Abstract
Diosgenin, a plant-derived steroid sapogenin, has been reported to have many health benefits, including antioxidant activity. Although oxidative stress is a major factor impeding the differentiation and homeostasis of skeletal muscles, its antioxidant activity in skeletal muscle cells has not been thoroughly studied. This study aimed to explore the protective mechanisms of diosgenin against oxidative damage in skeletal muscle cells. C2C12 murine myoblasts were pretreated with nontoxic concentrations of diosgenin and exposed to hydrogen peroxide (H2O2) to mimic oxidative stress. The results of this study showed that diosgenin significantly reduced H2O2-induced cytotoxicity, blocked the formation of comet tails, and increased the levels of 8-hydroxy-2’-deoxyguanosine, which are representative biomarkers of DNA damage. In addition, diosgenin counteracted H2O2-induced apoptosis by enhancing the Bax/Bcl-2 expression ratio and suppressing the activation of the caspase cascade, which is associated with the blockade of cytochrome c release into the cytoplasm by maintaining mitochondrial stability. Furthermore, diosgenin eliminated the production of intracellular and mitochondrial reactive oxygen species (ROS) by restoring glutathione (GSH) content and the activities of antioxidant enzymes such as GSH peroxidase 1 and manganese-dependent superoxide dismutase, which were inhibited by H2O2. Therefore, diosgenin protects C2C12 myoblasts from oxidative damage by attenuating mitochondrial ROS generation and regulating the mitochondrial apoptotic pathway.
Similar content being viewed by others
Avoid common mistakes on your manuscript.
Introduction
Diosgenin is a steroidal compound found in various plant parts, including the rhizome of Dioscorea, a member of the Dioscoreaceae family [1, 2]. Previous studies have shown that diosgenin has various pharmacological properties, such as anti-inflammatory, antidiabetic, immunostimulatory, anti-metabolic syndrome, hepatoprotective, neuroprotective, and cardiovascular protective effects, and is effective in treating numerous diseases due to its absence of side effects [3,4,5,6]. In addition, diosgenin has been shown to induce growth arrest and apoptosis in various types of cancer cells, inhibit angiogenesis, cancer cell invasion, and metastasis, and improve anticancer drug sensitivity, which is closely related to the induction of oxidative stress by targeting reactive oxygen species (ROS) production [7,8,9]. In contrast, the antioxidant activity of diosgenin has been shown to be involved in defense mechanisms that prevent damage to normal cells caused by various stimuli. For example, in a diabetic nephropathy model, diosgenin attenuated the apoptosis of renal proximal tubular epithelial cells by suppressing ROS production through the maintenance of mitochondrial homeostasis [10]. Diosgenin also protected against type II diabetes-associated non-alcoholic fatty liver disease by reducing ROS and improving β-oxidation and mitochondrial function by enhancing the activation of antioxidant enzymes such as catalase (CAT), glutathione (GSH) peroxidase (GPx), and superoxide dismutase (SOD) [11]. Similarly, studies showing that diosgenin possesses antioxidant-mediated antiapoptotic activity can also be found in its blocking effect on doxorubicin-induced cardiac injury [12] and hydrogen peroxide (H2O2)-induced apoptosis in human vein endothelial cells [13]. A common finding regarding the antioxidant activity of diosgenin found in similar studies [14, 15], including these, is that the blockade of ROS production is mostly associated with an improvement in mitochondrial dysfunction, which is the opposite of the anticancer activity of this compound in a number of cancer cell lines. In particular, Chen et al. [16] reported that diosgenin not only attenuates Fas-dependent and mitochondria-dependent apoptosis in H2O2-exposed cardiomyocytes through activation of the insulin-like growth factor-1 survival pathway, but also inhibits H2O2-induced cytotoxicity through estrogen receptor-activated phosphorylation of phosphoinositide 3-kinase/Akt and extracellular signal-regulated kinase signaling pathways in cardiomyocytes via estrogen receptor interaction. Diosgenin also prevents or inhibits death receptor-mediated and mitochondria-initiated apoptosis in ovariectomized cardiac cells [17]. These results support those of previous studies showing that diosgenin is effective in preventing diseases such as cardiac fibrosis [18] and myocardial inflammatory damage [19].
Furthermore, it has recently been reported that diosgenin can enhance the anabolic action of myotubes by interacting with the natural steroid hormone ecdysterone [20], and can attenuate glucocorticoid-induced skeletal muscle atrophy [21]. These results correlate well with those of Kusano et al. [22], who reported that diosgenin supplementation increased the area and diameter of thigh skeletal myofibers and promoted the differentiation and strengthening of skeletal muscle by activating the catabolic pathway. They concluded that this effect of diosgenin was due to the enhancement of the catabolic pathway through the activation of the AMP-activated protein kinase (AMPK) pathway, an enzyme that induces myoblast fusion and senses and regulates the intracellular energy status. Interestingly, in skeletal muscles exposed to oxidative stress, AMPK activation is closely linked to damage to mitochondria, the source of energy production [23, 24]. Skeletal muscles consume a lot of energy for muscle contraction, and this process consumes a lot of oxygen, which continuously produces oxidant species such as ROS. Since oxidative stress plays a key role in compromising muscle homeostasis by inhibiting skeletal muscle differentiation and increasing muscle loss [25, 26], it is evident that blocking mitochondrial pathological damage caused by oxidative stress is important for maintaining skeletal muscle homeostasis. However, the potential of diosgenin to protect against oxidative stress-induced skeletal muscle injury has not been properly evaluated. Therefore, this study aimed to establish the basis that diosgenin can block the damage to skeletal muscle cells caused by oxidative stress, wherein, C2C12 murine myoblasts were used, and H2O2 was used as an oxidative stress inducer. In particular, we focused on elucidating that the blockade of oxidative stress-induced cytotoxicity by diosgenin is due to the inhibition of mitochondrial ROS following mitochondrial dysfunction.
Materials and methods
Cell culture and diosgenin treatment
C2C12 cells (American Type Culture Collection, Manassas, VA, USA) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum and antibiotic mixtures at 37 °C and 5% CO2. All the materials used for cell culture were purchased from WelGENE (Gyeongsan, Republic of Korea). Diosgenin (Sigma-Aldrich Co., St. Louis, MO, USA) was solubilized in dimethyl sulfoxide (Sigma-Aldrich Co.) to prepare a stock solution, which was then diluted to various concentrations with culture medium before treating the cells.
Cytotoxicity assay
Cells were cultured in medium containing diosgenin or H2O2 at different concentrations for 48 h or pretreated with or without 25 µM diosgenin or 100 µM Mito-TEMPO for 1 h, and then treated with 0.5 mM H2O2 for 48 h. After treatment, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed as previously described [27]. The lactate dehydrogenase (LDH) release assay was performed using the LDH activity assay kit (Sigma-Aldrich Co.) according to the manufacturer’s protocol. Images were captured using an optical microscope (Carl Zeiss, Oberkochen, Germany).
Comet assay
The comet assay kit (Trevigen Inc., Gaithersburg, MD, USA) was used to assess DNA damage. Briefly, H2O2-exposed cells in the presence or absence of diosgenin were collected and the comet assay was performed as described by the manufacturer. Randomly selected images were acquired using a fluorescence microscope (Carl Zeiss).
Detection of 8-hydroxy-2’-deoxyguanosine (8-OHdG) levels
Levels of 8-OHdG, an oxidized nucleoside form of DNA were determined using an 8-OHdG ELISA kit (Abcam, Cambridge, UK). The cells were mixed with the reaction buffer and then allowed to react with the 8-OHdG antibody provided in the kit according to the manufacturer’s instructions. The cells were then washed with washing buffer and the absorbance was measured using an ELISA reader (BioTek, Winooski, VT, USA) at 405 nm, as previously reported [28].
Quantitative assessment of apoptosis
To investigate the degree of apoptosis induction, a FITC annexin V/propidium iodide (PI) apoptosis detection kit (BD Bioscience, Franklin Lakes, NJ, USA) was used. Briefly, Annexin V-FITC and PI buffer were added to cells suspended in binding buffer and it was allowed to react for 20 min, according to the manufacturer’s protocol [29]. The cell suspension was analyzed using a flow cytometer (Millipore Corporation, Hayward, CA, USA).
Protein extraction and immunoblotting
Whole cell lysates were prepared from cells cultured under various conditions as previously described [30]. Mitochondrial and cytoplasmic fractions were isolated using a Mitochondria/Cytosol Fractionation Kit (Sigma-Aldrich Co.). Equal amounts of protein extracted from the cells of each treatment group were fractionated by electrophoresis using sodium dodecyl sulfate-polyacrylamide gels and transferred to immunoblot membranes (Bio-Rad Lab. Inc., Hercules, CA, USA). The protein-transferred membranes were hybridized with primary antibodies against the target proteins and then incubated with secondary antibodies conjugated to horseradish peroxidase. Proteins were detected using an enhanced chemiluminescence detection kit (Sigma-Aldrich Co.). Antibodies were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA), Cell Signaling Technology, Inc. (Danvers, MA, USA), and Abcam, Inc. Cytochrome c oxidase subunit IV (COX IV) and β-actin were probed as loading controls for mitochondrial and cytosolic proteins, respectively.
Caspase-3 activity assay
The activity of caspase-3 in cells cultured under various conditions was calculated using a caspase-3 assay kit (Abcam, Inc.) based on the hydrolysis of fluorescent substrate peptides by activated caspases. Briefly, after resuspending the cells in the cell lysis buffer provided in the kit, the supernatants were allowed to react with the substrate according to the manufacturer’s instructions. Finally, the concentration of p-nitroaniline released from the substrates was determined using a microplate reader [31].
Mitochondrial Membrane Potential (MMP) assay
To analyze the MMP, 5,5,6,6’-tetrachloro‐1,1’,3,3’‐tetraethylbenzimi‐dazoylcarbocyanine iodide (JC-1) fluorescent dye (Abcam, Inc.) was used. Cells were stained with 10 µM JC-1 for 30 min according to the manufacturer’s protocol. Images of JC-1-stained cells were monitored using fluorescence microscopy, as previously described [32].
ROS generation assay
Intracellular ROS and mitochondrial ROS (mtROS) levels were analyzed using the dichlorodihydrofluorescein diacetate (DCF-DA) and MitoSOX staining technique, respectively. Briefly, harvested cells were incubated with 10 µM DCF-DA or 5 µM MitoSOX (Thermo Fisher Scientific, Waltham, MA, USA), and intracellular ROS and mtROS levels were measured using flow cytometry [33]. Additionally, fluorescence images of the cells stained with DCF-DA or MitoSOX were acquired using a fluorescence microscope. To visualize the location of nuclei in ROS fluorescence images, cells were additionally counterstained with 4′,6′-diamidino-2-phenylindole (DAPI, Sigma-Aldrich Co.) solution.
Assessment of the GSH/oxidized glutathione (GSSH) ratio
The antioxidant capacity of diosgenin was determined using the GSH assay kit (Sigma-Aldrich Co.), and the GSH/GSSG ratio was calculated using a standard curve containing known amounts of GSH and GSSG.
GPx and manganese SOD (MnSOD) activity measurement
The enzyme activities of GPx and MnSOD were analyzed using commercially available kits (Sigma-Aldrich Co.). Briefly, cell lysates were immunoprecipitated with antibodies against each enzyme, and the relative activity values were determined based on the measured absorbance according to the manufacturer’s protocol.
Statistical analysis
All data were statistically analyzed using GraphPad Prism 5.03 software (GraphPad Software Inc., La Jolla, CA, USA) using an unpaired two-tailed Student’s t-test and one-way analysis of variance. All results are presented as mean ± standard deviation (SD) of at least triplicate independent experiments. p was set at p < 0.05.
Results
Diosgenin reduced H2O2-induced cytotoxicity in C2C12 cells
An MTT assay was performed to study the protective ability of diosgenin against oxidative damage in C2C12 cells. According to the results shown in Fig. 1A, cell viability decreased in the 50 µM diosgenin treatment group; therefore, the pretreatment concentration of diosgenin was set to 25 µM or less. The concentration of H2O2 used to mimic oxidative stress (0.5 mM) resulted in approximately 60% cell viability as analyzed using the MTT assay (Fig. 1B). As shown in Fig. 1B, pretreatment with diosgenin concentration-dependently attenuated the H2O2-induced reduction in cell viability. Diosgenin also significantly blocked the leakage of LDH, which indicates cell damage in the H2O2-treated cells (Fig. 1C). H2O2-induced morphological changes and numerical reduction of cells were also suppressed in the presence of diosgenin (Data not shown) indicating that diosgenin attenuated H2O2-induced cytotoxicity in C2C12 cells.
Diosgenin inhibited H2O2-induced cytotoxicity in C2C12 cells. Cells were treated with different concentrations of diosgenin for 48 h (A) or treated with the indicated concentrations of diosgenin for 1 h and then treated with H2O2 for 48 h (B-D). Cell viability was analyzed using the MTT assay (A and B) or the relative level of LDH release (C) was investigated. **p < 0.01 and ***p < 0.001 vs. control cells; #p < 0.05 and ###p < 0.001 vs. H2O2-treated cells
Diosgenin inhibited H2O2-induced DNA damage in C2C12 cells
To explore whether the reduction in H2O2-induced cytotoxicity by diosgenin was associated with the inhibition of DNA damage, the comet assay was performed and 8-OHdG levels were examined. As shown in Fig. 2A, the formation of comet tails, which indicates DNA double-helix breakage, markedly increased in the H2O2-treated cells. Additionally, the level of 8-OHdG, a marker of oxidative DNA damage, significantly increased following H2O2 treatment (Fig. 2B). However, increase in the expression of these DNA damage markers was largely offset by the presence of diosgenin, suggesting that diosgenin protects DNA from oxidative stress.
Diosgenin attenuated H2O2-induced DNA damage and apoptosis in C2C12 cells. Cells were pretreated with or without diosgenin for 1 h and then stimulated with H2O2 for 48 h. (A and B) The extent of DNA damage was examined by Comet assay (A) or changes in 8-OHdG levels (B). (C) After treatment, the degree of apoptosis was analyzed using the Annexin V/PI staining technique, and the frequency of Annexin V-positive cells in each treatment group was presented. (D) The change in expression of the indicated proteins were investigated using total proteins isolated from cells. The loading control used was β-actin. (E) Caspase-3 activity was assessed using a commercially available kit. ***p < 0.001 vs. control cells; ###p < 0.001 vs. H2O2-treated cells
Diosgenin suppressed H2O2-induced apoptosis in C2C12 cells
To determine whether the H2O2-induced cytotoxicity suppressed by diosgenin was due to the inhibition of apoptosis, flow cytometric analysis was performed. As shown in Fig. 2C, the frequency of annexin-positive cells, indicating the induction of apoptosis, significantly increased in cells treated with H2O2, whereas it significantly reduced in the presence of diosgenin. In addition, the expression of Bcl-2 protein, an anti-apoptotic factor that reduced due to H2O2 treatment, and the increase in Bax, a pro-apoptotic protein, was maintained at control levels by pretreatment with diosgenin (Fig. 2D). In parallel, the expression of the pro-form of caspase-3, a key effector of caspase-dependent apoptosis, was downregulated in H2O2-treated cells, whereas the enzymatic activity and degradation of poly(ADP-ribose) polymerase (PARP), a representative substrate protein of caspase-3 increased (Fig. 2D and E). However, neither caspase-3 activation nor PARP degradation occurred in the presence of diosgenin (Fig. 2D and E), indicating that diosgenin inhibited the induction of apoptosis by preventing an increase in the Bax/Bcl-2 expression ratio and inactivating the caspase cascade due to H2O2 stimulation.
Diosgenin attenuated mitochondrial impairment in H2O2-treated C2C12 cells
To evaluate whether the protection of H2O2-trigerded apoptosis by diosgenin was due to the maintenance of mitochondrial homeostasis, we investigated changes in MMP, an indicator of mitochondrial stability. According to the fluorescence microscopy results after JC-1 staining, an increase in the green-to-red fluorescence ratio was observed in the H2O2-treated cells (Fig. 3A and B), indicating mitochondrial depolarization and a loss of MMP. The expression of cytochrome c, which exists in the space between the outer and inner mitochondrial membranes in normal cells, was upregulated in the cytosolic fraction of H2O2-treated cells but downregulated in the mitochondrial fraction (Fig. 3C). However, the translocation of cytochrome c into the cytoplasm was effectively blocked by diosgenin, suggesting that diosgenin blocked H2O2-induced disruption of mitochondrial membrane stability in the C2C12 cells.
Diosgenin counteracted H2O2-induced mitochondrial dysfunction in C2C12 cells. Cells were pretreated with or without diosgenin for 1 h and then exposed to H2O2 for 48 h. (A and B) After JC-1 staining, the fluorescence intensities of JC-1 aggregates (red) and monomers (green) were observed (A), and their relative intensities were indicated (B). ***p < 0.001 vs. control cells; ###p < 0.001 vs. H2O2-treated cells. (C) Change in expression of cytochrome c were investigated using mitochondrial and cytosolic fractions isolated from cells. COX IV and β-actin were used as loading controls for each fraction
Diosgenin restored ROS production and the decreased activity of antioxidant enzymes in H2O2-treated C2C12 cells
To confirm the antioxidant activity of diosgenin, we assessed whether this compound could block the increase in ROS production by H2O2. According to the results of DCF-DA staining, the increased intracellular ROS levels in H2O2-treated cells significantly reduced by pretreatment with diosgenin, similar to the effect of NAC, a well-known ROS scavenger (Fig. 4A). Moreover, a decrease in the GSH/GSSG ratio, a marker of oxidative stress, was observed in H2O2-treated cells. This decrease was significantly alleviated in the NAC and diosgenin pretreatment groups (Fig. 4B). Therefore, we examined the changes in the expression of GPx1, which plays a major role in removing intracellular ROS, and MnSOD, which is active in the mitochondrial matrix, and confirmed that their expression and activity, which were reduced by H2O2 treatment, were neutralized by diosgenin (Fig. 4C-E). These results suggest that the antioxidant activity of diosgenin involves the activation of these enzymes.
Diosgenin removed H2O2-induced oxidative stress in C2C12 cells. Cells were pretreated with or without diosgenin or NAC for 1 h and then treated with H2O2 for 1 h (A and B) or 48 h (C-F). (A) Changes in intracellular ROS levels were examined after DCF-DA staining, and the frequency of DCF-positive cells in each treatment group was presented. (B) Intracellular GSH/GSSG ratio was measured using a GSH assay kit. (C) Expression changes of GPx1 and MnSOD were examined using total proteins isolated from cells. (D and E) Enzyme activities of GPx (E) and MnSOD (F) were measured using the corresponding assay kits. ***p < 0.001 vs. control cells; ###p < 0.001 vs. H2O2-treated cells
Diosgenin alleviated H2O2-induced MtROS production in C2C12 cells
Because diosgenin restored the inactivation of MnSOD by H2O2, we examined whether its antioxidant activity was related to the inhibition of mtROS production. Staining with MitoSOX, a mitochondrial superoxide indicator, showed that diosgenin significantly blocked the production of mtROS induced by H2O2, similar to Mito-TEMPO, a mitochondrially targeted antioxidant (Fig. 5A). To confirm the ROS production inhibition ability of diosgenin, fluorescent images of cells stained with DCF-DA and MitoSOX were obtained. As shown in Fig. 5B-D, diosgenin significantly reduced the H2O2-induced fluorescence intensity, indicating not only the accumulation of intracellular ROS (green) but also the production of mtROS (red). Furthermore, when mtROS production by H2O2 was artificially blocked, both the increase in apoptosis and decrease in cell viability observed in H2O2-treated cells significantly improved (Fig. 6). Thus, maintenance of mitochondrial homeostasis by diosgenin significantly contributes to the inhibition of oxidative stress-mediated cytotoxicity in C2C12 cells.
Diosgenin restored H2O2-induced mtROS production in C2C12 cells. Cells were pretreated with or without diosgenin or Mito-TEMPO for 1 h and then treated with H2O2 for 1 h. (A) Changes in mtROS levels after MitoSOX staining were investigated, and the frequency of MitoSOX-positive cells in each treatment group was presented. (B-D) After treatment, cells were stained with DCF-DA (green) and MitoSOX (red), and then nuclei were additionally stained with DAPI (blue). Representative fluorescence images (B) and the relative values of each fluorescence intensity (C and D) are presented. ***p < 0.001 vs. control cells; ###p < 0.001 vs. H2O2-treated cells
The reverse effect of H2O2-induced apoptosis and cytotoxicity by diosgenin in C2C12 cells was comparable to that of the mtROS production inhibitor. Cells were pretreated with or without diosgenin or Mito-TEMPO for 1 h and then exposed to H2O2 for 48 h. (A) The degree of apoptosis was examined by Annexin V/PI staining, and the frequency of Annexin V-positive cells in each treatment group was presented. (B) The MTT assay was performed using cells cultured under the same conditions. ***p < 0.001 vs. control cells; ###p < 0.001 vs. H2O2-treated cells
Discussion
Recent studies have shown that diosgenin promotes skeletal muscle differentiation and inhibits muscle atrophy [20,21,22]. Oxidative stress can impair the two essential processes of skeletal muscle strengthening. Therefore, in this study, we investigated whether diosgenin protects against oxidative damage in myoblasts and cells prior to differentiation into myotubes. We first performed an MTT assay, a widely used method to measure in vitro cell viability based on changes in mitochondrial activity [34]. Since this assay reflects total mitochondrial activity as the number of viable cells, our finding that diosgenin protected against the decrease in cell viability in H2O2-treated cells may be related to the maintenance of mitochondrial homeostasis. Meanwhile, an increase in LDH, a cytosolic enzyme found in most cells in cell culture supernatants, indicates that the cell membrane is damaged, and the cells are undergoing necrosis, apoptosis, or other forms of cell death [35]. Our finding that diosgenin counteracted H2O2-induced extracellular release of LDH using this assay provides evidence that this blocked cell death. These results imply that the inhibition of cell death induced by oxidants, such as H2O2 in C2C12 cells by diosgenin is based on the preservation of mitochondrial function.
Most of the cell damage caused by oxidative stimuli is accompanied by DNA damage and unrepaired DNA damage contributes to cell transformation and death [32, 36]. Similar to our results, Son et al. [37] reported that diosgenin blocked lymphocyte DNA damage caused by H2O2 using a comet assay. The comet assay is a well-established method for assessing DNA damage based on the cleavage of the DNA double helix, and the formation of the comet tail is an indicator of apoptosis [38]. Diosgenin decreased the increase in the level of 8-OHdG, which indicates the level of nucleosides in oxidized DNA caused by H2O2, suggesting that this saponin enhanced the resistance to oxidative DNA damage in C2C12 cells. In addition, diosgenin reduced apoptosis by inhibiting the Bax/Bcl-2 expression ratio, caspase-3 activity, and PARP degradation in C2C12 cells treated with H2O2. These antiapoptotic effects of diosgenin are consistent with previously reported results [39,40,41]. Bcl-2 family proteins are major factors that regulate pore formation in the mitochondrial outer membrane and are composed of factors that can inhibit or promote apoptosis. Bax induces apoptosis by attacking mitochondria and increasing mitochondrial outer membrane permeability, whereas Bcl-2 is a pro-survival factor that supports cell survival by inhibiting apoptotic activity. Compared to Bcl-2, increased Bax weakens MMP, thereby releasing cytochrome c into the cytoplasm to activate the caspase cascade and terminate apoptosis [42, 43]. Therefore, our results suggest that diosgenin neutralizes the H2O2-induced increase in the Bax/Bcl-2 ratio, thereby maintaining mitochondrial stability and preventing cytochrome c release or caspase activation, ultimately blocking the mitochondria-mediated apoptotic pathway.
In a subsequent study, diosgenin inhibited ROS production and preserved the GSH/GSSG ratio in response to H2O2 treatment in C2C12 cells. Similar results have been reported in previous studies [12,13,14, 16] and we propose that the activities of MnSOD and GPx also contribute to the antioxidant potential of diosgenin. The results showed that the expression and activity of MnSOD, which reduced due to H2O2 stimulation, was the same in the presence of diosgenin, which may serve as evidence that it directly contributes to the preservation of mitochondrial function. This is because MnSOD is an enzyme that scavenges the mitochondrial superoxide present in the mitochondrial matrix [44], indicating that diosgenin inhibits the production of mtROS caused by H2O2 treatment. Dioscin, a natural steroid saponin composed of diosgenin, has been reported to increase the expression levels of MnSOD, CAT, and SOD to suppress oxidative injury induced by intestinal ischemia-reperfusion, thereby reducing ROS levels [45]. This is the first report to show that diosgenin itself increases the activity of MnSOD in an oxidative environment. Moreover, the alleviation of H2O2-induced apoptosis and cell viability reduction by mitochondria-targeting antioxidants in C2C12 cells was very similar to that of diosgenin, suggesting that the antioxidant capacity of diosgenin was closely associated with the inhibition of mtROS production.
In the current study, we propose that the preventive effect of diosgenin on C2C12 myoblasts in an oxidative environment mimicked by H2O2 treatment is due to the maintenance of mitochondrial homeostasis by ameliorating mtROS production. The ROS-scavenging effect of diosgenin may contribute to the inhibition of DNA damage and apoptosis during myoblast survival. However, as suggested by previous studies, various intracellular signaling pathways are involved in the antioxidant activity of diosgenin. For example, the improvement of mitochondrial dysfunction by this saponin was mediated through increased activity of AMPK, which suggests that mitochondrial biogenesis was enhanced [10, 16, 46, 47]. Nuclear factor erythroid-2-related factor 2, a regulator of redox homeostasis, plays a key role in ameliorating nonalcoholic fatty liver disease and renal inflammation by eliminating diosgenin-induced oxidative stress [48, 49]. And since diosgenin rescued H2O2-induced increase in mitochondrial superoxide and decrease in the expression and activity of MnSOD, it is difficult to predict that diosgenin completely blocked the production of mitochondrial ROS. Therefore, how the mtROS-scavenging efficacy of diosgenin is related to various signaling pathways involved in redox regulation and cell survival needs to be further studied. These include investigations of stress response pathways involving the role of the upstream transcription factors of MnSOD and studies in vivo models.
In conclusion, our results showed that diosgenin significantly suppressed DNA damage and apoptosis in H2O2-treated C2C12 myoblasts, which was related to the blockade of caspase cascade activation, and the Bax/Bcl-2 ratio increased following H2O2 treatment. In addition, diosgenin maintained the stability of mitochondrial membranes in H2O2-treated cells, as evidenced by an improvement in MMP loss and inhibition of cytochrome c efflux into the cytosol. Furthermore, diosgenin significantly maintained the GSH/GSSG ratio and GPx activity under H2O2-treated conditions, while eliminating ROS production. Diosgenin also attenuated H2O2-induced reduction in MnSOD activity and the production of mtROS, suggesting that the control of mtROS generation acts as an upstream event to preserve mitochondrial homeostasis for the suppression of oxidative damage by this saponin (Fig. 7). Therefore, the present results suggest that diosgenin has potential as a therapeutic agent to protect muscle cells from oxidative injury.
Data availability
All data is available in the main text.
References
Narzary C, Sarkar D, Das P, Papi D (2025) Ethanobotany, phytochemistry and Pharmacological activity of Dioscorea bulbifera: A comprehensive review. Chem Biodivers 22(1):e202401408. https://doi.org/10.1002/cbdv.202401408
Wang Z, Zhao S, Tao S, Hou G, Zhao F, Tan S et al (2023) Dioscorea spp.: Bioactive compounds and potential for the treatment of inflammatory and metabolic diseases. Molecules 28(6):2878. https://doi.org/10.3390/molecules28062878
Pathak N, Negi AS (2024) Plant based steroidal and triterpenoid sapogenins: chemistry on Diosgenin and biological aspects. Eur J Med Chem 279:116915. https://doi.org/10.1016/j.ejmech.2024.116915
Tak Y, Kaur M, Chitranashi A, Samota MK, Verma P, Bali M et al (2024) Fenugreek derived Diosgenin as an emerging source for diabetic therapy. Front Nutr 11:1280100. https://doi.org/10.3389/fnut.2024.1280100
Mohseni-Moghaddam P, Khanmohammadi M, Roghani M (2023) Literature review on hepatoprotective effects of Diosgenin: possible mechanisms of action. Front Pharmacol 14:1226548. https://doi.org/10.3389/fphar.2023.1226548
Zhang SZ, Liang PP, Feng YN, Yin GL, Sun FC, Ma CQ et al (2022) Therapeutic potential and research progress of Diosgenin for lipid metabolism diseases. Drug Dev Res 83(8):1725–1738. https://doi.org/10.1002/ddr.21991
Ren QL, Wang Q, Zhang XQ, Wang M, Hu H, Tang JJ et al (2023) Anticancer activity of Diosgenin and its molecular mechanism. Chin J Integr Med 29(8):738–749. https://doi.org/10.1007/s11655-023-3693-1
Wang Q, Zhang L, Xiao Y, Ghaemnezhad A (2022) Diosgenin promotes cisplatin-induced apoptosis through oxidative DNA damage in A549 non-small cell lung cells. Cell Biol Int 46(10):1571–1576. https://doi.org/10.1002/cbin.11862
Arya P, Kumar P (2021) Diosgenin a steroidal compound: an emerging way to cancer management. J Food Biochem 45(12):e14005. https://doi.org/10.1111/jfbc.14005
Zhong Y, Wang L, Jin R, Liu J, Luo R, Zhang Y et al (2023) Diosgenin inhibits ROS generation by modulating NOX4 and mitochondrial respiratory chain and suppresses apoptosis in diabetic nephropathy. Nutrients 15(9):2164. https://doi.org/10.3390/nu15092164
Zhong Y, Li Z, Jin R, Yao Y, He S, Lei M et al (2022) Diosgenin ameliorated type II diabetes-associated nonalcoholic fatty liver disease through inhibiting de Novo lipogenesis and improving fatty acid oxidation and mitochondrial function in rats. Nutrients 14(23):4994. https://doi.org/10.3390/nu14234994
Chen CT, Wang ZH, Hsu CC, Lin HH, Chen JH (2015) In vivo protective effects of Diosgenin against doxorubicin-induced cardiotoxicity. Nutrients 7(6):4938–4954. https://doi.org/10.3390/nu7064938
Gong G, Qin Y, Huang W, Zhou S, Wu X, Yang X et al (2010) Protective effects of Diosgenin in the hyperlipidemic rat model and in human vascular endothelial cells against hydrogen peroxide-induced apoptosis. Chem Biol Interact 184(3):366–375. https://doi.org/10.1016/j.cbi.2010.02.005
Tang X, Huang G, Zhang T, Li S (2020) Elucidation of colon-protective efficacy of Diosgenin in experimental TNBS-induced colitis: Inhibition of NF-kB/IkB-a and bax/caspase-1 signaling pathways. Biosci Biotechnol Biochem 84(9):1903–1912. https://doi.org/10.1080/09168451.2020.1776590
Chen MY, Tsai BC, Kuo WW, Kuo CH, Lin YM, Hsieh DJ et al (2023) Diosgenin attenuates myocardial cell apoptosis triggered by oxidative stress through Estrogen receptor to activate the PI3K/Akt and ERK axes. Am J Chin Med 51(5):1211–1232. https://doi.org/10.1142/S0192415X23500556
Zhong Y, Sun D, Yao Y, Liu Q, Guo T, Wang X et al (2022) Autophagy and mitochondrial dynamics contribute to the protective effect of Diosgenin against 3-MCPD induced kidney injury. Chem Biol Interact 355:109850. https://doi.org/10.1016/j.cbi.2022.109850
Wu XB, Lai CH, Ho YJ, Kuo CH, Lai PF, Tasi CY et al (2022) Anti-apoptotic effects of Diosgenin on ovariectomized hearts. Steroids 179:108980. https://doi.org/10.1016/j.steroids.2022.108980
Zhou HT, Yu XF, Zhou GM (2017) Diosgenin inhibits angiotensin II-induced extracellular matrix remodeling in cardiac fibroblasts through regulating the TGF-b1/Smad3 signaling pathway. Mol Med Rep 15(5):2823–2828. https://doi.org/10.3892/mmr.2017.6280
Wang HW, Liu HJ, Cao H, Qiao ZY, Xu YW (2018) Diosgenin protects rats from myocardial inflammatory injury induced by ischemia-reperfusion. Med Sci Monit 24:246–253. https://doi.org/10.12659/msm.907745
Kostov T, Diel P, Isenmann E (2024) Examination of the anabolic activity and mechanisms of action of the combination of Diosgenin and ecdysterone in C2C12 myotubes. Toxicol Lett 401:181–189. https://doi.org/10.1016/j.toxlet.2024.10.005
Yoshioka Y, Oishi S, Onoda K, Shibata K, Miyoshi N (2023) Diosgenin prevents dexamethasone-induced myotube atrophy in C2C12 cells. Arch Biochem Biophys 747:109759. https://doi.org/10.1016/j.abb.2023.109759
Kusano Y, Tsujihara N, Masui H, Shibata T, Uchida K, Takeuchi W (2019) Diosgenin supplementation prevents lipid accumulation and induces skeletal muscle-fiber hypertrophy in rats. J Nutr Sci Vitaminol (Tokyo) 65(5):421–429. https://doi.org/10.3177/jnsv.65.421
Bouviere J, Fortunato RS, Dupuy C, Werneck-de-Castro JP, Carvalho DP, Louzada RA (2021) Exercise-stimulated ROS sensitive signaling pathways in skeletal muscle. Antioxid (Basel) 10(4):537. https://doi.org/10.3390/antiox10040537
Alizadeh Pahlavani H, Laher I, Knechtle B, Zouhal H (2022) Exercise and mitochondrial mechanisms in patients with sarcopenia. Front Physiol 13:1040381. https://doi.org/10.3389/fphys.2022.1040381
Zhang H, Qi G, Wang K, Yang J, Shen Y, Yang X et al (2023) Roles in skeletal muscle atrophy. Biochem Pharmacol 214:115664. https://doi.org/10.1016/j.bcp.2023.115664
Lian D, Chen MM, Wu H, Deng S, Hu X (2022) The role of oxidative stress in skeletal muscle myogenesis and muscle disease. Antioxid (Basel) 11(4):755. https://doi.org/10.3390/antiox11040755
Kang JB, Son HK, Park DJ, Jin YB, Koh PO (2024) Chlorogenic acid regulates the expression of protein phosphatase 2A subunit B in the cerebral cortex of a rat stroke model and glutamate-exposed neurons. Lab Anim Res 40(1):8. https://doi.org/10.1186/s42826-024-00196-5
Hwangbo H, Park C, Bang E, Kim HS, Bae SJ, Kim E et al (2024) Morroniside protects C2C12 myoblasts from oxidative damage caused by ROS-mediated mitochondrial damage and induction of Endoplasmic reticulum stress. Biomol Ther (Seoul) 32(3):349–360. https://doi.org/10.4062/biomolther.2024.012
Jeon SJ, Jung GH, Choi EY, Han EJ, Lee JH, Han SH et al (2024) Kaempferol induces apoptosis through the MAPK pathway and regulates JNK-mediated autophagy in MC-3 cells. Toxicol Res 40(1):45–55. https://doi.org/10.1007/s43188-023-00206-z
Kang GS, Kim YE, Oh HR, Jo HJ, Bok S, Jeon YK et al (2024) Hypoxia-inducible factor-1a-deficient adipose-tissue macrophages produce the heat to mediate lipolysis of white adipose tissue through uncoupling protein-1. Lab Anim Res 40(1):37. https://doi.org/10.1186/s42826-024-00224-4
Park C, Cha HJ, Hwangbo H, Bang E, Kim HS, Yun SJ et al (2024) Activation of Heme oxygenase-1 by mangiferin in human retinal pigment epithelial cells contributes to blocking oxidative damage. Biomol Ther (Seoul) 32(3):329–340. https://doi.org/10.4062/biomolther.2023.175
Ni H, Hu X, Yang N, Liu X, Cai W, Zhong R et al (2024) Roundup® induces premature senescence of mouse granulosa cells via mitochondrial ROS-triggered NLRP3 inflammasome activation. Toxicol Res 40(3):377–387. https://doi.org/10.1007/s43188-024-00229-0
Jeon HJ, Seo JH, Jeong E, Son CY, Rawding PA, Hwang Y et al (2024) Carcinoembryonic antigen-positive Circulating epithelial cells as a biomarker for the diagnosis and prognosis of colorectal cancer. Biotechnol Bioprocess Eng 29(5):877–889. https://doi.org/10.1007/s12257-024-00115-4
Plumb JA (2004) Cell sensitivity assays: the MTT assay. Methods Mol Med 88:165–169. https://doi.org/10.1385/1-59259-406-9:165
Kaja S, Payne AJ, Naumchuk Y, Koulen P (2017) Quantification of lactate dehydrogenase for cell viability testing using cell lines and primary cultured astrocytes. Curr Protoc Toxicol 72(2261–). https://doi.org/10.1002/cptx.21
Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T et al (2021) The double-edged roles of ROS in cancer prevention and therapy. Theranostics 11(10):4839–4857. https://doi.org/10.7150/thno.56747
Son IS, Kim JH, Sohn HY, Son KH, Kim JS, Kwon CS (2007) Antioxidative and hypolipidemic effects of Diosgenin, a steroidal saponin of Yam (Dioscorea spp.), on high-cholesterol fed rats. Biosci Biotechnol Biochem 71(12):3063–3071. https://doi.org/10.1271/bbb.70472
Cordelli E, Bignami M, Pacchierotti F (2021) Comet assay: A versatile but complex tool in genotoxicity testing. Toxicol Res (Camb) 10(1):68–78. https://doi.org/10.1093/toxres/tfaa093
El-Far AH, Elghaity MM, Mohamed SA, Noreldin AE, Elewa YHA, Al Jaouni SK et al (2024) Diosgenin alleviates D-galactose-induced oxidative stress in rats’ brain and liver targeting aging and apoptotic marker genes. Front Mol Biosci 11:1303379. https://doi.org/10.3389/fmolb.2024.1303379
Liao WL, Huang CP, Wang HP, Lei YJ, Lin HJ, Huang YC (2023) Diosgenin, a natural steroidal Sapogenin, alleviates the progression of diabetic retinopathy in diabetic mice. Vivo 37(2):661–666. https://doi.org/10.21873/invivo.13126
Jin C, Miao X, Zhong Y, Han J, Liu Q, Zhu J et al (2020) The renoprotective effect of Diosgenin on aristolochic acid I-induced renal injury in rats: impact on apoptosis, mitochondrial dynamics and autophagy. Food Funct 11(9):7456–7467. https://doi.org/10.1039/d0fo00401d
Glover HL, Schreiner A, Dewson G, Tait SWG (2024) Mitochondria and cell death. Nat Cell Biol 26(9):1434–1446. https://doi.org/10.1038/s41556-024-01429-4
Sahoo G, Samal D, Khandayataray P, Murthy MK (2023) A review on caspases: key regulators of biological activities and apoptosis. Mol Neurobiol 60(10):5805–5837. https://doi.org/10.1007/s12035-023-03433-5
Liu M, Sun X, Chen B, Dai R, Xi Z, Xu H (2022) Insights into manganese superoxide dismutase and human diseases. Int J Mol Sci 23(24):15893. https://doi.org/10.3390/ijms232415893
Hu Y, Mao Z, Xu L, Yin L, Tao X, Tang Z et al (2018) Protective effect of Dioscin against intestinal ischemia/reperfusion injury via adjusting miR-351-5p-mediated oxidative stress. Pharmacol Res 137:56–63. https://doi.org/10.1016/j.phrs.2018.09.016
Fan JJ, Ding WD, Liang YF, Wei YX, Huang Y, Ma L et al (2024) Diosgenin derivative ML5 attenuates MPTP-induced neuronal impairment via regulating AMPK/PGC-1a-mediated mitochondrial biogenesis and fusion/fission. Am J Transl Res 16(8):3582–3598. https://doi.org/10.62347/JBRE5043
Yuan H, Sui H, Li S (2023) Diosgenin alleviates the inflammatory damage and insulin resistance in high glucose-induced podocyte cells via the AMPK/SIRT1/NF-kB signaling pathway. Exp Ther Med 25(6):259. https://doi.org/10.3892/etm.2023.11958
Zhang X, Yin G, Chen S, Meng D, Yu W, Liu H et al (2024) Diosgenin ameliorating non-alcoholic fatty liver disease via Nrf2-mediated regulation of oxidative stress and ferroptosis. Diabetes Obes Metab 26(12):5745–5756. https://doi.org/10.1111/dom.15945
Jia S, Si R, Liu G, Zhong Q (2024) Diosgenin protects against cationic bovine serum albumin-induced membranous glomerulonephritis by attenuating oxidative stress and renal inflammation via the NF-kB pathway. Pharm Biol 62(1):285–295. https://doi.org/10.1080/13880209.2024.2330602
Acknowledgements
The authors would like to thank Core-Facility Center for Tissue Regeneration, Dong-eui University (Busan, Republic of Korea), for letting us use their flow cytometer and fluorescence microscope.
Funding
This study was supported by the Basic Science Research Program grant (RS-2023-00217899) from the National Research Foundation (NRF) of the Republic of Korea.
Author information
Authors and Affiliations
Contributions
Conceived and designed the experiments: YHC. Performed the experiments: CP, GYK. Analyzed the data: CP, GYK YHC. Wrote the paper and reviewed the literature: GYK YHC. All authors have read and agreed to the published version of the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Park, C., Kim, GY. & Choi, Y. The antioxidant activity of diosgenin, a plant steroid sapogenin, in C2C12 myoblasts is achieved by blocking mitochondrial ROS production. Appl Biol Chem 68, 22 (2025). https://doi.org/10.1186/s13765-025-00996-w
Received:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1186/s13765-025-00996-w









