Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2020, Vol. 14 Issue (1) : 68-80    https://doi.org/10.1007/s11684-019-0690-z
RESEARCH ARTICLE
New incompatible pair of TCM: Epimedii Folium combined with Psoraleae Fructus induces idiosyncratic hepatotoxicity under immunological stress conditions
Yuan Gao1,2, Zhilei Wang2,3, Jinfa Tang4, Xiaoyi Liu2, Wei Shi2,5, Nan Qin2,5, Xiaoyan Wang4, Yu Pang6, Ruisheng Li7, Yaming Zhang2, Jiabo Wang2, Ming Niu2(), Zhaofang Bai2(), Xiaohe Xiao8()
1. School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
2. China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
3. School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
4. The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
5. School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Jiangxi 330004, China
6. National Institutes for Food and Drug Control, Beijing 100050, China
7. Research Center for Clinical and Translational Medicine, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
8. Integrative Medical Centre, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
 Download: PDF(3788 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Epimedii Folium (EF) combined with Psoraleae Fructus (PF) is a common modern preparation, but liver injury caused by Chinese patent medicine preparations containing EF and PF has been frequently reported in recent years. Zhuangguguanjiewan pills (ZGW), which contain EF and PF, could induce immune idiosyncratic liver injury according to clinical case reports and a nonhepatotoxic dose of lipopolysaccharide (LPS) model. This present study evaluated the liver injury induced by EF or PF alone or in combination and investigated the related mechanism by using the LPS model. Liver function indexes and pathological results showed that either EF or PF alone or in combination led to liver injury in normal rats; however, EF or PF alone could lead to liver injury in LPS-treated rats. Moreover, EF combined with PF could induce a greater degree of injury than that caused by EF or PF alone in LPS-treated rats. Furthermore, EF or PF alone or in combination enhanced the LPS-stimulated inflammatory cytokine production, implying that IL-1β, which is processed and released by activating the NLRP3 inflammasome, is a specific indicator of EF-induced immune idiosyncratic hepatotoxicity. Thus, EF may induce liver injury through enhancing the LPS-mediated proinflammatory cytokine production and activating the NLRP3 inflammasome. In addition, the metabolomics analysis results showed that PF affected more metabolites in glycerophospholipid and sphingolipid metabolic pathways compared with EF in LPS model, suggesting that PF increased the responsiveness of the liver to LPS or other inflammatory mediators via modulation of multiple metabolic pathways. Therefore, EF and PF combination indicates traditional Chinese medicine incompatibility, considering that it induces idiosyncratic hepatotoxicity under immunological stress conditions.

Keywords Epimedii Folium      Psoraleae Fructus      idiosyncratic hepatotoxicity      traditional Chinese medicine incompatibility     
Corresponding Author(s): Ming Niu,Zhaofang Bai,Xiaohe Xiao   
Just Accepted Date: 31 January 2019   Online First Date: 27 March 2019    Issue Date: 02 March 2020
 Cite this article:   
Yuan Gao,Zhilei Wang,Jinfa Tang, et al. New incompatible pair of TCM: Epimedii Folium combined with Psoraleae Fructus induces idiosyncratic hepatotoxicity under immunological stress conditions[J]. Front. Med., 2020, 14(1): 68-80.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-019-0690-z
https://academic.hep.com.cn/fmd/EN/Y2020/V14/I1/68
Fig.1  PF, EF, and PE can induce liver injury in LPS-treated rats. (A) Serum ALT activity in rats co-treated with EF, PF, or PE (normal saline or LPS-treated rats). (B) Serum AST activity in rats co-treated with EF, PF, or PE (normal saline- or LPS-treated rats). * P< 0.001, compared with that in the model group. (C) Typical histopathological section photographs of rat liver specimens for HE analysis (magnification 200×).
Fig.2  Positive TUNEL staining (indicative of cell apoptosis) was observed in liver tissues. A representative photomicrograph at 200× magnification is shown. Liver sections were stained for TUNEL (green) and nuclei (blue).*P<0.05, **P<0.001, compared with those in the model group.
Fig.3  Effect of PF, EF, or PE on inflammatory cytokines. (A) plasma concentrations of TNF-α; (B) plasma concentrations of IL-1β; (C) plasma concentrations of IL-6; (D) plasma concentrations of IFN-g. Mod, PL, EL, and PEL groups (n = 10); Con, PF, EF, and PE groups (n = 6); *P< 0.05, **P< 0.01, ***P< 0.001, compared with those in the model group. ###P<0.001, compared with that in the control group.
Fig.4  Correlation analysis between metabolites and immune factors in different groups. (A) Variations in the trends of RT19.35_M/Z330.276, RT17.14_M/Z703.572, RT7.93_M/Z560.3096, and RT13.96_M/Z301.297 are shown. *P<0.05, **P<0.01, ***P<0.001, compared with those in the control group. (B) Class 1–5 represents the control, model, PL, EL, and PEL groups.
Fig.5  Schematic diagram of the disturbed metabolic pathway. The size of the ball represents the relative degree of change between groups; the color of the ball represents the tendency of compounds to increase or decrease.
DATA FROM THE ESI+ MODE
Metabolite Mass
(Neutral)
Error
(ppm)
Formulate tR (min) FOLD
Con vs. Mod Con vs. PL Con vs. EL Con vs. PEL
PA (22:6(4Z,7Z,10Z,13Z,16Z,19Z)/14:1(9Z)) 690.4334 -10.63 C39H63O8P 16.05 1.5484 1.641 2.1565 22.931
PA (20:5(5Z,8Z,11Z,14Z,17Z)/13:0) 652.4192 -13.48 C36H61O8P 16.64 0.68069 6.817 4.5282 3.7127
PG (22:4(7Z,10Z,13Z,16Z)/0:0) 560.3096 3.24 C28H49O9P 7.93 0.96525 0.17944 0.33306 0.94477
PS (20:3(8Z,11Z,14Z)/0:0) 547.284 12.82 C26H46NO9P 9.40 5.49E-10 1.67E-11 1.00E-08 3.29E-10
MG (16:0/0:0/0:0)[rac] 330.276 3.05 C19H38O4 19.35 0.38613 0.35689 0.40864 0.23563
PI (12:0/18:1(9Z)) 780.485 -7.84 C39H73O13P 13.72 4.1323 0.32628 0.75197 0.26141
PI (19:1(9Z)/0:0) 612.3304 -4.79 C28H53O12P 14.78 0.42428 0.49225 0.55941 0.41603
PI (12:0/0:0) 516.2375 -7.62 C21H41O12P 9.15 1.19E-11 3.37E-11 2.33E-11 1.01E-12
Sphingosine-1-phosphate 379.244 12.55 C18H38NO5P 17.04 0.15385 0.049752 0.15583 0.038203
Cer (t18:0/20:0) 611.5902 -8.07 C38H77NO4 13.05 1.1437 0.58018 1.2547 1.5889
SM (d18:1/16:0) 703.5726 4.01 C39H80N2O6P 17.14 1.0255 1.0348 1.0243 1.0245
DATA FROM THE ESI− MODE
PS (20:0/0:0) 553.3361 3.37 C26H52NO9P 12.25 0.04056 0.008249 0.00953 0.008266
PI (16:0/12:0) 754.4657 -3.27 C37H71O13P 12.42 0.32803 0.25737 0.5367 0.003003
PI (20:0/0:0) 628.3617 -4.67 C29H57O12P 14.64 13.245 30.108 17.687 2227.6
PI (18:3(9Z,12Z,15Z)/0:0) 594.2807 -0.31 C27H47O12P 15.12 5.24E-12 6.89E-13 1.00E-12 1.72E-12
PS (15:0/12:0) 665.4275 -1.07 C33H64NO10P 17.01 0.39039 0.40176 0.45213 0.073747
PG (15:0/14:0) 680.4509 -13.51 C38H65O8P 18.43 8.61E-10 8.47E-11 2.41E-08 4.10E-10
LysoPE (20:2(11Z,14Z)/0:0) 505.3127 8.19 C25H48NO7P 9.95 0.092292 0.11025 0.26143 0.025222
Tab.1  Differentially identified metabolites for discrimination among the control, model, PL, EL, and PEL groups
Fig.6  Schematic diagram of liver injury induced by EF, PF, and PE under immunological stress conditions. (A) EF, PF or PE cannot induce liver injury under normal conditions. (B) EF, PF, or PE can lead to a serious liver injury under immunological stress conditions. The main mechanism of liver injury induced by EF, PF, and PE may be different: EF may regulate immune inflammation to induce liver injury; PF may regulate metabolism to induce liver injury; and PE may lead to a more serious liver injury by the comprehensive regulation of immune inflammation and metabolic dysfunction.
1 Y Xu, J Ding, JN An, YK Qu, X Li, XP Ma, YM Zhang, GJ Dai, N Lin. Effect of the interaction of Veratrum Nigrum with Panax Ginseng on estrogenic activity in vivo and in vitro. Sci Rep 2016; 6(1): 26924
https://doi.org/10.1038/srep26924 pmid: 27229740
2 W Long, XD Zhang, HY Wu, J Jin, GY Yu, X He, H Wang, X Shen, ZW Zhou, PX Liu, SJ Fan. Study on incompatibility of traditional chinese medicine: evidence from formula network, chemical space, and metabolism room. Evid Based Complement Alternat Med 2013; 2013: 352145
https://doi.org/10.1155/2013/352145 pmid: 24369478
3 JF Tang, XY Wang, Q Wen, S Tang, F Sang, WX Li, YH Li, CN Zhang, M Niu, ZF Bai, JB Wang, XH Xiao. Idiosyncratic hepatotoxicity evaluation of Zhuangguguanjie wan mediated by immune stress. Acta Pharmaceutica Sinica (Yao Xue Xue Bao) 2017; 52: 1033–1040 (in Chinese)
4 PJ Shaw, PE Ganey, RA Roth. Idiosyncratic drug-induced liver injury and the role of inflammatory stress with an emphasis on an animal model of trovafloxacin hepatotoxicity. Toxicol Sci 2010; 118(1): 7–18
https://doi.org/10.1093/toxsci/kfq168 pmid: 20538741
5 K Zhou, Z Dai, ZB Liu, Y Tan, LM Hu, BL Zhang, XM Gao. Experimental study of screening the hepatotoxic medicinal materials in Zhuangguguanjie Pill. Chin J Pharmacovigilance (Zhongguo Yao Wu Jing Jie) 2009; 11: 641–648 (in Chinese)
6 JF Tang, XY Wang, Q Wen, S Tang, F Sang, WX Li, YH Li, CY Li, CE Zhang, M Niu, ZF Bai, JB Wang, XH Xiao. Cytokine analysis of Zhuangguguanjie wan-induced idiosyncratic liver injury based on mathematical modeling.Acta Pharmaceutica Sinica ( Yao Xue Xue Bao) 2018; 53: 574–584 (in Chinese)
7 JF Tang, XY Wang, WX Li, YH Li, JB Wang, ZF Bai, XH Xiao, W Yang. Correlation analysis on idiosyncratic hepatotoxicity of Zhuangguguanjie Wan and 27 cytokines based on related visualization. Chin Tradit Herbal Drugs 2018; 49: 1624–1633
8 Y Cheng, Y Liu, H Wang, J Li, J Ren, L Zhu, L Gong. A 26-week repeated dose toxicity study of Xian-ling-gu-bao in Sprague-Dawley rats. J Ethnopharmacol 2013; 145(1): 85–93
https://doi.org/10.1016/j.jep.2012.09.055 pmid: 23123261
9 H Wu, QX Zhong, J Wang, M Wang, F Fang, Z Xia, RL Zhong, HC Huang, ZC Ke, YJ Wei, L Feng, ZQ Shi, E Sun, J Song, XB Jia. Beneficial effects and toxicity studies of Xian-ling-gu-bao on bone metabolism in ovariectomized rats. Front Pharmacol 2017; 8:273–284
10 WI Cheung, ML Tse, T Ngan, J Lin, WK Lee, WT Poon, TW Mak, VK Leung, TN Chau. Liver injury associated with the use of Fructus Psoraleae (Bol-gol-zhee or Bu-gu-zhi) and its related proprietary medicine. Clin Toxicol (Phila) 2009; 47(7): 683–685
https://doi.org/10.1080/15563650903059136 pmid: 19640237
11 SW Nam, JT Baek, DS Lee, SB Kang, BM Ahn, KW Chung. A case of acute cholestatic hepatitis associated with the seeds of Psoralea corylifolia (Boh-Gol-Zhee). Clin Toxicol (Phila) 2005; 43(6): 589–591
https://doi.org/10.1081/CLT-200068863 pmid: 16255343
12 D Melchart, S Hager, S Albrecht, J Dai, W Weidenhammer, R Teschke. Herbal traditional Chinese medicine and suspected liver injury: a prospective study. World J Hepatol 2017; 9(29): 1141–1157
https://doi.org/10.4254/wjh.v9.i29.1141 pmid: 29085558
13 G Danan, R Teschke. RUCAM in drug and herb induced liver injury: the update. Int J Mol Sci 2015; 17(1): 14–47
https://doi.org/10.3390/ijms17010014 pmid: 26712744
14 DM Li, XF Yin, JH Liu, DW Cai. Long term toxicity of total flavonoids from Epimedii Folium. Chin J of Exp Tradit Med Formulae ( Zhongguo Shi Yan Fang Ji Xue Za Zhi ) 2008; 14(7): 60–62 (in Chinese)
15 R Teschke, D Larrey, D Melchart, G Danan. Traditional Chinese medicine (TCM) and herbal hepatotoxicity: RUCAM and the role of novel diagnostic biomarkers such as microRNAs. Medicines (Basel) 2016; 3(3): 18
https://doi.org/10.3390/medicines3030018 pmid: 28930128
16 J Jing, R Teschke. Traditional Chinese medicine and herb-induced liver injury: comparison with drug-induced liver injury. J Clin Transl Hepatol 2018; 6(1): 57–68
https://doi.org/10.14218/JCTH.2017.00033 pmid: 29577033
17 R Teschke. Traditional Chinese medicine induced liver injury. J Clin Transl Hepatol 2014; 2(2): 80–94
pmid: 26357619
18 R Teschke. Kava hepatotoxicity: pathogenetic aspects and prospective considerations. Liver Int 2010; 30(9): 1270–1279
https://doi.org/10.1111/j.1478-3231.2010.02308.x pmid: 20630022
19 C Li, M Niu, Z Bai, C Zhang, Y Zhao, R Li, C Tu, H Li, J Jing, Y Meng, Z Ma, W Feng, J Tang, Y Zhu, J Li, X Shang, Z Zou, X Xiao, J Wang. Screening for main components associated with the idiosyncratic hepatotoxicity of a tonic herb, Polygonum multiflorum. Front Med 2017; 11(2): 253–265
https://doi.org/10.1007/s11684-017-0508-9 pmid: 28315126
20 JB Wang. LI CY, Zhu Y, Song HB, Bai ZF, Xiao XH. Integrated evidence chain-based identification of Chinese herbal medicine- induced hepatotoxicity and rational usage: exemplification by Polygonum Multiflorum (He shou wu). Chin Sci Bull (Ke Xue Tong Bao) 2016; 61(9): 971–980 (in Chinese)
21 JB Wang, HR Cui, ZF Bai, XH Xiao. Precision medicine-oriented safety assessment strategy for traditional Chinese medicines: disease-syndrome-based toxicology. Acta Pharmaceutica Sinica (Yao Xue Xue Bao) 2016; 51(11): 1681–1688 (in Chinese)
pmid: 29908110
22 G Danan, R Teschke. Drug-induced liver injury: why is the Roussel Uclaf Causality Assessment Method (RUCAM) still used 25 years after its launch? Drug Saf 2018; 41(8): 735–743
https://doi.org/10.1007/s40264-018-0654-2 pmid: 29502198
23 R Teschke, G Danan. Drug induced liver injury with analysis of alternative causes as confounding variables. Br J Clin Pharmacol 2018; 84(7): 1467–1477
https://doi.org/10.1111/bcp.13593 pmid: 29607530
24 R Teschke, J Schulze, A Eickhoff, G Danan. Drug induced liver injury: can biomarkers assist RUCAM in causality assessment? Int J Mol Sci 2017; 18(4): 803
https://doi.org/10.3390/ijms18040803 pmid: 28398242
25 J Wang, Z Ma, M Niu, Y Zhu, Q Liang, Y Zhao, J Song, Z Bai, Y Zhang, P Zhang, N Li, Y Meng, Q Li, L Qin, G Teng, J Cao, B Li, S Chen, Y Li, Z Zou, H Zhou, X Xiao. Evidence chain-based causality identification in herb-induced liver injury: exemplification of a well-known liver-restorative herb Polygonum multiflorum. Front Med 2015; 9(4): 457–467
https://doi.org/10.1007/s11684-015-0417-8 pmid: 26459430
26 R Teschke, R Bahre. Severe hepatotoxicity by Indian Ayurvedic herbal products: a structured causality assessment. Ann Hepatol 2009; 8(3): 258–266
pmid: 19841509
27 PX Deng, M Xu. Comparision on hepatotoxicity in rats by using Fructus Psoralea alone and in combination with other herbs. Guangxi J Tradit Chin Med (Guangxi Zhong Yi Yao) 2005; 28(2): 49–50 (in Chinese)
28 Y Yang, X Tang, F Hao, Z Ma, Y Wang, L Wang, Y Gao. Bavachin induces apoptosis through mitochondrial regulated ER stress pathway in HepG2 cells. Biol Pharm Bull 2018; 41(2): 198–207
https://doi.org/10.1248/bpb.b17-00672 pmid: 29187671
29 JH Cheng, HD Cai. Adverse reactions to Zhuangguguanjie Wan and cause analysis. Adverse Drug Reactions J (Yao Wu Bu Liang Fan Ying Za Zhi) 2000; 1: 15–19 (in Chinese)
30 SZ Qi, N Li, ZD Tuo, JL Li, SS Xing, BB Li, L Zhang, HS Lee, JG Chen, L Cui. Effects of Morus root bark extract and active constituents on blood lipids in hyperlipidemia rats. J Ethnopharmacol 2016; 180: 54–59
https://doi.org/10.1016/j.jep.2016.01.024 pmid: 26806569
31 Z Sarhadynejad, F Sharififar, A Pardakhty, MH Nematollahi, S Sattaie-Mokhtari, A Mandegary. Pharmacological safety evaluation of a traditional herbal medicine “Zereshk-e-Saghir” and assessment of its hepatoprotective effects on carbon tetrachloride induced hepatic damage in rats. J Ethnopharmacol 2016; 190: 387–395
https://doi.org/10.1016/j.jep.2016.07.043 pmid: 27426508
32 JW Lou, LL Cao, Q Zhang, L Zhang, AW Ding. Comparative study on acute toxicity of different active ingredient fractions from Kansui stir-baked with vinegar on zebrafish embryos. China J Chin Materia Medica (Zhongguo Zhong Yao Za Zhi) 2017; 42(18): 3516–3522 (in Chinese)
pmid: 29218936
33 A Eguchi, A Wree, AE Feldstein. Biomarkers of liver cell death. J Hepatol 2014; 60(5): 1063–1074
https://doi.org/10.1016/j.jhep.2013.12.026 pmid: 24412608
34 V Kolarski, A Todorov, D Petrova. Cytokines and the liver in health and disease. Vutr Boles 2000; 32(1): 19–24
pmid: 11195192
35 D Schmidt-Arras, S Rose-John. IL-6 pathway in the liver: from physiopathology to therapy. J Hepatol 2016; 64(6): 1403–1415
https://doi.org/10.1016/j.jhep.2016.02.004 pmid: 26867490
36 A Kwak, Y Lee, H Kim, S Kim. Intracellular interleukin (IL)-1 family cytokine processing enzyme. Arch Pharm Res 2016; 39(11): 1556–1564
https://doi.org/10.1007/s12272-016-0855-0 pmid: 27826753
37 JF Waring, MJ Liguori, JP Luyendyk, JF Maddox, PE Ganey, RF Stachlewitz, C North, EA Blomme, RA Roth. Microarray analysis of lipopolysaccharide potentiation of trovafloxacin-induced liver injury in rats suggests a role for proinflammatory chemokines and neutrophils. J Pharmacol Exp Ther 2006; 316(3): 1080–1087
https://doi.org/10.1124/jpet.105.096347 pmid: 16299187
38 JP Luyendyk, LD Lehman-McKeeman, DM Nelson, VM Bhaskaran, TP Reilly, BD Car, GH Cantor, X Deng, JF Maddox, PE Ganey, RA Roth. Coagulation-dependent gene expression and liver injury in rats given lipopolysaccharide with ranitidine but not with famotidine. J Pharmacol Exp Ther 2006; 317(2): 635–643
https://doi.org/10.1124/jpet.105.096305 pmid: 16401727
39 X Deng, RF Stachlewitz, MJ Liguori, EA Blomme, JF Waring, JP Luyendyk, JF Maddox, PE Ganey, RA Roth. Modest inflammation enhances diclofenac hepatotoxicity in rats: role of neutrophils and bacterial translocation. J Pharmacol Exp Ther 2006; 319(3): 1191–1199
https://doi.org/10.1124/jpet.106.110247 pmid: 16990511
40 S Lacour, JC Gautier, M Pallardy, R Roberts. Cytokines as potential biomarkers of liver toxicity. Cancer Biomark 2005; 1(1): 29–39
https://doi.org/10.3233/CBM-2005-1105 pmid: 17192030
41 RA Roth, PE Ganey. Animal models of idiosyncratic drug-induced liver injurycurrent status. Crit Rev Toxicol 2011; 41(9): 723–739
https://doi.org/10.3109/10408444.2011.575765 pmid: 21726137
42 AA Jesus, R Goldbach-Mansky. IL-1 blockade in autoinflammatory syndromes. Annu Rev Med 2014; 65(1): 223–244
https://doi.org/10.1146/annurev-med-061512-150641 pmid: 24422572
43 CA Dinarello, A Simon, JWM van der Meer. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012; 11(8): 633–652
https://doi.org/10.1038/nrd3800 pmid: 22850787
44 MSJ Mangan, EJ Olhava, WR Roush, HM Seidel, GD Glick, E Latz. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discov 2018; 17(8): 588–606
https://doi.org/10.1038/nrd.2018.97 pmid: 30026524
45 H Guo, JB Callaway, JPY Ting. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 2015; 21(7): 677–687
https://doi.org/10.1038/nm.3893 pmid: 26121197
46 O Gross, CJ Thomas, G Guarda, J Tschopp. The inflammasome: an integrated view. Immunol Rev 2011; 243(1): 136–151
https://doi.org/10.1111/j.1600-065X.2011.01046.x pmid: 21884173
47 A Wree, MD McGeough, ME Inzaugarat, A Eguchi, S Schuster, CD Johnson, CA Peña, LJ Geisler, BG Papouchado, HM Hoffman, AE Feldstein. NLRP3 inflammasome driven liver injury and fibrosis: Roles of IL-17 and TNF in mice. Hepatology 2018; 67(2): 736–749
https://doi.org/10.1002/hep.29523 pmid: 28902427
48 JF Brouwers. Liquid chromatographic-mass spectrometric analysis of phospholipids. Chromatography, ionization and quantification. Biochim Biophys Acta 2011; 1811(11): 763–775
https://doi.org/10.1016/j.bbalip.2011.08.001 pmid: 21851861
49 K Sims, CA Haynes, S Kelly, JC Allegood, E Wang, A Momin, M Leipelt, D Reichart, CK Glass, MC Sullards, AH Merrill Jr. Kdo2-lipid A, a TLR4-specific agonist, induces de novo sphingolipid biosynthesis in RAW264.7 macrophages, which is essential for induction of autophagy. J Biol Chem 2010; 285(49): 38568–38579
https://doi.org/10.1074/jbc.M110.170621 pmid: 20876532
50 GF Nixon. Sphingolipids in inflammation: pathological implications and potential therapeutic targets. Br J Pharmacol 2009; 158(4): 982–993
https://doi.org/10.1111/j.1476-5381.2009.00281.x pmid: 19563535
51 H Le Stunff, S Milstien, S Spiegel. Generation and metabolism of bioactive sphingosine-1-phosphate. J Cell Biochem 2004; 92(5): 882–899
https://doi.org/10.1002/jcb.20097 pmid: 15258913
52 NA Babenko, EG Shakhova. Effects of flavonoids on sphingolipid turnover in the toxin-damaged liver and liver cells. Lipids Health Dis 2008; 7(1): 1–11
https://doi.org/10.1186/1476-511X-7-1 pmid: 18226198
53 SJ Sacket, DS Im. Activity change of sphingomyelin catabolic enzymes during dimethylnitrosamine-induced hepatic fibrosis in rats. Biomol Ther (Seoul) 2008; 16(16): 34–39
https://doi.org/10.4062/biomolther.2008.16.1.034
54 CA Rouzer, PT Ivanova, MO Byrne, HA Brown, LJ Marnett. Lipid profiling reveals glycerophospholipid remodeling in zymosan-stimulated macrophages. Biochemistry 2007; 46(20): 6026–6042
https://doi.org/10.1021/bi0621617 pmid: 17458939
55 FA Fitzpatrick, R Soberman. Regulated formation of eicosanoids. J Clin Invest 2001; 107(11): 1347–1351
https://doi.org/10.1172/JCI13241 pmid: 11390414
56 G Pérez-Chacón, AM Astudillo, D Balgoma, MA Balboa, J Balsinde. Control of free arachidonic acid levels by phospholipases A2 and lysophospholipid acyltransferases. Biochim Biophys Acta 2009; 1791(12): 1103–1113
https://doi.org/10.1016/j.bbalip.2009.08.007 pmid: 19715771
57 N Kawada, Y Mizoguchi, K Kobayashi, S Yamamoto, S Morisawa. Arachidonic acid metabolites in carbon tetrachloride-induced liver injury. Gastroenterol Jpn 1990; 25(3): 363–368
https://doi.org/10.1007/BF02779452 pmid: 2113499
58 N Piao, K Ikejima, K Kon, T Aoyama, T Osada, Y Takei, N Sato, S Watanabe. Synthetic triglyceride ocontaining an arachidonic acid branch (8A8) prevents lipopolysaccharide-induced liver injury. Life Sci 2009; 85(17-18): 617–624
https://doi.org/10.1016/j.lfs.2009.07.013 pmid: 19647752
59 A Ghigo, F Damilano, L Braccini, E Hirsch. PI3K inhibition in inflammation: Toward tailored therapies for specific diseases. BioEssays 2010; 32(3): 185–196
https://doi.org/10.1002/bies.200900150 pmid: 20162662
60 R Dhand, I Hiles, G Panayotou, S Roche, MJ Fry, I Gout, NF Totty, O Truong, P Vicendo, K Yonezawa. PI 3-kinase is a dual specificity enzyme: autoregulation by an intrinsic protein-serine kinase activity. EMBO J 1994; 13(3): 522–533
https://doi.org/10.1002/j.1460-2075.1994.tb06290.x pmid: 8313897
61 K Lehmann, JP Müller, B Schlott, P Skroblin, D Barz, J Norgauer, R Wetzker. PI3Kγ controls oxidative bursts in neutrophils via interactions with PKCα and p47phox. Biochem J 2009; 419(3): 603–610
https://doi.org/10.1042/BJ20081268 pmid: 18983267
[1] FMD-18057-OF-BZF_suppl_1 Download
[1] Chunyu Li, Ming Niu, Zhaofang Bai, Congen Zhang, Yanling Zhao, Ruiyu Li, Can Tu, Huifang Li, Jing Jing, Yakun Meng, Zhijie Ma, Wuwen Feng, Jinfa Tang, Yun Zhu, Jinjie Li, Xiaoya Shang, Zhengsheng Zou, Xiaohe Xiao, Jiabo Wang. Screening for main components associated with the idiosyncratic hepatotoxicity of a tonic herb, Polygonum multiflorum[J]. Front. Med., 2017, 11(2): 253-265.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed