Volume 13 Issue 8
Aug.  2023
Turn off MathJax
Article Contents
Qiuyan Guo, Jiangpeng Wu, Qixin Wang, Yuwen Huang, Lin Chen, Jie Gong, Maobo Du, Guangqing Cheng, Tianming Lu, Minghong Zhao, Yuan Zhao, Chong Qiu, Fei Xia, Junzhe Zhang, Jiayun Chen, Feng Qiu, Jigang Wang. Single-cell transcriptome analysis uncovers underlying mechanisms of acute liver injury induced by tripterygium glycosides tablet in mice[J]. Journal of Pharmaceutical Analysis, 2023, 13(8): 908-925. doi: 10.1016/j.jpha.2023.03.004
Citation: Qiuyan Guo, Jiangpeng Wu, Qixin Wang, Yuwen Huang, Lin Chen, Jie Gong, Maobo Du, Guangqing Cheng, Tianming Lu, Minghong Zhao, Yuan Zhao, Chong Qiu, Fei Xia, Junzhe Zhang, Jiayun Chen, Feng Qiu, Jigang Wang. Single-cell transcriptome analysis uncovers underlying mechanisms of acute liver injury induced by tripterygium glycosides tablet in mice[J]. Journal of Pharmaceutical Analysis, 2023, 13(8): 908-925. doi: 10.1016/j.jpha.2023.03.004

Single-cell transcriptome analysis uncovers underlying mechanisms of acute liver injury induced by tripterygium glycosides tablet in mice

doi: 10.1016/j.jpha.2023.03.004
Funds:

This work is supported by the National Key Research and Development Program of China (Grant Nos.: 2020YFA0908000, and 2022YFC2303600), the Establishment of Sino-Austria “Belt and Road” Joint Laboratory on Traditional Chinese Medicine for Severe Infectious Diseases and Joint Research (Grant No.: 2020YFE0205100), the National Natural Science Foundation of China (Grant Nos.: 82104480, 82004248, 82141001, 82274182, 82074098, and 82173914), the Fundamental Research Funds for the Central public welfare research institutes (Grant Nos.: ZZ14-YQ-055, ZZ14-YQ-059, ZZ14-YQ-060, ZXKT19018, ZXKT19021, ZXKT19022, ZZ14-YQ-050, ZZ14-YQ-051, ZZ14-YQ-052, ZZ14-FL-002, ZZ14-ND-010, ZZ15-ND-10, and ZZ16-ND-10-19), the Beijing Municipal Natural Science Foundation (Grant No.: 7214287), the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (Grant No.: ZYYCXTD-C-202002), the Young Elite Scientists Sponsorship Program by CACM (Grant No.: 2021QNRC2B29), the CACMS Innovation Fund (Grant Nos.: CI2021A05101, and CI2021A05104), the Scientific and Technological Innovation Project of China Academy of Chinese Medical Sciences (Grant No.: CI2021B014), the Science and Technology Foundation of Shenzhen (Grant No.: JCYJ20210324115800001), the Science and Technology Foundation of Shenzhen (Shenzhen Clinical Medical Research Center for Geriatric Diseases), Shenzhen Governmental Sustainable Development Fund (Grant No.: KCXFZ20201221173612034), Shenzhen key Laboratory of Kidney Diseases (Grant No.: ZDSYS201504301616234), Shenzhen Fund for Guangdong Provincial High-level Clinical Key Specialties (Grant No.: SZGSP001), the Distinguished Expert Project of Sichuan Province Tianfu Scholar(Grant No.: CW202002), and the State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process Open Fund (Grant No.: SKL2020Z0302). Authors gratefully acknowledge the financial support from the above findings.

  • Received Date: Oct. 24, 2022
  • Rev Recd Date: Feb. 17, 2023
  • Tripterygium glycosides tablet (TGT), the classical commercial drug of Tripterygium wilfordii Hook. F. has been effectively used in the treatment of rheumatoid arthritis, nephrotic syndrome, leprosy, Behcet's syndrome, leprosy reaction and autoimmune hepatitis. However, due to its narrow and limited treatment window, TGT-induced organ toxicity (among which liver injury accounts for about 40% of clinical reports) has gained increasing attention. The present study aimed to clarify the cellular and molecular events underlying TGT-induced acute liver injury using single-cell RNA sequencing (scRNA-seq) technology. The TGT-induced acute liver injury mouse model was constructed through short-term TGT exposure and further verified by hematoxylin-eosin staining and liver function-related serum indicators, including alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and total bilirubin. Using the mouse model, we identified 15 specific subtypes of cells in the liver tissue, including endothelial cells, hepatocytes, cholangiocytes, and hepatic stellate cells. Further analysis indicated that TGT caused a significant inflammatory response in liver endothelial cells at different spatial locations; led to marked inflammatory response, apoptosis and fatty acid metabolism dysfunction in hepatocytes; activated hepatic stellate cells; brought about the activation, inflammation, and phagocytosis of liver capsular macrophages cells; resulted in immune dysfunction of liver lymphocytes; disturbed the intercellular crosstalk in liver microenvironment by regulating various signaling pathways. Thus, these findings elaborate the mechanism underlying TGT-induced acute liver injury, provide new insights into the safe and rational applications in the clinic, and complement the identification of new biomarkers and therapeutic targets for liver protection.
  • loading
  • Q. Lv, W. Zhang, Q. Shi, et al., Comparison of Tripterygium wilfordii Hook F with methotrexate in the treatment of active rheumatoid arthritis (TRIFRA): A randomised, controlled clinical trial, Ann. Rheum. Dis. 74 (2015) 1078-1086.
    Y. Tian, X. Su, L. Liu, et al., Overview of hepatotoxicity studies on Tripterygium wilfordii in recent 20 years, Zhongguo Zhong Yao Za Zhi 44 (2019) 3399-3405.
    N. Lin, Y. Zhang, Q. Jiang, et al., Clinical practice guideline for Tripterygium glycosides/Tripterygium wilfordii tablets in the treatment of rheumatoid arthritis, Front. Pharmacol. 11 (2021), 608703.
    C.F. Liu, J.X. Zhang, Y.Q. Li, et al., Effects of Tripterygium glycosides tablets from 6 different manufacturers on acute liver injury of normal mice, Zhongguo Zhong Yao Za Zhi 44 (2019) 3494-3501.
    Z. Yu, Z. Feng, L. Fu, et al., Qingluotongbi formula regulates the LXR-ERS-SREBP-1c pathway in hepatocytes to alleviate the liver injury caused by Tripterygium wilfordii Hook. F, J. Ethnopharmacol. 287 (2022), 114952.
    Q.L. Shi, Q.X. Wang, J.Y. Chen, et al., Transcriptome and lipid metabolomics-based discovery: Glycyrrhizic acid alleviates Tripterygium glycoside tablet-induced acute liver injury by regulating the activities of CYP and the metabolism of phosphoglycerides, Front. Pharmacol. 12 (2022), 822154.
    W. Peng, M. Dai, L. Bao, et al., FXR activation prevents liver injury induced by Tripterygium wilfordii preparations, Xenobiotica 51 (2021) 716-727.
    Z. An, Y. Sun, C. Shi, et al., Metabonomic and transcriptomic analyses of Tripterygium glycosides tablet-induced hepatotoxicity in rats, Drug Chem. Toxicol. 46 (2023) 650-664.
    M. Dai, W. Peng, T. Zhang, et al., Metabolomics reveals the role of PPAR in Tripterygium Wilfordii-induced liver injury, J. Ethnopharmacol. 289 (2022), 115090.
    A. Regev, S.A. Teichmann, E.S. Lander, et al., The human cell atlas, eLife 6 (2017), 27041.
    X. Xiong, H. Kuang, S. Ansari, et al., Landscape of intercellular crosstalk in healthy and NASH liver revealed by single-cell secretome gene analysis, Mol. Cell 75 (2019) 644-660.e5.
    J. Wang, W. Hu, Z. Shen, et al., Dissecting the single-cell transcriptome underlying chronic liver injury, Mol. Ther. Nucleic Acids 26 (2021) 1364-1373.
    C.W. Wernberg, K. Ravnskjaer, M.M. Lauridsen, et al., The role of diagnostic biomarkers, omics strategies, and single-cell sequencing for nonalcoholic fatty liver disease in severely obese patients, J. Clin. Med. 10 (2021), 930.
    Q. Su, S.Y. Kim, F. Adewale, et al., Single-cell RNA transcriptome landscape of hepatocytes and non-parenchymal cells in healthy and NAFLD mouse liver, iScience 24 (2021), 103233.
    Z. Wang, A. Keogh, A. Waldt, et al., Single-cell and bulk transcriptomics of the liver reveals potential targets of NASH with fibrosis, Sci. Rep. 11 (2021), 19396.
    C. Zheng, L. Zheng, J.K. Yoo, et al., Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing, Cell 169 (2017) 1342-1356.e16.
    X. Su, L. Zhao, Y. Shi, et al., Clonal evolution in liver cancer at single-cell and single-variant resolution, J. Hematol. Oncol. 14 (2021), 22.
    P. Ramachandran, K.P. Matchett, R. Dobie, et al., Single-cell technologies in hepatology: New insights into liver biology and disease pathogenesis, Nat. Rev. Gastroenterol. Hepatol. 17 (2020) 457-472.
    B.J. Pepe-Mooney, M.T. Dill, A. Alemany, et al., Single-cell analysis of the liver epithelium reveals dynamic heterogeneity and an essential role for YAP in homeostasis and regeneration, Cell Stem Cell 25 (2019) 23-38.e8.
    S.R. Gross, C.G.T. Sin, R. Barraclough, et al., Joining S100 proteins and migration: For better or for worse, in sickness and in health, Cell. Mol. Life Sci. 71 (2014) 1551-1579.
    S. Wang, R. Song, Z. Wang, et al., S100A8/A9 in inflammation, Front. Immunol. 9 (2018), 1298.
    J. Nemeth, I. Stein, D. Haag, et al., S100A8 and S100A9 are novel nuclear factor kappa B target genes during malignant progression of murine and human liver carcinogenesis, Hepatology 50 (2009) 1251-1262.
    R. Schierwagen, F.E. Uschner, C. Ortiz, et al., The role of macrophage-inducible C-type lectin in different stages of chronic liver disease, Front. Immunol. 11 (2020), 1352.
    S.A. MacParland, J.C. Liu, X. Ma, et al., Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations, Nat. Commun. 9 (2018), 4383.
    E. Papachristoforou, P. Ramachandran, Macrophages as key regulators of liver health and disease, Int. Rev. Cell. Mol. Biol. 368 (2022) 143-212.
    H. Zheng, Y. Pomyen, M.O. Hernandez, et al., Single-cell analysis reveals cancer stem cell heterogeneity in hepatocellular carcinoma, Hepatology 68 (2018) 127-140.
    C. Brenner, L. Galluzzi, O. Kepp, et al., Decoding cell death signals in liver inflammation, J. Hepatol. 59 (2013) 583-594.
    C. Dai, X. Xiao, D. Li, et al., Chloroquine ameliorates carbon tetrachloride-induced acute liver injury in mice via the concomitant inhibition of inflammation and induction of apoptosis, Cell Death Dis. 9 (2018), 1164.
    LiverTox: Clinical and Research Information on Drug-Induced Liver Injury [Internet], Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases, 2012.
    C. Prelli Bozzo, R. Nchioua, M. Volcic, et al., IFITM proteins promote SARS-CoV-2 infection and are targets for virus inhibition in vitro, Nat. Commun. 12 (2021), 4584.
    F. Wrensch, G. Ligat, L. Heydmann, et al., Interferon-induced transmembrane proteins mediate viral evasion in acute and chronic hepatitis C virus infection, Hepatology 70 (2019) 1506-1520.
    J. Chen, P. Luo, C. Wang, et al., Integrated single-cell transcriptomics and proteomics reveal cellular-specific responses and microenvironment remodeling in aristolochic acid nephropathy, JCI Insight 7 (2022), e157360.
    L. Goodla, X. Xue, The role of inflammatory mediators in colorectal cancer hepatic metastasis, Cells 11 (2022), 2313.
    S. Bose, P. Saha, B. Chatterjee, et al., Chemokines driven ovarian cancer progression, metastasis and chemoresistance: Potential pharmacological targets for cancer therapy, Semin. Cancer Biol. 86 (2022) 568-579.
    B.A. David, P. Kubes, Exploring the complex role of chemokines and chemoattractants in vivo on leukocyte dynamics, Immunol. Rev. 289 (2019) 9-30.
    S.J. Burke, J.J. Collier, Transcriptional regulation of chemokine genes: A link to pancreatic islet inflammation? Biomolecules 5 (2015) 1020-1034.
    N. Lou, M.L. Lennard Richard, J. Yu, et al., The Fli-1 transcription factor is a critical regulator for controlling the expression of chemokine C-X-C motif ligand 2 (CXCL2), Mol. Immunol. 81 (2017) 59-66.
    M.H. Zhao, T.M. Lu, L. Liu, et al., Mechanism of Licorice alleviating Tripterygium glycosides tablet induced liver injury, Zhongguo ShiYan FangJiXue ZaZhi 44 (2019) 3399-3405.
    Y. Zhang, X. Mao, W. Li, et al., Tripterygium wilfordii: An inspiring resource for rheumatoid arthritis treatment, Med. Res. Rev. 41 (2021) 1337-1374.
    L. Luo, Z. Jiang, L. Zhang, Research progress of hepatotoxicity mechanism and attenuation of Tripterygium wilfordii glycoside, Drug Eval. Res. 40 (2017) 1504-1509.
    J. Sun, J. Zhang, X. Wang, et al., Gut-liver crosstalk in sepsis-induced liver injury, Crit. Care 24 (2020), 614.
    J. Poisson, S. Lemoinne, C. Boulanger, et al., Liver sinusoidal endothelial cells: Physiology and role in liver diseases, J. Hepatol. 66 (2017) 212-227.
    S. Shetty, P.F. Lalor, D.H. Adams, Liver sinusoidal endothelial cells-Gatekeepers of hepatic immunity, Nat. Rev. Gastroenterol. Hepatol. 15 (2018) 555-567.
    M.E. Guicciardi, H. Malhi, J.L. Mott, et al., Apoptosis and necrosis in the liver, Compr. Physiol. 3 (2013) 977-1010.
    S. Gaul, A. Leszczynska, F. Alegre, et al., Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis, J. Hepatol. 74 (2021) 156-167.
    V.V. Kalinichenko, D. Bhattacharyya, Y. Zhou, et al., Foxf1 mice exhibit defective stellate cell activation and abnormal liver regeneration following CCl4 injury, Hepatology 37 (2003) 107-117.
    W. Zhang, S.J. Conway, Y. Liu, et al., Heterogeneity of hepatic stellate cells in fibrogenesis of the liver: Insights from single-cell transcriptomic analysis in liver injury, Cells 10 (2021), 2129.
    T. Tsuchida, S.L. Friedman, Mechanisms of hepatic stellate cell activation, Nat. Rev. Gastroenterol. Hepatol. 14 (2017) 397-411.
    S. Schuster, D. Cabrera, M. Arrese, et al., Triggering and resolution of inflammation in NASH, Nat. Rev. Gastroenterol. Hepatol. 15 (2018) 349-364.
    Z. Liu, M. Wang, X. Wang, et al., XBP1 deficiency promotes hepatocyte pyroptosis by impairing mitophagy to activate mtDNA-cGAS-STING signaling in macrophages during acute liver injury, Redox Biol. 52 (2022), 102305.
    X. Tian, Y. Wang, Y. Lu, et al., Conditional depletion of macrophages ameliorates cholestatic liver injury and fibrosis via lncRNA-H19, Cell Death Dis. 12 (2021), 646.
    E. Papachristoforou, P. Ramachandran, Macrophages as key regulators of liver health and disease. International Review of Cell and Molecular Biology. 368 (2022) 143-212.
    F. Marra, F. Tacke, Roles for chemokines in liver disease, Gastroenterology 147 (2014) 577-594.e1.
    H. Malhi, R.J. Kaufman, Endoplasmic reticulum stress in liver disease, J. Hepatol. 54 (2011) 795-809.
    Y. Saiman, S.L. Friedman, The role of chemokines in acute liver injury, Front. Physio. 3 (2012), 213.
    P.E. Marques, S.S. Amaral, D.A. Pires, et al., Chemokines and mitochondrial products activate neutrophils to amplify organ injury during mouse acute liver failure, Hepatology 56 (2012) 1971-1982.
    Y. Koda, N. Nakamoto, P.S. Chu, et al., Plasmacytoid dendritic cells protect against immune-mediated acute liver injury via IL-35, J. Clin. Investig. 129 (2019) 3201-3213.
    S. Zhou, J. Gu, R. Liu, et al., Spermine alleviates acute liver injury by inhibiting liver-resident macrophage pro-inflammatory response through ATG5-dependent autophagy, Front. Immunol. 9 (2018), 948.
  • 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 (432) PDF downloads(60) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return