Open Access

Vitamin E (α‑tocopherol) ameliorates aristolochic acid‑induced renal tubular epithelial cell death by attenuating oxidative stress and caspase‑3 activation

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

  • Published online on: October 27, 2017     https://doi.org/10.3892/mmr.2017.7921
  • Pages: 31-36
  • Copyright: © Wu 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

Aristolochic acid (AA) is a component identified in traditional Chinese remedies for the treatment of arthritic pain, coughs and gastrointestinal symptoms. However, previous studies have indicated that AA can induce oxidative stress in renal cells leading to nephropathy. α‑tocopherol exists in numerous types of food, such as nuts, and belongs to the vitamin E isoform family. It possesses antioxidant activities and has been used previously for clinical applications. Therefore, the aim of the present study was to determine whether α‑tocopherol could reduce AA‑induced oxidative stress and renal cell cytotoxicity, determined by cell survival rate, reactive oxygen species detection and apoptotic features. The results indicated that AA markedly induced H2O2 levels and caspase‑3 activity in renal tubular epithelial cells. Notably, the presence of α‑tocopherol inhibited AA‑induced H2O2 and caspase‑3 activity. The present study demonstrated that antioxidant mechanisms of α‑tocopherol may be involved in the increased survival rates from AA‑induced cell injury.

Introduction

Aristolochic acid (AA) is a component present in Chinese herbs (for example Asarum and Aristolochia) from remedies for the treatment of arthritis pain, coughs and gastrointestinal symptoms (14). Previous studies have indicated that AA can lead to renal injury (5,6) and this finding has led to further studies (7,8). Previous studies have indicated that renal damage from renal cell death and renal fibrosis is associated with AA treatment (9,10).

AA-induced oxidative stress may serve an important role in the development of renal injury (1113). Previous studies have demonstrated that oxidative stress causes lipid peroxidation, DNA damage and protein peroxidation, and results in cell damage (1416). O2 and H2O2 are key reactive oxygen species (ROS) identified in cells (17,18). Normally, O2 and H2O2 are produced in the mitochondria via electron transport chain (19,20) and these ROS are removed by cellular superoxide dismutase (SOD), glutathione peroxidase (Gpx) and catalase (CAT) (2123). However, various toxins also induce O2 and H2O2 production (2426). The excessive O2 and H2O2 lead to cell injury (27,28) and it has additionally been reported that AA-induced H2O2 leads to renal damage (29).

Various studies have demonstrated that oxidative stress can induce cell apoptosis or cell necrosis (3032), and consequently AA-induced oxidative stress can cause apoptosis or necrosis of renal cells (29,3335). Concerning apoptosis, caspase-dependent and caspase-independent pathways have been reported previously (36,37). Although certain mechanisms of AA-induced cell death remain unclear, the caspase activation may be associated with AA-induced apoptosis (38,39). Previous studies indicated that AA can activate caspase-9 and caspase-3 leading to cell apoptosis (4042).

The isoforms of vitamin E consist of α-tocopherol, β-tocopherol, δ-tocotrienol and γ-tocotrienol (43). Among them, α-tocopherol possesses anti-oxidative activities and has been used in a clinical setting (44,45). In addition, previous studies have suggested that α-tocopherol can inhibit renal fibrosis (46,47). Due to the fact that AA-induced renal injury was associated with oxidative damage and fibrotic renal injury (9,1113), the effects of α-tocopherol on AA-induced renal cell cytotoxicity were studied. The results of the present study demonstrated that α-tocopherol can inhibit the H2O2 level and caspase-3 activities to attenuate renal tubular epithelial cell death under AA treatment.

Materials and methods

Materials

The MTT assay kit was obtained from Bio Basic Canada, Inc. (Markham, OT, Canada). Vitamin E (α-tocopherol), luminol, lucigenin, tubulin polyclonal antibody and Hoechst 33342 were obtained from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Caspase-3 and cleaved caspase-3 polyclonal antibodies were obtained from Cell Signaling Technology, Inc. (9662; 1:1,000; Danvers, MA, USA). Fetal bovine serum, DMEM, non-essential amino acid, L-glutamine, and penicillin/streptomycin were obtained from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Cell culture

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

ROS detection

H2O2 and O2 levels were measured by using the lucigenin-amplified chemiluminescence method (48,49). The culture supernatant (200 µl) were added with 0.2 mmol/l of luminol solution (100 µl) and measured subsequently by using a chemiluminescence analyzing system (CLA-FSI; Tohoko Electronic Industrial Co., Ltd., Sendai, Japan) for the determination of H2O2 levels. The samples (200 µl) were treated with 0.1 mmol/l lucigenin solution (200 µl) and then O2 levels were measured using the CLA-FSI chemiluminescence analyzing system.

Cell survival rates determination

The cell survival rates were determined using the MTT assay kit according to the manufacturer's instructions. In brief, NRK-52E cells were cultured into 96-well plates at a density of 8×103 cells/well and incubated for 24 h in 100 µl DMEM medium. The suitable concentration and optimum exposure time of AAI were determined as 5, 10, 20 and 100 µM at 6 h time intervals. Cells were treated with MTT assay kit for 3 h at 37°C and were measured at 570 nm absorbance using a Multiskan™ FC microplate photometer (Molecular Devices, Inc., Sunnyvale, CA, USA). The cell survival rate was calculated as the following formula: Optical density (OD) 570 experimental group/OD 570 control group ×100%.

Observation of apoptotic features

Apoptotic features containing DNA fragmentation and nuclear condensation were observed by using Hoechst 33342 (23491-52-3; Sigma-Aldrich; Merck KGaA) nuclear staining (49,50). Control and experimental cells were treated with Hoechst 33342 (10 µg/ml) at 37°C 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).

Western blotting

Cells were treated with radioimmunoprecipitation assay buffer (20–188; EMD Millipore, Billerica, MA, USA). Following 10 min centrifugation (16,000 × g) at 4°C, proteins were obtained from the supernatant layer and the concentration was determined by using the protein assay kit (23200; Thermo Fischer Scientific, Inc.). Equal quantities of samples were separated on a 13.3% SDS-PAGE (Bio-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. Next, the membranes were washed with phosphate-buffered saline (PBS) then incubated with the primary antibodies for 4 h. Following that, membranes were washed with PBS and treated with anti-rabbit-horseradish peroxidase secondary antibodies (NA934; 1:1,000 Amersham; GE Healthcare Life Sciences, Chalfont, UK) for 1 h at room temperature. Finally, proteins were observed by using Western Lightning Chemiluminescence Reagent Plus (PerkinElmer, Inc., Waltham, MA, USA).

Statistical analysis

Student's t-test and two-way analysis of variance were utilized for the analysis of the data using SPSS version 18.0 (SPSS, Inc., Chicago, IL, USA). Values are expressed as the mean ± standard error. P<0.05 was considered statistically significant different between values.

Results

Increases of H2O2 and O2- levels by different concentrations of AA treatment

Previous studies have demonstrated that AA induced ROS generation in renal tubular cells (13,51). H2O2 and O2, two major types of ROS, were measured in AA-treated renal tubular cells. Experimental results indicated that H2O2 levels were increased dose-dependently in the AA-treated cells (Fig. 1A). Compared with H2O2 levels, O2 levels were increased only at the 100 µM AA treatment however not at 20–50 µM AA concentrations (Fig. 1B). The data suggested low-dose AA (5–20 µM) can induce increases in H2O2 levels, but not O2 levels. Additionally, high-dose AA (100 µM) can induce increases of H2O2 and O2 levels.

AA decreased cell survival rates in dose- and time- dependent manners

In order to determine the cytotoxic effects on AA-treated renal tubular cells, various concentrations (5–100 µM) of AA were studied. As presented in Fig. 2, the cell survival rates were below 50% at 100 µM AA (6 h), 20 µM AA (12 h), 10 µM AA (24 h) and 5 µM AA (48 h) treatment. These results demonstrated AA-induced cell cytotoxicity was dose- and time-dependent.

Apoptotic characteristics in AA-treated renal tubular cells

Cell death can be described as apoptosis or necrosis (52,53). Apoptotic cells can be removed by macrophages in order to prevent serious inflammatory responses (54,55), and previous studies have indicated that nuclear condensation and DNA fragmentation are key apoptotic characteristics (49,56). In the present study, the cell nuclei were observed by Hoechst 33342 staining (49,50). As presented in Fig. 3, compared with control cell, the nuclear condensation and DNA fragmentation were identified in AA-treated renal tubular cells. The results indicated that AA-induced cell death was associated with the apoptotic death pathway.

α-tocopherol attenuated H2O2 levels and increased cell survival in AA-treated cells

The antioxidant stress activities of vitamin E (α-tocopherol) has been demonstrated in clinical cases (44,45). Due to the fact that AA elevated H2O2 levels (Fig. 1A), it was investigated whether α-tocopherol could inhibit H2O2 in AA-treated cells with various concentrations of α-tocopherol (5, 10, 20 and 100 µM). The data indicated that α-tocopherol attenuated AA-induced H2O2 levels (Fig. 4). Notably, it was observed that 10 µM α-tocopherol appeared to have a more marked effect on AA-induced H2O2 compared with other concentrations. Subsequently, the effects of α-tocopherol on AA-induced renal cell death were investigated. As presented in Fig. 5, the cell survival rates were increased in AA-treated renal tubular cells undergoing 10 or 20 µM α-tocopherol treatment. These results first demonstrated that α-tocopherol attenuated AA-induced H2O2 levels and increased cell survival of AA-treated cells.

α-tocopherol reduced AA-activated caspase-3

Caspase-3 activation is associated with the apoptotic death pathway (4042). Due to the fact that apoptotic characteristics were predominantly identified in AA-treated renal tubular cells (Fig. 3B), whether AA could activate caspase-3 was investigated. As presented in Fig. 6, compared with the control group (lane 1), the level of cleaved caspase-3 was markedly increased in the AA-treated group (lane 2). AA was identified to induce caspase-3 activity and the effect of α-tocopherol on AA-induced caspase-3 was further determined. AA was reported to decrease mitochondrial membrane depolarization and to lead to an increase of caspase-3 (42). The results demonstrated that cleaved caspase-3 levels were reduced in the AA plus α-tocopherol group (lane 5 and 6) compared with the AA treatment group (lane 2). Due to the fact that AA-induced H2O2 levels are also reduced in the AA + α-tocopherol group (Fig. 4), this suggested that α-tocopherol attenuation of AA-induced cell cytotoxicity may be associated with caspase-3 activity from the reduce of H2O2 levels.

Discussion

The results of the current study indicated that AA causes increases in H2O2 levels and a reduction in cell survival rates in renal tubular cells and α-tocopherol (10 and 20 µM) attenuated AA-induced H2O2 levels and inhibited AA-induced cytotoxicity in these cells. These data suggested that AA-induced cell cytotoxicity may be associated with H2O2 levels. By contrast, α-tocopherol could not inhibit AA-induced cytotoxicity under high-dose (100 µM) AA treatment (data not shown), however it effectively ameliorated AA-induced cytotoxicity under low-dose (5–20 µM) AA treatment. This suggested that α-tocopherol ameliorated AA-induced renal cell damage was dependent on AA dosage. Furthermore, high-dose AA alone elevated both H2O2 and O2 levels. Therefore, this may partially explain why α-tocopherol could not inhibit high-dose AA-induced cell cytotoxicity.

CAT, Gpx and SOD are major cellular antioxidant enzyme systems (57,58). CAT is a tetrameric iron-porphyrin protein in peroxisomes that converts H2O2 to H2O and O2. CAT and Cu/Zn-SOD are expressed constitutively, whereas Mn-SOD expression within the mitochondria is induced by oxidative stress. GSH is a sulfhydryl peptide that may directly react with O2 or N2 containing free radicals, or is able to donate electrons in the enzymatic dismutation of H2O2 to H2O and O2 by GPx (59,60). Cellular CAT and Gpx can remove H2O2, whereas SOD removes O2 (61). In the present study, the data indicated that AA induced increases in H2O2 in a dose-dependent manner, however the 100 µM AA alone was capable of increasing O2 levels. This result suggested that low-dose AA may influence the activity of CAT and Gpx while high-dose AA may influence CAT, Gpx and SOD activities. However, further studies are required to confirm this hypothesis.

Studies have indicated that antioxidant can attenuate AA-induced renal damage (11,29,59) and it has been additionally demonstrated that vitamin C attenuated AA-induced renal damage (29). However, the effect of antioxidant α-tocopherol on AA-induced renal damage remains to be reported. The current study demonstrated that α-tocopherol ameliorated AA-induced renal cell cytotoxicity.

Vitamin C and α-tocopherol are common antioxidants and have been used in clinical cases (44,45,62,63). Vitamin C and α-tocopherol both have antioxidant activities, however their antioxidant mechanisms differ (6466). These studies indicated that α-tocopherol (lipid-soluble material) is able to pass through the cell membrane and catch the free radicals to protect the cell from oxidative damage. However, vitamin C (water-soluble material) cannot pass through cell membrane to remove free radicals directly. Based on the results of a previous study (29) and the current study, it is suggested that both vitamin C and α-tocopherol scavenge H2O2 produced by AA-treated renal cells, leading to an increase of survival rate. It was suggested that AA-induced H2O2 existed not only in the cytosol however additionally in the cell membrane. In addition, another key antioxidant function of vitamin C is converting the oxidized α-tocopherol radical back to α-tocopherol (62). It was suggested that the combination of vitamin C and α-tocopherol may be more powerful for protection of AA-induced renal damage. In patients with coronary artery bypass surgery, vitamin cocktail (ascorbic acid and α-tocopherol) effectively attenuated oxidative stress than control (44). In summary, the results demonstrated that α-tocopherol attenuates AA-induced H2O2 and caspase-3 and ameliorated AA-induced renal cell cytotoxicity.

Acknowledgements

The present study was supported by grants from the Ministry of Science and Technology, Taiwan (grant nos. MOST103 2320-B-039-052-MY3 and MOST104-2321-B-039-005) and Ministry of Health and Welfare (MOHW104-TDU-B-212-124-002).

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

2 

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

3 

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 :

4 

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

5 

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

6 

Luciano RL and Perazella MA: Aristolochic acid nephropathy: Epidemiology, clinical presentation, and treatment. Drug Saf. 38:55–64. 2015. View Article : Google Scholar

7 

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

8 

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

9 

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

10 

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 :

11 

Matsui K, Kamijo-Ikemorif A, Sugaya T, Yasuda T and Kimura K: Renal liver-type fatty acid binding protein (L-FABP) attenuates acute kidney injury in aristolochic acid nephrotoxicity. Am J Pathol. 178:1021–1032. 2011. View Article : Google Scholar :

12 

Bunel V, Antoine MH, Nortier J, Duez P and Stévigny C: In vitro effects of Panax ginseng in aristolochic acid-mediated renal tubulotoxicity: Apoptosis versus regeneration. Planta Med. 81:363–372. 2015. View Article : Google Scholar

13 

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

14 

Singh M, Kapoor A and Bhatnagar A: Oxidative and reductive metabolism of lipid-peroxidation derived carbonyls. Chem Biol Interact. 234:261–273. 2015. View Article : Google Scholar :

15 

Dur A, Kocaman O, Koçyiğit A, Türkdoğan KA, Sönmez E, Keskin S, Yiğit M, Gülen B, Kılıç E and Uysal Ö: Oxidative status and lymphocyte DNA damage in patients with acute pancreatitis and its relationship with severity of acute pancreatitis. Turk J Gastroenterol. 27:68–72. 2016. View Article : Google Scholar

16 

Kruk J, Kubasik-Kladna K and Aboul-Enein HY: The role oxidative stress in the pathogenesis of eye diseases: Current status and a dual role of physical activity. Mini Rev Med Chem. 16:241–257. 2015. View Article : Google Scholar

17 

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 :

18 

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

19 

Goncalves RL, Quinlan CL, Perevoshchikova IV, Hey-Mogensen M and Brand MD: Sites of superoxide and hydrogen peroxide production by muscle mitochondria assessed ex vivo under conditions mimicking rest and exercise. J Biol Chem. 290:209–227. 2015. View Article : Google Scholar

20 

Goncalves RL, Rothschild DE, Quinlan CL, Scott GK, Benz CC and Brand MD: Sources of superoxide/H2O2 during mitochondrial proline oxidation. Redox Biol. 2:901–909. 2014. View Article : Google Scholar :

21 

Prasad AK and Mishra PC: Mechanism of action of sulforaphane as a superoxide radical anion and hydrogen peroxide scavenger by double hydrogen transfer: A model for iron superoxide dismutase. J Phys Chem B. 119:7825–7836. 2015. View Article : Google Scholar

22 

Ludwig E and Eyer P: Reactivity of glutathione adducts of 4-(dimethylamino)phenol. Involvement of reactive oxygen species during the interaction with oxyhemoglobin. Chem Res Toxicol. 8:363–368. 1995. View Article : Google Scholar

23 

Tulunoglu O, Alacam A, Bastug M and Yavuzer S: Superoxide dismutase activity in healthy and inflamed pulp tissues of permanent teeth in children. J Clin Pediatr Dent. 22:341–345. 1998.

24 

Gölz L, Memmert S, Rath-Deschner B, Jäger A, Appel T, Baumgarten G, Götz W and Frede S: LPS from P. gingivalis and hypoxia increases oxidative stress in periodontal ligament fibroblasts and contributes to periodontitis. Mediators Inflamm. 2014:9862642014. View Article : Google Scholar :

25 

Magdalan J, Piotrowska A, Gomulkiewicz A, Sozański T, Szelag A and Dziegiel P: Influence of commonly used clinical antidotes on antioxidant systems in human hepatocyte culture intoxicated with alpha-amanitin. Hum Exp Toxicol. 30:38–43. 2011. View Article : Google Scholar

26 

Peng XL, Xu WT, Wang Y, Huang KL, Liang ZH, Zhao WW and Luo YB: Mycotoxin Ochratoxin A-induced cell death and changes in oxidative metabolism of Arabidopsis thaliana. Plant Cell Rep. 29:153–161. 2010. View Article : Google Scholar

27 

Maruf AA and O'Brien P: Inflammation-enhanced drug-induced liver injury. Free Radic Biol Med. 75 Suppl 1:S402014. View Article : Google Scholar

28 

Pisoschi AM and Pop A: The role of antioxidants in the chemistry of oxidative stress: A review. Eur J Med Chem. 97:55–74. 2015. View Article : Google Scholar

29 

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. View Article : Google Scholar

30 

Gomez C, Martinez L, Mesa A, Duque JC, Escobar LA, Pham SM and Vazquez-Padron RI: Oxidative stress induces early-onset apoptosis of vascular smooth muscle cells and neointima formation in response to injury. Biosci Rep. 35:pii: e00227. 2015. View Article : Google Scholar :

31 

Hu XL, Niu YX, Zhang Q, Tian X, Gao LY, Guo LP, Meng WH and Zhao QC: Neuroprotective effects of Kukoamine B against hydrogen peroxide-induced apoptosis and potential mechanisms in SH-SY5Y cells. Environ Toxicol Pharmacol. 40:230–240. 2015. View Article : Google Scholar

32 

Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, et al: CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 22:175–182. 2016. View Article : Google Scholar

33 

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

34 

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

35 

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

36 

Seo HS, Ku JM, Choi HS, Woo JK, Jang BH, Go H, Shin YC and Ko SG: Apigenin induces caspase-dependent apoptosis by inhibiting signal transducer and activator of transcription 3 signaling in HER2-overexpressing SKBR3 breast cancer cells. Mol Med Rep. 12:2977–2984. 2015. View Article : Google Scholar

37 

Göke A, Göke R, Ofner A, Herbst A and Lankat-Buttgereit B: The FGFR inhibitor NVP-BGJ398 induces NSCLC cell death by activating caspase-dependent pathways as well as caspase-independent apoptosis. Anticancer Res. 35:5873–5879. 2015.

38 

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

39 

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

40 

Yuan SY, Yang CR, Cheng CL, Hsu SL, Liao JW, Lin CC, Chou YY and Cheng YW: Comparative nephrotoxicity of aristolochic acid and tetrandrine in vitro and in vivo. Int J Toxicol. 30:35–46. 2011. View Article : Google Scholar

41 

Kwak DH, Park JH, Lee HS, Moon JS and Lee S: Aristolochic acid I induces ovarian toxicity by inhibition of akt phosphorylation. Chem Res Toxicol. 27:2128–35. 2014. View Article : Google Scholar

42 

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

43 

Cook-Mills JM: Isoforms of vitamin E differentially regulate PKC α and inflammation: A review. J Clin Cell Immunol. 4:pii: 1000137. 2013. View Article : Google Scholar :

44 

Stanger O, Aigner I, Schimetta W and Wonisch W: Antioxidant supplementation attenuates oxidative stress in patients undergoing coronary artery bypass graft surgery. Tohoku J Exp Med. 232:145–154. 2014. View Article : Google Scholar

45 

Park OJ, Kim HY, Kim WK, Kim YJ and Kim SH: Effect of vitamin E supplementation on antioxidant defense systems and humoral immune responses in young, middle-aged and elderly Korean women. J Nutr Sci Vitaminol (Tokyo). 49:94–99. 2003. View Article : Google Scholar

46 

Kutlubay R, Oğuz EO, Güven C, Can B, Sinik Z and Tuncay OL: Histological and ultrastructural evidence for protective effects on aluminium-induced kidney damage by intraperitoneal administration of alpha-tocopherol. Int J Toxicol. 26:95–101. 2007. View Article : Google Scholar

47 

Tasanarong A, Kongkham S, Duangchana S, Thitiarchakul S and Eiam-Ong S: Vitamin E ameliorates renal fibrosis by inhibition of TGF-beta/Smad2/3 signaling pathway in UUO mice. J Med Assoc Thai 94 Suppl. 7:S1–S9. 2011.

48 

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 :

49 

Yiang GT, Yu YL, Lin KT, Chen JN, Chang WJ and Wei CW: Acetaminophen induces JNK/p38 signaling and activates the caspase-9-3-dependent cell death pathway in human mesenchymal stem cells. Int J Mol Med. 36:485–492. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Yu YL, Yiang GT, Chou PL, Tseng HH, Wu TK, Hung YT, Lin PS, Lin SY, Liu HC, Chang WJ and Wei CW: Dual role of acetaminophen in promoting hepatoma cell apoptosis and kidney fibroblast proliferation. Mol Med Rep. 9:2077–2084. 2014. View Article : Google Scholar : PubMed/NCBI

51 

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

52 

Kataki A, Skandami V, Memos N, Nikolopoulou M, Oikonomou V, Androulis A, Konstadoulakis MM and Zografos CG: Similar immunity profiles in patients with meningioma and glioma tumors despite differences in the apoptosis and necrosis of circulating lymphocyte and monocyte populations. J Neurosurg Sci. 58:9–15. 2014.PubMed/NCBI

53 

Song AS, Najjar AM and Diller KR: Thermally induced apoptosis, necrosis and heat shock protein expression in 3D culture. J Biomech Eng. 136:2014. View Article : Google Scholar :

54 

Wang Q, Zeng P, Liu Y, Wen G, Fu X and Sun X: Inhibition of autophagy ameliorates atherogenic inflammation by augmenting apigenin-induced macrophage apoptosis. Int Immunopharmacol. 27:24–31. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Martin KR, Ohayon D and Witko-Sarsat V: Promoting apoptosis of neutrophils and phagocytosis by macrophages: Novel strategies in the resolution of inflammation. Swiss Med Wkly. 145:w140562015.PubMed/NCBI

56 

Yiang GT, Chou PL, Hung YT, Chen JN, Chang WJ, Yu YL and Wei CW: Vitamin C enhances anticancer activity in methotrexate-treated Hep3B hepatocellular carcinoma cells. Oncol Rep. 32:1057–1063. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Valle A, Oliver J and Roca P: Role of uncoupling proteins in cancer. Cancers (Basel). 2:567–591. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Cai H: Hydrogen peroxide regulation of endothelial function: Origins, mechanisms, and consequences. Cardiovasc Res. 68:26–36. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Zelko IN, Mariani TJ and Folz RJ: Superoxide dismutase multigene family: A comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expressio. Free Radic Biol Med. 33:337–349. 2002. View Article : Google Scholar : PubMed/NCBI

60 

Flora SJ: Structural, chemical and biological aspects of antioxidants for strategies against metal and metalloid exposure. Oxid Med Cell Longev. 2:191–206. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Costa A, Scholer-Dahirel A and Mechta-Grigoriou F: The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin Cancer Biol. 25:23–32. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Anichini C, Lotti F, Pietrini A, Lo Rizzo C, Longini M, Proietti F, Felici C and Buonocore G: Antioxidant effects of potassium ascorbate with ribose in costello syndrome. Anticancer Res. 33:691–695. 2013.PubMed/NCBI

63 

Moore DF, Ye F, Brennan ML, Gupta S, Barshop BA, Steiner RD, Rhead WJ, Brady RO, Hazen SL and Schiffmann R: Ascorbate decreases Fabry cerebral hyperperfusion suggesting a reactive oxygen species abnormality: An arterial spin tagging study. J Magn Reson Imaging. 20:674–683. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Packer L, Weber SU and Rimbach G: Molecular aspects of alpha-tocotrienol antioxidant action and cell signalling. J Nutr. 131:369S–373S. 2001.PubMed/NCBI

65 

Chaudière J and Ferrari-Iliou R: Intracellular antioxidants: From chemical to biochemical mechanisms. Food Chem Toxicol. 37:949–962. 1999. View Article : Google Scholar : PubMed/NCBI

66 

Singh M, Singh S and Kale RK: Chemomodulatory potential of Asparagus adscendens against murine skin and forestomach papillomagenesis. Eur J Cancer Prev. 20:240–247. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 Issue 1

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, Pan YR, Wang HF, Wei CW and Yu YL: Vitamin E (α‑tocopherol) ameliorates aristolochic acid‑induced renal tubular epithelial cell death by attenuating oxidative stress and caspase‑3 activation. Mol Med Rep 17: 31-36, 2018
APA
Wu, T., Pan, Y., Wang, H., Wei, C., & Yu, Y. (2018). Vitamin E (α‑tocopherol) ameliorates aristolochic acid‑induced renal tubular epithelial cell death by attenuating oxidative stress and caspase‑3 activation. Molecular Medicine Reports, 17, 31-36. https://doi.org/10.3892/mmr.2017.7921
MLA
Wu, T., Pan, Y., Wang, H., Wei, C., Yu, Y."Vitamin E (α‑tocopherol) ameliorates aristolochic acid‑induced renal tubular epithelial cell death by attenuating oxidative stress and caspase‑3 activation". Molecular Medicine Reports 17.1 (2018): 31-36.
Chicago
Wu, T., Pan, Y., Wang, H., Wei, C., Yu, Y."Vitamin E (α‑tocopherol) ameliorates aristolochic acid‑induced renal tubular epithelial cell death by attenuating oxidative stress and caspase‑3 activation". Molecular Medicine Reports 17, no. 1 (2018): 31-36. https://doi.org/10.3892/mmr.2017.7921