Volume 14 Issue 2
Feb.  2024
Turn off MathJax
Article Contents
Baixi Shan, Haoyan Zhou, Congying Guo, Xiaolu Liu, Mingyu Wu, Rao Zhai, Jun Chen. Tanshinone IIA ameliorates energy metabolism dysfunction of pulmonary fibrosis using 13C metabolic flux analysis[J]. Journal of Pharmaceutical Analysis, 2024, 14(2): 244-258. doi: 10.1016/j.jpha.2023.09.008
Citation: Baixi Shan, Haoyan Zhou, Congying Guo, Xiaolu Liu, Mingyu Wu, Rao Zhai, Jun Chen. Tanshinone IIA ameliorates energy metabolism dysfunction of pulmonary fibrosis using 13C metabolic flux analysis[J]. Journal of Pharmaceutical Analysis, 2024, 14(2): 244-258. doi: 10.1016/j.jpha.2023.09.008

Tanshinone IIA ameliorates energy metabolism dysfunction of pulmonary fibrosis using 13C metabolic flux analysis

doi: 10.1016/j.jpha.2023.09.008
Funds:

This work was financially supported by the National Natural Science Foundation of China (Grant No.: 82174100). The authors thank Nian Wang and Zi-Yuan Wang for technical support from the Center for Analysis and Testing of China Pharmaceutical University.

  • Received Date: May 26, 2023
  • Accepted Date: Sep. 18, 2023
  • Rev Recd Date: Sep. 06, 2023
  • Publish Date: Feb. 29, 2024
  • Evidence indicates that metabolic reprogramming characterized by the changes in cellular metabolic patterns contributes to the pathogenesis of pulmonary fibrosis (PF). It is considered as a promising therapeutic target anti-PF. The well-documented against PF properties of Tanshinone IIA (Tan IIA) have been primarily attributed to its antioxidant and anti-inflammatory potency. Emerging evidence suggests that Tan IIA may target energy metabolism pathways, including glycolysis and tricarboxylic acid (TCA) cycle. However, the detailed and advanced mechanisms underlying the anti-PF activities remain obscure. In this study, we applied [U-13C]-glucose metabolic flux analysis (MFA) to examine metabolism flux disruption and modulation nodes of Tan IIA in PF. We identified that Tan IIA inhibited the glycolysis and TCA flux, thereby suppressing the production of transforming growth factor-β1 (TGF-β1)-dependent extracellular matrix and the differentiation and proliferation of myofibroblasts in vitro. We further revealed that Tan IIA inhibited the expression of key metabolic enzyme hexokinase 2 (HK2) by inhibiting phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α) pathway activities, which decreased the accumulation of abnormal metabolites. Notably, we demonstrated that Tan IIA inhibited ATP citrate lyase (ACLY) activity, which reduced the collagen synthesis pathway caused by cytosol citrate consumption. Further, these results were validated in a mouse model of bleomycin-induced PF. This study was novel in exploring the mechanism of the occurrence and development of Tan IIA in treating PF using 13C-MFA technology. It provided a novel understanding of the mechanism of Tan IIA against PF from the perspective of metabolic reprogramming.
  • loading
  • [1]
    R. Rajesh, R. Atallah, T. Barnthaler, Dysregulation of metabolic pathways in pulmonary fibrosis, Pharmacol. Ther. 246 (2023) 108436.
    [2]
    H. Zhao, P.A. Dennery, H. Yao, Metabolic reprogramming in the pathogenesis of chronic lung diseases, including BPD, COPD, and pulmonary fibrosis, Am. J. Physiol. Lung Cell Mol. Physiol. 314 (2018) L544-L554.
    [3]
    T. Kinoshita, T. Goto, Molecular mechanisms of pulmonary fibrogenesis and its progression to lung cancer: a review, Int. J. Mol. Sci. 20 (2019) 1461.
    [4]
    B. Selvarajah, I. Azuelos, D. Anastasiou, et al., Fibrometabolism-An emerging therapeutic frontier in pulmonary fibrosis, Sci. Signal. 14 (2021) eaay1027.
    [5]
    S. Saito, A. Alkhatib, J.K. Kolls, et al., Pharmacotherapy and adjunctive treatment for idiopathic pulmonary fibrosis (IPF), J. Thorac. Dis. 11 (2019) S1740-S1754.
    [6]
    L.A.J. O'Neill, R.J. Kishton, J. Rathmell, A guide to immunometabolism for immunologists, Nat. Rev. Immunol. 16 (2016) 553-565.
    [7]
    B. Selvarajah, I. Azuelos, M. Plate, et al., mTORC1 amplifies the ATF4-dependent de novo serine-glycine pathway to supply glycine during TGF-β1-induced collagen biosynthesis, Sci. Signal. 12 (2019) eaav3048.
    [8]
    R.C. Chambers, P.F. Mercer, Mechanisms of alveolar epithelial injury, repair, and fibrosis, Ann. Am. Thorac. Soc. 12 (2015) S16-S20.
    [9]
    C. Vancheri, Common pathways in idiopathic pulmonary fibrosis and cancer, Eur. Respir. Rev. 22 (2013) 265-272.
    [10]
    N. Xie, Z. Tan, S. Banerjee, et al., Glycolytic reprogramming in myofibroblast differentiation and lung fibrosis, Am. J. Respir. Crit. Care Med. 192 (2015) 1462-1474.
    [11]
    M. Andrianifahanana, D.M. Hernandez, X. Yin, et al., Profibrotic up-regulation of glucose transporter 1 by TGF-β involves activation of MEK and mammalian target of rapamycin complex 2 pathways, Faseb. J. 30 (2016) 3733-3744.
    [12]
    H. He, H. Tang, L. Gao, et al., Tanshinone IIA attenuates bleomycin-induced pulmonary fibrosis in rats, Mol. Med. Rep. 11 (2015) 4190-4196.
    [13]
    F. Feng, N. Li, P. Cheng, et al., Tanshinone IIA attenuates silica-induced pulmonary fibrosis via inhibition of TGF-β1-Smad signaling pathway, Biomed. Pharmacother. 121 (2020) 109586.
    [14]
    L. An, L.Y. Peng, N.Y. Sun, et al., Tanshinone IIA activates nuclear factor-erythroid 2-related factor 2 to restrain pulmonary fibrosis via regulation of redox homeostasis and glutaminolysis, Antioxidants Redox Signal. 30 (2019) 1831-1848.
    [15]
    Z. Wang, F. Zhang, W. Liu, et al., Impaired tricarboxylic acid cycle flux and mitochondrial aerobic respiration during isoproterenol induced myocardial ischemia is rescued by bilobalide, J. Pharm. Anal. 11 (2021) 764-775.
    [16]
    M. Yuan, D.M. Kremer, H. Huang, et al., Ex vivo and in vivo stable isotope labelling of central carbon metabolism and related pathways with analysis by LC-MS/MS, Nat. Protoc. 14 (2019) 313-330.
    [17]
    Y. Toya, N. Kono, K. Arakawa, et al., Metabolic flux analysis and visualization, J. Proteome Res. 10 (2011) 3313-3323.
    [18]
    M.R. Antoniewicz, A guide to 13C metabolic flux analysis for the cancer biologist, Exp. Mol. Med. 50 (2018) 1-13.
    [19]
    C.P. Long, M.R. Antoniewicz, High-resolution 13C metabolic flux analysis, Nat. Protoc. 14 (2019) 2856-2877.
    [20]
    Y. Li, Y.C. Li, X.T. Liu, et al., Blockage of citrate export prevents TCA cycle fragmentation via Irg1 inactivation, Cell Rep. 38 (2022) 110391.
    [21]
    Y. Zhang, G. Yu, H. Chu, et al., Macrophage-associated PGK1 phosphorylation promotes aerobic glycolysis and tumorigenesis, Mol. Cell 71 (2018) 201-215.
    [22]
    G.F. Zhang, M.V. Jensen, S.M. Gray, et al., Reductive TCA cycle metabolism fuels glutamine- and glucose-stimulated insulin secretion, Cell Metab. 33 (2021) 804-817.
    [23]
    B. Shan, M. Wu, T. Chen, et al., Berberine attenuates hyperuricemia by regulating urate transporters and gut microbiota, Am. J. Chin. Med. 50 (2022) 2199-2221.
    [24]
    B. Shan, Z. Ai, S. Zeng, et al., Gut microbiome-derived lactate promotes to anxiety-like behaviors through GPR81 receptor-mediated lipid metabolism pathway, Psychoneuroendocrinology 117 (2020) 104699.
    [25]
    H. Li, M. Wu, C. Guo, et al., Tanshinone IIA regulates Keap1/Nrf2 signal pathway by activating Sestrin2 to restrain pulmonary fibrosis, Am. J. Chin. Med. 50 (2022) 2125-2151.
    [26]
    R. Nigdelioglu, R.B. Hamanaka, A.Y. Meliton, et al., Transforming growth factor (TGF)-β promotes de novo serine synthesis for collagen production, J. Biol. Chem. 291 (2016) 27239-27251.
    [27]
    D. DeWaal, V. Nogueira, A.R. Terry, et al., Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin, Nat. Commun. 9 (2018) 446.
    [28]
    H. Liu, X. Zhang, Y. Shao, et al., Danshensu alleviates bleomycin-induced pulmonary fibrosis by inhibiting lung fibroblast-to-myofibroblast transition via the MEK/ERK signaling pathway, Bioengineered 12 (2021) 3113-3124.
    [29]
    H. Liu, C. Liu, M. Wang, et al., Tanshinone IIA affects the malignant growth of Cholangiocarcinoma cells by inhibiting the PI3K-Akt-mTOR pathway, Sci. Rep. 11 (2021) 19268.
    [30]
    T. Zhang, X. Zhu, H. Wu, et al., Targeting the ROS/PI3K/AKT/HIF-1α/HK2 axis of breast cancer cells: combined administration of Polydatin and 2-Deoxy-d-glucose, J. Cell Mol. Med. 23 (2019) 3711-3723.
    [31]
    A. Mamazhakypov, R.T. Schermuly, L. Schaefer, et al., Lipids - two sides of the same coin in lung fibrosis, Cell. Signal. 60 (2019) 65-80.
    [32]
    P.K. Arnold, B.T. Jackson, K.I. Paras, et al., A non-canonical tricarboxylic acid cycle underlies cellular identity, Nature 603 (2022) 477-481.
    [33]
    J. Li, X. Zhai, X. Sun, et al., Metabolic reprogramming of pulmonary fibrosis, Front. Pharmacol. 13 (2022) 1031890.
    [34]
    F. Feng, P. Cheng, S. Xu, et al., Tanshinone IIA attenuates silica-induced pulmonary fibrosis via Nrf2-mediated inhibition of EMT and TGF-β1/Smad signaling, Chem. Biol. Interact. 319 (2020) 109024.
    [35]
    N. Wang, H. Liu, G. Liu, et al., Yeast β-D-glucan exerts antitumour activity in liver cancer through impairing autophagy and lysosomal function, promoting reactive oxygen species production and apoptosis, Redox Biol. 32 (2020) 101495.
    [36]
    Z. Chen, M. Liu, L. Li, et al., Involvement of the Warburg effect in non-tumor diseases processes, J. Cell. Physiol. 233 (2018) 2839-2849.
    [37]
    X. Zhao, P. Psarianos, L.S. Ghoraie, et al., Metabolic regulation of dermal fibroblasts contributes to skin extracellular matrix homeostasis and fibrosis, Nat. Metab. 1 (2019) 147-157.
    [38]
    K. Bernard, N.J. Logsdon, S. Ravi, et al., Metabolic reprogramming is required for myofibroblast contractility and differentiation, J. Biol. Chem. 290 (2015) 25427-25438.
    [39]
    N. Zamboni, S.M. Fendt, M. Ruhl, et al., 13C-based metabolic flux analysis, Nat. Protoc. 4 (2009) 878-892.
    [40]
    X. Yin, M. Choudhury, J.H. Kang, et al., Hexokinase 2 couples glycolysis with the profibrotic actions of TGF-β, Sci. Signal. 12 (2019) eaax4067.
    [41]
    W. Qian, X. Cai, Q. Qian, et al., Astragaloside IV modulates TGF-β1-dependent epithelial-mesenchymal transition in bleomycin-induced pulmonary fibrosis, J. Cell Mol. Med. 22 (2018) 4354-4365.
    [42]
    J. Wang, K. Hu, X. Cai, et al., Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis, Acta Pharm. Sin. B 12 (2022) 18-32.
    [43]
    X. Hu, Q. Xu, H. Wan, et al., PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis, Lab. Invest. 100 (2020) 801-811.
    [44]
    Q. Lou, M. Zhang, K. Zhang, et al., Arsenic exposure elevated ROS promotes energy metabolic reprogramming with enhanced AKT-dependent HK2 expression, Sci. Total Environ. 836 (2022) 155691.
    [45]
    Y. Ji, Y.N. Dou, Q.W. Zhao, et al., Paeoniflorin suppresses TGF-β mediated epithelial-mesenchymal transition in pulmonary fibrosis through a Smad-dependent pathway, Acta Pharmacol. Sin. 37 (2016) 794-804.
    [46]
    H.S. Hsu, C.C. Liu, J.H. Lin, et al., Involvement of ER stress, PI3K/AKT activation, and lung fibroblast proliferation in bleomycin-induced pulmonary fibrosis, Sci. Rep. 7 (2017) 14272.
    [47]
    S. Wang, J.L. Fu, H.F. Hao, et al., Metabolic reprogramming by traditional Chinese medicine and its role in effective cancer therapy, Pharmacol. Res. 170 (2021) 105728.
    [48]
    M. Imamura, J.S. Moon, K.P. Chung, et al., RIPK3 promotes kidney fibrosis via AKT-dependent ATP citrate lyase, JCI Insight 3 (2018) e94979.
    [49]
    G. Hatzivassiliou, F. Zhao, D.E. Bauer, et al., ATP citrate lyase inhibition can suppress tumor cell growth, Cancer Cell 8 (2005) 311-321.
  • 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 (346) PDF downloads(29) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return