Volume 13 Issue 11
Nov.  2023
Turn off MathJax
Article Contents
Hansong Bai, Jiahua Lyu, Xinyu Nie, Hao Kuang, Long Liang, Hongyuan Jia, Shijie Zhou, Churong Li, Tao Li. Ginsenoside Rg5 enhances the radiosensitivity of lung adenocarcinoma via reducing HSP90-CDC37 interaction and promoting client protein degradation[J]. Journal of Pharmaceutical Analysis, 2023, 13(11): 1296-1308. doi: 10.1016/j.jpha.2023.06.004
Citation: Hansong Bai, Jiahua Lyu, Xinyu Nie, Hao Kuang, Long Liang, Hongyuan Jia, Shijie Zhou, Churong Li, Tao Li. Ginsenoside Rg5 enhances the radiosensitivity of lung adenocarcinoma via reducing HSP90-CDC37 interaction and promoting client protein degradation[J]. Journal of Pharmaceutical Analysis, 2023, 13(11): 1296-1308. doi: 10.1016/j.jpha.2023.06.004

Ginsenoside Rg5 enhances the radiosensitivity of lung adenocarcinoma via reducing HSP90-CDC37 interaction and promoting client protein degradation

doi: 10.1016/j.jpha.2023.06.004
Funds:

This work was supported by grants from the Project of Sichuan Science and Technology Department, China (Grant No.: 2021YJ0010).

  • Received Date: Mar. 15, 2023
  • Accepted Date: Jun. 05, 2023
  • Rev Recd Date: Jun. 02, 2023
  • Publish Date: Jun. 07, 2023
  • Ginsenoside Rg5 is a rare ginsenoside showing promising tumor-suppressive effects. This study aimed to explore its radio-sensitizing effects and the underlying mechanisms. Human lung adenocarcinoma cell lines A549 and Calu-3 were used for in vitro and in vivo analysis. Bioinformatic molecular docking prediction and following validation by surface plasmon resonance (SPR) technology, cellular thermal shift assay (CETSA), and isothermal titration calorimetry (ITC) were conducted to explore the binding between ginsenoside Rg5 and 90 kD heat shock protein alpha (HSP90α). The effects of ginsenoside Rg5 on HSP90-cell division cycle 37 (CDC37) interaction, the client protein stability, and the downstream regulations were further explored. Results showed that ginsenoside Rg5 could induce cell-cycle arrest at the G1 phase and enhance irradiation-induced cell apoptosis. It could bind to HSP90α with a high affinity, but the affinity was drastically decreased by HSP90α Y61A mutation. Co-immunoprecipitation (Co-IP) and ITC assays confirmed that ginsenoside Rg5 disrupts the HSP90-CDC37 interaction in a dose-dependent manner. It reduced irradiation-induced upregulation of the HSP90-CDC37 client proteins, including SRC, CDK4, RAF1, and ULK1 in A549 cell-derived xenograft (CDX) tumors. Ginsenoside Rg5 or MRT67307 (an IKKε/TBK1 inhibitor) pretreatment suppressed irradiation-induced elevation of the LC3-II/β ratio and restored irradiation-induced downregulation of p62 expression. In A549 CDX tumors, ginsenoside Rg5 treatment suppressed LC3 expression and enhanced irradiation-induced DNA damage. In conclusion, ginsenoside Rg5 may be a potential radiosensitizer for lung adenocarcinoma. It interacts with HSP90α and reduces the binding between HSP90 and CDC37, thereby increasing the ubiquitin-mediated proteasomal degradation of the HSP90-CDC37 client proteins.
  • loading
  • A.G. Nicholson, M.S. Tsao, M.B. Beasley, et al., The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015, J. Thorac. Oncol. 17 (2022) 362-387.
    S.K. Vinod, E. Hau, Radiotherapy treatment for lung cancer: Current status and future directions, Respirology 25 Suppl 2 (2020) 61-71.
    G.P. Delaney, M.B. Barton, Evidence-based estimates of the demand for radiotherapy, Clin. Oncol. 27 (2015) 70-76.
    J. Shafiq, T.P. Hanna, S.K. Vinod, et al., A Population-based Model of Local Control and Survival Benefit of Radiotherapy for Lung Cancer, Clin. Oncol. 28 (2016) 627-638.
    M. Krause, A. Dubrovska, A. Linge, et al., Cancer stem cells: Radioresistance, prediction of radiotherapy outcome and specific targets for combined treatments, Adv. Drug Deliv. Rev. 109 (2017) 63-73.
    S. Rey, L. Schito, M. Koritzinsky, et al., Molecular targeting of hypoxia in radiotherapy, Adv. Drug Deliv. Rev. 109 (2017) 45-62.
    H. Wang, X. Mu, H. He, et al., Cancer Radiosensitizers, Trends Pharmacol. Sci. 39 (2018) 24-48.
    Y. Chen, Q. Liu, P. An, et al., Ginsenoside Rd: A promising natural neuroprotective agent, Phytomedicine Int. J. Phytother. Phytopharm. 95 (2022), 153883.
    Y. Yang, Z. Ju, Y. Yang, et al., Phytochemical analysis of Panax species: A review, J. Ginseng Res. 45 (2021) 1-21.
    S. Yoo, B.I. Park, D.H. Kim, et al., Ginsenoside absorption rate and extent enhancement of black ginseng (CJ EnerG) over red ginseng in healthy adults, Pharmaceutics 13 (2021), 487.
    H. Kim, P. Choi, T. Kim, et al., Ginsenosides Rk1 and Rg5 inhibit transforming growth factor-β1-induced epithelial-mesenchymal transition and suppress migration, invasion, anoikis resistance, and development of stem-like features in lung cancer, J. Ginseng Res. 45 (2021) 134-148.
    X. Yang, G. Wang, J. You, et al., High expression of cancer-IgG is associated with poor prognosis and radioresistance via PI3K/AKT/DNA-PKcs pathway regulation in lung adenocarcinoma, Front. Oncol. 11 (2021), 675397.
    A. Tsolou, M. Liousia, D. Kalamida, et al., Inhibition of IKK-NFkappaB pathway sensitizes lung cancer cell lines to radiation, Cancer Biol. Med. 14 (2017) 293-301.
    K. Park, A.E. Cho, Using reverse docking to identify potential targets for ginsenosides, Journal of Ginseng Research 41 (2017) 534-539.
    A.E. Kabakov, V.A. Kudryavtsev, V.L. Gabai, Hsp90 inhibitors as promising agents for radiotherapy, J. Mol. Med. 88 (2010) 241-247.
    E. Amatya, B.S.J. Blagg, Recent advances toward the development of Hsp90 C-terminal inhibitors, Bioorg. Med. Chem. Lett. 80 (2023), 129111.
    Q. Wang, Y. Chen, H. Chang, et al., The role and mechanism of ATM-mediated autophagy in the transition from hyper-radiosensitivity to induced radioresistance in lung cancer under low-dose radiation, Front. Cell Dev. Biol. 9 (2021), 650819.
    J. Elegheert, E. Behiels, B. Bishop, et al., Lentiviral transduction of mammalian cells for fast, scalable and high-level production of soluble and membrane proteins, Nat. Protoc. 13 (2018) 2991-3017.
    N.A.P. Franken, H.M. Rodermond, J. Stap, et al., Clonogenic assay of cells in vitro, Nat. Protoc. 1 (2006) 2315-2319.
    L. Bodgi, N. Foray, The nucleo-shuttling of the ATM protein as a basis for a novel theory of radiation response: Resolution of the linear-quadratic model, Int. J. Radiat. Biol. 92 (2016) 117-131.
    I. Lakshmanan, S.K. Batra, Protocol for apoptosis assay by flow cytometry using annexin V staining method, Bio. Protoc. 3 (2013), e374.
    Q. Song, J. Wen, W. Li, et al., HSP90 promotes radioresistance of cervical cancer cells via reducing FBXO6 mediated CD147 polyubiquitination, Cancer. Sci. 113 (2022) 1463-1474.
    A. Daina, O. Michielin, V. Zoete, SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules, Nucleic Acids Res. 47 (2019) W357-W364.
    Z. Yao, J. Dong, Y. Che, et al., TargetNet: A web service for predicting potential drug-target interaction profiling via multi-target SAR models, J. Comput. Aided Mol. Des. 30 (2016) 413-424.
    L. Wright, X. Barril, B. Dymock, et al., Structure-activity relationships in purine-based inhibitor binding to HSP90 isoforms, Chem. Biol. 11 (2004) 775-785.
    Y. Liu, M. Grimm, W. Dai, et al., CB-Dock: A web server for cavity detection-guided protein-ligand blind docking, Acta Pharmacol. Sin. 41 (2020) 138-144.
    C. Zhou, C. Zhang, H. Zhu, et al., Allosteric regulation of Hsp90α’s activity by small molecules targeting the middle domain of the chaperone, iScience 23 (2020), 100857.
    D. Raghu, P. Hamill, A. Banaji, et al., Assessment of the binding interactions of SARS-CoV-2 spike glycoprotein variants, J. Pharm. Anal. 12 (2022) 58-64.
    L. Wang, L. Zhang, L. Li, et al., Small-molecule inhibitor targeting the Hsp90-Cdc37 protein-protein interaction in colorectal cancer, Sci. Adv. 5 (2019), eaax2277.
    T. Li, H. Jiang, Y. Tong, et al., Targeting the Hsp90-Cdc37-client protein interaction to disrupt Hsp90 chaperone machinery, J. Hematol. Oncol. 11 (2018), 59.
    R.C. Russell, Y. Tian, H. Yuan, et al., ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase, Nat. Cell Biol. 15 (2013) 741-750.
    W. Wu, X. Wang, N. Berleth, et al., The autophagy-initiating kinase ULK1 controls RIPK1-mediated cell death, Cell Rep. 31 (2020), 107547.
    X. Chen, P. Wang, F. Guo, et al., Autophagy enhanced the radioresistance of non-small cell lung cancer by regulating ROS level under hypoxia condition, Int. J. Radiat. Biol. 93 (2017) 764-770.
    H. Chaachouay, P. Ohneseit, M. Toulany, et al., Autophagy contributes to resistance of tumor cells to ionizing radiation, Radiother. Oncol. 99 (2011) 287-292.
    S. Nisar, T. Masoodi, K.S. Prabhu, et al., Natural products as chemo-radiation therapy sensitizers in cancers, Biomed. Pharmacother. 154 (2022), 113610.
    Q. Zhang, F. Wang, K. Jia, et al., Natural product interventions for chemotherapy and radiotherapy-induced side effects, Front. Pharmacol. 9 (2018), 1253.
    L. Yang, X. Zhang, K. Li, et al., Protopanaxadiol inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by targeting STAT3 pathway, Cell Death Dis. 10 (2019) 630.
    Y. Liu, D. Fan, The preparation of ginsenoside Rg5, its antitumor activity against breast cancer cells and its targeting of PI3K, Nutrients 12 (2020), 246.
    Z. Niu, W. Zhang, J. Shi, et al., Effect of silencing C-erbB-2 on esophageal carcinoma cell biological behaviors by inhibiting IGF-1 pathway activation, J. Cardiothorac. Surg. 16 (2021), 194.
    M.A. Serwetnyk, B.S.J. Blagg, The disruption of protein-protein interactions with co-chaperones and client substrates as a strategy towards Hsp90 inhibition, Acta Pharm. Sin. B. 11 (2021) 1446-1468.
    T.T. Koll, S.S. Feis, M.H. Wright, et al., HSP90 inhibitor, DMAG, synergizes with radiation of lung cancer cells by interfering with base excision and ATM-mediated DNA repair, Mol. Cancer Ther. 7 (2008) 1985-1992.
    Y. Wang, H. Liu, L. Diao, et al., Hsp90 inhibitor ganetespib sensitizes non-small cell lung cancer to radiation but has variable effects with chemoradiation, Clin. Cancer Res. 22 (2016) 5876-5886.
    M. Provencio, A. Sanchez, P. Garrido, et al., New molecular targeted therapies integrated with radiation therapy in lung cancer, Clin. Lung Cancer 11 (2010) 91-97.
    L. Li, L. Wang, Q. You, et al., Heat Shock Protein 90 Inhibitors: An update on achievements, challenges, and future directions, J. Med. Chem. 63 (2020) 1798-1822.
    Y. Xiao, Y. Liu, Recent advances in the discovery of novel HSP90 inhibitors: An update from 2014, Curr. Drug Targets 21 (2020) 302-317.
    A. Wu, B. Wu, J. Guo, et al., Elevated expression of CDK4 in lung cancer, J. Transl. Med. 9 (2011), 38.
    S. Goel, J.S. Bergholz, J. Zhao, Targeting CDK4 and CDK6 in cancer, Nat. Rev. Cancer 22 (2022) 356-372.
    S. Biade, C.C. Stobbe, J.D. Chapman, The intrinsic radiosensitivity of some human tumor cells throughout their cell cycles, Radiat. Res. 147 (1997) 416-421.
    S.Y. Tam, V.W. Wu, H.K. Law, Influence of autophagy on the efficacy of radiotherapy, Radiat. Oncol. 12 (2017), 57.
    J. Gao, F. Lu, J. Yan, et al., The role of radiotherapy-related autophagy genes in the prognosis and immune infiltration in lung adenocarcinoma, Front. Immunol. 13 (2022), 992626.
    M. Dai, C. Zhang, A. Ali, et al., CDK4 regulates cancer stemness and is a novel therapeutic target for triple-negative breast cancer, Sci. Rep. 6 (2016), 35383.
    M.A. Ortiz, T. Mikhailova, X. Li, et al., Src family kinases, adaptor proteins and the actin cytoskeleton in epithelial-to-mesenchymal transition, Cell Commun. Signal. 19 (2021), 67.
    S. Gao, H. Kushida, T. Makino, Ginsenosides, ingredients of the root of Panax ginseng, are not substrates but inhibitors of sodium-glucose transporter 1, J. Nat. Med. 71 (2017) 131-138.
    X. Wang, W. Zheng, Q. Shen, et al., Identification and construction of a novel biomimetic delivery system of paclitaxel and its targeting therapy for cancer, Signal. Transduct. Target. Ther. 6 (2021), 33.
    C. Hong, D. Wang, J. Liang, et al., Novel ginsenoside-based multifunctional liposomal delivery system for combination therapy of gastric cancer, Theranostics 9 (2019) 4437-4449.
    Y.W. Koh, S. Lee, S.Y. Park, Differential expression and prognostic significance of GLUT1 according to histologic type of non-small-cell lung cancer and its association with volume-dependent parameters, Lung Cancer 104 (2017) 31-37.
    Y. Weng, X. Fan, Y. Bai, et al., SLC2A5 promotes lung adenocarcinoma cell growth and metastasis by enhancing fructose utilization, Cell Death Discov. 4 (2018), 38.
    Y. Zhu, J. Liang, C. Gao, et al., Multifunctional ginsenoside Rg3-based liposomes for glioma targeting therapy, J. Control. Release 330 (2021) 641-657.
    M. Wang, Y. Xu, J. Xie, et al., Ginsenoside as a new stabilizer enhances the transfection efficiency and biocompatibility of cationic liposome, Biomater. Sci. 9 (2021) 8373-8385.
    J. Xia, S. Ma, X. Zhu, et al., Versatile ginsenoside Rg3 liposomes inhibit tumor metastasis by capturing circulating tumor cells and destroying metastatic niches, Sci. Adv. 8 (2022), eabj1262.
  • 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 (270) PDF downloads(24) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return