Vitamin C attenuates the toxic effect of aristolochic acid on renal tubular cells via decreasing oxidative stress‑mediated cell death pathways

  • Authors:
    • Tsai‑Kun Wu
    • Chyou‑Wei Wei
    • Ying‑Ru Pan
    • Shur‑Hueih Cherng
    • Wei‑Jung Chang
    • Hsueh‑Fang Wang
    • Yung‑Luen Yu
  • View Affiliations

  • Published online on: August 3, 2015     https://doi.org/10.3892/mmr.2015.4167
  • Pages: 6086-6092
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aristolochic acid (AA) is a component of Chinese medicinal herbs, including asarum and aristolochia and has been used in Traditional Chinese Medicine for a long time. Recent studies found that AA has a cytotoxic effect resulting in nephropathy. These studies indicated that AA‑induced cytotoxicity is associated with increases in oxidative stress and caspase‑3 activation. The present study further demonstrated that AA mainly elevates the H2O2 ratio, leading to increases in oxidative stress. Furthermore, the results indicated that AA induces cell death can via caspase‑dependent and ‑independent pathways. It is desirable to identify means of inhibiting AA‑induced renal damage; therefore, the present study applied an anti‑oxidative nutrient, vitamin C, to test whether it can be employed to reduce AA‑induced cell cytotoxicity. The results showed that vitamin C decreased AA‑induced H2O2 levels, caspase‑3 activity and cytotoxicity in renal tubular cells. In conclusion, the present study was the first to demonstrate that AA‑induced increases of the H2O2 ratio resulted in renal tubular cell death via caspase‑dependent and ‑independent pathways, and that vitamin C can decrease AA‑induced increases in H2O2 levels and caspase‑3 activity to attenuate AA‑induced cell cytotoxicity.

Introduction

Aristolochic acid (AA) is contained in the Chinese medicinal herbs asarum and aristolochia (13). Numerous studies have demonstrated that AA can cause nephropathy (46). In traditional Chinese medicine, asarum and aristolochia are usual components of complex Chinese remedies applied to treat arthritic pain, cough and gastrointestinal symptoms (79). Therefore, AA-induced nephropathy is widely discussed in China (10,11). Previous studies showed that AA can induce renal tubular cell death and fibrosis, leading to nephropathy (1214). As shown in numerous studies, AA-induced tubular cell injury was associated with apoptosis (12,1517). However, certain studies reported that AA induced cell death through necrosis (1820).

Apoptosis can be induced via a caspase-dependent or caspase-independent pathway (21,22). AA-induced renal tubular cell death was reported to proceed via the caspase-dependent apoptotic pathway by numerous studies (13,23,24). In general, AA mainly induces caspase-3 activation, resulting in tubular cell apoptosis (2527). The present study further investigated the cytotoxic effects of AA on renal tubular cells; dose- and time-dependency were examined and apoptotic pathways were tested by using a caspase inhibitor.

Although AA-induced renal damage has been reported in numerous studies (46), the mechanisms of AA-induced renal tubular cell death have remained to be clarified. Previous studies reported increases of oxidative stress in AA-treated renal tubular cells and suggested that the increase of reactive oxygen species (ROS) is a possible mechanism for AA-induced renal damage (16,27,28). However, it has remained elusive which ROS is elevated by AA treatment. O2 and H2O2 belong to ROS families which are common in cells (29,30). Thus, O2 and H2O2 levels in renal cells following treatment with AA were examined in the present study.

Based on the observation that the cytotoxicity of AA is mediated via the induction of oxidative stress (16,27,28), N-acetyl cysteine (NAC) and glutathione (GSH), anti-oxidative agents, were studied to prevent cell death through AA treatment (16). Vitamin C is a common nutrient with anti-oxidative properties (3133). In the present study, it was hypothesized that vitamin C is able to inhibit AA-induced cytotoxicity. Thus, AA-treated renal tubular cells were co-treated with vitamin C, and its effects on cell viability, caspase-3 activation and H2O2 levels were tested.

The present study indicated that co-administration of vitamin C may be employed to reduce the nephrotoxic effects of the Chinese medicinal herbs asarum and aristolochia.

Materials and methods

Materials

Luminol, lucigenin, aristolochic acid vitamin C, tubulin antibody and Hoechst 33342 were purchased from Sigma-Aldrich (St. Louis, MO, USA). TGF-β was obtained from R&D Systems (Minneapolis, MN, USA; cat. no. AB-100-NA). An MTT assay kit was purchased from Bio-Basic Canada Inc. (Markham, OT, Canada). Caspase inhibitor Z-Val-Ala-Asp-fluoromethylketone (Z-VAD-FMK) was purchased from BD Pharmingen (Franklin Lakes, NJ, USA). Fetal bovine serum, Dulbecco's modified Eagle's medium (DMEM), non-essential amino acids, l-glutamine and penicillin/streptomycin were obtained from Gibco-BRL (Invitrogen Life Technologies, Carlsbad, CA, USA). Caspase-3 and cleaved caspase-3 antibodies (cat. no. 9662) were purchased from Cell Signaling Technology, Inc. (Beverly, MD, USA).

Cell culture

The rat kidney tubular epithelial cell line NRK-52E was obtained from the Bioresource Collection and Research Center (Shin Chu, Taiwan). NRK-52E cells were cultured in a DMEM medium supplemented with 10% fetal bovine serum, 2 mM l-glutamine, 100 IU/ml penicillin/streptomycin and 0.1 mM non-essential amino acids and maintained at 37°C in a humidified atmosphere containing 5% CO2.

Cell survival assay

The cell survival rate of NRK-52E cells was analyzed using the MTT assay according to the manufacturer's instructions. Briefly, NRK 52E cells were seeded into 96-well plates at a density of 8×103 cells/well, and incubated for 24 h in 100 µl culture medium. The suitable concentration and optimum exposure time of AAI were determined as 5, 10, 20 and 100 µM at 6 h time intervals. At the 6 h time intervals, the control group and experimental groups were examined with the MTT assay kit. Cells were incubated with MTT solution at 37°C for 3 h and the survival rate was then measured at 570 nm absorbance using a Multiskan™ FC Microplate Photometer (Molecular Devices, Sunnyvale, CA, USA). The cell survival rate was calculated using the following formula: A570 experimental group/A570 control group ×100% (34).

Observation of cell morphology and apoptotic features

Cell morphology was observed under a phase-contrast microscope. Apoptotic features were observed by Hoechst 33342 (cat. no. 23491-52-3; Sigma-Aldrich) nuclear staining (3537). In brief, cells were treated with Hoechst 33342 (10 µg/ml) for 10 min. DNA fragmentation and nuclear condensation were observed under an Olympus DP71 fluorescence microscope (excitation, 352 nm; emission, 450 nm; Olympus Corporation, Tokyo, Japan).

Caspase inhibition assay

Z-VAD-FMK is a general caspase inhibitor. In the present study, NRK-52E cells were pre-treated with 20 µM Z-VAD-FMK prior to AA treatment (0, 5, 10, 20 or 100 µM). The survival rates of NRK-52E cells were determined using an MTT assay as described above (38).

Determination of H2O2 and O2− ratios

H2O2 and O2 ratios were determined by using the lucigenin-amplified chemiluminescence method (39,40). In brief, the culture supernatant (200 µl) was treated with 0.2 mmol/l of luminol solution (100 µl) and subsequently evaluated using a chemiluminescence analyzing system (CLA-FSI; Tohoko Electronic Industrial Co., Ltd., Sendai, Japan) for the determination of H2O2 levels. Samples (200 µl) were treated with 0.1 mmol/l lucigenin solution (500 µl) and were then measured using the CLA-FSI chemiluminescence analyzing system for the determination of O2 levels. The ratios of H2O2 and O2 were calculated as (experimental group counts/control group counts) ×100%.

Western blot analysis

Western blot analysis was performed as in a previous study (34). Briefly, cells were lysed with radioimmunoprecipitation assay buffer (cat. no. 20-188; EMD Millipore, Billerica, MA, USA). After centrifugation at 16,000 × g for 10 min at 4°C, proteins were obtained and their concentration was determined using the Bradford assay (cat. no. 23200; Thermo Fischer Scientific, Inc., Waltham, MA, USA). Equal amounts of samples were separated by 13.3% SDS-PAGE (Bio-Rad Rad Laboratories, Inc., Hercules, CA, USA), and then transferred onto polyvinylidene difluoride membranes (EMD Millipore). The membranes were blocked with 5% milk for 2 h at room temperature. After washing with phosphate-buffered saline (PBS), samples were incubated with the primary antibodies for 4 h overnight. Next, samples were washed with PBS and treated with anti-rabbit-HRP secondary antibodies (cat. no. NA934; Amersham Biosciences Inc., Beverly, MD, USA) for 1 h at room temperature. Finally, samples were visualized by using Western Lightning Chemiluminescence Reagent Plus (Perkin Elmer, Waltham, MA, USA) and quantified using a densitometer (41,42).

Statistical analysis

Student's t-test was utilized for the analysis of data. Values are expressed as the mean ± standard error. P<0.05 was considered to indicate a statistically significant difference between values.

Results

AA induces cytotoxicity in a dose- and time-dependent manner

Previous studies have demonstrated that AA exerts cytotoxic effects in renal tubular cells (16,43,44). Similarly, the results of the present study also indicated that AA can exert cytotoxic effects on renal tubular cells (Fig. 1). The cell survival rate was <50% by the following treatments: 100 µM AA (6 h), 20 µM AA (6 h), 10 µM AA (24 h) and 5 µM AA (48 h). The results indicated that high-dose AA treatment exerted a higher cytotoxic effect on renal tubular cells than low-dose AA treatment. In addition, long durations of incubation had a greater cytotoxic effect as compared with short incubations with AA. These results suggested that AA induced cytotoxicity in a dose- and time-dependent manner.

AA-induced cell death is associated with the apoptotic pathway

Cell death can be associated with apoptotic or necrotic pathways (45,46). In the present study, cell morphology was observed under a phase-contrast and a fluorescent microscope (Fig. 2). Compared with control cells (Fig. 2A), shrunken types were observed among AA-treated cells (Fig. 2B) under a phase-contrast microscope. In addition, apoptotic characteristics, including nuclear condensation and DNA fragmentation, were determined by Hoechst nuclear staining (35,36). Compared with the control cells (Fig. 2C), nuclear condensation and DNA fragmentation were observed in AA-treated cells under a fluorescent microscope (Fig. 2D). These observations indicated that AA-induced cell death was associated with the apoptotic pathway.

AA-induced apoptosis involves caspase-dependent and -independent signaling

AA can activate caspase, resulting in cell cytotoxicity (2527). However, whether AA-induced apoptosis is dependent on caspases has remained elusive. Therefore, in the present study, caspase inhibitor was applied to cells prior to AA treatment in order to investigate the association between caspase activity and AA-induced cytotoxicity. The results showed that blocking of caspase activity significantly inhibited the cytotoxic effects of low doses of AA (5–20 µM) AA treatment (Fig. 3); however, blocking of caspase activity did not inhibit the cytotoxicity of AA at a high dose (100 µM). These results indicated that caspase activity is an important factor in low-dose AA-induced cytotoxicity. In addition, caspase-independent apoptosis signaling, which remains to be further investigated, is involved in the cytotoxic effects of high doses of AA.

AA increases H2O2 but not O2− levels in kidney cells

H2O2 and O2, distinct species of the family of ROS, are generally present in cells. Previous studies have demonstrated that AA can induce increases in ROS in renal tubular cells (16,27,28). However, it has remained elusive which ROS are elevated by AA treatment. Therefore, the present study examined H2O2 and O2 levels in kidney cells treated with AA by using the lucigenin-amplified method (39,40). As shown in Fig. 4A, H2O2 levels were elevated in renal tubular cells after AA treatment. The results also indicated that AA induced increases in H2O2 levels in a dose-dependent manner. By contrast, the O2 ratio was not significantly altered in renal cells following AA treatment (Fig. 4B). Therefore, the results of the present study indicated that H2O2, but not O2, was among the ROS increased in kidney cells by AA treatment.

Vitamin C reduces AA-induced increases in H2O2 levels and inhibits AA-induced cytotoxicity

As AA treatment increased H2O2 levels (Fig. 4A), it was further investigated whether H2O2 levels were associated with AA-induced cytotoxicity in renal tubular cells. Vitamin C, an anti-oxidative nutrient, was applied in AA-treated renal tubular cells. The results showed that vitamin C was able to attenuate the increases in H2O2 levels in AA-treated renal cells (Fig. 5). Furthermore, the cell survival rate of renal cells was assessed following combined treatment with AA and vitamin C. The results showed that vitamin C can inhibit AA-induced cytotoxicity in renal tubular cells (Fig. 6). These results suggested that H2O2 was an important factor in AA-induced toxicity to renal tubular cells. In addition, vitamin C effectively reduced H2O2 levels alongside cytotoxicity in AA-treated cells.

Vitamin C decreases AA-induced caspase-3 activation to attenuate AA-induced cytotoxicity

The results of the present study demonstrated that vitamin C decreased AA-induced increases in H2O2 levels to inhibit AA-induced cytotoxicity (Figs. 5 and 6). In addition, previous studies (2527) and the results of the present study (Fig. 3) indicated that caspase-3 activity was an important factor associated with AA-induced cytotoxicity. Therefore, the present study further investigated whether vitamin C can decrease caspase-3 activity to attenuate AA-induced cytotoxicity. For this, caspase-3 (inactivated caspase-3) and cleaved caspase-3 (activated caspase-3) levels were determined by western blot analysis (Fig. 7A) and the caspase-3 activity ratio (cleaved caspase-3/caspase-3) was obtained by densitometric analysis (Fig. 7B). Compared with the control group, the caspase-3 activity ratio was obviously increased in the AA-treated group (43.9 vs. 89.8%). Of note, compared with that in the AA-treated group, the caspase-3 activity ratio was decreased to 68.6% in the AA plus vitamin C-treated group. These results indicated that vitamin C decreased AA-induced caspase-3 activation. Regarding the association between AA-induced cytotoxicity and vitamin C (Figs. 5Figure 67), the results suggested that vitamin C can attenuate AA-induced cytotoxicity, at least in part via decreasing AA-induced H2O2 levels and caspase-3 activity.

Discussion

Numerous studies have demonstrated that AA can induce renal tubular cell death (46) and kidney fibrosis (14,47), resulting in nephropathy. Regarding AA-induced cell death, the apoptotic pathway was widely identified in AA-treated cells (12,1517); however, necrosis was also reported by certain studies on AA-treated cells (1820). DNA fragmentation and nuclear condensation were observed by fluorescent microscopic observation. Therefore, the results of the present study indicated that AA-induced cytotoxicity was mediated via the apoptotic pathway. Previous studies have shown that AA can activate caspase-3 activity leading to apoptosis (2527). However, whether AA-induced cytotoxicity depends on caspase-3 activation had remained elusive. The results of the present study showed that AA induced caspase-3 activation in accordance with the results of previous studies (2527). Furthermore, the present study found that the cytotoxicity of low doses of AA was attenuated by a caspase inhibitor, whereas that of high doses of AA was not inhibited. These data suggested that the cytotoxicity of AA at low doses is mediated via caspase activation signaling, while the cytotoxicity of AA at high doses involves caspase-dependent as well as -independent signaling, which remains to be further elucidated in the future.

Previous studies have demonstrated that AA induces increases of ROS levels, resulting in renal injury (16,27,28). However, these studies did not determine which type of ROS is generated by AA treatment. O2 and H2O2 are ROS which are common in cells, and O2 can be converted into H2O2 by superoxide dismutase (48,49). Furthermore, H2O2 can be converted into H2O by GSH and GSH peroxidase (50,51). In the present study, O2 and H2O2 levels were determined using the lucigenin-amplified method (39,40). The results showed that AA treatment induced increases of H2O2 levels but not of O2 levels. From these results it can therefore be deduced that AA may be able to decrease GSH levels or GSH peroxidase activities. This would also explain why NAC, which is required for GSH synthesis, or GSH pre-treatment can inhibit AA-induced cell death (16,27). A major function of SOD is converting O2 to H2O2. The present study demonstrated that AA is not able to influence O2 levels. These results suggested that AA may not be associated with SOD activity.

Because AA is a component of various Traditional Chinese Medicinal plants (7,5256), it is important to discover means of preventing AA-induced renal damage. Though studies have developed a monoclonal antibody against AA-induced cytotoxicity (3,57,58), most approaches are directed against oxidative stress to suppress AA-induced cytotoxicity. Based on the above findings, elucidating whether vitamin C inhibits AA-induced cytotoxicity in animal models will require further study. To date, anti-oxidant agents including Tiron, NAC, GSH, bone morphogenetic protein 7 and darbepoetin have been studied for their ability to reduce AA-induced cytotoxicity (13,27,5961). However, these substances are not easily available. Vitamin C has anti-oxidative activities and is contained in numerous vegetables and fruits (3133). The present study therefore assessed whether vitamin C can suppress AA-induced cytotoxicity to kidney cells. The results demonstrated that vitamin C markedly attenuated increases of H2O2 levels, caspase-3 activation and cytotoxicity following AA treatment. These results therefore suggested that supplementation of vitamin C may be beneficial for reducing AA-induced renal damage when using Traditional Chinese Medicines.

In conclusion, the present study demonstrated that: 1) AA activates caspase-3 activity and induces increases of H2O2 levels, resulting in renal tubular cell death; 2) AA-induced cell death is mediated via caspase-dependent and -independent pathways, depending on the dose; and 3) Vitamin C can decrease AA-induced increases in H2O2 levels and caspase-3 activity to attenuate AA-induced cytotoxicity.

Acknowledgments

The present study was supported by Ministry of Science and Technology (grant no. MOST-103-2320-B-039) and the National Health Research Institutes (grant no. NHRI-EX102-10245BI).

References

1 

Schaneberg BT and Khan IA: Analysis of products suspected of containing Aristolochia or Asarum species. J Ethnopharmacol. 94:245–249. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Schaneberg BT, Applequist WL and Khan IA: Determination of aristolochic acid I and II in North American species of Asarum and Aristolochia. Pharmazie. 57:686–689. 2002.PubMed/NCBI

3 

Li XW, Morinaga O, Tian M, Uto T, Yu J, Shang MY, Wang X, Cai SQ and Shoyama Y: Development of an Eastern blotting technique for the visual detection of aristolochic acids in Aristolochia and Asarum species by using a monoclonal antibody against aristolochic acids I and II. Phytochem Anal. 24:645–653. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Feng C, Xie X, Wu M, Li C, Gao M, Liu M, Qi X and Ren J: Tanshinone I protects mice from aristolochic acid I-induced kidney injury by induction of CYP1A. Environ Toxicol Pharmacol. 36:850–857. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Grollman AP: Aristolochic acid nephropathy: Harbinger of a global iatrogenic disease. Environ Mol Mutagen. 54:1–7. 2013. View Article : Google Scholar

6 

Yang L, Su T, Li XM, Wang X, Cai SQ, Meng LQ, Zou WZ and Wang HY: Aristolochic acid nephropathy: Variation in presentation and prognosis. Nephrol Dial Transplant. 27:292–298. 2012. View Article : Google Scholar

7 

Tsai DM, Kang JJ, Lee SS, Wang SY, Tsai IL, Chen GY, Liao HW, Wei-Chu L, Kuo CH and Tseng YJ: Metabolomic analysis of complex chinese remedies: Examples of induced nephrotoxicity in the mouse from a series of remedies containing aristolochic Acid. Evid Based Complement Alternat Med. 2013:2637572013. View Article : Google Scholar : PubMed/NCBI

8 

Arlt VM, Stiborova M and Schmeiser HH: Aristolochic acid as a probable human cancer hazard in herbal remedies: A review. Mutagenesis. 17:265–277. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Heinrich M, Chan J, Wanke S, Neinhuis C and Simmonds MS: Local uses of Aristolochia species and content of nephrotoxic aristolochic acid 1 and 2 - a global assessment based on bibliographic sources. J Ethnopharmacol. 125:108–144. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Zhang J, Zhang L, Wang W and Wang H; China National Survey of Chronic Kidney Disease Working Group: Association between aristolochic acid and CKD: A cross-sectional survey in China. Am J Kidney Dis. 61:918–922. 2013. View Article : Google Scholar : PubMed/NCBI

11 

De Broe ME: Chinese herbs nephropathy and Balkan endemic nephropathy: Toward a single entity, aristolochic acid nephropathy. Kidney Int. 81:513–515. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Pozdzik AA, Salmon IJ, Debelle FD, Decaestecker C, Van den Branden C, Verbeelen D, Deschodt-Lanckman MM, Vanherweghem JL and Nortier JL: Aristolochic acid induces proximal tubule apoptosis and epithelial to mesenchymal transformation. Kidney Int. 73:595–607. 2008. View Article : Google Scholar

13 

Wang Z, Zhao J, Zhang J, Wei J, Zhang J and Huang Y: Protective effect of BMP-7 against aristolochic acid-induced renal tubular epithelial cell injury. Toxicol Lett. 198:348–357. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Lin TC, Lee TC, Hsu SL and Yang CS: The molecular mechanism of leptin secretion and expression induced by aristolochic acid in kidney fibroblast. PloS One. 6:e166542011. View Article : Google Scholar : PubMed/NCBI

15 

Yang H, Dou Y, Zheng X, Tan Y, Cheng J, Li L, Du Y, Zhu D and Lou Y: Cysteinyl leukotrienes synthesis is involved in aristolochic acid I-induced apoptosis in renal proximal tubular epithelial cells. Toxicology. 287:38–45. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Zhu S, Wang Y, Jin J, Guan C, Li M, Xi C, Ouyang Z, Chen M, Qiu Y, Huang M, et al: Endoplasmic reticulum stress mediates aristolochic acid I-induced apoptosis in human renal proximal tubular epithelial cells. Toxicol In Vitro. 26:663–671. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Wang W and Zhang J: Protective effect of erythropoietin against aristolochic acid-induced apoptosis in renal tubular epithelial cells. Eur J Pharmacol. 588:135–140. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Baudoux TE, Pozdzik AA, Arlt VM, De Prez EG, Antoine MH, Quellard N, Goujon JM and Nortier JL: Probenecid prevents acute tubular necrosis in a mouse model of aristolochic acid nephropathy. Kidney Int. 82:1105–1113. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Cao J, Yang XD, Wang XY, Qu L, Liu G and Li XM: Differential changes of intrarenal oxygenation in rat models of acute tubular necrosis caused by aristolochic acid and gentamicin. Chinese Medical Journal. 90:1208–1212. 2010.In Chinese.

20 

Yang L, Li X and Wang H: Possible mechanisms explaining the tendency towards interstitial fibrosis in aristolochic acid-induced acute tubular necrosis. Nephrol Dial Transplant. 22:445–456. 2007. View Article : Google Scholar

21 

Zhou Y, Bi Y, Yang C, Yang J, Jiang Y, Meng F, Yu B, Khan M, Ma T and Yang H: Magnolol induces apoptosis in MCF-7 human breast cancer cells through G2/M phase arrest and caspase-independent pathway. Pharmazie. 68:755–762. 2013.PubMed/NCBI

22 

An FF, Liu YC, Zhang WW and Liang L: Dihydroartemisinine enhances dictamnine-induced apoptosis via a caspase dependent pathway in human lung adenocarcinoma A549 cells. Asian Pac J Cancer Prev. 14:5895–5900. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Wang Y, Fu W, Wang H, Liang Y, Wang Y, Yao W, Chen W, Li Q, Ying PH, Shi X, et al: Renal microvascular injury in chronic aristolochic acid nephropathy and protective effects of Cozaar. Ren Fail. 34:60–67. 2012. View Article : Google Scholar

24 

Zhang L, Li J, Jiang Z, Sun L, Mei X, Yong B and Zhang L: Inhibition of aquaporin-1 expression by RNAi protects against aristolochic acid I-induced apoptosis in human proximal tubular epithelial (HK-2) cells. Biochem Biophys Res Commun. 405:68–73. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Li J and Zhang L, Jiang Z, Shu B, Li F, Bao Q and Zhang L: Toxicities of aristolochic acid I and aristololactam I in cultured renal epithelial cells. Toxicol In Vitro. 24:1092–1097. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Qi X, Cai Y, Gong L, Liu L, Chen F, Xiao Y, Wu X, Li Y, Xue X and Ren J: Role of mitochondrial permeability transition in human renal tubular epithelial cell death induced by aristolochic acid. Toxicol Appl Pharmacol. 222:105–110. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Yu FY, Wu TS, Chen TW and Liu BH: Aristolochic acid I induced oxidative DNA damage associated with glutathione depletion and ERK1/2 activation in human cells. Toxicol In Vitro. 25:810–816. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Chen YY, Chung JG, Wu HC, Bau DT, Wu KY, Kao ST, Hsiang CY, Ho TY and Chiang SY: Aristolochic acid suppresses DNA repair and triggers oxidative DNA damage in human kidney proximal tubular cells. Oncol Rep. 24:141–153. 2010.PubMed/NCBI

29 

Yao CW, Piao MJ, Kim KC, Zheng J, Cha JW and Hyun JW: 6′-o-galloylpaeoniflorin protects human keratinocytes against oxidative stress-induced cell damage. Biomol Ther (Seoul). 21:349–357. 2013. View Article : Google Scholar

30 

Sen S, Kawahara B, Fry NL, Farias-Eisner R, Zhang D, Mascharak PK and Chaudhuri G: A light-activated NO donor attenuates anchorage independent growth of cancer cells: Important role of a cross talk between NO and other reactive oxygen species. Arch Biochem Biophys. 540:33–40. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Kataoka T, Nishiyama Y, Yamato K, Teraoka J, Morii Y, Sakoda A, Ishimori Y, Taguchi T and Yamaoka K: Comparative study on the inhibitory effects of antioxidant vitamins and radon on carbon tetrachloride-induced hepatopathy. J Radiat Res (Tokyo). 53:830–839. 2012. View Article : Google Scholar :

32 

Al-Rejaie SS, Abuohashish HM, Alkhamees OA, Aleisa AM and Alroujayee AS: Gender difference following high cholesterol diet induced renal injury and the protective role of rutin and ascorbic acid combination in Wistar albino rats. Lipids Health Dis. 11:412012. View Article : Google Scholar : PubMed/NCBI

33 

Taniguchi M, A rai N, Kohno K, Ushio S and Fukuda S: Anti-oxidative and anti-aging activities of 2-O-α-glucopyranosyl-L-ascorbic acid on human dermal fibroblasts. Eur J Pharmacol. 674:126–131. 2012. View Article : Google Scholar

34 

Yu YL, Su KJ, Chen CJ, Wei CW, Lin CJ, Yiang GT, Lin SZ, Harn HJ and Chen YL: Synergistic anti-tumor activity of isochaihulactone and paclitaxel on human lung cancer cells. J Cell Physiol. 227:213–222. 2012. View Article : Google Scholar

35 

Yiang GT, Chen YH, Chou PL, Chang WJ, Wei CW and Yu YL: The NS3 protease and helicase domains of Japanese encephalitis virus trigger cell death via caspase-dependent and -independent pathways. Mol Med Rep. 7:826–830. 2013.PubMed/NCBI

36 

Yiang GT, Yu YL, Chou PL, Tsai HF, Chen LA, Chen YH, Su KJ, Wang JJ, Bau DT and Wei CW: The cytotoxic protein can induce autophagocytosis in addition to apoptosis in MCF-7 human breast cancer cells. In Vivo. 26:403–409. 2012.PubMed/NCBI

37 

Yiang GT, Chou PL, Tsai HF, Chen LA, Chang WJ, Yu YL and Wei CW: Immunotherapy for SV40 T/t antigen-induced breast cancer by recombinant adeno-associated virus serotype 2 carrying interleukin-15 in mice. Int J Mol Med. 29:809–814. 2012.PubMed/NCBI

38 

Wu CS, Yen CJ, Chou RH, Li ST, Huang WC, Ren CT, Wu CY and Yu YL: Cancer-associated carbohydrate antigens as potential biomarkers for hepatocellular carcinoma. PloS One. 7:e394662012. View Article : Google Scholar : PubMed/NCBI

39 

Chen KH, Li PC, Lin WH, Chien CT and Low BH: Depression by a green tea extract of alcohol-induced oxidative stress and lipogenesis in rat liver. Biosci Biotechnol Biochem. 75:1668–1676. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Lin BR, Yu CJ, Chen WC, Lee HS, Chang HM, Lee YC, Chien CT and Chen CF: Green tea extract supplement reduces D-galactosamine-induced acute liver injury by inhibition of apoptotic and proinflammatory signaling. J Biomed Sci. 16:352009. View Article : Google Scholar : PubMed/NCBI

41 

Yu YL, Wei CW, Chen YL, Chen MH and Yiang GT: Immunotherapy of breast cancer by single delivery with rAAV2-mediated interleukin-15 expression. Int J Oncol. 36:365–370. 2010.PubMed/NCBI

42 

Yiang GT, Harn HJ, Yu YL, Hu SC, Hung YT, Hsieh CJ, Lin SZ and Wei CW: Immunotherapy: rAAV2 expressing interleukin-15 inhibits HeLa cell tumor growth in mice. J Biomed Sci. 16:472009. View Article : Google Scholar : PubMed/NCBI

43 

Yang CC, Wu CT, Chen LP, Hung KY, Liu SH and Chiang CK: Autophagy induction promotes aristolochic acid-I-induced renal injury in vivo and in vitro. Toxicology. 312:63–73. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Zeng Y, Yang X, Wang J, Fan J, Kong Q and Yu X: Aristolochic acid I induced autophagy extenuates cell apoptosis via ERK 1/2 pathway in renal tubular epithelial cells. PloS one. 7:e303122012. View Article : Google Scholar : PubMed/NCBI

45 

Hanus J, Zhang H, Wang Z, Liu Q, Zhou Q and Wang S: Induction of necrotic cell death by oxidative stress in retinal pigment epithelial cells. Cell Death Dis. 4:e9652013. View Article : Google Scholar : PubMed/NCBI

46 

Suzuki-Karasaki M, Ochiai T and Suzuki-Karasaki Y: Crosstalk between mitochondrial ROS and depolarization in the potentiation of TRAIL-induced apoptosis in human tumor cells. Int J Oncol. 44:616–628. 2014.

47 

Fragiadaki M, Witherden AS, Kaneko T, Sonnylal S, Pusey CD, Bou-Gharios G and Mason RM: Interstitial fibrosis is associated with increased COL1A2 transcription in AA-injured renal tubular epithelial cells in vivo. Matrix Biol. 30:396–403. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Shearer J: Dioxygen and superoxide stability of metallopeptide based mimics of nickel containing superoxide dismutase: The influence of amine/amidate vs. bis-amidate ligation. J Inorg Biochem. 129:145–149. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Foresman EL and Miller FJ Jr: Extracellular but not cytosolic superoxide dismutase protects against oxidant-mediated endothelial dysfunction. Redox Biol. 1:292–296. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Hatem E, Berthonaud V, Dardalhon M, Lagniel G, Baudouin-Cornu P, Huang ME, Labarre J and Chédin S: Glutathione is essential to preserve nuclear function and cell survival under oxidative stress. Free Radic Biol Med. 67:103–114. 2014. View Article : Google Scholar

51 

Sullivan-Gunn MJ and Lewandowski PA: Elevated hydrogen peroxide and decreased catalase and glutathione peroxidase protection are associated with aging sarcopenia. BMC Geriatr. 13:1042013. View Article : Google Scholar : PubMed/NCBI

52 

Lin HH, Chou SA, Yang HY, Hwang YH, Kuo CH, Kao TW, Lo TC and Chen PC: Association of blood lead and mercury with estimated GFR in herbalists after the ban of herbs containing aristolochic acids in Taiwan. Occup Environ Med. 70:545–551. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Ma B, Li N and Lin G: Importance of metabolic activation study to the safe use of Chinese herbal medicines. Curr Drug Metab. 13:652–658. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Lai JN, Tang JL and Wang JD: Observational studies on evaluating the safety and adverse effects of traditional Chinese medicine. Evid Based Complement Alternat Med. 2013:6978932013. View Article : Google Scholar : PubMed/NCBI

55 

Yang HY, Wang JD, Lo TC and Chen PC: Occupational kidney disease among Chinese herbalists exposed to herbs containing aristolochic acids. Occup Environ Med. 68:286–290. 2011. View Article : Google Scholar

56 

Eaton L: Traditional Chinese practitioner breaches Medicine Act. BMJ. 340(feb19 1): c10282010. View Article : Google Scholar : PubMed/NCBI

57 

Shang MY, Tian M, Tanaka H, Li XW, Cai SQ and Shoyama Y: Quality control of traditional chinese medicine by monoclonal antibody method. Curr Drug Discov Technol. 8:60–65. 2011. View Article : Google Scholar

58 

Tian M, Tanaka H, Shang MY, Karashima S, Chao Z, Wang X, Cai SQ and Shoyama Y: Production, characterization of a monoclonal antibody against aristolochic acid-II and development of its assay system. Am J Chin Med. 36:425–436. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Hamano Y, Aoki T, Shirai R, Hatano M, Kimura R, Ogawa M, Yokosuka O and Ueda S: Low-dose darbepoetin alpha attenuates progression of a mouse model of aristolochic acid nephropathy through early tubular protection. Nephron Exp Nephrol. 114:e69–e81. 2010. View Article : Google Scholar

60 

Hsing CH, Chou W, Wang JJ, Chen HW and Yeh CH: Propofol increases bone morphogenetic protein-7 and decreases oxidative stress in sepsis-induced acute kidney injury. Nephrol Dial Transplant. 26:1162–1172. 2011. View Article : Google Scholar

61 

Parissis JT, Kourea K, Andreadou I, Ikonomidis I, Markantonis S, Ioannidis K, Paraskevaidis I, Iliodromitis E, Filippatos G and Kremastinos DT: Effects of Darbepoetin Alfa on plasma mediators of oxidative and nitrosative stress in anemic patients with chronic heart failure secondary to ischemic or idiopathic dilated cardiomyopathy. Am J Cardiol. 103:1134–1138. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 12 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu TK, Wei CW, Pan YR, Cherng SH, Chang WJ, Wang HF and Yu YL: Vitamin C attenuates the toxic effect of aristolochic acid on renal tubular cells via decreasing oxidative stress‑mediated cell death pathways. Mol Med Rep 12: 6086-6092, 2015
APA
Wu, T., Wei, C., Pan, Y., Cherng, S., Chang, W., Wang, H., & Yu, Y. (2015). Vitamin C attenuates the toxic effect of aristolochic acid on renal tubular cells via decreasing oxidative stress‑mediated cell death pathways. Molecular Medicine Reports, 12, 6086-6092. https://doi.org/10.3892/mmr.2015.4167
MLA
Wu, T., Wei, C., Pan, Y., Cherng, S., Chang, W., Wang, H., Yu, Y."Vitamin C attenuates the toxic effect of aristolochic acid on renal tubular cells via decreasing oxidative stress‑mediated cell death pathways". Molecular Medicine Reports 12.4 (2015): 6086-6092.
Chicago
Wu, T., Wei, C., Pan, Y., Cherng, S., Chang, W., Wang, H., Yu, Y."Vitamin C attenuates the toxic effect of aristolochic acid on renal tubular cells via decreasing oxidative stress‑mediated cell death pathways". Molecular Medicine Reports 12, no. 4 (2015): 6086-6092. https://doi.org/10.3892/mmr.2015.4167