Open Access

Ursolic acid protects against cisplatin‑induced ototoxicity by inhibiting oxidative stress and TRPV1‑mediated Ca2+‑signaling

  • Authors:
    • Yang Di
    • Tao Xu
    • Yuan Tian
    • Tingting Ma
    • Donghao Qu
    • Yan Wang
    • Yuhan Lin
    • Dongyan Bao
    • Li Yu
    • Shuangyue Liu
    • Aimei Wang
  • View Affiliations

  • Published online on: June 4, 2020     https://doi.org/10.3892/ijmm.2020.4633
  • Pages: 806-816
  • Copyright: © Di 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

Cisplatin (CDDP) is widely used in clinical settings for the treatment of various cancers. However, ototoxicity is a major side effect of CDDP, and there is an associated risk of irreversible hearing loss. We previously demonstrated that CDDP could induce ototoxicity via activation of the transient receptor potential vanilloid receptor 1 (TRPV1) pathway and subsequent induction of oxidative stress. The present study investigated whether ursolic acid (UA) treatment could protect against CDDP‑induced ototoxicity. UA is a triterpenoid with strong antioxidant activity widely used in China for the treatment of liver diseases. This traditional Chinese medicine is mainly isolated from bearberry, a Chinese herb. The present results showed that CDDP increased auditory brainstem response threshold shifts in frequencies associated with observed damage to the outer hair cells. Moreover, CDDP increased the expression of TRPV1, calpain 2 and caspase‑3 in the cochlea, and the levels of Ca2+ and 4‑hydroxynonenal. UA co‑treatment significantly attenuated CDDP‑induced hearing loss and inhibited TRPV1 pathway activation. In addition, UA enhanced CDDP‑induced growth inhibition in the human ovarian cancer cell line SKOV3, suggesting that UA synergizes with CDDP in vitro. Collectively, the present data suggested that UA could effectively attenuate CDDP‑induced hearing loss by inhibiting the TRPV1/Ca²+/calpain‑oxidative stress pathway without impairing the antitumor effects of CDDP.

Introduction

Cisplatin (CDDP) is a divalent platinum compound with broad-spectrum anticancer activity that synergizes with a variety of antitumor drugs without cross-resistance, making it a first-line anticancer drug (1). CDDP is commonly used for the treatment of various cancers including bladder cancer, lung cancer, ovarian cancer and other soft tissue tumors. In children, CDDP is also effective for the treatment of various solid tumors (2,3). However, CDDP has serious side effects, including irreversible hearing loss, especially in juvenile patients treated with CDDP and radiotherapy for at least six months (4-6).

The mechanism of CDDP-induced ototoxicity remains to be elucidated. CDDP could induce apoptosis of cochlear cells, which led to hearing loss (7-9). Several studies also showed that CDDP increased NADPH oxidase 3 (NOX 3) expression in the cochlea, which increased the production of reactive oxygen species (ROS) (10,11). ROS directly attacked the cochlear cell membrane and produced the toxic lipid peroxidation product 4-hydroxynonenal (4-HNE), which might also account for cochlear cell apoptosis (12). A study showed that CDDP-induced ototoxicity in the cochlea was related to calcium (Ca2+) over-load (13). As an important second messenger, Ca2+ is involved in various functions, including auditory signal transduction (14). Moreover, maintenance of a low intracellular Ca2+ concentration in the normal resting state plays an important role in sustaining homeostasis (15). Abnormal influx of free Ca2+ in cells (referred to as Ca2+ overload) can cause structural damage and disruption of intracellular metabolism of hair cells (HCs), eventually leading to apoptosis of HCs (16). In the cochlea, Ca2+ overload was related to high expression of the transient receptor potential vanilloid receptor 1 (TRPV1) (17). TRP channels are a family of non-selective cation ion channels that control important cellular functions and signaling pathways (18). TRP channels are also important targets for a wide range of drugs, such as menthol, which adjust human TRP melastatin 8 (hTRPM8)-induced signaling, and could alleviate hyperglycemia in alloxantreated mice (a model of type 1 diabetes) or reverse muscle atrophy in dexamethasone-treated mice (a model of muscle wasting) (19). TRP channel activation can lead to changes in Ca2+ homeostasis, resulting in long lasting modulation of Ca2+ levels (20,21). TRPV1 is expressed in many organs, including the heart, kidney, brain, dorsal root ganglions and sensory neurons (22). TRPV1 plays a critical role in the maintenance of cellular homeostasis, particularly in cochlear cells (23). Studies have suggested that CDDP induced Ca2+ overload by increasing TRPV1 expression, and that Ca2+ subsequently activates calpain, a neutral cysteine protease, to cause cochlear apoptosis (24). However, the association between Ca2+ overload and TRPV1 remains to be confirmed.

Ursolic acid (UA) is a plant-derived triterpene compound with strong antioxidant effects (25-27). UA is the main ingredient of several traditional Chinese herbal medicines used in the treatment of liver diseases and in lowering blood lipid (28,29). In PC12 cells, UA could assert its neuroprotective effects by inhibiting ROS production induced by amyloid beta peptide (30,31). In addition, UA could capture oxygen free radicals in the P450 monoamine oxidase system and liver microsome, thus exerting a strong inhibitory effect on lipid peroxidation (32). However, to the best of our knowl-edge, there are currently no reports on the anti-apoptotic and anti-oxidative effects of UA on ototoxicity induced by CDDP. The present study evaluated the protective effects of UA on CDDP-induced ototoxicity and the underlying mechanisms were investigated. It was hypothesized that UA could protect against CDDP-induced ototoxicity by inhibiting oxidative stress and TRPV1-mediated Ca2+-signaling in the cochlear cells.

Materials and methods

Animal and experimental protocols

A total of 48 BALB/c mice (3 days) were purchased from the Animal Experimental Center, Jinzhou Medical University, China. A total of 48 BALB/c mice (20-22 g, 4 weeks, male and female) with normal Preyer's reflexes were purchased from the Animal Experimental Center, Dalian Medical University, China [license no. SCXK (Liao) 2008-0002]. All animal studies complied with the regulations for the Management of Laboratory Animals published by the Ministry of Science and Technology of the People's Republic of China, and all animal protocols were approved by the Animal Care and Use Committee of Jinzhou Medical University. All mice were fed a standard commercial diet and housed at an ambient temperature of 20-24°C with a relative humidity of 50±5% under a 12/12 h light-dark cycle in a specific pathogen-free facility during the experiment. All animals were allowed 1 week to acclimate before treatment and then were randomly divided into four groups (n=12 in each group) and treated for five days as follows: i) Normal control group, where the mice received physiological saline [0.5 ml/100 g, intra-peritoneally (i.p)]; ii) UA group, where the mice received UA (purity ≥98%; cat. no. N1823, APExBIO Technology LLC; 80 mg/kg/day, i.p); iii) CDDP group, where the mice received CDDP (Sigma-Aldrich; Merck KGaA; 4.5 mg/kg/day, i.p,) and iv) UA plus CDDP (UA+CDDP) group, where the mice received UA (80 mg/kg/day, i.p) followed by CDDP (4.5 mg/kg/day, i.p) 30 min later each day (Fig. 1A).

During the treatment period, the weight of the experimental animals was measured every day (Fig. S1). Although CDDP is toxic, the survival rate of mice is 100% during the administration (33).

Measurement of auditory brainstem response (ABR)

For the analysis of the auditory threshold, ABR was recorded 1 day before and 5 days after related drug treatment with tone bursts of 8, 12 and 24 kHz using the Smart EP & OAE auditory evoked potential recording system (Intelligent Hearing Systems). The mice were anesthetized using pentobarbital sodium (90 mg/kg) and kept warm with a heating pad during ABR recording. A subdermal (active) needle electrode was inserted at the vertex, while ground and reference electrodes were inserted subdermally in the loose skin beneath the pinnae of opposite ears. The technique used to record ABRs has been previously described in detail (34). The ABR waveforms were averaged over a 10 msec time window using the Smart EP & OAE auditory evoked potential recording system software (Launch Pad 2.32 HIS program). The sound intensity was varied at 5 dB intervals near the hearing threshold. The differences in ABR thresholds shift for each frequency between the starting and the terminal points of the experimental time course were noted. The threshold was determined off-line by two independent, experimentally blinded observers based on the ABR records. In addition, the mice were euthanized, and double cochleae were removed for further analysis.

Outer hair cell (OHC) counting

Immediately after completion of the ABR test, the animals were killed by cervical dislocation under anesthesia with an intraperitoneal injection of pentobarbital sodium (90 mg/kg). An array of criteria was used to verify the death of animals, including absence of respiration and heartbeat and loss of corneal and palpebral reflexes.

The temporal bones were removed, and the cochlea was immersed in 4% paraformaldehyde and incubated overnight at 4°C. The cochlea was decalcified in 4% EDTA for 7 days at 4°C. Subsequently, the basilar membrane was dissected under a dissecting microscope, and the stria vascularis and tectorial membrane were removed. The basilar membrane was then stained with phalloidin-tetramethylrhodamine (TRITC; Sigma-Aldrich; Merck KGaA) for 20 min at 20-24°C and protected from light. The specimens were then rinsed three times with 0.01 M phosphate-buffered saline (PBS; pH 7.4) and then mounted on slides. Images were obtained using the TCS-SP5 II laser-scanning confocal microscope (Leica Biosystems GmbH). Outer hair cells were counted from the captured images using a fluorescent BX41 microscope (Olympus Corporation) at ×200 magnification in 20 consecutive fields from the apex to the basal turn along the entire length of the cochlear epithelium. The percentages of outer hair cell loss in each 0.5-mm length of epithelium were plotted as a function of the cochlear length as a cytocochleogram (34).

Immunohistochemical staining of paraffin sections of the cochlea

After the last auditory brainstem response test, five mice from each group were decapitated under abdominal anesthesia using pentobarbital sodium (90 mg/kg), and the temporal bones were removed immediately. The round and oval windows were opened, cochleae were perfused with 40 g/l paraformaldehyde (pH 7.4), and the specimens were immersed in the 4% paraformaldehyde for 24 h at 4°C. Following decalcification in 40 g/l EDTA solution (pH 7.4) for 5 days at 4°C, the cochleae were dehydrated with ethanol, cleared with xylene and embedded in paraffin wax. Serial sections (5-µm thickness) were prepared, dewaxed, rehydrated in descending alcohol series, rinsed with distilled water and treated with high pressure (125°C; 103 kPa; 3 min) to retrieve antigens. Endogenous peroxidase activity was blocked with 3 g/l H2O2 for 10 min at 20-24°C, followed by a PBS wash. After incubation with normal goat serum (cat. no. C0265; Beyotime Institute of Biotechnology) for 15 min at 20-24°C, tissue sections were stained overnight at 4°C with primary antibodies for TRPV1 (1:500; cat. no. ab6166; Abcam) and calpain 2 (1:1,000; cat. no. sc-373967; Santa Cruz Biotechnology, Inc.). After washing with PBS, sections were stained with horseradish peroxidase-conjugated-biotinylated goat anti-rabbit immunoglobulin G (IgG) secondary anti-body (1:1,000; cat. no. ab205718; Abcam) for 1 h at 20-24°C. Following washing with PBS, color was developed with 0.05% 3,3'-diaminobenzidine for 3 min at 20-24°C. Sections were counterstained with hematoxylin for 10 min at 20-24°C, dehydrated with gradient concentrations of ethanol, vitrified with xylene and mounted with neutral gum. PBS was used as a negative control throughout the study. The sections were observed by using a BX41 fluorescent microscope at ×400 magnification (Olympus Corporation). Relative optical intensity was determined using Image-Pro Plus 6.0 software (Media Cybernetics, Inc.).

Western blot analysis

Total cochlea tissue was homogenized in RIPA buffer (cat. no. P0013B; Beyotime Institute of Biotechnology) containing protease inhibitor cocktail tablets and phosphatase inhibitors cocktail (Roche Diagnostics GmbH). The tissue homogenate was sonicated for 30 sec and centrifuged at 14,000 × g at 4°C for 30 min to extract the supernatant. Protein concentrations were determined using a bicinchoninic acid kit. The protein samples (50 µg/lane) were separated using SDS-PAGE (7.5% for TRPV1 and calpain 2; 10% for 4-HNE and procaspase 3 and 12.5% for cleaved caspase 3). Following electrophoresis, the proteins were transferred onto nitrocellulose membranes (Thermo Fisher Scientific, Inc.). The membranes were then blocked with 10% bovine serum albumin for 1 h at 4°C and incubated overnight at 4°C with the following primary antibodies: Anti-TRPV1 (1:500; cat. no. ab6166; Abcam), anti-calpain 2 (1:1,000; cat. no. sc-373967; Santa Cruz Biotechnology, Inc.), anti-4-HNE (1:800; cat. no. ab48506; Abcam), anti-cleaved caspase-3 (1:1,000; cat. no. ab13847; Abcam) and anti-caspase-3 (1:1,000; cat. no. sc-7272; Santa Cruz Biotechnology, Inc.). After washing three times with TBS-0.05% Tween-20, membranes were incubated with IRDye 800CW goat anti-rabbit IgG secondary antibody (1:10,000; cat. no. 925-32211; LI-COR Biosciences.) for 1 h at 4°C. Following extensive washing, the immunoreactive bands were visualized by ECL. The Odyssey CLx Infrared imaging system (LI-COR Biosciences) was used to analyze the optical density value of immunoreactive bands. β-actin (1:1,000; cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) was used as a loading control.

Organotypic cultures of postnatal organ of Corti

The organ culture procedures were modified from a previously published report (35). In brief, Cochlear explants were isolated from newborn 3-day-old BALB/c mice. BALB/c pups were euthanized following antisepsis using 75% ethanol. The two otic capsules were extracted from surrounding tissue and immersed ice-cold Hank's balanced salt solution (HBSS; cat. nos. CC014 and CC016; M&C Gene Biotechnology). The lateral wall tissues (stria vascularis and spiral ligament) and the auditory nerve bundle were micro-dissected for sterile dissection under a binocular microscope (Olympus SZX7; Olympus Corporation). The explants were carefully placed onto a prepared culture dish containing a 15-µl polymerized drop of rat tail collagen in 1 ml culture medium consisting of DMEM/F12 (Invitrogen; Thermo Fisher Scientific, Inc.), 1% bovine serum albumin (Invitrogen; Thermo Fisher Scientific, Inc.), and 10 U/ml penicillin G. Following 4 h of incubation (37°C, 5% CO2), an additional 1 ml medium was added to submerge the explants.

Measurement of Ca2+ levels in OHCs

Cell-permeable Ca2+ indicator Fluo-4 AM (cat. no. F14201; Invitrogen; Thermo Fisher Scientific, Inc.) were used to monitor levels of intracellular Ca2+, based on the literature (36). Following immersion in HBSS without Ca2+ and Mg2+ for 20 min at 20-24°C, the organ explants were immersed in dye (cat. no. F14201; Invitrogen; Thermo Fisher Scientific, Inc.) for 25 min following Fluo4-AM loading at 24°C. Subsequently, the explants were fixed in 4% paraformaldehyde for 30 min at 20-24°C. Phalloidin (1:200) was applied for 1 h at 20-24°C and protected from light. For measurement of Fluo-4 Ca2+ signals, organs were imaged under a BX41 microscope at ×400 magnification (Olympus Corporation) with epifluorescence, and the images were obtained using a TCS-SP5II laser-scanning confocal micro-scope (×400 magnification; Leica Biosystems GmbH) in 20 consecutive fields.

Immunofluorescence for assessment of 4-HNE

Preparation of cochlear paraffin sections (5-µm thickness), deparaffinization, rehydration, antigen retrieval and blocking nonspecific binding were performed as described in a previous study (34). Tissue sections were incubated with rabbit anti-4-HNE antibody (1:200; cat. no. ab46545; Abcam), followed by Cy3-labeled goat anti-rabbit IgG secondary antibody (1:500; cat. no. A0516; Beyotime Institute of Biotechnology) incubation for 1 h at 24°C. Immunofluorescence images were obtained via TCS-SP5II laser-scanning confocal microscopy (×400 magnification; Leica Biosystems GmbH). Relative optical intensity was determined using Image-Pro Plus 6.0 software (Media Cybernetics, Inc.).

Measurement of cell growth inhibition by MTT assay

The human ovarian cancer cell line SKOV3 (Peking Union Medical College, Beijing) were seeded at a density of 3×103 cells/well in 96-well plates and treated with different concentrations (1, 2, 3, 4, 8, 10 and 16 µg/ml) of CDDP. Each concentration treatment was repeated in six separate wells. Cells treated with RPMI 1640 media (100 ul; Sigma-Aldrich; Merck KGaA) alone acted as a control. The cells were incubated at 37°C with 5% CO2 for 24 h, and MTT reagent (20 µl; 5 mg/ml in PBS; Sigma-Aldrich; Merck KGaA) was added to each well and incubated at 37°C with 5% CO2 for 4 h. The absorbance in each well was measured at a wavelength of 570 nm using an enzyme-linked immunoassay microplate reader. According to the tumor cell growth inhibition rate, 4 µg/ml of CDDP was determined as optimal inhibitory concentration. Subsequently, SKOV3 cells were seeded at a density of 3×103 cells/well in a 96-well plate and treated with CDDP (4 µg/ml). Different concentrations (20, 40, 80 and 160 mol/l) of UA were added later. Each concentration of UA treatment was repeated in six separate wells. The cells were incubated at 37°C with 5% CO2 for 24 h, and MTT reagent (20 µl; 5 mg/ml in PBS; Sigma-Aldrich; Merck KGaA) was added to each well and incubated at 37°C with 5% CO2 for 4 h. The absorbance in each well was measured at a wavelength of 570 nm using an enzyme-linked immunoassay microplate reader (BioTek Instruments, Inc.).

Statistical analysis

Data are presented as the mean ± SD and were analyzed using SPSS 17.0 software (SPSS, Inc.). Intergroup differences in mean values were compared by one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

UA prevents CDDP-induced hearing loss and hair cell loss in vivo

To investigate whether UA could prevent CDDP-induced hearing loss, the ABR test was performed on mice. In the control group, ABR thresholds at 8, 12, and 24 kHz were observed to be stable (0.48±1.43, 0.50±1.37 and 0.67±1.44 dB, respectively). However, ABR threshold shifts in the CDDP group were significantly increased at 8, 12 and 24 kHz (25.78±2.06, 27.59±2.86 and 28.51±2.32 dB, respectively), compared with the control group (P<0.01). In the UA + CDDP group, ABR threshold shifts significantly reduced at 8, 12 and 24 kHz (P<0.01; 3.93±1.19, 4.63±1.16 and 5.38±1.36 dB, respectively), compared with the CDDP group. In the UA group, UA treatment alone did not affect the ABR threshold shifts compared with the control group. These results suggested that UA can mitigate CDDP-induced hearing loss in mice (Fig. 1B).

To further determine the in vivo protective effects of UA against CDDP, TRITC staining of OHCs was performed on the basilar membrane of mouse cochlea and the number of surviving OHCs was counted. In the control and UA groups, OHCs on the basilar membrane appeared normal in structure and the staining pattern was homogeneous. Moreover, the loss rate of OHCs at each turn of the basilar membrane was <10%. In the CDDP group, the stereocilia of OHCs were characterized by loss and disorder. OHCs exhibited regional variation in their susceptibility to cisplatin, with gradual severity of OHC loss from the apical to the basal turn, and loss rates of OHCs at apical, middle and basal turns reaching 16, 41 and 61%, respectively, which was significantly higher compared with the control group (P<0.01; Fig. 1C and D). However, compared with the CDDP group, animals receiving UA + CDDP demonstrated significantly fewer OHC loss, especially at basal and middle turns, with the loss rate of OHCs at apical, middle and basal turns reaching 13, 25 and 32%, respectively (P<0.01; Fig. 1C and D). These results suggested that UA effectively protects against CDDP-induced OHC structural damage. Numerous studies showed that CDDP mainly damaged OHCs, while the damage of inner hair cells was weak (8). Hence, the present study focused on the outer hair cell change in and inner hair cell counting was not investigated.

Western blot analysis showed that the CDDP group expressed significantly higher levels of cleaved caspase-3 protein (a marker for apoptosis) compared with the control group (P<0.01; Fig. 1E). By contrast, cleaved caspase-3 expression levels in the UA + CDDP group were significantly lower compared with the CDDP group (P<0.01; Fig. 1E). However, cleaved caspase-3 expression levels in the UA group were not significantly different compared with the control group (Fig. 1E). Again, these results are consistent with the notion that UA can prevent the auditory loss observed in mice treated with CDDP.

UA inhibits CDDP-induced activation of the TRPV1/Ca2+/calpain 2 signaling pathway in the cochleae

Based on immunohistochemistry results (Figs. 2A and 3A), TRPV1 and calpain 2 were uniformly distributed on the OHCs, spiral ganglion (SG), and stria vascularis (SV) of the cochlea in the control group. By contrast, TRPV1 and calpain 2 staining was unevenly distributed in the CDDP group, and the expression levels of these proteins significantly increased compared with the control group (P<0.01; Figs. 2B and 3B). In the UA + CDDP group, TRPV1 and calpain 2 staining was uniformly distributed, and expression significantly decreased compared with the CDDP group (P<0.01, respectively; Figs. 2B and 3B). UA treatment alone did not affect TRPV1 and calpain 2 expression in the mouse cochlea (Figs. 2A and B and 3A and B). These results were consistent with the results of western blot analysis (P<0.01; Figs. 2C and 3C).

In the control group, Ca2+ staining was uniformly distributed in OHCs of the basement membrane (Fig. 4A). In the CDDP group, Ca2+ staining was unevenly distributed (Fig. 4A), and its intensity significantly increased compared with the control group (P<0.01; Fig. 4B). In the UA + CDDP group, Ca2+ staining was more uniformly distributed (Fig. 4A), with significantly lower intensity compared with the CDDP group (P<0.01; Fig. 4B). The results suggested that CDDP could cause Ca2+ overload, and that UA could effectively inhibit Ca2+ influx in mice cochleae.

UA alleviates oxidative stress in mice with CDDP-induced ototoxicity

To investigate whether UA could alleviate oxidative stress, the levels of 4-HNE were analyzed. Based on immunofluorescence results, CDDP significantly enhanced production of 4-HNE in the OHCs, SG, and SV of mouse cochlea compared with the control group (P<0.01; Fig. 5A and B). However, 4-HNE production was significantly inhibited in the UA + CDDP group compared with the CDDP group (P<0.01; Fig. 5A and B). Moreover, compared with the control group, UA treatment alone did not affect 4-HNE production in the mouse cochleae (Fig. 5B). These results were confirmed by western blot analysis of 4-HNE levels (P<0.01; Fig. 5C). The results suggested that UA could prevent an increase in oxidative stress-related markers in the cochlea of mouse treated with CDDP.

The effect of UA on the antitumor effect of CDDP

Since CDDP is used as an anticancer therapeutic, it was investigated whether UA co-treatment would impair its antitumor effects. MTT assay was performed to evaluate the viability of the human ovarian cancer cell line SKVO3 in response to CDDP, UA, and UA + CDDP. While CDDP inhibited the growth of SKVO3 cells in a dose-dependent manner (Fig. 6A), growth inhibition of SKVO3 cells was significantly enhanced following UA + CDDP (4 g/ml) treatment. Moreover, UA inhibited the growth of SKOV3 cells in a dose-dependent manner (P<0.01; Fig. 6B), suggesting that UA enhanced the antitumor effect of CDDP in vitro.

Discussion

UA is a triterpenoid compound found in >60 varieties of plants, including bearberry, Chinese elder herb and hawthorn (37). The therapeutic effects of UA include sedative, anti-inflammatory, anti-ulcer and anti-diabetic effects (38,39). A previous study demonstrated that UA significantly alleviated hydrogen peroxide-induced apoptosis against HEI-OC1 cells in vitro (40). In the present study, UA was found to inhibit CDDP-induced cochlear OHC damage, which in turn significantly attenuated hearing loss at low and high frequencies in mice. Therefore, based on the present findings, it was proposed that UA could provide therapeutic benefits for CDDP-induced ototoxicity.

TRPV1 channels are highly permeable to intracellular calcium influx, and overexpression of TRPV1 induces Ca2+ overload and the activation of calpains, especially calpain 2 (41). Activation of calpain 2 might mediate caspase-3-dependent apoptosis of cochlear cells by down-regulating Bcl-xl and up-regulating Bax (42-45). The TRPV1 channel was previously reported to play a critical role in CDDP-induced ototoxicity (46). Moreover, several studies have shown that TRPV1 expression is significantly increased in OHC, SG and SV in the cochlea in response to CDDP treatment (47-49). The present study found that UA decreased the levels of CDDP-mediated TRPV1 protein expression in the cochlear OHC, SG, and SV. In addition, CDDP dysregulated Ca2+ homeostasis in the cochlea, which is accompanied by increased expression of calpain 2. UA was previously shown to alleviate coughing via TRPV1 channel desensitization (50). Whether a similar mechanism can account for inhibition of CDDP-mediated TRPV1 protein expression by UA remains to be elucidated. TRPV1 is also activated by heat (>42°C), capsaicin, low extracellular pH and oxidative stress products (51). Thus, the antioxidant activity of UA might inhibit activation of the TRPV1 channel by reducing oxidative stress products in the cochlea. The present results suggested that UA significantly blocked CDDP-induced Ca2+ overload in hair cells and also inhibited calpain 2 expression. These results suggested an association between the TRPV1 channel and the Ca2+/calpain signaling pathway, which might explain the decrease in CDDP-induced Ca2+ influx observed in the cochlea following UA treatment.

Oxidative stress is considered to be an important cause of CDDP-induced hearing loss (52). The main source of ROS is NADPH oxidase (NOX) (53). CDDP induced expression of NOX3 in the cochlea, resulting in the formation of ROS, and consequently lipid peroxidation of the cochlea (10). In the present study, the levels of 4-HNE, a highly toxic lipid peroxidation product of aldehydes (54), were observed to increase as a result. High 4-HNE levels can induce the release of cytochrome c from the mitochondria by activating the mitogen-activated protein kinase/JNK cell death signal cascade (55). In the cytoplasm, cytochrome c activates caspase-9 and caspase-3, eventually leading to apoptosis of cochlear cells (56,57). Studies have shown that UA exerts significant antioxidant effects, inhibiting excessive free radical production by upregulation of endothelial nitric oxide synthase expression and downregulation of inducible nitric oxide synthase and NOX expression (58-60). In addition, UA might play an antioxidant role by enhancing free radical scavenging (61). Consistent with these studies, the present study found that UA inhibited the production of the highly toxic lipid peroxidation product 4-HNE in the cochlea, indicating that UA could inhibit CDDP-induced lipid peroxidation.

Antioxidants such as tiopronin, resveratrol, and vitamin E can alleviate inner ear damage caused by CDDP (62-64). However, most of these antioxidants have been found to attenuate the antitumor effects of CDDP, making them a poor therapeutic option for reducing CDDP-induced ototoxicity (65). Previous research has shown that UA can exert antitumor effects by promoting cancer cell apoptosis, inhibiting tumor angiogenesis and reducing cytotoxicity, and that it might have therapeutic effects on several tumors, including gastric, liver and ovarian cancers (66-68). The present study found that UA enhanced CDDP-induced tumor cell apoptosis in vitro. It was hypothesized that UA may complement cisplatin in its antitumor effect. These results suggested the potential of UA and CDDP co-treatment for the treatment of solid tumors.

In conclusion, the present study found a role in the TRPV1/Ca2+/calpain signaling pathway, which may facilitate CDDP-induced hearing loss. UA can reduce the extent of calcium overload and oxidative stress, and subsequently decrease the induction of apoptotic pathways associated with OHC loss (Fig. 7). These results suggested that UA can function as a lead compound for the pharmacological control of drug-induced ototoxicity.

Supplementary Data

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81674036 and 81704127), the Liaoning Natural Science Foundation (grant nos. 2019-ZD-0832 and 20180550402), the Doctoral Scientific Research Foundation of Liaoning Province (grant no. 20170520045) and College Students' Innovation and Entrepreneurship Training Program (grant nos. 201710160000214, 201710160000154 and 201910160112).

Availability of data and materials

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

Authors' contributions

YD was responsible for the study design and implementation, data analysis and manuscript preparation. TX interpreted the data and wrote the manuscript. YT, TM, DQ, YW and DB were responsible for data collection and statistical analysis. YL, LY, SL and AW played an important role in study design and guidance and were responsible for the revision of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal studies complied with the regulations for the Management of Laboratory Animals published by the Ministry of Science and Technology of the People's Republic of China, and all animal protocols were approved by the Animal Care and Use Committee of Jinzhou Medical University

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

We thank Professor Ming-Sheng Zhou from the Department of Physiology, Shenyang Medical College, China for helpful discussions and comments.

References

1 

Basu A, Bhattacharjee A, Samanta A and Bhattacharya S: An oxovanadium(IV) complex protects murine bone marrow cells against cisplatin-induced myelotoxicity and DNA damage. Drug Chem Toxicol. 40:359–367. 2017. View Article : Google Scholar

2 

Vera O, Jimenez J, Pernia O, Rodriguez-Antolin C, Rodriguez C, Sanchez Cabo F, Soto J, Rosas R, Lopez-Magallon S, Esteban Rodriguez I, et al: DNA methylation of miR-7 is a mechanism involved in platinum response through MAFG overexpression in cancer cells. Theranostics. 7:4118–4134. 2017. View Article : Google Scholar :

3 

van As JW, van den Berg H and van Dalen EC: Different infusion durations for preventing platinum-induced hearing loss in children with cancer. Cochrane Database Syst Rev. 7:CD0108852018.PubMed/NCBI

4 

Klumpers MJ, Coenen MJ, Gidding CE and Te Loo DM: The role of germline variants in chemotherapy outcome in brain tumors: A systematic review of pharmacogenetic studies. Pharmacogenomics. 18:501–513. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Pang J, Xiong H, Zhan T, Cheng G, Jia H, Ye Y, Su Z, Chen H, Lin H, Lai L, et al: Sirtuin 1 and autophagy attenuate cisplatin-induced hair cell death in the mouse cochlea and zebrafish lateral line. Front Cell Neurosci. 12:5152019. View Article : Google Scholar : PubMed/NCBI

6 

Ryu NG, Moon IJ, Chang YS, Kim BK, Chung WH, Cho YS and Hong SH: Cochlear implantation for profound hearing loss after multimodal treatment for neuroblastoma in children. Clin Exp Otorhinolaryngol. 8:329–334. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Youm I, West MB, Li W, Du X, Ewert DL and Kopke RD: siRNA-loaded biodegradable nanocarriers for therapeutic MAPK1 silencing against cisplatin-induced ototoxicity. Int J Pharm. 528:611–623. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Guo X, Bai X, Li L, Li J and Wang H: Forskolin protects against cisplatin-induced ototoxicity by inhibiting apoptosis and ROS production. Biomed Pharmacother. 99:530–536. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Martín-Saldaña S, Palao-Suay R, Aguilar MR, Ramírez-Camacho R and San Román J: Polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate to prevent cisplatin-induced ototoxicity. Acta Biomater. 53:199–210. 2017. View Article : Google Scholar

10 

Mukherjea D, Jajoo S, Sheehan K, Kaur T, Sheth S, Bunch J, Perro C, Rybak LP and Ramkumar V: NOX3 NADPH oxidase couples transient receptor potential vanilloid 1 to signal transducer and activator of transcription 1-mediated inflammation and hearing loss. Antioxid Redox Signal. 14:999–1010. 2011. View Article : Google Scholar :

11 

Kim SJ, Park C, Lee JN and Park R: Protective roles of fenofibrate against cisplatin-induced ototoxicity by the rescue of peroxisomal and mitochondrial dysfunction. Toxicol Appl Pharmacol. 353:43–54. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Fetoni AR, Eramo SL, Paciello F, Rolesi R, Podda MV, Troiani D and Paludetti G: Curcuma longa (curcumin) decreases in vivo cisplatin-induced ototoxicity through heme oxygenase-1 induction. Otol Neurotol. 35:e169–e177. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Harris MS, Gilbert JL, Lormore KA, Musunuru SA and Fritsch MH: Cisplatin ototoxicity affecting cochlear implant benefit. Otol Neurotol. 32:969–972. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Ceriani F, Hendry A, Jeng JY, Johnson SL, Stephani F, Olt J, Holley MC, Mammano F, Engel J, Kros CJ, et al: Coordinated calcium signalling in cochlear sensory and non-sensory cells refines afferent innervation of outer hair cells. EMBO J. 38:e998392019. View Article : Google Scholar : PubMed/NCBI

15 

Hegedűs L, Zámbó B, Pászty K, Padányi R, Varga K, Penniston JT and Enyedi Á: Molecular diversity of plasma membrane Ca2+ transporting ATPases: Their function under normal and pathological conditions. Adv Exp Med Biol. 1131:2020.

16 

Wang X, Zhu Y, Long H, Pan S, Xiong H, Fang Q, Hill K, Lai R, Yuan H and Sha SH: Mitochondrial calcium transporters mediate sensitivity to noise-induced losses of hair cells and cochlear synapses. Front Mol Neurosci. 11:4692018. View Article : Google Scholar

17 

Brito R, Sheth S, Mukherjea D, Rybak LP and Ramkumar V: TRPV1: A potential drug target for treating various diseases. Cells. 3:517–545. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Samanta A, Hughes TET and Moiseenkova-Bell VY: Transient receptor potential (TRP) channels. Subcell Biochem. 87:141–165. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Bai P, Liu Y, Xue S, Hamri GC, Saxena P, Ye H, Xie M and Fussenegger M: A fully human transgene switch to regulate therapeutic protein production by cooling sensation. Nat Med. 25:1266–1273. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Övey IS and Naziroğlu M: Homocysteine and cytosolic GSH depletion induce apoptosis and oxidative toxicity through cytosolic calcium overload in the hippocampus of aged mice: Involvement of TRPM2 and TRPV1 channels. Neuroscience. 284:225–233. 2015. View Article : Google Scholar

21 

Kaneko Y and Szallasi A: Transient receptor potential (TRP) channels: A clinical perspective. Br J Pharmacol. 171:2474–2507. 2014. View Article : Google Scholar :

22 

Lima B, Sánchez M, Luna L, Agüero MB, Zacchino S, Filippa E, Palermo JA, Tapia A and Feresin GE: Antimicrobial and anti-oxidant activities of Gentianella multicaulis collected on the Andean Slopes of San Juan Province, Argentina. Z Naturforsch C J Biosci. 67:29–38. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Kaur T, Borse V, Sheth S, Sheehan K, Ghosh S, Tupal S, Jajoo S, Mukherjea D, Rybak LP and Ramkumar V: Adenosine A1 receptor protects against cisplatin ototoxicity by suppressing the NOX3/STAT1 inflammatory pathway in the cochlea. J Neurosci. 36:3962–3977. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Marwaha L, Bansal Y, Singh R, Saroj P, Bhandari R and Kuhad A: TRP channels: Potential drug target for neuropathic pain. Inflammopharmacology. 24:305–317. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Jabeen M, Ahmad S, Shahid K, Sadiq A and Rashid U: Ursolic acid hydrazide based organometallic complexes: Synthesis, characterization, antibacterial, antioxidant, and docking studies. Front Chem. 6:3432018. View Article : Google Scholar

26 

Iqbal J, Abbasi BA, Ahmad R, Mahmood T, Kanwal S, Ali B, Khalil AT, Shah SA, Alam MM and Badshah H: Ursolic acid a promising candidate in the therapeutics of breast cancer: Current status and future implications. Biomed Pharmacother. 108:752–756. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Silva FS, Oliveira PJ and Duarte MF: Oleanolic, ursolic, and betulinic acids as food supplements or pharmaceutical agents for type 2 diabetes: Promise or illusion? J Agric Food Chem. 64:2991–3008. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Wan SZ, Liu C, Huang CK, Luo FY and Zhu X: Ursolic acid improves intestinal damage and bacterial dysbiosis in liver fibrosis mice. Front Pharmacol. 10:13212019. View Article : Google Scholar : PubMed/NCBI

29 

Kim GH, Kan SY, Kang H, Lee S, Ko HM, Kim JH and Lim JH: Ursolic acid suppresses cholesterol biosynthesis and exerts anti-cancer effects in hepatocellular carcinoma cells. Int J Mol Sci. 20:47672019. View Article : Google Scholar :

30 

Yoon JH, Youn K, Ho CT, Karwe MV, Jeong WS and Jun M: p-Coumaric acid and ursolic acid from Corni fructus attenuated β-amyloid (25-35)-induced toxicity through regulation of the NF-κB signaling pathway in PC12 cells. J Agric Food Chem. 62:4911–4916. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Hong SY, Jeong WS and Jun M: Protective effects of the key compounds isolated from corni fructus against β-amyloid- induced neurotoxicity in PC12 cells. Molecules. 17:10831–10845. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Misra RC, Sharma S, Sandeep, Garg A, Chanotiya CS and Ghosh S: Two CYP716A subfamily cytochrome P450 mono-oxygenases of sweet basil play similar but nonredundant roles in ursane- and oleanane-type pentacyclic triterpene biosynthesis. New Phytol. 214:706–720. 2017. View Article : Google Scholar : PubMed/NCBI

33 

DeBacker JR, Harrison RT and Bielefeld EC: Cisplatin-induced threshold shift in the CBA/CaJ, C57BL/6J, BALB/cJ mouse models of hearing loss. Hear Res. 387:1078782020. View Article : Google Scholar : PubMed/NCBI

34 

Liu S, Xu T, Wu X, Lin Y, Bao D, Di Y, Ma T, Dang Y, Jia P, Xian J, et al: Pomegranate peel extract attenuates D-galactose-induced oxidative stress and hearing loss by regu-lating PNUTS/PP1 activity in the mouse cochlea. Neurobiol Aging. 59:30–40. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Matt T, Ng CL, Lang K, Sha SH, Akbergenov R, Shcherbakov D, Meyer M, Duscha S, Xie J, Dubbaka SR, et al: Dissociation of antibacterial activity and aminoglycoside ototoxicity in the 4-monosubstituted 2-deoxystreptamine apramycin. Proc Natl Acad Sci USA. 109:10984–10989. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Spinelli KJ and Gillespie PG: Monitoring intracellular calcium ion dynamics in hair cell populations with Fluo-4 AM. PLoS One. 7:e5.18742012. View Article : Google Scholar

37 

Woźniak Ł, Skąpska S and Marszałek K: Ursolic acid-a penta-cyclic triterpenoid with a wide spectrum of pharmacological activities. Molecules. 20:20614–20641. 2015. View Article : Google Scholar

38 

Jinhua W: Ursolic acid: Pharmacokinetics process in vitro and in vivo, a mini review. Arch Pharm (Weinheim). 352:e18002222019. View Article : Google Scholar

39 

Xu HL, Wang XT, Cheng Y, Zhao JG, Zhou YJ, Yang JJ and Qi MY: Ursolic acid improves diabetic nephropathy via suppression of oxidative stress and inflammation in streptozotocin-induced rats. Biomed Pharmacother. 105:915–921. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Yu HH, Hur JM, Seo SJ, Moon HD, Kim HJ, Park RK and You YO: Protective effect of ursolic acid from Cornus officinalis on the hydrogen peroxide-induced damage of HEI-OC1 auditory cells. Am J Chin Med. 37:735–746. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Bao D, Zhao W, Dai C, Wan H and Cao Y: H89 dihydrochloride hydrate and calphostin C lower the body temperature through TRPV1. Mol Med Rep. 17:1599–1608. 2018.

42 

Huang KF, Ma KH, Chang YJ, Lo LC, Jhap TY, Su YH, Liu PS and Chueh SH: Baicalein inhibits matrix metalloproteinase 1 expression via activation of TRPV1-Ca-ERK pathway in ultra-violet B-irradiated human dermal fibroblasts. Exp Dermatol. 28:568–575. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Berekméri E, Deák O, Téglás T, Sághy É, Horváth T, Aller M, Fekete Á, Köles L and Zelles T: Targeted single-cell electroporation loading of Ca indicators in the mature hemicochlea preparation. Hear Res. 371:75–86. 2019. View Article : Google Scholar

44 

Chang L and Wang A: Calpain mediated cisplatin-induced ototoxicity in mice. Neural Regen Res. 8:1995–2002. 2013.PubMed/NCBI

45 

Yu L, Tang H, Jiang XH, Tsang LL, Chung YW and Chan HC: Involvement of calpain-I and microRNA34 in kanamycin- induced apoptosis of inner ear cells. Cell Biol Int. 34:1219–1225. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Bhatta P, Dhukhwa A, Sheehan K, Al Aameri RFH, Borse V, Ghosh S, Sheth S, Mamillapalli C, Rybak L, Ramkumar V and Mukherjea D: Capsaicin protects against cisplatin ototoxicity by changing the STAT3/STAT1 ratio and activating cannabinoid (CB2) receptors in the cochlea. Sci Rep. 9:41312019. View Article : Google Scholar : PubMed/NCBI

47 

Mukherjea D, Jajoo S, Whitworth C, Bunch JR, Turner JG, Rybak LP and Ramkumar V: Short interfering RNA against transient receptor potential vanilloid 1 attenuates cisplatin-induced hearing loss in the rat. J Neurosci. 28:13056–13065. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Rybak LP, Mukherjea D, Jajoo S, Kaur T and Ramkumar V: siRNA-mediated knock-down of NOX3: Therapy for hearing loss? Cell Mol Life Sci. 69:2429–2434. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Sheth S, Mukherjea D, Rybak LP and Ramkumar V: Mechanisms of cisplatin-induced ototoxicity and otoprotection. Front Cell Neurosci. 11:3382017. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Y, Sreekrishna K, Lin Y, Huang L, Eickhoff D, Degenhardt C and Xu T: Modulation of transient receptor potential (TRP) channels by chinese herbal extracts. Phytother Res. 25:1666–1670. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Wang S, Wang S, Asgar J, Joseph J, Ro JY, Wei F, Campbell JN and Chung MK: Ca2+ and calpain mediate capsaicin-induced ablation of axonal terminals expressing transient receptor poten-tial vanilloid 1. J Biol Chem. 292:8291–8303. 2017. View Article : Google Scholar : PubMed/NCBI

52 

González-García JA, Nevado J, García-Berrocal JR, Sánchez-Rodríguez C, Trinidad A, Sanz R and Ramírez-Camacho R: Endogenous protection against oxidative stress caused by cisplatin: Role of superoxide dismutase. Acta Otolaryngol. 130:453–457. 2010. View Article : Google Scholar

53 

Peng X, Yang Y, Tang L, Wan J, Dai J, Li L, Huang J, Shen Y, Lin L, Gong X and Zhang L: Therapeutic benefits of apocynin in mice with lipopolysaccharide/D-galactosamine-induced acute liver injury via suppression of the late stage pro-apoptotic AMPK/JNK pathway. Biomed Pharmacother. 125:1100202020. View Article : Google Scholar : PubMed/NCBI

54 

Zhang Y, Xu K, Kerwin T, LaManna JC and Puchowicz M: Impact of aging on metabolic changes in the Ketotic rat brain: Glucose, oxidative and 4-HNE metabolism. Adv Exp Med Biol. 1072:21–25. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Wang L, He T, Wan B, Wang X and Zhang L: Orexin A ameliorates HBV X protein-induced cytotoxicity and inflam-matory response in human hepatocytes. Artif Cells Nanomed Biotechnol. 47:2003–2009. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Hsu CL, Hong BH, Yu YS and Yen GC: Antioxidant and anti-inflammatory effects of Orthosiphon aristatus and its bioactive compounds. J Agric Food Chem. 58:2150–2156. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Yang EJ, Moon JY, Lee JS, Koh J, Lee NH and Hyun CG: Acanthopanax koreanum fruit waste inhibits lipopolysaccha-ride-induced production of nitric oxide and prostaglandin E2 in RAW 264.7 macrophages. J Biomed Biotechnol. 2010:7157392010. View Article : Google Scholar

58 

Mu H, Liu H, Zhang J, Huang J, Zhu C, Lu Y, Shi Y and Wang Y: Ursolic acid prevents doxorubicin-induced cardiac toxicity in mice through eNOS activation and inhibition of eNOS uncoupling. J Cell Mol Med. 23:2174–2183. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Zhang C, Wang C, Li W, Wu R, Guo Y, Cheng D, Yang Y, Androulakis IP and Kong AN: Pharmacokinetics and pharmacodynamics of the triterpenoid ursolic acid in regulating the antioxidant, anti-inflammatory, and epigenetic gene responses in rat leukocytes. Mol Pharm. 14:3709–3717. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Yu SS, Chen B, Huang CK, Zhou JJ, Huang X, Wang AJ, Li BM, He WH and Zhu X: Ursolic acid suppresses TGF-β1-induced quiescent HSC activation and transformation by inhibiting NADPH oxidase expression and Hedgehog signaling. Exp Ther Med. 14:3577–3582. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Antônio E: Poly(lactic acid) nanoparticles loaded with ursolic acid: Characterization and in vitro evaluation of radical scavenging activity and cytotoxicity. Mater Sci Eng C Mater Biol Appl. 71:156–166. 2017. View Article : Google Scholar

62 

Fetoni AR, Sergi B, Ferraresi A, Paludetti G and Troiani D: Protective effects of alpha-tocopherol and tiopronin against cisplatin-induced ototoxicity. Acta Otolaryngol. 124:421–426. 2004. View Article : Google Scholar : PubMed/NCBI

63 

Chirtes F and Albu S: Prevention and restoration of hearing loss associated with the use of cisplatin. Biomed Res Int. 2014:9254852014. View Article : Google Scholar : PubMed/NCBI

64 

Luo J, Hu YL and Wang H: Ursolic acid inhibits breast cancer growth by inhibiting proliferation, inducing autophagy and apoptosis, and suppressing inflammatory responses via the PI3K/AKT and NF-κB signaling pathways in vitro. Exp Ther Med. 14:3623–3631. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Chang HY, Chen CJ, Ma WC, Cheng WK, Lin YN, Lee YR, Chen JJ and Lim YP: Modulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation by ursolic acid (UA) attenuates rifampin-isoniazid cytotoxicity. Phytomedicine. 36:37–49. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Kim SH, Jin H, Meng RY, Kim DY, Liu YC, Chai OH, Park BH and Kim SM: Activating hippo pathway via Rassf1 by ursolic acid suppresses the tumorigenesis of gastric cancer. Int J Mol Sci. 20:47092019. View Article : Google Scholar :

67 

Wang WJ, Sui H, Qi C, Li Q, Zhang J, Wu SF, Mei MZ, Lu YY, Wan YT, Chang H and Guo PT: Ursolic acid inhibits proliferation and reverses drug resistance of ovarian cancer stem cells by downregulating ABCG2 through suppressing the expression of hypoxia-inducible factor-1α in vitro. Oncol Rep. 36:428–440. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Zhou M, Yi Y, Liu L, Lin Y, Li J, Ruan J and Zhong Z: Polymeric micelles loading with ursolic acid enhancing anti-tumor effect on hepatocellular carcinoma. J Cancer. 10:5820–5831. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 46 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Di Y, Xu T, Tian Y, Ma T, Qu D, Wang Y, Lin Y, Bao D, Yu L, Liu S, Liu S, et al: Ursolic acid protects against cisplatin‑induced ototoxicity by inhibiting oxidative stress and TRPV1‑mediated Ca2+‑signaling. Int J Mol Med 46: 806-816, 2020
APA
Di, Y., Xu, T., Tian, Y., Ma, T., Qu, D., Wang, Y. ... Wang, A. (2020). Ursolic acid protects against cisplatin‑induced ototoxicity by inhibiting oxidative stress and TRPV1‑mediated Ca2+‑signaling. International Journal of Molecular Medicine, 46, 806-816. https://doi.org/10.3892/ijmm.2020.4633
MLA
Di, Y., Xu, T., Tian, Y., Ma, T., Qu, D., Wang, Y., Lin, Y., Bao, D., Yu, L., Liu, S., Wang, A."Ursolic acid protects against cisplatin‑induced ototoxicity by inhibiting oxidative stress and TRPV1‑mediated Ca2+‑signaling". International Journal of Molecular Medicine 46.2 (2020): 806-816.
Chicago
Di, Y., Xu, T., Tian, Y., Ma, T., Qu, D., Wang, Y., Lin, Y., Bao, D., Yu, L., Liu, S., Wang, A."Ursolic acid protects against cisplatin‑induced ototoxicity by inhibiting oxidative stress and TRPV1‑mediated Ca2+‑signaling". International Journal of Molecular Medicine 46, no. 2 (2020): 806-816. https://doi.org/10.3892/ijmm.2020.4633