Determination of the Effect of Thymoquinone on DNA Damage in Kidney Cells Treated to High Glucose Depending on Time and Dosage by Comet Assay

Yazarlar

DOI:

https://doi.org/10.24925/turjaf.v12i8.1359-1365.6573

Anahtar Kelimeler:

comet tahlili- genotoksisite- NRK-52E- timokinon- yüksek glukoz

Özet

The purpose of this study was to assess the anti-genotoxic potential of thymoquinone (TQ) against DNA damage in NRK-52E cells treated with high glucose using the comet assay technique single cell gel electrophoresis method. Cells were propagated by regular passages in in vitro conditions. TQ proliferative concentration (10μM) and IC25 (3rd-hour: 550 mM, 12th-hour: 240 mM, 24th-hour: 200 mM) and IC50 (3rd-hour: 760 mM, 12th-hour: 400 mM, 24th-hour: 280 mM) values for each hour of high glucose and were determined separately with MTT method. At these concentrations, the cells were divided into control(C), Thymoquinone (TQ), high glucose(G) and high glucose plus thymoquinone (GT) groups; It was incubated with the indicated substances for 3, 12, 24 hours. DNA damage was evaluated by applying the comet assay protocol and the results were calculated as DNA damage index (DDI). While DDI levels were observed to be significantly higher (p<0.05) in all groups administered high glucose compared to the control, a significant decrease was determined in all groups in which TQ was added along with high glucose. It was determined that high concentrations of glucose had genotoxic effects on kidney cells, and TQ administration together with high glucose, depending on concentration and time, had a significant effect on reducing DNA damage. However, it was concluded that the application of only thymoquinone significantly increased the DDI value compared to the control, and this was a data worth investigating in future studies. Additionally, TQ inhibited DNA damage. These results demonstrated the importance of TQ against nephrotic syndrome with its high antioxidant properties.

Referanslar

Abdelmeguid, N.E., Fakhoury, R., Kamal, S.M., & Al Wafai, R.J. (2010). Effects of Nigella sativa and thymoquinone on biochemical and subcellular changes in pancreatic β-cells of streptozotocin-induced diabetic rats. J Diabetes, 2(4), 256-266. https://doi.org/ 10.1111/j.1753-0407.2010.00091.x

Afanasieva, K., & Sivolob, A. (2018). Physical principles and new applications of comet assay. Biophys Chem, 238(1), 1–7. https://doi.org/10.1016/j.bpc.2018.04.003

Alam, M.M., Iqbal, S., & Naseem, I. (2015). Ameliorative effect of riboflavin on hyperglycemia, oxidative stress and DNA damage in type-2 diabetic mice: Mechanistic and therapeutic strategies. Arch Biochem Biophys, 584(1), 10-19. https://doi.org/10.1016/j.abb.2015.08.013

Al-Aubaidy, H.A., & Jelinek, H.F. (2011). Oxidative DNA damage and obesity in type 2 diabetes mellitus. Eur J Endocrinol, 164(6), 899-904. https://doi.org/10.1530/EJE-11-0053

Aybastıer, Ö., Dawbaa, S., Demir, C., Akgün, O., Ulukaya, E., & Arı, F. (2018). Quantification of DNA damage products by gas chromatography-tandem mass spectrometry in lung cell lines and prevention effect of thyme antioxidants on oxidative induced DNA damage. Mutat Res, 808 (1), 1-9. https://doi.org/10.1016/j.mrfmmm.2018.01.004

Bazyel, B., Dede, S., Çetin, S., Yüksek, V., & Taşpinar, M. (2019). In vitro evaluation of the effects of lycopene on caspase system and oxidative DNA damage in high-glucose condition. Phcog Mag, 15(1), 30-33. https://doi.org/10.4103/pm.pm_488_18

Bhatt, S., Gupta, M.K., Khamaisi, M., Martinez, R., Gritsenko, M.A., & Wagner, B.K, et al. (2015). Preserved DNA Damage Checkpoint Pathway Protects against Complications in Long-Standing Type 1 Diabetes. Cell Metab, 22(2), 239-252. https://doi.org/10.1016/j.cmet.2015.07.015

Boutet-Robinet, E., Trouche, D., & Canitrot, Y. (2013). Neutral Comet Assay. Bio-protocol. Bio-protocol LCC, 3(18), 1-6. https://doi.org/10.21769/BioProtoc.915

Cui, B., & Yu, J.M. (2018). Autophagy: a new pathway for traditional Chinese medicine. J Asian Nat Prod Res, 20(1), 14-26. https://doi.org/10.1080/10286020.2017.1374948

Çelebioğlu, H.G.N., Becit-Kizilkaya, M., Çağlayan, A., & Aydın Dilsiz, S. (2022). Effects of thymoquinone and etoposide combination on cell viability and genotoxicity in human cervical cancer hela cells. Istanbul J Pharm, 52(3), 258–264. https://doi.org/10.26650/IstanbulJPharm.2022.1105443

Çetin, S., Usta, A., & Yüksek, V. (2021). The effect of lycopene on dna damage and repair in fluoride-treated NRK-52E cell line. Biol Trace Elem Res, 199(1), 1979-1985. https://doi.org/10.1007/s12011-020-02288-4

Devi, T.S., Hosoya, K., Terasaki, T., & Singh, L.P. (2013). Critical role of TXNIP in oxidative stress, DNA damage and retinal pericyte apoptosis under high glucose: implications for diabetic retinopathy. Exp Cell Res, 319(7), 1001-1012. https://doi.org/10.1016/j.yexcr.2013.01.012

El-Shemi, A.G., Kensara, O.A., Alsaegh, A., & Mukhtar, M.H. (2018). Pharmacotherapy with Thymoquinone Improved Pancreatic β-Cell Integrity and Functional Activity, Enhanced Islets Revascularization, and Alleviated Metabolic and Hepato-Renal Disturbances in Streptozotocin-Induced Diabetes in Rats. Pharmacology, 101(1-2), 9-21. https://doi.org/10.1159/000480018

Garcia, O., Romero, I., González, J.E., Moreno, D.L., Cuétara, E., & Rivero, Y., et al. (2011). Visual estimation of the percentage of DNA in the tail in the comet assay: evaluation of different approaches in an intercomparison exercise. Mutat Res, 720(1-2), 14-21. https://doi.org/10.1016/j.mrgentox.2010.11.011

Gondi, M., Basha, S.A., Bhaskar, J.J., Salimath, P.V., & Rao, U.J. (2015). Anti-diabetic effect of dietary mango (Mangifera indica L.) peel in streptozotocin-induced diabetic rats. J Sci Food Agric, 95(5), 991-999. https://doi.org/10.1002/jsfa.6778

Gümüş, A., Dede, S., Yüksek, V., Çetin, S., & Taşpınar, M. (2018). In vitro evaluation of thymoquinone on apoptosis and oxidative DNA damage in high glucose condition. Cell Mol Biol (Noisy-le-grand), 64(1), 79-83. https://doi.org/10.14715/cmb/2018.64.14.13

Habib, S.L., Yadav, A., Kidane, D., Weiss, R.H., & Liang, S. (2016). Novel protective mechanism of reducing renal cell damage in diabetes: Activation AMPK by AICAR increased NRF2/OGG1 proteins and reduced oxidative DNA damage. Cell Cycle, 15(22), 3048-3059. https://doi.org/10.1080/15384101.2016.1231259

Hazman, Ö., Sarıova, A., Bozkurt, M.F., & Ciğerci, İ.H. (2021). The anticarcinogen activity of β-arbutin on MCF-7 cells: Stimulation of apoptosis through estrogen receptor-α signal pathway, inflammation and genotoxicity. Mol Cell Biochem, 476(1), 349-360. https://doi.org/10.1007/s11010-020-03911-7

Hou, S., Zheng, F., Li, Y., Gao, L., & Zhang, J. (2014). The protective effect of glycyrrhizic acid on renal tubular epithelial cell injury induced by high glucose. Int J Mol Sci, 15(9), 15026-15043. https://doi.org/10.3390/ijms150915026

Karahan, F., Dede, S., & Ceylan E. (2018). The Effect of Lycopene Treatment on Oxidative DNA Damage of Experimental Diabetic Rats. The Open Clinical Biochemistry Journal, 8(1), 1-6. https://doi.org/10.2174/2588778501808010001

Kim, K.M., Kim, Y.S., Jung, D.H., Lee, J., & Kim, J.S. (2012). Increased glyoxalase I levels inhibit accumulation of oxidative stress and an advanced glycation end product in mouse mesangial cells cultured in high glucose. Exp Cell Res, 318(2), 152-159. https://doi.org/10.1016/j.yexcr.2011.10.013

Kumaravel, T.S., Vilhar, B., Faux, S.P., & Jha, A.N. (2009). Comet Assay measurements: a perspective. Cell Biol Toxicol, 25(1), 53–64. https://doi.org/10.1007/s10565-007-9043-9

Kurt, E., Dede, S., & Ragbetli, C. (2015). The investigations of total antioxidant status and biochemical serum profile in thymoquinone-treated rats. Afr J Tradit Complement Altern Med, 12(2), 68-72. https://doi.org/10.4314/ajtcam.v12i2.13

Lopez-Sanz, L., Bernal, S., Recio, C., Lazaro, I., Oguiza, A., & Melgar, A., et al. (2018). SOCS1-targeted therapy ameliorates renal and vascular oxidative stress in diabetes via STAT1 and PI3K inhibition. Lab Invest, 98(10), 1276-1290. https://doi.org/10.1038/s41374-018-0043-6

Maynard, S., Schurman, S.H., Harboe, C., de Souza-Pinto, N.C., & Bohr, V.A. (2009). Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis, 30(1), 2–10. https://doi.org/10.1093/carcin/bgn250

Nakamichi, R., Hayashi, K., & Itoh, H. (2021). Effects of high glucose and lipotoxicity on diabetic podocytes. Nutrients, 13(1), 1-11. https://doi.org/10.3390/nu13010241

Orsolic, N., Gajski, G., Garaj-Vrhovac, V., Dikić, D., Prskalo, Z.Š., & Sirovina, D. (2011). DNA-protective effects of quercetin or naringenin in alloxan-induced diabetic mice. Eur J Pharmacol, 656(1-3), 110-118. https://doi.org/10.1016/j.ejphar.2011.01.021

Othman, E.M., Kreissl, M.C., Kaiser, F.R., Arias-Loza, P.A., & Stopper, H. (2013). Insulin-mediated oxidative stress and DNA damage in LLC-PK1 pig kidney cell line, female rat primary kidney cells, and male ZDF rat kidneys. in vivo. Endocrinology, 154(4), 1434-1443. https://doi.org/10.1210/en.2012-1768

Pari, L., & Sankaranarayanan, C. (2009). Beneficial effects of thymoquinone on hepatic key enzymes in Spironolactone improves nephropathy by enhancing glucose-6-phosphate dehydrogenase activity and reducing oxidative stress in diabetic hypertensive rat. Life Sci, 85(23-26), 830–834. https://doi.org/10.1016/j.lfs.2009.10.021

Pessôa, B.S., Peixoto, E.B., Papadimitriou, A., Lopes de Faria, J.M., & Lopes de Faria, J.B. (2012). Spironolactone improves nephropathy by enhancing glucose-6-phosphate dehydrogenase activity and reducing oxidative stress in diabetic hypertensive rat. J Renin Angiotensin Aldosterone Syst, 13(1), 56-66. https://doi.org/10.1177/1470320311422581

Ping, K.Y., Darah, I., Chen, Y., Sreeramanan, S., & Sasidharan, S. (2013). Acute and subchronic toxicity study of Euphorbia hirta L. methanol extract in rats. Biomed Res Int, 2013(182064), 1-13. https://doi.org/10.1155/2013/182064

Poetsch, A.R. (2020). The genomics of oxidative DNA damage, repair, and resulting mutagenesis. Computational and Structural Biotechnology Journal, 18(1), 207–219. https://doi.org/10.1016/j.csbj.2019.12.013

Samikkannu, T., Thomas, J.J., Bhat, G.J., Wittman, V., & Thekkumkara, T.J. (2006). Acute effect of high glucose on long-term cell growth: a role for transient glucose increase in proximal tubule cell injury. Am J Physiol Renal Physiol, 291(1), 162-175. https://doi.org/10.1152/ajprenal.00189.2005

Simone, S., Gorin, Y., Velagapudi, C., Abboud, H.E., & Habib, S.L. (2008). Mechanism of oxidative DNA damage in diabetes tuberin inactivation and downregulation of DNA Repair Enzyme 8-Oxo-7,8-Dihydro-2′-Deoxyguanosine-DNA Glycosylase. Diabetes, 57(10), 2626–2636. https://doi.org/10.2337/db07-1579

Smail, H.O. (2023). Identification of micronuclei in the lymphocytes of the type 2 diabetes mellitus according to the period of diagnosis as a biomarker. Polytechnic Journal, 13(2), 23-27. https://doi.org/10.59341/2707-7799.1720

Subramaniyan, S.D., Natarajan, A.K., & Citral, A. (2017). Monoterpene protect against high glucose induced oxidative injury in Hepg2 cell in vitro-an experimental study. J Clin Diagn Res, 11(8), 10-15. https://doi.org/10.7860/JCDR/2017/28470.10377

Sun, L., Dutta, R.K., Xie, P., & Kanwar, Y.S. (2016). Myo-inositol oxygenase overexpression accentuates generation of reactive oxygen species and exacerbates cellular injury following high glucose ambience: A new mechanism relevant to the pathogenesis of diabetic nephropathy. J Biol Chem, 291(11), 5688-5707. https://doi.org/10.1074/jbc.M115.669952

Talebi, M., Talebi, M., Farkhondeh, T., & Samarghandian, S. (2021). Biological and therapeutic activities of thymoquinone: Focus on the Nrf2 signaling pathway. Phytother Res, 35(4), 1739-1753. https://doi.org/10.1002/ptr.6905

Usta, A., & Dede, S. (2017). The effect of thymoquinone on nuclear factor kappa b levels and oxidative DNA damage on experimental diabetic rats. Pharmacogn Mag, 13 (3), 458-461. https://doi.org/10.4103/pm.pm_134_17

Usta, A., Yüksek, V., Çetin, S., & Dede, S. (2024). Lycopene prevents cell death in NRK‐52E cells by inhibition of high glucose‐activated DNA damage and apoptotic, autophagic, and necrotic pathways. Journal of Biochemical and Molecular Toxicology, 38(1), 1-13. https://doi.org/10.1002/jbt.23678

Varun, K., Zoltan, K., Alba, S., Manuel, B., Elisabeth, K., & Dimitrios, T., et al. (2023). Elevated markers of DNA damage and senescence are associated with the progression of albuminuria and restrictive lung disease in patients with type 2 diabetes. EBioMedicine, 90:104516, 1-17. https://doi.org/10.1016/j.ebiom.2023.104516

Verma, R., Rigatti, M.J., Belinsky, G.S., Godman, C.A., & Giardina, C. (2010). DNA damage response to the Mdm2 inhibitor nutlin-3. Biochem Pharmacol, 79(1), 565–574. https://doi.org/10.1016/j.bcp.2009.09.020

Woo, C.C., Kumar, A.P., Sethi, G., & Tan, K.H. (2012). Thymoquinone: potential cure for inflammatory disorders and cancer. Biochem Pharmacol, 83(4), 443-451. https://doi.org/10.1016/j.bcp.2011.09.029

Wu, T., Ding, L., Andoh, V., Zhang, J., & Chen, L. (2023). The mechanism of hyperglycemia-induced renal cell injury in diabetic nephropathy disease: an update. Life (Basel), 13(2), 539, 1-18. https://doi.org/10.3390/life13020539

Xiong, Y., & Zhou, L. (2019). The signaling of cellular senescence in diabetic nephropathy. Oxid Med Cell Longev, 2019;3:2019:7495629, 1-16. https://doi.org/10.1155/2019/7495629

Yaycı, G., Dede, S., Usta, A., Yüksek, V., & Çetin, S. (2021). In vitro evaluation of the effect of glutathione on caspase system and oxidative DNA damage in high glucose condition. Eskişehir Techn Univ J of Sci and Tech C - Life Sci and Biotech, 10(2), 132-137. https://doi.org/10.18036/estubtdc.802794

Yılmaz, O., Yüksek, V., Çetin, S., Dede, S., & Tuğrul, T. (2021). The effects of thymoquinone on DNA damage, apoptosis and oxidative stress in an osteoblast cell line exposed to ionizing radiation. Radiat Eff Defect S, 176(5-6), 575-589. https://doi.org/10.1080/10420150.2021.1898394

Yu, J., Liu, C., Li, Z., Zhang, C., Wang, Z., & Liu, X. (2016). Inhibitory effects and mechanism of 25-OH-PPD on glomerular mesangial cell proliferation induced by high glucose. Environ Toxicol Pharmacol, 44(1), 93-98. https://doi.org/10.1016/j.etap.2016.04.013

Yüksek, V., Dede, S., Usta, A., Çetin, S., & Taşpınar, M. (2020a). DNA damage induced by sodium fluoride (NaF) and the effect of cholecalciferol. Biocell, 44(2), 263-268. https://doi.org/10.32604/biocell.2020.09172

Yüksek, V., Çetin, S., & Usta, A. (2020b). The effect of vitamin E and selenium combination in repairing fuoride‑induced DNA damage to NRK‑52E cells. Molecular Biology Reports, 47(1), 7761–7770. https://doi.org/10.1007/s11033-020-05852-2

Yayınlanmış

2024-08-24

Nasıl Atıf Yapılır

Usta, A., Dede, S., Yüksek, V., & Öner, A. C. (2024). Determination of the Effect of Thymoquinone on DNA Damage in Kidney Cells Treated to High Glucose Depending on Time and Dosage by Comet Assay. Türk Tarım - Gıda Bilim Ve Teknoloji Dergisi, 12(8), 1359–1365. https://doi.org/10.24925/turjaf.v12i8.1359-1365.6573

Sayı

Bölüm

Araştırma Makalesi