Volume 12 Issue 4
Sep.  2022
Turn off MathJax
Article Contents
Qian Zhang, Piao Luo, Liuhai Zheng, Jiayun Chen, Junzhe Zhang, Huan Tang, Dandan Liu, Xueling He, Qiaoli Shi, Liwei Gu, Jiahao Li, Qiuyan Guo, Chuanbin Yang, Yin Kwan Wong, Fei Xia, Jigang Wang. 18beta-glycyrrhetinic acid induces ROS-mediated apoptosis to ameliorate hepatic fibrosis by targeting PRDX1/2 in activated HSCs[J]. Journal of Pharmaceutical Analysis, 2022, 12(4): 570-582. doi: 10.1016/j.jpha.2022.06.001
Citation: Qian Zhang, Piao Luo, Liuhai Zheng, Jiayun Chen, Junzhe Zhang, Huan Tang, Dandan Liu, Xueling He, Qiaoli Shi, Liwei Gu, Jiahao Li, Qiuyan Guo, Chuanbin Yang, Yin Kwan Wong, Fei Xia, Jigang Wang. 18beta-glycyrrhetinic acid induces ROS-mediated apoptosis to ameliorate hepatic fibrosis by targeting PRDX1/2 in activated HSCs[J]. Journal of Pharmaceutical Analysis, 2022, 12(4): 570-582. doi: 10.1016/j.jpha.2022.06.001

18beta-glycyrrhetinic acid induces ROS-mediated apoptosis to ameliorate hepatic fibrosis by targeting PRDX1/2 in activated HSCs

doi: 10.1016/j.jpha.2022.06.001
Funds:

The authors gratefully acknowledge the financial support from the Innovation Team and Talents Cultivation Program of the National Administration of Traditional Chinese Medicine, China (Grant No.: ZYYCXTD-C-202002), the National Key Research and Development Program of China, China (Grant No.: 2020YFA0908000), the National Natural Science Foundation of China, China (Grant Nos.: 81803389, 81903588, 32101219, 81702580, 82074098, 81903866, and 81803456), and the Fundamental Research Funds for the Central Public Welfare Research Institutes, China (Grant Nos.: ZZ14-YQ-050, ZZ14-YQ-059, ZZ15-ND-10, ZZ15-YQ-063, ZZ14-ND-010, and ZZ14-FL-002).

  • Received Date: Dec. 15, 2021
  • Accepted Date: Jun. 01, 2022
  • Rev Recd Date: May 31, 2022
  • Publish Date: Jun. 08, 2022
  • Hepatic stellate cells (HSCs) are essential drivers of fibrogenesis. Inducing activated-HSC apoptosis is a promising strategy for treating hepatic fibrosis. 18beta-glycyrrhetinic acid (18β-GA) is a natural compound that exists widely in herbal medicines, such as Glycyrrhiza uralensis Fisch, which is used for treating multiple liver diseases, especially in Asia. In the present study, we demonstrated that 18β-GA decreased hepatic fibrosis by inducing the apoptosis in activated HSCs. 18β-GA inhibited the expression of α-smooth muscle actin and collagen type I alpha-1. Using a chemoproteomic approach derived from activity-based protein profiling, together with cellular thermal shift assay and surface plasmon resonance, we found that 18β-GA covalently targeted peroxiredoxin 1 (PRDX1) and peroxiredoxin 2 (PRDX2) proteins via binding to active cysteine residues and thereby inhibited their enzymatic activities. 18β-GA induced the elevation of reactive oxygen species (ROS), resulting in the apoptosis of activated HSCs. PRDX1 knockdown also led to ROS-mediated apoptosis in activated HSCs. Collectively, our findings revealed the target proteins and molecular mechanisms of 18β-GA in ameliorating hepatic fibrosis, highlighting the future development of 18β-GA as a novel therapeutic drug for hepatic fibrosis.
  • loading
  • M.M. Aydin, K.C. Akcali, Liver fibrosis, Turk. J. Gastroenterol. 29 (2018) 14-21
    P. Ginès, A. Krag, J.G. Abraldes, et al., Liver cirrhosis, Lancet 398 (2021) 1359-1376
    E. Seki, D.A. Brenner, Recent advancement of molecular mechanisms of liver fibrosis, J. Hepatobiliary Pancreat. Sci. 22 (2015) 512-518
    T. Higashi, S.L. Friedman, Y. Hoshida, Hepatic stellate cells as key target in liver fibrosis, Adv. Drug Deliv. Rev. 121 (2017) 27-42
    T. Tsuchida, S.L. Friedman, Mechanisms of hepatic stellate cell activation, Nat. Rev. Gastroenterol. Hepatol. 14 (2017) 397-411
    Y. Jia, F. Wang, Q. Guo, et al., Curcumol induces RIPK1/RIPK3 complex-dependent necroptosis via JNK1/2-ROS signaling in hepatic stellate cells, Redox Biol. 19 (2018) 375-387
    Z. Zhang, Z. Yao, S. Zhao, et al., Interaction between autophagy and senescence is required for dihydroartemisinin to alleviate liver fibrosis, Cell Death Dis. 8 (2017), e2886
    S. Bi, F. Chu, M. Wang, et al., Ligustrazine-oleanolic acid glycine derivative, G-TOA, selectively inhibited the proliferation and induced apoptosis of activated HSC-T6 cells, Molecules 21 (2016), 1599
    B.N. Whitley, E.A. Engelhart, S. Hoppins, Mitochondrial dynamics and their potential as a therapeutic target, Mitochondrion 49 (2019) 269-283
    G. Lenaz, The mitochondrial production of reactive oxygen species:mechanisms and implications in human pathology, IUBMB Life 52 (2001) 159-164
    T. Luangmonkong, S. Suriguga, H.A.M. Mutsaers, et al., Targeting oxidative stress for the treatment of liver fibrosis, Rev. Physiol. Biochem. Pharmacol. 175 (2018) 71-102
    S. Liang, T. Kisseleva, D.A. Brenner, The role of NADPH oxidases (NOXs) in liver fibrosis and the activation of myofibroblasts, Front. Physiol. 7 (2016), 17
    H.J. Lin, C.P. Tseng, C.F. Lin, et al., A Chinese herbal decoction, modified Yi Guan Jian, induces apoptosis in hepatic stellate cells through an ROS-mediated mitochondrial/caspase pathway, Evid. Based Complement Alternat. Med. 2011 (2011), 459531
    A. Marti-Rodrigo, F. Alegre, Á.B. Moragrega, et al., Rilpivirine attenuates liver fibrosis through selective STAT1-mediated apoptosis in hepatic stellate cells, Gut 69 (2020) 920-932
    H. Zheng, X. Wang, Y. Zhang, et al., Pien-Tze-Huang ameliorates hepatic fibrosis via suppressing NF-κB pathway and promoting HSC apoptosis, J. Ethnopharmacol. 244 (2019), 111856
    D. Meng, Z. Li, G. Wang, et al., Carvedilol attenuates liver fibrosis by suppressing autophagy and promoting apoptosis in hepatic stellate cells, Biomed. Pharmacother. 108 (2018) 1617-1627
    C. Bailly, G. Vergoten, Glycyrrhizin:An alternative drug for the treatment of COVID-19 infection and the associated respiratory syndrome? Pharmacol. Ther. 214 (2020), 107618
    L. Wang, R. Yang, B. Yuan, et al., The antiviral and antimicrobial activities of licorice, a widely-used Chinese herb, Acta Pharm. Sin. B 5 (2015) 310-315
    Y.-H. Luo, C. Wang, W.-T. Xu, et al., 18β-glycyrrhetinic acid has anti-cancer effects via inducing apoptosis and G2/M cell cycle arrest, and inhibiting migration of A549 lung cancer cells, Onco. Targets Ther. 14 (2021) 5131-5144
    A. Roohbakhsh, M. Iranshahy, M. Iranshahi, Glycyrrhetinic acid and its derivatives:anti-cancer and cancer chemopreventive properties, mechanisms of action and structure-cytotoxic activity relationship, Curr. Med. Chem. 23 (2016) 498-517
    X. Tian, Y. Liu, X. Liu, et al., Glycyrrhizic acid ammonium salt alleviates Concanavalin A-induced immunological liver injury in mice through the regulation of the balance of immune cells and the inhibition of hepatocyte apoptosis, Biomed. Pharmacother. 120 (2019), 109481
    X. Huo, X. Meng, J. Zhang, et al., Hepatoprotective effect of different combinations of 18α-and 18β-Glycyrrhizic acid against CCl4-induced liver injury in rats, Biomed. Pharmacother. 122 (2020), 109354
    J.-Y. Li, H.-Y. Cao, P. Liu, Glycyrrhizic acid in the treatment of liver diseases:literature review, Biomed. Res. Int. 2014 (2014), 872139
    C.G. Tag, S. Sauer-Lehnen, S. Weiskirchen, et al., Bile duct ligation in mice:induction of inflammatory liver injury and fibrosis by obstructive cholestasis, J. Vis. Exp. (2015), 52438
    N. Liu, J. Feng, X. Lu, et al., Isorhamnetin inhibits liver fibrosis by reducing autophagy and inhibiting extracellular matrix formation via the TGF-β1/Smad3 and TGF-β1/p38 MAPK Pathways, Mediators Inflamm. 2019 (2019), 6175091
    Y. Sun, Y. Qiao, Y. Liu, et al., ent-Kaurane diterpenoids induce apoptosis and ferroptosis through targeting redox resetting to overcome cisplatin resistance, Redox Biol. 43 (2021), 101977
    X. Chen, W. Li, C. Xu, et al., Comparative profiling of analog targets:a case study on resveratrol for mouse melanoma metastasis suppression, Theranostics 8 (2018) 3504-3516
    P. Luo, D. Liu, Q. Zhang, et al., Celastrol induces ferroptosis in activated HSCs to ameliorate hepatic fibrosis via targeting peroxiredoxins and HO-1, Acta Pharm. Sin. B 2021https://doi.org/10.1016/j.apsb.2021.12.007
    Y.T. Lim, N. Prabhu, L. Dai, et al., An efficient proteome-wide strategy for discovery and characterization of cellular nucleotide-protein interactions, PLoS One 13 (2018), e0208273
    Y. Huang, S.-H. Yu, W.-X. Zhen, et al., Tanshinone I, a new EZH2 inhibitor restricts normal and malignant hematopoiesis through upregulation of MMP9 and ABCG2, Theranostics 11 (2021) 6891-6904
    C.-X. Liu, Q.-Q. Yin, H.-C. Zhou, et al., Adenanthin targets peroxiredoxin I and II to induce differentiation of leukemic cells, Nat. Chem. Biol. 8 (2012) 486-493
    X. Chen, Y. Zhao, W. Luo, et al., Celastrol induces ROS-mediated apoptosis via directly targeting peroxiredoxin-2 in gastric cancer cells, Theranostics 10 (2020) 10290-10308
    D. Martinez Molina, R. Jafari, M. Ignatushchenko, et al., Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay, Science 341 (2013) 84-87
    S.G. Rhee, S.W. Kang, T.S. Chang, et al., Peroxiredoxin, a novel family of peroxidases, IUBMB Life 52 (2001) 35-41
    S.G. Rhee, H.Z. Chae, K. Kim, Peroxiredoxins:a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling, Free Radic. Biol. Med. 38 (2005) 1543-1552
    S.K. Asrani, H. Devarbhavi, J. Eaton, et al., Burden of liver diseases in the world, J. Hepatol. 70 (2019) 151-171
    F.-S. Wang, J.-G. Fan, Z. Zhang, et al., The global burden of liver disease:the major impact of China, Hepatology 60 (2014) 2099-2108
    C. Peng, A.G. Stewart, O.L. Woodman, et al., Non-alcoholic steatohepatitis:a review of its mechanism, models and medical treatments, Front. Pharmacol. 11 (2020), 603926
    R.J. Chen, H.H. Wu, Y.J. Wang, Strategies to prevent and reverse liver fibrosis in humans and laboratory animals, Arch. Toxicol. 89 (2015) 1727-1750
    X. Ma, Y. Jiang, J. Wen, et al., A comprehensive review of natural products to fight liver fibrosis:Alkaloids, terpenoids, glycosides, coumarins and other compounds, Eur. J. Pharmacol. 888 (2020), 173578
    L. Shan, Z. Liu, L. Ci, et al., Research progress on the anti-hepatic fibrosis action and mechanism of natural products, Int. Immunopharmacol. 75 (2019), 105765
    K.K. Wai, Y. Liang, L. Zhou, et al., The protective effects of Acanthus ilicifolius alkaloid A and its derivatives on pro- and anti-inflammatory cytokines in rats with hepatic fibrosis, Biotechnol. Appl. Biochem. 62 (2015) 537-546
    H. Wang, Y. Zhang, R. Bai, et al., Baicalin attenuates alcoholic liver injury through modulation of hepatic oxidative stress, inflammation and sonic hedgehog pathway in rats, Cell Physiol. Biochem. 39 (2016) 1129-1140
    S.Y. Wu, W.J. Wang, J.H. Dou, et al., Research progress on the protective effects of licorice-derived 18β-glycyrrhetinic acid against liver injury, Acta Pharmacol. Sin. 42 (2021) 18-26
    X. Li, R. Sun, R. Liu, Natural products in licorice for the therapy of liver diseases:Progress and future opportunities, Pharmacol. Res. 144 (2019) 210-226
    J. Wang, X.F. Tan, V.S. Nguyen, et al., A quantitative chemical proteomics approach to profile the specific cellular targets of andrographolide, a promising anticancer agent that suppresses tumor metastasis, Mol. Cell. Proteomics 13 (2014) 876-886
    J. Zhang, X. Sun, L. Wang, et al., Artesunate-induced mitophagy alters cellular redox status, Redox Biol. 19 (2018) 263-273
    J. Wang, C.J. Zhang, W.N. Chia, et al., Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum, Nat. Commun. 6 (2015), 10111
    J. Wang, J. Zhang, C.J. Zhang, et al., In situ proteomic profiling of curcumin targets in HCT116 colon cancer cell line, Sci. Rep. 6 (2016), 22146
    B. Knoops, V. Argyropoulou, S. Becker, et al., Multiple roles of peroxiredoxins in inflammation, Mol. Cells 39 (2016) 60-64
    C. Ding, X. Fan, G. Wu, Peroxiredoxin 1-an antioxidant enzyme in cancer, J. Cell Mol. Med. 21 (2017) 193-202
    S. Wang, Z. Chen, S. Zhu, et al., PRDX2 protects against oxidative stress induced by H. pylori and promotes resistance to cisplatin in gastric cancer, Redox Biol. 28 (2020), 101319
    Z. Zou, H. Chang, H. Li, et al., Induction of reactive oxygen species:an emerging approach for cancer therapy, Apoptosis 22 (2017) 1321-1335
    D.B. Zorov, M. Juhaszova, S.J. Sollott, Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release, Physiol. Rev. 94 (2014) 909-950
    K. Pant, A.K. Yadav, P. Gupta, et al., Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells, Redox Biol. 12 (2017) 340-349
    Y. Lu, J. Liu, C. Lin, et al., Peroxiredoxin 2:a potential biomarker for early diagnosis of hepatitis B virus related liver fibrosis identified by proteomic analysis of the plasma, BMC Gastroenterol. 10 (2010), 115
    J. Zhang, X. Guo, T. Hamada, et al., Protective Effects of peroxiredoxin 4 (PRDX4) on cholestatic liver injury, Int. J. Mol. Sci. 19 (2018), 2509
    M.H. Kim, J.B. Seong, J.W. Huh, et al., Peroxiredoxin 5 ameliorates obesity-induced non-alcoholic fatty liver disease through the regulation of oxidative stress and AMP-activated protein kinase signaling, Redox Biol. 28 (2020), 101315
    B.L. Hopkins, M. Nadler, J.J. Skoko, et al., A peroxidase peroxiredoxin 1-specific redox regulation of the novel FOXO3 microRNA target let-7, Antioxid. Redox Signal. 28 (2018) 62-77
    Z.A. Wood, L.B. Poole, P.A. Karplus, Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling, Science 300 (2003) 650-653
    Q. Zhao, Y. Ding, Z. Deng, et al., Natural products triptolide, celastrol, and withaferin A inhibit the chaperone activity of peroxiredoxin I, Chem. Sci. 6 (2015) 4124-4130
    Y. Zhang, C. Sun, G. Xiao, et al., S-nitrosylation of the Peroxiredoxin-2 promotes S-nitrosoglutathione-mediated lung cancer cells apoptosis via AMPK-SIRT1 pathway, Cell Death Dis. 10 (2019), 329
    B. Turner-Ivey, Y. Manevich, J. Schulte, et al., Role for Prdx1 as a specific sensor in redox-regulated senescence in breast cancer, Oncogene 32 (2013) 5302-5314
    N.M. Mazure, VDAC in cancer, Biochim. Biophys. Acta Bioenerg. 1858 (2017) 665-673
    V. Shoshan-Barmatz, V. De Pinto, M. Zweckstetter, et al., VDAC, a multi-functional mitochondrial protein regulating cell life and death, Mol. Aspects Med. 31 (2010) 227-285
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (586) PDF downloads(46) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return