دورية أكاديمية

Isobavachalcone induces hepatotoxicity in zebrafish embryos and HepG2 cells via the System Xc - -GSH-GPX4 signaling pathway in ferroptosis response.

التفاصيل البيبلوغرافية
العنوان: Isobavachalcone induces hepatotoxicity in zebrafish embryos and HepG2 cells via the System Xc - -GSH-GPX4 signaling pathway in ferroptosis response.
المؤلفون: Ni X; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China., Gao C; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China., Zhu X; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China., Zhang X; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China., Fang Y; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China., Hao Z; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.; Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China.
المصدر: Journal of applied toxicology : JAT [J Appl Toxicol] 2024 Aug; Vol. 44 (8), pp. 1139-1152. Date of Electronic Publication: 2024 Apr 06.
نوع المنشور: Journal Article
اللغة: English
بيانات الدورية: Publisher: John Wiley And Sons Country of Publication: England NLM ID: 8109495 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1099-1263 (Electronic) Linking ISSN: 0260437X NLM ISO Abbreviation: J Appl Toxicol Subsets: MEDLINE
أسماء مطبوعة: Publication: : Chichester : John Wiley And Sons
Original Publication: [Philadelphia, Pa. : Heyden & Son, c1981-
مواضيع طبية MeSH: Zebrafish*/embryology , Chalcones*/toxicity , Chalcones*/pharmacology , Ferroptosis*/drug effects , Signal Transduction*/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase*/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase*/genetics, Animals ; Humans ; Hep G2 Cells ; Embryo, Nonmammalian/drug effects ; Glutathione/metabolism ; Chemical and Drug Induced Liver Injury/metabolism ; Chemical and Drug Induced Liver Injury/etiology ; Oxidative Stress/drug effects ; Membrane Potential, Mitochondrial/drug effects
مستخلص: Isobavachalcone (IBC) is a flavonoid component of the traditional Chinese medicine Psoraleae Fructus, with a range of pharmacological properties. However, IBC causes some hepatotoxicity, and the mechanism of toxicity is unclear. The purpose of this paper was to investigate the possible mechanism of toxicity of IBC on HepG2 cells and zebrafish embryos. The results showed that exposure to IBC increased zebrafish embryo mortality and decreased hatchability. Meanwhile, IBC induced liver injury and increased expression of ALT and AST activity. Further studies showed that IBC caused the increase of ROS and MDA the decrease of CAT, GSH, and GSH-Px; the increase of Fe 2+ content; and the changes of ferroptosis related genes (acsl4, gpx4, and xct) and iron storage related genes (tf, fth, and fpn) in zebrafish embryos. Through in vitro verification, it was found that IBC also caused oxidative stress and increased Fe 2+ content in HepG2 cells. IBC caused depolarization of mitochondrial membrane potential (MMP) and reduction of mitochondrial ATP, as well as altered expression of ACSl4, SLC7A11, GPX4, and FTH1 proteins. Treatment of HepG2 cells with ferrostatin-1 could reverse the effect of IBC. Targeting the System Xc - -GSH-GPX4 pathway of ferroptosis and preventing oxidative stress damage might offer a theoretical foundation for practical therapy and prevention of IBC-induced hepatotoxicity.
(© 2024 John Wiley & Sons Ltd.)
References: Asrani, S. K., Devarbhavi, H., Eaton, J., & Kamath, P. S. (2019). Burden of liver diseases in the world. Journal of Hepatology, 70(1), 151–171. https://doi.org/10.1016/j.jhep.2018.09.014.
Bhalla, V. K., Nayak, U. R., & Dev, S. (1968). Some new flavonoids from psoralea corylifolia. Tetrahedron Letters, 9(20), 2401–2406. https://doi.org/10.1016/S0040-4039(00)76141-7.
Chen, H., Wang, F., Ni, X., Rigui, Y., Bai, Y., Xu, L., Yang, J., Zhang, X., Deng, J., Li, J., Yin, X., Ao, W., Kwok, K. W. H., & Dong, W. (2021). Aconitine disrupts serotonin neurotransmission via 5‐hydroxytryptamine receptor in zebrafish embryo. Journal of Applied Toxicology, 41(3), 483–492. https://doi.org/10.1002/jat.4059.
Dixon, S. J., Lemberg, K. M., Lamprecht, M. R., Skouta, R., Zaitsev, E. M., Gleason, C. E., Patel, D. N., Bauer, A. J., Cantley, A. M., Yang, W. S., & Stockwell, B. R. (2012). Ferroptosis: An iron‐dependent form of nonapoptotic cell death. Cell, 149(5), 1060–1072. https://doi.org/10.1016/j.cell.2012.03.042.
Driessen, M., Vitins, A. P., Pennings, J. L., Kienhuis, A. S., Water, B., & van der Ven, L. T. (2015). A transcriptomics‐based hepatotoxicity comparison between the zebrafish embryo and established human and rodent in vitro and in vivo models using cyclosporine A, amiodarone and acetaminophen. Toxicology Letters, 232(2), 403–412. https://doi.org/10.1016/j.toxlet.2014.11.020.
Dzoyem, J. P., Nkuete, A. H. L., Ngameni, B., & Eloff, J. N. (2017). Anti‐inflammatory and anticholinesterase activity of six flavonoids isolated from Polygonum and Dorstenia species. Archives of Pharmacal Research, 40(10), 1129–1134. https://doi.org/10.1007/s12272-015-0612-9.
Fang, X., Ardehali, H., Min, J., & Wang, F. (2023). The molecular and metabolic landscape of iron and ferroptosis in cardiovascular disease. Nature Reviews Cardiology, 20(1), 7–23. https://doi.org/10.1038/s41569-022-00735-4.
Fang, X., Cai, Z., Wang, H., Han, D., Cheng, Q., Zhang, P., Gao, F., Yu, Y., Song, Z., Wu, Q., An, P., Huang, S., Pan, J., Chen, H.‐Z., Chen, J., Linkermann, A., Min, J., & Wang, F. (2020). Loss of cardiac ferritin H facilitates cardiomyopathy via Slc7a11‐mediated ferroptosis. Circulation Research, 127(4), 486–501. https://doi.org/10.1161/CIRCRESAHA.120.316509.
Gao, D., Liu, F., Li, Z., & Guan, Y. (2019). Isobavachalcone attenuates Sephadex‐induced lung injury via activation of A20 and NRF2/HO‐1 in rats. European Journal of Pharmacology, 848, 49–54. https://doi.org/10.1016/j.ejphar.2019.01.034.
Gautheron, J., Gores, G. J., & Rodrigues, C. M. P. (2020). Lytic cell death in metabolic liver disease. Journal of Hepatology, 73(2), 394–408. https://doi.org/10.1016/j.jhep.2020.04.001.
Goessling, W., & Sadler, K. C. (2015). Zebrafish: An important tool for liver disease research. Gastroenterology, 149(6), 1361–1377. https://doi.org/10.1053/j.gastro.2015.08.034.
Green, D. R. (2019). The coming decade of cell death research: Five riddles. Cell, 177(5), 1094–1107. https://doi.org/10.1016/j.cell.2019.04.024.
Hassannia, B., Vandenabeele, P., & vanden Berghe, T. (2019). Targeting ferroptosis to iron out cancer. Cancer Cell, 35(6), 830–849. https://doi.org/10.1016/j.ccell.2019.04.002.
He, H., Wang, C., Liu, G., Ma, H., Jiang, M., Li, P., Lu, Q., Li, L., & Qi, H. (2021). Isobavachalcone inhibits acute myeloid leukemia: Potential role for ROS‐dependent mitochondrial apoptosis and differentiation. Phytotherapy Research, 35(6), 3337–3350. https://doi.org/10.1002/ptr.7054.
He, Z., Liao, W., Song, Q., Li, B., Liu, J., Xiong, Y., Song, C., & Yang, S. (2021). Role of ferroptosis induced by a high concentration of calcium oxalate in the formation and development of urolithiasis. International Journal of Molecular Medicine, 47(1), 289–301. https://doi.org/10.3892/ijmm.2020.4770.
Hill, A., Mesens, N., Steemans, M., Xu, J. J., & Aleo, M. D. (2012). Comparisons between in vitro whole cell imaging and in vivo zebrafish‐based approaches for identifying potential human hepatotoxicants earlier in pharmaceutical development. Drug Metabolism Reviews, 44(1), 127–140. https://doi.org/10.3109/03602532.2011.645578.
Jiang, X., Stockwell, B. R., & Conrad, M. (2021). Ferroptosis: Mechanisms, biology and role in disease. Nature Reviews Molecular Cell Biology, 22(4), 266–282. https://doi.org/10.1038/s41580-020-00324-8.
Jin, X., & Shi, Y. I. (2016). Isobavachalcone induces the apoptosis of gastric cancer cells via inhibition of the Akt and Erk pathways. Experimental and Therapeutic Medicine, 11(2), 403–408. https://doi.org/10.3892/etm.2015.2904.
Jo, S., Kim, H., Kim, S., Shin, D. H., & Kim, M. S. (2019). Characteristics of flavonoids as potent MERS‐CoV 3C‐like protease inhibitors. Chemical Biology and Drug Design, 94(6), 2023–2030. https://doi.org/10.1111/cbdd.13604.
Koppula, P., Zhang, Y., Zhuang, L., & Gan, B. (2018). Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Communications, 38(1), 12–13. https://doi.org/10.1186/s40880-018-0288-x.
Kuete, V., Mbaveng, A. T., Zeino, M., Fozing, C. D., Ngameni, B., Kapche, G. D., Ngadjui, B. T., & Efferth, T. (2015). Cytotoxicity of three naturally occurring flavonoid derived compounds (artocarpesin, cycloartocarpesin and isobavachalcone) towards multi‐factorial drug‐resistant cancer cells. Phytomedicine, 22(12), 1096–1102. https://doi.org/10.1016/j.phymed.2015.07.006.
Kuete, V., Ngameni, B., Mbaveng, A. T., Ngadjui, B., Meyer, J. J., & Lall, N. (2010). Evaluation of flavonoids from Dorstenia barteri for their antimycobacterial, antigonorrheal and anti‐reverse transcriptase activities. Acta Tropica, 116(1), 100–104. https://doi.org/10.1016/j.actatropica.2010.06.005.
Lee, H., Li, H., Kweon, M., Choi, Y., Kim, M. J., & Ryu, J. H. (2018). Isobavachalcone from Angelica keiskei inhibits adipogenesis and prevents lipid accumulation. International Journal of Molecular Sciences, 19(6), 1693. https://doi.org/10.3390/ijms19061693.
Lei, G., Zhang, Y., Koppula, P., Liu, X., Zhang, J., Lin, S. H., Ajani, J. A., Xiao, Q., Liao, Z., Wang, H., & Gan, B. (2020). The role of ferroptosis in ionizing radiation‐induced cell death and tumor suppression. Cell Research, 30(2), 146–162. https://doi.org/10.1038/s41422-019-0263-3.
Li, A., Gao, M., Zhao, N., Li, P., Zhu, J., & Li, W. (2019). Acute liver failure associated with Fructus Psoraleae: A case report and literature review. BMC Complementary Medicine and Therapies, 19(1), 84. https://doi.org/10.1186/s12906-019-2493-9.
Li, J., Liu, X., Ning, Q., Li, Y., Shi, Z., & Wei, Y. (2021). Effective identification of toxic related components of Psoraleae Fructus based on zebrafish model. Chinese Traditional and Herbal Drugs, 52(1), 129–136. https://kns.cnki.net/kcms/detail/12.1108.R.20201027.1747.004.html.
Li, Y., Qin, X., Li, P., Zhang, H., Lin, T., Miao, Z., & Ma, S. (2019). Isobavachalcone isolated from Psoralea corylifolia inhibits cell proliferation and induces apoptosis via inhibiting the AKT/GSK‐3beta/beta‐catenin pathway in colorectal cancer cells. Drug Design, Development and Therapy, 13, 1449–1460. https://doi.org/10.2147/DDDT.S192681.
Liu, J. J., Zhang, J., & Chen, Z. L. (2013). Chiral separation of bavachinin in Fructus Psoraleae and rat plasma by liquid chromatography using permethylated‐beta‐CD as a chiral selector. Journal of Pharmaceutical Analysis, 3(5), 349–353. https://doi.org/10.1016/j.jpha.2012.12.010.
Mao, C., Liu, X., Zhang, Y., Lei, G., Yan, Y., Lee, H., Koppula, P., Wu, S., Fang, B., Poyurovsky, M. V., Olswezki, K., & Gan, B. (2021). DHODH‐mediated ferroptosis defence is a targetable vulnerability in cancer. Nature, 593(7860), 586–590. https://doi.org/10.1038/s41586-021-03539-7.
Nair, A. B., & Jacob, S. (2016). A simple practice guide for dose conversion between animals and human. Journal of Basic and Clinical Pharmacy, 7(2), 27–31. https://doi.org/10.4103/0976-0105.177703.
Nikolova, G., Ananiev, J., Ivanov, V., Petkova‐Parlapanska, K., Georgieva, E., & Karamalakova, Y. (2022). The Azadirachta indica (neem) seed oil reduced chronic redox‐homeostasis imbalance in a mice experimental model on Ochratoxine A‐induced hepatotoxicity. Antioxidants, 11(9), 1678. https://doi.org/10.3390/antiox11091678.
Park, J. Y., Jeong, H. J., Kim, Y. M., Park, S. J., Rho, M. C., Park, K. H., Rho, M. C., Park, K. H., Ryu, Y. B., Lee, W. S., & Lee, W. S. (2011). Characteristic of alkylated chalcones from Angelica keiskei on influenza virus neuraminidase inhibition. Bioorganic & Medicinal Chemistry Letters, 21(18), 5602–5604. https://doi.org/10.1016/j.bmcl.2011.06.130.
Pavlik, L., Regev, A., Ardayfio, P. A., & Chalasani, N. P. (2019). Drug‐induced steatosis and steatohepatitis: The search for novel serum biomarkers among potential biomarkers for non‐alcoholic fatty liver disease and non‐alcoholic steatohepatitis. Drug Safety, 42(6), 701–711. https://doi.org/10.1007/s40264-018-00790-2.
Qin, Z., Wang, P., Duan, S., Wan, X., Xing, H., Yang, J., Zhang, X., Yao, Z., & Yao, X. (2021). Potential determinants for metabolic fates and inhibitory effects of isobavachalcone involving in human cytochrome P450, UDP‐glucuronosyltransferase enzymes, and efflux transporters. Journal of Pharmaceutical Sciences, 110(5), 2285–2294. https://doi.org/10.1016/j.xphs.2021.02.013.
Rochette, L., Dogon, G., Rigal, E., Zeller, M., Cottin, Y., & Vergely, C. (2022). Lipid peroxidation and iron metabolism: Two corner stones in the homeostasis control of ferroptosis. International Journal of Molecular Sciences, 24(1), 449. https://doi.org/10.3390/ijms24010449.
Scarpellini, C., Klejborowska, G., Lanthier, C., Hassannia, B., vanden Berghe, T., & Augustyns, K. (2023). Beyond ferrostatin‐1: A comprehensive review of ferroptosis inhibitors. Trends in Pharmacological Sciences, 44(12), 902–916. https://doi.org/10.1016/j.tips.2023.08.012.
Shi, G. Z., Song, D., Li, P. Y., Chen, S. S., Zhang, L., Li, S. S., Xiao, X.‐H., Qin, X.‐H., & Wang, J. B. (2021). Screening of hepatotoxic compounds in Psoralea corylifolia L., a traditional Chinese herbal and dietary supplement, using high‐resolution mass spectrometry and high‐content imaging. Biomedical Chromatography, 35(9), e5140. https://doi.org/10.1002/bmc.5140.
Shojaie, L., Iorga, A., & Dara, L. (2020). Cell death in liver diseases: A review. International Journal of Molecular Sciences, 21(24), 9682. https://doi.org/10.3390/ijms21249682.
State Pharmacopoeia Committee. (2020). The pharmacopoeia of People's Republic of China. Chemical Industry Press.
Tang, D., Chen, X., Kang, R., & Kroemer, G. (2021). Ferroptosis: Molecular mechanisms and health implications. Cell Research, 31(2), 107–125. https://doi.org/10.1038/s41422-020-00441-1.
Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P., & Kroemer, G. (2019). The molecular machinery of regulated cell death. Cell Research, 29(5), 347–364. https://doi.org/10.1038/s41422-019-0164-5.
Tong, X., Tang, R., Xiao, M., Xu, J., Wang, W., Zhang, B., Liu, J., Yu, X., & Shi, S. (2022). Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. Journal of Hematology & Oncology, 15(1), 174. https://doi.org/10.1186/s13045-022-01392-3.
Wang, H. M., Liu, T. X., Wang, T. Y., Wang, G., Liu, Y. G., Liu, S. G., Tang, Y.‐D., & Cai, X. H. (2018). Isobavachalcone inhibits post‐entry stages of the porcine reproductive and respiratory syndrome virus life cycle. Archives of Virology, 163(5), 1263–1270. https://doi.org/10.1007/s00705-018-3755-4.
Wang, H. M., Zhang, L., Liu, J., Yang, Z. L., Zhao, H. Y., Yang, Y., Shen, D., Lu, K., Fan, Z.‐C., Yao, Q.‐Y., Zhang, Y.‐M., Teng, Y.‐O., & Peng, Y. (2015). Synthesis and anti‐cancer activity evaluation of novel prenylated and geranylated chalcone natural products and their analogs. European Journal of Medicinal Chemistry, 92, 439–448. https://doi.org/10.1016/j.ejmech.2015.01.007.
Wang, M., Lin, L., Lu, J. J., & Chen, X. (2021). Pharmacological review of isobavachalcone, a naturally occurring chalcone. Pharmacological Research, 165, 105483. https://doi.org/10.1016/j.phrs.2021.105483.
Wang, X., Li, W., Zhang, H., Zhang, S., Song, S., Wang, Y., & Tang, J. (2020). Study on injury of Psoraleae and its main components on human normal hepatocytes L02. Traditional Chinese Medicinal Research, 33(4), 59–63.
Wei, L., Zuo, Z., Yang, Z., Yin, H., Yang, Y., Fang, J., Cui, H., Du, Z., Ouyang, P., Chen, X., Chen, J., Geng, Y., Zhu, Y., Chen, Z., Huang, C., Wamh, F., & Guo, H. (2022). Mitochondria damage and ferroptosis involved in Ni‐induced hepatotoxicity in mice. Toxicology, 466, 153068. https://doi.org/10.1016/j.tox.2021.153068.
Wu, D., Wang, W., Chen, W., Lian, F., Lang, L., Huang, Y., Xu, Y., Zhang, N., Chen, Y., Liu, M., Nussinov, R., Cheng, F., Lu, W., & Huang, J. (2018). Pharmacological inhibition of dihydroorotate dehydrogenase induces apoptosis and differentiation in acute myeloid leukemia cells. Haematologica, 103(9), 1472–1483. https://doi.org/10.3324/haematol.2018.188185.
Yuan, H., Li, X., Zhang, X., Kang, R., & Tang, D. (2016). Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochemical and Biophysical Research Communications, 478(3), 1338–1343. https://doi.org/10.1016/j.bbrc.2016.08.124.
Zhang, H. L., Hu, B. X., Li, Z. L., Du, T., Shan, J. L., Ye, Z. P., Peng, X.‐D., Li, X., Huang, Y., Zhu, X.‐Y., Chen, Y.‐H., Feng, G.‐K., Yang, D., Deng, R., & Zhu, X. F. (2022). PKCbetaII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nature Cell Biology, 24(1), 88–98. https://doi.org/10.1038/s41556-021-00818-3.
Zhao, L., Yu, Y., Li, L., Wang, J., Wang, J., Su, S., Ding, J., Zhang, Y., Wamg, A., & Zhou, K. (2022). Isobavachalcone disrupts mitochondrial respiration and induces cytotoxicity through ROS accumulation and Akt suppression. Toxicon, 216, 28–36. https://doi.org/10.1016/j.toxicon.2022.06.018.
Zhuang, C., Zhang, W., Sheng, C., Zhang, W., Xing, C., & Miao, Z. (2017). Chalcone: A privileged structure in medicinal chemistry. Chemical Reviews, (12), 7762–7810. https://doi.org/10.1021/acs.chemrev.7b00020.
معلومات مُعتمدة: 2060302 Key Project at Central Government Level; 2022YFD1801105 State Key Research and Development Plan, China; 2023KFKT001 Key Laboratory of Animal Disease Prevention Control and Food Safety of Sichuan Province
فهرسة مساهمة: Keywords: HepG2 cell; ferroptosis; hepatotoxicity; isobavachalcone; zebrafish embryo
المشرفين على المادة: 0 (Chalcones)
EC 1.11.1.12 (Phospholipid Hydroperoxide Glutathione Peroxidase)
20784-50-3 (isobavachalcone)
GAN16C9B8O (Glutathione)
تواريخ الأحداث: Date Created: 20240406 Date Completed: 20240715 Latest Revision: 20240715
رمز التحديث: 20240715
DOI: 10.1002/jat.4607
PMID: 38581191
قاعدة البيانات: MEDLINE
الوصف
تدمد:1099-1263
DOI:10.1002/jat.4607