2023 Impact Factor
The liver plays a central role in metabolizing alcohol, with chronic alcohol consumption increasing the risk of liver diseases, such as fatty liver, hepatitis, and cirrhosis (Contreras-Zentella
Chronic alcohol consumption activates the microsomal ethanol oxidation system (MEOS), involving cytochrome P450 2E1 (CYP2E1). This shift leads to an increase in reactive oxygen species (ROS) production and depletes glutathione (GSH), a crucial antioxidant in hepatocytes (Chen
Glutathione (GSH), a tripeptide consisting of glutamate, cysteine, and glycine, is a vital antioxidant that maintains cellular redox balance (Wu
Glutathione was obtained from Sigma-Aldrich Corporation (St. Louis, MO, USA). The enzyme analyses were carried out using commercially available kits in accordance with the manufacturer’s instructions (BioVision, Milpitas, CA, USA). Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were assessed with commercial kits from Asan Pharma, Hwasung, Korea. LR-grade chemicals and reagents were utilized in the investigations to ensure accuracy and consistency.
Yeast extract (YE) with above 20% GSH (w/w) was prepared using a fermentative process with Candida utilis (Torula). In this investigation, the produced YE contains 21.25% of GSH. Thus, the concentrations used in the
The animal investigations were conducted strictly with the Principles of Laboratory Animal Care of the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC) of the Jeonbuk National University Hospital. The Institutional Animal Care and Use Committee (IACUC) of the Jeonbuk National University Hospital, Jeonju, Korea, approved the protocols utilized in this study. The approval number is CUH-IACUC-2021-7.
Seven-week-old male Sprague Dawley (SD) rats were procured from Central Lab Animal Inc., Seoul, Korea. All the animals were cared for and acclimated to standard laboratory living conditions, with a 12 h L/D cycle and ad libitum food and water. Experimental animals were acclimatized for seven days prior to the investigation. Acclimatized rats were randomly divided into six groups, each with 10 animals. Control, rats received water; Ethanol, rats received 3 g/kg ethanol; GSH-L, Glutathione low group rats received 3 g/kg ethanol+glutathione 0.52 mg/kg (equal to the glutathione 5 mg/60 kg human dose); GSH-M, Glutathione medium group rats received 3 g/kg ethanol+glutathione 5.17 mg/kg (equal to the glutathione 50 mg/60 kg human dose); GSH-H, Glutathione high group rats received 3 g/kg ethanol+glutathione 25.83 mg/kg (equal to the glutathione 250 mg/60 kg human dose); YE, yeast extract group rats received 3 g/kg ethanol+24.31 mg/kg of yeast extract (equal to the YE dose of 235.29 mg/60 kg human). In the behavioral testing, the medium dose used in the experiments was selected based on the dose and glutathione content, with the YE dose determined by its glutathione content of 21.25%. The glutathione and yeast extract were dissolved in water and administered separately via oral gavage 30 min prior to ethanol administration. Whole blood samples were collected at specific time points (0, 0.5, 1, 3, 5, and 8 h) following ethanol administration. Tissue samples were collected from euthanized animals. The liver tissue was homogenized with a buffer solution having 250 mM sucrose, 50 mM Tris-HCl (pH 7.4), and 1 mM EDTA and stored with 4% paraformaldehyde (PFA) or at –80°C.
The Laboratory Animal Behaviour Observation, Registration, and Analysis System (LABORAS™, Metris B.V., Hoofddorp, Netherlands) was used to investigate rat locomotor movements, following the protocols previously outlined by Castagne
Blood alcohol levels were measured using an ethanol quantification assay kit, following the manufacturer’s guidelines (Megazyme International, Wicklow, Ireland). Briefly, 20X diluted serum was mixed with 25 μL of ALDH solution, 100 μL of 0.02% sodium azide, and 100 μL NAD+ reagent. The mixture was incubated at room temperature for 2 min, and absorbance (A1) was measured at 340 nm. Further, 10 μL ADH was added, and then the mixture was incubated at room temperature for 5 min, and absorbance (A2) was measured at 340 nm. The change in absorbance (A2-A1) was used to calculate the blood alcohol levels.
The ADH activity of the supernatant were measured using an Alcohol Dehydrogenase Assay kit (Abcam, Cambridge, UK). All the measurements were performed as suggested by the manufacturer. Briefly, the reaction mixture was prepared by combining 1 part of supernatant obtained from liver homogenates or whole blood with 10 parts of buffer containing ADH assay buffer, developer, and isopropanol. Subsequently, a standard curve was produced using concentrations of 0, 2, 4, 6, 8, and 10 nmol. The concentrations were prepared using a 1 mM NADH standard. Subsequently, 50 μL of the reaction mixture was transferred to the standard and sample wells in a 96-well plate and incubated at ambient temperature for three minutes. The absorbance was measured at 450 nm at regular intervals of 5 min over a duration of 120 min.
ALDH activity was determined using a colorimetric assay Kit (#K731-100, BioVision, Cambridge, UK). A 10X diluted sample was thoroughly mixed with reaction buffer having ALDH assay buffer, substrate mix, and acetaldehyde. Then, 50 μL of the resulting mixture was incubated at 22°C-24°C for 5 min, and the absorbance was measured at regular intervals of 5 min for up to 1 h at 450 nm. The resulting reaction time and sample volume was divided by the NADH released to determine ALDH activity (mU/mL). A standard curve was prepared using 0-10 nmol concentrations. The concentrations were prepared using a 1 mM NADH standard.
All the enzyme activities were measured using commercially available test kits. Catalase activity (#K773, BioVision), SOD (#K335, BioVision), and activity of glutathione peroxidase (#K762, BioVision). All the protocols and analyses were performed as recommended by the manufacturers.
ROS was determined using fixed tissues following a previously reported protocol (Lee
Homogenized liver tissues were mixed with 40 mM Tris-HCl buffer and centrifuged at 12,000× g for 10 min at 4°C to obtain supernatant. 50 μL of supernatant was mixed with 450 μL of 40 mM Tris-HCl buffer and 10 μL of 10 μM 2′,7′-dichlorofluorescein diacetate and incubated at 37°C for 30 min in the dark. The ROS levels were determined by fluorescence spectroscopy, where excitation and emission wavelengths were set at 482 nm and 535 nm, respectively. Further, MDA levels were assessed with OxiTec™ TBARS assay kit (#BO-TBR-200, BIOMAX Inc., Suwon, Korea) by following manufacturer guidelines.
Well-characterized and standardized human hepatoblastoma (HepG2) cells were obtained from Korea Cell Line Bank (Seoul, Korea). HepG2 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) with 1% penicillin-streptomycin and 10% fetal bovine serum (FBS), incubated in a standard humidified incubator. 1×105 cells were seeded in each well in a 96-well plate and cultured for 24 h. These cells were treated with different concentrations of GSH and 600 mM of ethanol for 24 h to assess the influence of GSH against ethanol-induced cytotoxicity. The cell viability was measured using an 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay kit (Sigma-Aldrich Corporation) following manufacturer guidelines.
The RNA isolation and quantitative RT-PCR were performed as described earlier (Choi
Table 1 Primer sequences used for reverse transcription-polymerase chain reaction
Gene name | Direction | Sequence (5′-3′) | |
---|---|---|---|
Forward Reverse | GAGCACCATCAATCTCTGGACC CACGGTGATACCGTCCATTGTG | Human | |
Forward Reverse | TGGCTACAAGGCTGTCAAGG AGGCTGGCCTTTGGTCTTTT | Rat | |
Forward Reverse | CGGCACATGAATGGCTATGGATC AAGCCTTCCTGCCTCTCCAACA | Human | |
Forward Reverse | CCAGCGACCAGATGAAGCA TGGTCAGGACATCGGGTTTC | Rat | |
Forward Reverse | CTCACTCTCAGGAGACCATTGC CCACAAGCCAAACGACTTCCAG | Human | |
Forward Reverse | AAGCGGTGAACCAGTTGTG CCAGGTCTCCAACATGCC | Rat | |
Forward Reverse | GTGCTCGGCTTCCCGTGCAAC CTCGAAGAGCATGAAGTTGGGC | Human | |
Forward Reverse | TATAGAAGCCCTGCTGTCCA CAAGCCCAGATACCAGGAA | Rat | |
Forward Reverse | GAAACCCATCCAGGAGGTGCTA GACACTTGTGCCACATGCCTCA | Human | |
Forward Reverse | CGGGAAAAGCAATCTGAAGAGGG GATGCGGCTATACAACACTGGC | Human | |
Forward Reverse | CACATCCAGTCAGAAACCAGTGG GGAATGTCTGCGCCAAAAGCTG | Human | |
Forward Reverse | GAGCGGGAGAAATCACACAGAATG CAGGAGCTGCATGCACTCATCG | Rat | |
Forward Reverse | CCAGGCAGAGAATGCTGAGTTC AAGACTGGGCTCTCCTTGTTGC | Human | |
Forward Reverse | CGACAGCATGTCCCAGGATT TCGCTCTATCTCCTCTTCCAGG | Rat |
Protein S-glutathionylation and protein content in tissue and cells were assayed by Western blot analysis as described (Han
Liver frozen sections and cells were performed as described previously (Seidel
The data presented in the charts and tables are expressed as the mean ± standard error of the mean (SEM). All statistical analyses were carried out with GraphPad Prism version 10.1.1 (GraphPad Software, San Diego, CA, USA). The data were examined using one-way analysis of variance (ANOVA) and subsequently underwent Tukey’s multiple comparisons test, where
This investigation used LABORAS to monitor the change in behavioural patterns in GSH and GSH-rich YE-treated alcoholic rats. The locomotor activities of the alcohol treatment group were significantly reduced relative to those of the control group. However, the locomotor activities of rats treated with 5.17 mg/kg of GSH and YE with 5.17 mg/kg of endogenously produced GSH were significantly enhanced (Fig. 1). These findings strongly indicate that GSH and GSH-rich YE effectively improved locomotor activities in alcoholic rats and suggest the positive influence of GSH in alleviating alcohol-induced behavioral changes.
Multiple concentrations of GSH were administered to rats to evaluate its positive impact on alcohol toxicity. The evaluation of hepatocyte damage typically involves measuring serum levels of GOT and GPT (Giannini
To further demonstrate that alcohol disrupts redox balance and contributes to cellular oxidative stress and damage, oxidative stress markers in ethanol-treated rats were examined. The groups treated with GSH and YE exhibited a notable reduction in ROS production compared to the control group exposed to ethanol alone (Fig. 3A-3C). In addition, levels of MDA, a marker of lipid peroxidation and free radical damage to hepatocytes, were significantly increased in the alcohol treatment group, highlighting the oxidative stress imposed by alcohol administration (Fig. 3D). Notably, the reduction of MDA levels was correlated with an increase in the dose of GSH, indicating the protection against liver oxidative stress induced by acute alcohol consumption. A significant effect was observed at GSH concentrations above the medium level (>5.17 mg/kg). Further, the expression levels of
This study evaluated the activity of SOD and Catalase, as well as the mRNA expression of
The pro-apoptotic protein Bax was upregulated in the rats treated with ethanol, while the anti-apoptotic protein Bcl-2 was downregulated. GSH dose-dependently downregulated Bax, whereas Bcl-2 was upregulated. Moreover, cleaved caspase-3 in the alcohol treatment group was enhanced, while the administration of GSH appeared to prevent cleavage (Fig. 5A, 5B). Moreover, real-time PCR showed an increase in pro-inflammatory cytokines, including
The MTT assay indicated that GSH has a limited influence on cell viability (Fig. 6A). This observation suggests that GSH has no cytotoxic effect on HepG2 cells at concentrations below 1 mM. Interestingly, Ethanol treatment significantly reduced the viability of HepG2 cells, while GSH dose-dependently showed a positive influence on cell viability. Specifically, GSH at 0.5 mM and above significantly improved the cell viability (Fig. 6B). These data clearly indicate that GSH effectively protects cells against ethanol-induced cytotoxicity. Additionally, the mRNA expression of ADH and ALDH in HepG2 cells was significantly reduced in ethanol treatment, whereas GSH significantly increased ADH and ALDH mRNA expression (Fig. 6C, 6D). On the contrary, the expression of CYP2E1 was substantially elevated with ethanol treatment, whereas GSH significantly reduced CYP2E1 expression (Fig. 6E). Similarly, ethanol treatment stimulated MDA accumulation, while GSH administration significantly inhibited MDA (Fig. 6F). Furthermore, the mRNA expression of antioxidant enzymes, such as
The impact of ethanol metabolism on protein S-glutathionylation was investigated in HepG2 cells and liver tissues. Liver tissue was collected and subjected to biochemical analysis to examine protein S-glutathionylation and its regulatory system (Fig. 7A). In this study, the ethanol-treated group exhibited a significant increase in protein S-glutathionylation in liver tissues. However, treatment with GSH and GSH-rich YE significantly reduced the levels of protein S-glutathionylation (Fig. 7B). The immunofluorescence analysis of liver cross-sections also confirmed that the ethanol administration stimulated protein S-glutathionylation, while GSH significantly regulated protein S-glutathionylation (Fig. 7C, 7D). Additionally, the Grx1 levels were significantly improved with GSH treatment (Fig. 7C, 7D). Moreover, the protein levels of Grx1 and GR, which are key enzymes that regulate S-glutathionylation, were substantially reduced in the alcohol treatment group compared to the control group. However, GSH administration restored the levels of these proteins and positively regulated the S-glutathionylation. On the contrary, GST-π levels were enhanced in the alcohol treatment group, whereas GSH significantly reduced GST-π levels (Fig. 7E, 7F). Similarly, the
The study demonstrates that GSH and GSH-rich YE effectively controlled hangover symptoms in rats exposed to binge drinking by enhancing alcohol metabolism. The GSH administration lowered ROS production and lipid peroxidation, indicating reduced oxidative stress. Additionally, GSH and GSH-rich YE restored the activity of antioxidant enzymes and upregulated the Nrf2 signaling pathway, highlighting their protective role against ethanol-induced liver damage. These findings underscore the potential of GSH and YE in mitigating alcohol-induced liver toxicity and oxidative stress (Fig. 9).
In this investigation, an alcohol hangover was induced in rats by administering a single acute dose of alcohol. GSH and GSH-rich YE effectively inhibited hangover patterns and improved motor coordination (Fig. 1). The findings of this study, combined with previous investigations (Choi
Furthermore, the study results demonstrate that GSH treatments significantly reduce ROS accumulation in ethanol-treated HepG2 cells and
Additionally, the findings of the study indicate that the efficacy of GSH is contingent upon the concentration at which it is administered, with rats exhibiting significant effects at a dose of 5.17 mg/kg and higher. The significance of this specific dose was further demonstrated by its notable regulation of key biomarkers associated with alcohol metabolism and oxidative stress. When this specific concentration is converted to human intake standards, an effective concentration corresponds to the consumption of 50 mg of GSH per 60 kg of body weight. Further, alcohol-induced oxidative stress in the liver was also regulated by protein modifications such as protein S-glutathionylation. GSH, in its reduced (GSH) and oxidized (GSSG) forms, plays a critical role in oxidative stress regulation and detoxification (McBean, 2017). The study findings indicate that ethanol stimulates protein S-glutathionylation in the liver and HepG2 cells (Fig. 7A, 7B, 8). GSH administration markedly decreased protein S-glutathionylation (Fig. 7C-7E). Protein levels of key regulatory enzymes, such as Grx1 and GST-π, were altered by ethanol treatment and antioxidant intervention (Fig. 7E, 7F). Our data suggest that glutathione and yeast extract containing glutathione effectively control liver protein S-glutathionylation induced by binge drinking, possibly through regulating glutathionylation/deglutathionylation enzyme levels. This study offers a potential therapeutic approach for mitigating alcohol-induced hepatic oxidative stress and alcohol metabolite accumulation, while addressing alcohol-associated hangover symptoms. Although glutathione has limited oral bioavailability due to gastrointestinal degradation, studies show that orally administered GSH can still elevate tissue glutathione levels and improve antioxidant defence. For example, it was demonstrated that GSH raised glutathione levels in the liver and plasma (Favilli
In summary, glutathione and yeast extract containing glutathione alleviate hangovers by reducing alcohol and acetaldehyde concentrations in the blood, thereby highlighting their role in regulating oxidative stress. Given that glutathione is an efficacious compound for alcohol metabolism and antioxidant actions, and that yeast extract containing glutathione shows equivalent effects, this study suggests that glutathione, a natural ingredient from yeast extract, can alleviate hangover symptoms through its antioxidant activity and ability to enhance alcohol metabolism. Therefore, glutathione and yeast extract containing glutathione are proposed as alternative natural functional food products to prevent the negative effects of binge drinking.
This work was supported by Samyang Corp., Seongnam-si, Republic of Korea.
The authors declare the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: Yeast extract (YE) was obtained from Samyang Corporation. These contributions could be perceived as potential conflicts of interest. However, the authors affirm that the data interpretation and the conclusions drawn in this study are based solely on the scientific evidence presented.