the lightest and diffusible gas molecule that has been shown to confer potent antioxidant and anti-inflammatory properties (10,11). It can be absorbed into the blood circulation, such that it reaches the target organ either by blood circulation or free diffusion (11). Therefore, treatments involving exogenous H2, including breathing H2 gas, injection with H2-rich saline and drinking H2-rich water, may protect against excessive oxidative stress- and inflammation-related liver damage, including liver injury induced by drugs, sepsis, bile duct ligation, ischemia/reperfusion (I/R), CO2 pneumoperitoneum and chronic intermittent hypoxia, in addition to non-alcoholic fatty liver disease (NAFLD) (12-17). Furthermore, drinking H2-rich water has been indicated to protect against chronic ethanol-induced hepatotoxicity (18). These previous observations suggest that H2 serves an important role in modulating hepatic redox, immune and inflammatory homeostasis (11).

Previous studies have demonstrated that supplementation with exogenous H2 by intraperitoneal injection may improve lipopolysaccharide (LPS)-induced cardiac dysfunction, isoproterenol-induced cardiac hypertrophy, pressure overload-induced vascular hypertrophy, and cardiopulmonary cerebral resuscitation in a cardiac arrest rabbit model (19-23). These were mediated by the suppression of excessive oxidative stress and inflammatory responses (19-23). However, the effects of intraperitoneal injection of H2 on ALD remain unclear.

Therefore, the aim of the present study was to investigate the effect of H2 intraperitoneal injection on acute alcohol-induced liver injury in a mouse model and to elucidate the potential underlying mechanisms.

Materials and methods

Drugs

In total, 99,999% H2 (Dalian Special Gases Co., Ltd.) was injected into a vacuumed aseptic soft plastic infusion bag (100 ml; Hebei Tiancheng Pharmaceutical Co., Ltd.) from a seamless steel gas cylinder under sterile conditions, as previously described (Fig. 1) (20-23). Anhydrous ethanol (Guangzhou Chemical Reagent Factory) was dissolved in double-distilled water to obtain 33% (v/v) ethanol (0.26 g/ml) (24).

Animal model of acute alcohol-induced liver injury and treatment protocol

Male C57BL/6J mice (Animal license no. 44007200061823) were purchased from the Guangdong Medical Laboratory Animal Center (Foshan, China). A total of 28 mice aged 8-10 weeks were used in this study. All animals were housed in a temperature-controlled animal facility (21-24˚C) with a 12-hour light-dark cycle, and the animals had access to rodent chow and water ad libitum (20). All mice were provided with humane care according to the Principles of Laboratory Animal Care formulated by the National Society of Medical Research and the Guide for the Care and Use of Laboratory Animals published by the NIH (8th Edition, Revised 2011) (20,25). All animal procedures were approved by the Institutional Animal Care and Use Committee of Zhongshan School of Medicine, Sun Yat-sen University (Guangzhou, China).

Mice were randomly assigned into the following four groups (n=7 mice per group): i) Control; ii) Alcohol; iii) Alcohol+H2; and iv) H2. In the Alcohol+H2 and H2 groups, H2 was administered daily at the dose of 1.0 ml/100 g by intraperitoneal injection for 4 days. On day 4, mice in each group were fasted for 6 h before the mice in the Alcohol and Alcohol+H2 groups were orally administered with 33% (v/v) ethanol at a cumulative dose of 4.5 g/kg body weight (17.3 ml/kg body weight) by four equally divided gavages at 20-min intervals (24). Mice in the Alcohol+H2 and H2 groups were administered with an intraperitoneal injection (1.0 ml/100 g) of H2 30 min before the first ethanol administration (20). In addition, mice in the Control and H2 groups received the same volume of double-distilled water (17.3 ml/kg body weight) by four equally divided gavages at 20 min intervals. The animals were anesthetized by intraperitoneal injection of ketamine (100 mg/kg) and xylazine (10 mg/kg) after 16 h of the first ethanol gavage, cardiac puncture was performed when the animals reached the surgical plane of anesthesia and ~0.4-0.5 ml blood was collected from each mouse (26-28). Euthanasia was then performed by cervical dislocation in a state of deep anesthesia before liver tissues were extracted (Fig. 2) (24). The body weight of animals in each group was recorded at the baseline (Control, 25.19±1.58 g; Alcohol, 24.63±1.42 g; Alcohol+H2, 25.17±1.15 g; H2, 23.9±0.70 g) and after intervention (Control, 24.90±1.36 g; Alcohol, 25.16±1.53 g; Alcohol+H2, 25.00±0.93 g; H2, 23.71±0.63 g).

Biochemical analysis

Blood biochemical analysis was performed as previously described (29). Briefly, the blood samples were centrifuged at the speed of 986 x g for 10 min at 4˚C to separate the serum. Levels of liver function markers alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in addition to the levels of lipid markers triglyceride (TG) and total cholesterol (TC) in the serum were analyzed using an Automatic Clinical Analyzer (Hitachi 7600; Hitachi High-Technologies Corporation) at the Department of Clinical Laboratory, The Third Affiliated Hospital of Sun Yat-sen University.

Western blotting

JNK antibody (cat. no. 9252S), phosphorylated (p-) JNK antibody (cat. no. 9255S), anti-rabbit IgG horseradish peroxidase (HRP)-conjugated secondary antibody (cat. no. 7074S) and anti-mouse IgG HRP-conjugated secondary antibody (cat. no. 7076S) were purchased from Cell Signaling Technology, Inc. GAPDH antibody (cat. no. MB001) was obtained from Bioworld Technology, Inc. BSA (cat. no. ST023-200g) was purchased from Beyotime Institute of Biotechnology.

Western blotting was performed as previously described (30). The proteins were transferred onto polyvinylidene fluoride membranes (EMD Millipore). Membranes were incubated in blocking buffer (1X TBST with 5% BSA) for 30 min at room temperature. For antibody incubations, membranes were incubated with primary antibodies in antibody dilution buffer (1X TBST with 5% BSA) with gentle agitation overnight at 4˚C (p-JNK and JNK: 1:2,000; GAPDH: 1:10,000), and with secondary antibodies in antibody dilution buffer (1X TBST with 5% BSA) with gentle agitation for 1 h at room temperature (anti-rabbit IgG HRP-conjugated secondary antibody and anti-mouse IgG HRP-conjugated secondary antibody: 1:2,000). Immobilon™ Western Chemiluminescent HRP Substrate (ECL; cat. no. WBKLS0100, EMD Millipore) was used to reveal the bands using the ChemiDoc™ Touch Imaging System (Bio-Rad Laboratories, Inc.). Image J software (Version 1.52; National Institutes of Health) was used for estimating the ‘IntDen’ value of the western blot bands for quantification.

Statistical analysis

All statistical analyses were performed using GraphPad Prism 5.0 (GraphPad Software, Inc.). Data were expressed as the mean ± SD. For biochemical analysis, n=7 mice in each group. For western blotting, n=5 mice in each group. Statistical analysis was performed by one-way ANOVA followed by Bonferroni's post hoc test. P<0.05 was considered to indicate statistically significant differences.

Results

H2 treatment prevents acute alcohol-induced liver damage

To investigate the effects of H2 on alcohol-induced liver injury, an acute alcohol-induced hepatotoxicity mouse model was established as previously described (24). This model was shown to closely mimic excessive ethanol consumption in humans in terms of blood alcohol levels, behavioral and physiological effects (24). Compared with those in the Control group, serum ALT and AST levels in the Alcohol group were significantly higher following acute alcohol gavage (Fig. 3), whilst intraperitoneal injection of H2 significantly prevented these elevations in serum ALT and AST levels in mice following alcohol gavage (Fig. 3). However, H2 alone exerted no effects on serum ALT and AST levels compared with those in the Control group (Fig. 3). Therefore, these observations suggest that intraperitoneal injection of H2 effectively alleviated acute alcohol-induced liver injury in mice.

Effects of H2 on serum lipid levels in acute alcohol-induced liver injury

Long-term chronic alcohol consumption may lead to disruptions in lipid metabolism (6). Therefore, the serum levels of TC and TG were next examined. Although acute alcohol treatment slightly increased serum TC levels, there was no significant difference between those in the Control and Alcohol groups (Fig. 4A). Serum TC levels in the Alcohol+H2 group were significantly lower compared with those in the Alcohol group (Fig. 4A). However, there were no significant differences in TG levels among the four groups (Fig. 4B). These data suggest that intraperitoneal injection of H2 may modulate blood TC levels in an acute alcohol-induced liver injury mice model.

H2 alleviates acute alcohol-induced hepatic JNK activation

Acute ethanol loading causes oxidative stress and activates cell-death signaling through the JNK pathway in the liver (31). In addition, JNK is a key mediator in hepatic steatosis, where it regulates transcription factor activity associated with lipid metabolism (32,33). In the present study, the upregulation of serum ALT and AST levels after ethanol treatment indicated hepatocytes damage. Therefore, the role of hepatic JNK activation in the potentially protective effects of H2 against acute alcohol-induced liver injury was examined. Western blotting data demonstrated that acute ethanol treatment significantly increased hepatic JNK phosphorylation, which was significantly prevented by intraperitoneal injection with H2 (Fig. 5). Therefore, these data suggest that intraperitoneal injection with H2 conferred protective effects against acute alcohol-induced liver injury by preventing hepatic JNK activation.

Discussion

Inflammatory and cytokine signaling, oxidative stress and mitochondrial dysfunction, alterations in hepatic metabolism and hepatocyte cell death, and abnormalities in immunity and dysbiosis are all key to the pathogenesis of liver diseases, including ALD and NAFLD (2,34-36). Therefore, strategies preventing fatty liver disease progression were previously developed using combinations of naturally occurring compounds products in animal models. For example, the combination of docosahexaenoic acid and the antioxidant hydroxytyrosol was demonstrated to prevent high-fat diet-induced liver steatosis, by inhibiting oxidative stress, mitochondrial dysfunction and inflammation associated with steatosis (34,35). H2 was first reported to alleviate skin tumors by neutralizing toxic free radicals in 1975(37). In 2007, H2 was demonstrated to act as a therapeutic antioxidant by selectively reducing cytotoxic hydroxyl radical levels to improve focal I/R-induced brain injury in rats (38). Since then, H2 has been extensively investigated, where it has been shown to be an able antioxidant, anti-inflammatory and anti-apoptotic agent (10). The present study was undertaken to determine the effect of supplementation with exogenous H2 on acute alcohol-induced liver injury in mice.

Exogenous H2 can be supplied by 2% H2 gas inhalation (38), drinking H2-rich water (39), intraperitoneal injection of H2-rich saline (12) and intraperitoneal injection of H2 gas (19). Previous studies found that intraperitoneal injection of H2 gas can alleviate vascular remodeling (23), cardiac hypertrophy and dysfunction (20,21), and display neuroprotective effects in rabbits experiencing cardiac arrest (19). In the present study, it was observed that ethanol consumption induced hepatocyte injury as indicated by the upregulation of serum ALT and AST levels. Intraperitoneal injection with H2 gas was found to effectively protect against acute alcohol-induced liver injury and reduced serum TC levels. However, body weight was not influenced by the acute intraperitoneal injection of H2 gas or acute alcohol feeding. The lack of food intake records, liver weight, liver histological analysis and steatosis score and hepatic TG analysis are limitations of the present study. It has been documented that chronic alcohol over consumption may lead to hepatic steatosis, fibrosis and cirrhosis and eventually HCC (2,5,6). The long-term effects of intraperitoneal H2 injection on pathological features, such as hepatic fibrosis, and the effects of other H2 delivery methods, such as drinking H2-rich alcohol, on ALD, require further study.

Acute alcohol feeding activates cytochrome P450 2E1 and causes oxidative stress to activate JNK in hepatocytes, and JNK, in turn, reciprocally increases oxidative stress (33). JNK activation can cause programmed cell death, and increases the expression of lipogenic transcription factor sterol regulatory element binding protein (SREBP)-activated lipid synthesis enzymes, resulting in hepatic steatosis (31,33,40,41). Additionally, JNK may suppress hepatic peroxisome proliferator-activated receptor-α (PPAR-α) activation, which is a transcription factor and a positive regulator of intracellular free fatty acid and TG metabolism by regulating gene transcription involved in fatty acid transport and degradation in mitochondria and peroxisomes (32). Therefore, hepatic JNK activation increases oxidative stress, leads to hepatocyte apoptosis and injury, and contributes to hepatic steatosis via modulating lipid metabolic transcription factor activation. H2 has been shown to inhibit JNK activation in numerous liver disease and cardiovascular disease animal models (11,20,22,23). JNK inhibitor can improve acute alcohol-induced fatty liver and oxidative stress in mice (33). In the present study, phosphorylation of JNK in the liver induced by acute alcohol consumption was inhibited by the intraperitoneal injection of H2. Therefore, the protective effect of H2 against acute alcohol-induced liver injury is hypothesized to be partially mediated by reducing JNK phosphorylation. In addition to the inhibition of JNK activation, H2 has been shown to attenuate the activation of NF-κB in an LPS-induced cardiac dysfunction mice model (20). H2 has also been shown to increase the hepatic expression of nuclear factor erythroid 2-related factor 2 (Nrf2) (42) and PPAR-α (43) and reduced the hepatic expression of SREBP-1c (44) in NAFLD animal models. These transcription factors are essential mediators of inflammation (NF-κB), oxidative stress (Nrf2) and lipid metabolism (SREBP-1c and PPAR-α), where they have been reported to serve key roles in the pathogenesis of both ALD and NAFLD (35). Therefore, strategies modulating the expression or activation of these transcription factors may alleviate ALD and NAFLD (35,45-51). However, whether the protective effects of intraperitoneal injection of H2 against acute alcohol-induced liver injury is mediated by regulating the expression and/or activation of these transcription factors aforementioned requires further investigation.

The gut microbiome serves an important role in liver homeostasis, intestinal dysbiosis, including quantitative (such as intestinal bacterial overgrowth) and qualitative (such as colonic Firmicutes and Bacteroidetes levels) changes in the gut microbiota, and pathological bacterial translocation are fundamental for the pathogenesis of ALD (8,52). A number of these intestinal microbiota are affected by alcohol, which express hydrogenases and act as the main producers of endogenous H2 in humans and animals (10,20,52,53). Endogenous H2 is crucial for hepatic redox homeostasis, glucose and lipid homeostasis, in addition to immune and inflammatory homeostasis (11,54-57). Therefore, it remains of importance to investigate the effects of endogenous H2 in the pathogenesis of ALD.

In summary, findings of the present study indicated that intraperitoneal injection with exogenous H2 attenuated acute alcohol-induced liver injury in mice by inhibiting hepatic JNK activation. Therefore, it would be possible to treat alcohol-induced liver injury with H2, such as drinking H2-rich alcohol or H2-rich water, where H2 can be a potentially useful natural agent for the treatment of ALD.

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 81900376), Natural Science Foundation of Guangdong Province (grant no. 2018A030313657), the Project funded by China Postdoctoral Science Foundation (grant no. 2019M653238) and Guangdong famous Traditional Chinese Medicine inheritance studio construction project (grant no. 20180137).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YZ, WG, MD and HY conceived and designed the experiments. YZ, GZ, ZC, MB and YH performed the experiments and analyzed the data. YZ drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal procedures were approved by the Institutional Animal Care and Use Committee of Sun Yat-sen University (IACUC code no: 2018-057).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Osna NA, Donohue TM Jr and Kharbanda KK: Alcoholic liver disease: Pathogenesis and current management. Alcohol Res. 38:147–161. 2017.PubMed/NCBI

2 

Seitz HK, Bataller R, Cortez-Pinto H, Gao B, Gual A, Lackner C, Mathurin P, Mueller S, Szabo G and Tsukamoto H: Alcoholic liver disease. Nat Rev Dis Primers. 4(16)2018.PubMed/NCBI View Article : Google Scholar

3 

Asrani SK, Devarbhavi H, Eaton J and Kamath PS: Burden of liver diseases in the world. J Hepatol. 70:151–171. 2019.PubMed/NCBI View Article : Google Scholar

4 

Lieber CS: Alcoholic liver disease: New insights in pathogenesis lead to new treatments. J Hepatol. 32 (Suppl):113–128. 2000.PubMed/NCBI View Article : Google Scholar

5 

Ding WX and Yang L: Alcohol and drug-induced liver injury: Metabolism, mechanisms, pathogenesis and potential therapies. Liver Res. 3:129–131. 2019.PubMed/NCBI View Article : Google Scholar

6 

Gao B and Bataller R: Alcoholic liver disease: Pathogenesis and new therapeutic targets. Gastroenterology. 141:1572–1585. 2011.PubMed/NCBI View Article : Google Scholar

7 

Louvet A and Mathurin P: Alcoholic liver disease: Mechanisms of injury and targeted treatment. Nat Rev Gastroenterol Hepatol. 12:231–242. 2015.PubMed/NCBI View Article : Google Scholar

8 

Bajaj JS: Alcohol, liver disease and the gut microbiota. Nat Rev Gastroenterol Hepatol. 16:235–246. 2019.PubMed/NCBI View Article : Google Scholar

9 

Wang FS, Fan JG, Zhang Z, Gao B and Wang HY: The global burden of liver disease: The major impact of China. Hepatology. 60:2099–2108. 2014.PubMed/NCBI View Article : Google Scholar

10 

Zhang Y, Tan S, Xu J and Wang T: Hydrogen therapy in cardiovascular and metabolic diseases: From bench to bedside. Cell Physiol Biochem. 47:1–10. 2018.PubMed/NCBI View Article : Google Scholar

11 

Zhang Y, Xu J and Yang H: Hydrogen: An endogenous regulator of liver homeostasis. Front Pharmacol. 11(877)2020.PubMed/NCBI View Article : Google Scholar

12 

Sun H, Chen L, Zhou W, Hu L, Li L, Tu Q, Chang Y, Liu Q, Sun X, Wu M, et al: The protective role of hydrogen-rich saline in experimental liver injury in mice. J Hepatol. 54:471–480. 2011.PubMed/NCBI View Article : Google Scholar

13 

Liu Q, Li BS, Song YJ, Hu MG, Lu JY, Gao A, Sun XJ, Guo XM and Liu R: Hydrogen-rich saline protects against mitochondrial dysfunction and apoptosis in mice with obstructive jaundice. Mol Med Rep. 13:3588–3596. 2016.PubMed/NCBI View Article : Google Scholar

14 

Fukuda K, Asoh S, Ishikawa M, Yamamoto Y, Ohsawa I and Ohta S: Inhalation of hydrogen gas suppresses hepatic injury caused by ischemia/reperfusion through reducing oxidative stress. Biochem Biophys Res Commun. 361:670–674. 2007.PubMed/NCBI View Article : Google Scholar

15 

Yang SC, Chen LL, Fu T, Li WY and Ji ES: Improvement of hydrogen on liver oxidative stress injury in chronic intermittent hypoxia rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 34:61–64. 2018.PubMed/NCBI View Article : Google Scholar : (In Chinese).

16 

Chen M, Jiang L, Li Y, Bai G, Zhao J, Zhang M and Zhang J: Hydrogen protects against liver injury during CO2 pneumoperitoneum in rats. Oncotarget. 9:2631–2645. 2017.PubMed/NCBI View Article : Google Scholar

17 

Kawai D, Takaki A, Nakatsuka A, Wada J, Tamaki N, Yasunaka T, Koike K, Tsuzaki R, Matsumoto K, Miyake Y, et al: Hydrogen-rich water prevents progression of nonalcoholic steatohepatitis and accompanying hepatocarcinogenesis in mice. Hepatology. 56:912–921. 2012.PubMed/NCBI View Article : Google Scholar

18 

Lin CP, Chuang WC, Lu FJ and Chen CY: Anti-oxidant and anti-inflammatory effects of hydrogen-rich water alleviate ethanol-induced fatty liver in mice. World J Gastroenterol. 23:4920–4934. 2017.PubMed/NCBI View Article : Google Scholar

19 

Huang G, Zhou J, Zhan W, Xiong Y, Hu C, Li X, Li X, Li Y and Liao X: The neuroprotective effects of intraperitoneal injection of hydrogen in rabbits with cardiac arrest. Resuscitation. 84:690–695. 2013.PubMed/NCBI View Article : Google Scholar

20 

Tan S, Long Z, Hou X, Lin Y, Xu J, You X, Wang T and Zhang Y: H2 protects against lipopolysaccharide-induced cardiac dysfunction via blocking TLR4-mediated cytokines expression. Front Pharmacol. 10(865)2019.PubMed/NCBI View Article : Google Scholar

21 

Zhang Y, Long Z, Xu J, Tan S, Zhang N, Li A, Wang L and Wang T: Hydrogen inhibits isoproterenol induced autophagy in cardiomyocytes in vitro and in vivo. Mol Med Rep. 16:8253–8258. 2017.PubMed/NCBI View Article : Google Scholar

22 

Zhang Y, Xu J, Long Z, Wang C, Wang L, Sun P, Li P and Wang T: Hydrogen (H2) inhibits isoproterenol-induced cardiac hypertrophy via antioxidative pathways. Front Pharmacol. 7(392)2016.PubMed/NCBI View Article : Google Scholar

23 

Zhang YX, Xu JT, You XC, Wang C, Zhou KW, Li P, Sun P, Wang L and Wang TH: Inhibitory effects of hydrogen on proliferation and migration of vascular smooth muscle cells via down-regulation of mitogen/activated protein kinase and ezrin-radixin-moesin signaling pathways. Chin J Physiol. 59:46–55. 2016.PubMed/NCBI View Article : Google Scholar

24 

Ding WX, Li M, Chen X, Ni HM, Lin CW, Gao W, Lu B, Stolz DB, Clemens DL and Yin XM: Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice. Gastroenterology. 139:1740–1752. 2010.PubMed/NCBI View Article : Google Scholar

25 

Polhemus DJ, Trivedi RK, Gao J, Li Z, Scarborough AL, Goodchild TT, Varner KJ, Xia H, Smart FW, Kapusta DR, et al: Renal sympathetic denervation protects the failing heart via inhibition of neprilysin activity in the kidney. J Am Coll Cardiol. 70:2139–2153. 2017.PubMed/NCBI View Article : Google Scholar

26 

Hill JA, Karimi M, Kutschke W, Davisson RL, Zimmerman K, Wang Z, Kerber RE and Weiss RM: Cardiac hypertrophy is not a required compensatory response to short-term pressure overload. Circulation. 101:2863–2869. 2000.PubMed/NCBI View Article : Google Scholar

27 

Frankenberg L: Cardiac puncture in the mouse through the anterior thoracic aperture. Lab Anim. 13:311–312. 1979.PubMed/NCBI View Article : Google Scholar

28 

Kaur H, Fisher K and Othman M: Thromboelastography testing in mice following blood collection from facial vein and cardiac puncture. Blood Coagul Fibrinolysis. 30:366–369. 2019.PubMed/NCBI View Article : Google Scholar

29 

Zhang Y, Zhou G, Chen Z, Guan W, Zhang J, Bi M, Wang F, You X, Liao Y, Zheng S, et al: Si-Wu-Tang alleviates nonalcoholic fatty liver disease via blocking TLR4-JNK and Caspase-8-GSDMD signaling pathways. Evid Based Complement Alternat Med. 2020(8786424)2020.PubMed/NCBI View Article : Google Scholar

30 

Zhang Y, Zhang J, Xu K, Chen Z, Xu X, Xu J, Zheng S, Dai M and Yang H: Helium protects against lipopolysaccharide-induced cardiac dysfunction in mice via suppressing Toll-like receptor 4-nuclear factor κB-tumor necrosis factor-alpha/interleukin-18 signaling. Chin J Physiol. 63:276–285. 2020.PubMed/NCBI View Article : Google Scholar

31 

Nishitani Y and Matsumoto H: Ethanol rapidly causes activation of JNK associated with ER stress under inhibition of ADH. FEBS Lett. 580:9–14. 2006.PubMed/NCBI View Article : Google Scholar

32 

Manieri E, Folgueira C, Rodríguez ME, Leiva-Vega L, Esteban-Lafuente L, Chen C, Cubero FJ, Barrett T, Cavanagh-Kyros J, Seruggia D, et al: JNK-mediated disruption of bile acid homeostasis promotes intrahepatic cholangiocarcinoma. Proc Natl Acad Sci USA. 117:16492–16499. 2020.PubMed/NCBI View Article : Google Scholar

33 

Yang L, Wu D, Wang X and Cederbaum AI: Cytochrome P4502E1, oxidative stress, JNK, and autophagy in acute alcohol-induced fatty liver. Free Radic Biol Med. 53:1170–1180. 2012.PubMed/NCBI View Article : Google Scholar

34 

Ortiz M, Soto-Alarcón SA, Orellana P, Espinosa A, Campos C, López-Arana S, Rincón MA, Illesca P, Valenzuela R and Videla LA: Suppression of high-fat diet-induced obesity-associated liver mitochondrial dysfunction by docosahexaenoic acid and hydroxytyrosol co-administration. Dig Liver Dis. 52:895–904. 2020.PubMed/NCBI View Article : Google Scholar

35 

Valenzuela R and Videla LA: Impact of the co-administration of N-3 fatty acids and olive oil components in preclinical nonalcoholic fatty liver disease models: A mechanistic view. Nutrients. 12(499)2020.PubMed/NCBI View Article : Google Scholar

36 

Nevzorova YA, Boyer-Diaz Z, Cubero FJ and Gracia-Sancho J: Animal models for liver disease-A practical approach for translational research. J Hepatol. 73:423–440. 2020.PubMed/NCBI View Article : Google Scholar

37 

Dole M, Wilson FR and Fife WP: Hyperbaric hydrogen therapy: A possible treatment for cancer. Science. 190:152–154. 1975.PubMed/NCBI View Article : Google Scholar

38 

Ohsawa I, Ishikawa M, Takahashi K, Watanabe M, Nishimaki K, Yamagata K, Katsura K, Katayama Y, Asoh S and Ohta S: Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat Med. 13:688–694. 2007.PubMed/NCBI View Article : Google Scholar

39 

Sun Q, Kawamura T, Masutani K, Peng X, Sun Q, Stolz DB, Pribis JP, Billiar TR, Sun X, Bermudez CA, et al: Oral intake of hydrogen-rich water inhibits intimal hyperplasia in arterialized vein grafts in rats. Cardiovasc Res. 94:144–153. 2012.PubMed/NCBI View Article : Google Scholar

40 

Chang L, Kamata H, Solinas G, Luo JL, Maeda S, Venuprasad K, Liu YC and Karin M: The E3 ubiquitin ligase itch couples JNK activation to TNFalpha-induced cell death by inducing c-FLIP(L) turnover. Cell. 124:601–613. 2006.PubMed/NCBI View Article : Google Scholar

41 

Kamata H, Honda S, Maeda S, Chang L, Hirata H and Karin M: Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. 120:649–661. 2005.PubMed/NCBI View Article : Google Scholar

42 

Wang X and Wang J: High-content hydrogen water-induced downregulation of miR-136 alleviates non-alcoholic fatty liver disease by regulating Nrf2 via targeting MEG3. Biol Chem. 399:397–406. 2018.PubMed/NCBI View Article : Google Scholar

43 

Zhai X, Chen X, Lu J, Zhang Y, Sun X, Huang Q and Wang Q: Hydrogen-rich saline improves non alcoholic fatty liver disease by alleviating oxidative stress and activating hepatic PPARα and PPARγ. Mol Med Rep. 15:1305–1312. 2017.PubMed/NCBI View Article : Google Scholar

44 

Liu B, Xue J, Zhang M, Wang M, Ma T, Zhao M, Gu Q and Qin S: Hydrogen inhalation alleviates nonalcoholic fatty liver disease in metabolic syndrome rats. Mol Med Rep. 22:2860–2868. 2020.PubMed/NCBI View Article : Google Scholar

45 

Hernández-Rodas MC, Valenzuela R, Echeverría F, Rincón-Cervera MÁ, Espinosa A, Illesca P, Muñoz P, Corbari A, Romero N, Gonzalez-Mañan D, et al: Supplementation with docosahexaenoic acid and extra virgin olive oil prevents liver steatosis induced by a high-fat diet in mice through PPAR-α and Nrf2 upregulation with concomitant SREBP-1c and NF-κB downregulation. Mol Nutr Food Res. 61(61)2017.PubMed/NCBI View Article : Google Scholar

46 

Li L, Fu J, Liu D, Sun J, Hou Y, Chen C, Shao J, Wang L, Wang X, Zhao R, et al: Hepatocyte-specific Nrf2 deficiency mitigates high-fat diet-induced hepatic steatosis: Involvement of reduced PPARγ expression. Redox Biol. 30(101412)2020.PubMed/NCBI View Article : Google Scholar

47 

Ambade A, Catalano D, Lim A, Kopoyan A, Shaffer SA and Mandrekar P: Inhibition of heat shock protein 90 alleviates steatosis and macrophage activation in murine alcoholic liver injury. J Hepatol. 61:903–911. 2014.PubMed/NCBI View Article : Google Scholar

48 

Lamlé J, Marhenke S, Borlak J, von Wasielewski R, Eriksson CJ, Geffers R, Manns MP, Yamamoto M and Vogel A: Nuclear factor-eythroid 2-related factor 2 prevents alcohol-induced fulminant liver injury. Gastroenterology. 134:1159–1168. 2008.PubMed/NCBI View Article : Google Scholar

49 

Ji C, Chan C and Kaplowitz N: Predominant role of sterol response element binding proteins (SREBP) lipogenic pathways in hepatic steatosis in the murine intragastric ethanol feeding model. J Hepatol. 45:717–724. 2006.PubMed/NCBI View Article : Google Scholar

50 

Nakajima T, Kamijo Y, Tanaka N, Sugiyama E, Tanaka E, Kiyosawa K, Fukushima Y, Peters JM, Gonzalez FJ and Aoyama T: Peroxisome proliferator-activated receptor alpha protects against alcohol-induced liver damage. Hepatology. 40:972–980. 2004.PubMed/NCBI View Article : Google Scholar

51 

Valenzuela R and Videla LA: Crosstalk mechanisms in hepatoprotection: Thyroid hormone-docosahexaenoic acid (DHA) and DHA-extra virgin olive oil combined protocols. Pharmacol Res. 132:168–175. 2018.PubMed/NCBI View Article : Google Scholar

52 

Hartmann P, Seebauer CT and Schnabl B: Alcoholic liver disease: The gut microbiome and liver cross talk. Alcohol Clin Exp Res. 39:763–775. 2015.PubMed/NCBI View Article : Google Scholar

53 

Wolf PG, Biswas A, Morales SE, Greening C and Gaskins HR: H2 metabolism is widespread and diverse among human colonic microbes. Gut Microbes. 7:235–245. 2016.PubMed/NCBI View Article : Google Scholar

54 

Tanabe H, Sasaki Y, Yamamoto T, Kiriyama S and Nishimura N: Suppressive effect of high hydrogen generating high amylose cornstarch on subacute hepatic ischemia-reperfusion injury in rats. Biosci Microbiota Food Health. 31:103–108. 2012.PubMed/NCBI View Article : Google Scholar

55 

Kajiya M, Sato K, Silva MJ, Ouhara K, Do PM, Shanmugam KT and Kawai T: Hydrogen from intestinal bacteria is protective for concanavalin A-induced hepatitis. Biochem Biophys Res Commun. 386:316–321. 2009.PubMed/NCBI View Article : Google Scholar

56 

Yu J, Zhang W, Zhang R, Ruan X, Ren P and Lu B: Lactulose accelerates liver regeneration in rats by inducing hydrogen. J Surg Res. 195:128–135. 2015.PubMed/NCBI View Article : Google Scholar

57 

Zhao L, Wang Y, Zhang G, Zhang T, Lou J and Liu J: L-arabinose elicits gut-derived hydrogen production and ameliorates metabolic syndrome in C57BL/6J mice on high-fat-diet. Nutrients. 11(3054)2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Bi M, Chen Z, Dai M, Zhou G, Hu Y, Yang H and Guan W: Hydrogen gas alleviates acute alcohol‑induced liver injury by inhibiting JNK activation. Exp Ther Med 21: 453, 2021
APA
Zhang, Y., Bi, M., Chen, Z., Dai, M., Zhou, G., Hu, Y. ... Guan, W. (2021). Hydrogen gas alleviates acute alcohol‑induced liver injury by inhibiting JNK activation. Experimental and Therapeutic Medicine, 21, 453. https://doi.org/10.3892/etm.2021.9884
MLA
Zhang, Y., Bi, M., Chen, Z., Dai, M., Zhou, G., Hu, Y., Yang, H., Guan, W."Hydrogen gas alleviates acute alcohol‑induced liver injury by inhibiting JNK activation". Experimental and Therapeutic Medicine 21.5 (2021): 453.
Chicago
Zhang, Y., Bi, M., Chen, Z., Dai, M., Zhou, G., Hu, Y., Yang, H., Guan, W."Hydrogen gas alleviates acute alcohol‑induced liver injury by inhibiting JNK activation". Experimental and Therapeutic Medicine 21, no. 5 (2021): 453. https://doi.org/10.3892/etm.2021.9884