AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (23.7 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Identification and mechanism elucidation of medicative diet for food therapy XQCSY in NAFLD prevention: an integrative in silico study

Chen-Xi Ren1Ming-Yuan Gao1Ning Li1Chao Tang1Gang-Hui Chu2Abdulla Yusuf2Li-Xia Xiao1Zhen-Quan Yang1Tian-Zhu Guan1,2( )
School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashi University, Kashi 844000, China
Show Author Information

Highlights

(1) Network pharmacology and molecular docking were performed to study the role of XingQiChuShiYin (XQCSY) in preventing non-alcoholic fatty liver disease (NAFLD).

(2) The compound-target-disease network screened out six potential hub targets based on the cluster screening, GO, and KEGG analysis.

(3) Observations of the targets-active ingredients docking behavior and affinity indicate that XQCSY is well affinitive to the potential hub genes and thus greatly affects NAFLD prevention.

(4) This work may present a theoretical proof to deeply clarify the main bioactive constituents and mechanisms of XQCSY and its multi-component, multi-target and multi-pathway properties in NAFLD prevention.

Graphical Abstract

With remarkable efficacy and small side effects, Conventional Chinese medicine dietary therapy is an ideal treatment for various diseases. As a conventional Chinese medicine dietary therapy, the effective constituents and functional mechanisms of XingQiChuShiYin (XQCSY) in preventing non-alcoholic fatty liver disease (NAFLD) are yet vague. Network pharmacology and molecular docking reveal that XQCSY plays a pivotal role in preventing NAFLD with a “multicomponent-multitarget-multichannel” mechanism. Molecular docking also confirms active ingredients from XQCSY are well affinitive to the key genes and thus greatly affect NAFLD prevention. In conclusion, this research may provide a vital theoretical basis for determining the prevention mechanism of XQCSY on NAFLD.

Abstract

XingQiChuShiYin (XQCSY), a conventional Chinese medicine dietary therapy, is effective in preventing non-alcoholic fatty liver disease (NAFLD), but its effective constituents and functional mechanisms are yet vague for the reason conventional modes are not suitable. Therefore, this study is centered on exploring the NAFLD prevention mechanisms of XQCSY via an in-silico method. The potential targets of XQCSY and NAFLD are obtained from public databases, and there are 81 intersected targets. Then six key targets including RAC-alpha serine/threonine-protein kinase, tumor necrosis factor, interleukin-6, interleukin-1 beta, cellular tumor antigen p53, and epidermal growth factor receptor are screened from the “herb-compound-common target” network. Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis reveals that 513 of biological processes, 51 of cellular components, and 90 of molecular functions in XQCSY are related to the treatment of NAFLD. Results from network pharmacology show that XQCSY plays a pivotal role in preventing NAFLD with a “multicomponent-multitarget-multichannel” mechanism. Molecular docking also confirms active ingredients from XQCSY are well affinitive to the key genes and thus greatly affect NAFLD prevention. In conclusion, this research may provide a vital theoretical basis for determining the prevention mechanism of XQCSY on NAFLD.

Electronic Supplementary Material

Download File(s)
FMH-2024-9420015_ESM.pdf (106.9 KB)

References

[1]

Klein, R. J., Viana, R. G. M., Rotman, Y., et al. Divergent pathways of liver fat accumulation, oxidation, and secretion in lipodystrophy versus obesity-associated NAFLD. Liver International: Official Journal of the International Association for the Study of the Liver, 2023, 43: 2692–2700. https://doi.org/10.1111/LIV.15707

[2]

Fan, J. G., Farrell, G. C. Epidemiology of non-alcoholic fatty liver disease in China. Journal of Hepatology, 2009, 50: 204–210. https://doi.org/10.1016/j.jhep.2008.10.010

[3]

Diehl, A. M., Day, C. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. The New England Journal of Medicine, 2017, 377: 2063–2072. https://doi.org/10.1056/NEJMra1503519

[4]

Wang, F. S., Fan, J. G., Zhang, Z., et al. The global burden of liver disease: the major impact of China. Hepatology (Baltimore, Md.), 2014, 60: 2099–2108. https://doi.org/10.1002/hep.27406

[5]

Christian, T., Christopher, P. D., Michael, I. T. Lifestyle interventions for the treatment of non-alcoholic fatty liver disease in adults: a systematic review. Journal of Hepatology, 2012, 56: 255–266. https://doi.org/10.1016/j.jhep.2011.06.010

[6]

Li, H., Wang, Q. L., Chen, P. Z., et al. Ursodeoxycholic acid treatment restores gut microbiota and alleviates liver inflammation in non-alcoholic steatohepatitic mouse model. Frontiers in Pharmacology, 2021, 12: 788558. https://doi.org/10.3389/FPHAR.2021.788558

[7]

Wang, M. Y., Prabahar, K., Găman, M. A., et al. Vitamin E supplementation in the treatment on non-alcoholic fatty liver disease (NAFLD): evidence from an umbrella meta-analysis on randomized controlled trials. Journal of Digestive Diseases, 2023, 24: 380–389. https://doi.org/10.1111/1751-2980.13210

[8]

Lombardi, R., Onali, S., Thorburn, D., et al. Pharmacological interventions for non-alcohol related fatty liver disease (NAFLD): an attempted network meta-analysis. The Cochrane Database of Systematic Reviews, 2017, 3: CD011640. https://doi.org/10.1002/14651858.CD011640.PUB2

[9]

Li, G. D., Liu, H., Feng, R. B., et al. A bioactive ligand-conjugated iridium(III) metal-based complex as a Keap1-Nrf2 protein-protein interaction inhibitor against acetaminophen-induced acute liver injury. Redox Biology, 2021, 48: 102129. https://doi.org/10.1016/J.REDOX.2021.102129

[10]

Chen, F. F., Huang, Y. F. Observation on therapeutic effect of JiaWeiZhaQuPingWeiSan on nonalcoholic fatty liver disease. Chinese Journal of Integrated Traditional and Western Medicine on Digestion, 2015, 23: 817–819.

[11]

Fan, Y. M., Zhao, Q. Y., Wei, Y. Y., et al. Pingwei San ameliorates spleen deficiency-induced diarrhea through intestinal barrier protection and gut microbiota modulation. Antioxidants, 2023, 12: 1122. https://doi.org/10.3390/ANTIOX12051122

[12]
Zhang, W. J., Zhao, Z. Y., Chang, L. K., et al. Atractylodis rhizoma: a review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control. Journal of Ethnopharmacology, 2021 , 266: 113415. https://doi.org/10.1016/j.jep.2020.113415
[13]

Chen, X. M., Tait, A. R., Kitts, D. D. Flavonoid composition of orange peel and its association with antioxidant and anti-inflammatory activities. Food Chemistry, 2017, 218: 15–21. https://doi.org/10.1016/j.foodchem.2016.09.016

[14]
Kim, K. H., Moon, E., Kim, S. Y., et al. 4-Methylthio-butanyl derivatives from the seeds of Raphanus sativus and their biological evaluation on anti-inflammatory and antitumor activities. Journal of Ethnopharmacology, 2014 , 151: 503–508. https://doi.org/10.1016/j.jep.2013.11.003
[15]

Sham, T. T., Ailsa, C. Y. Y., Ng, Y. F., et al. A review of the phytochemistry and pharmacological activities of raphani semen. Evidence-based Complementary and Alternative Medicine : eCAM, 2013: 636194. https://doi.org/10.1155/2013/636194

[16]

Ye, Q. Y., Jiang, Y., Wu, D., et al. Atractylodin alleviates nonalcoholic fatty liver disease by regulating Nrf2-mediated ferroptosis. Heliyon, 2023, 9: e18321. https://doi.org/10.1016/J.HELIYON.2023.E18321

[17]

Ke, Z. L., Zhao, Y. Y., Tan, S., et al. Citrus reticulata blanco peel extract ameliorates hepatic steatosis, oxidative stress and inflammation in HF and MCD diet-induced NASH C57BL/6 J mice. The Journal of Nutritional Biochemistry, 2020, 83: 108426. https://doi.org/10.1016/j.jnutbio.2020.108426

[18]

Park, W. Y., Song, G., Noh, J. H., et al. Raphani Semen ( Raphanus sativus L.) ameliorates alcoholic fatty liver disease by regulating de novo lipogenesis. Nutrients, 2021, 13: 4448. https://doi.org/10.3390/NU13124448

[19]

Yuen, H., Hong, Y. A. W., Hung, A., et al. How does traditional knowledge of Cassiae Semen shed light on weight management?–a classical and modern literature review. Journal of Ethnopharmacology, 2020, 268: 113572. https://doi.org/10.1016/j.jep.2020.113572

[20]

Zhang, R., Wu, H. P., Lian, Z. X. Bioinformatics analysis of evolutionary characteristics and biochemical structure of FGF5 gene in sheep. Gene, 2019, 702: 123–132. https://doi.org/10.1016/j.gene.2019.03.040

[21]

Xu, Z. J., Lin, S. S., Gong, J. J., et al. Exploring the protective effects and mechanism of crocetin from saffron against NAFLD by network pharmacology and experimental validation. Frontiers in Medicine, 2021, 8: 681391. https://doi.org/10.3389/FMED.2021.681391

[22]

Hong, W., Li, S. S., Wu, L. Y., et al. Prediction of VEGF-C as a key target of pure total flavonoids from citrus against NAFLD in mice via network pharmacology. Frontiers in Pharmacology, 2019, 10: 582. https://doi.org/10.3389/fphar.2019.00582

[23]

Jing, C. Y., Sun, Z. L., Xie, X. F., et al. Network pharmacology-based identification of the key mechanism of Qinghuo Rougan Formula acting on uveitis. Biomedicine & Pharmacotherapy, 2019, 120: 109381. https://doi.org/10.1016/j.biopha.2019.109381

[24]
Krau, S. D. The role of pharmacogenomics: the same medications do not work the same on everyone. Nursing Clinics of North America, 2016 , 51. https://doi.org/10.1016/j.cnur.2015.12.002
[25]

Ng, C. Y. J., Bun, H. H., Zhao, Y., et al. TCM “medicine and food homology” in the management of post-COVID disorders. Frontiers in Immunology, 2023, 14: 1234307. https://doi.org/10.3389/fimmu.2023.1234307

[26]

Crichton, G. E., Howe, P. R. C., Buckley, J. D., et al. Long-term dietary intervention trials: critical issues and challenges. Trials, 2012, 13: 111. https://doi.org/10.1186/1745-6215-13-111

[27]

Shi, Y., Wu, L. Q., Zhang, N. N., et al. Exploring the mechanism of Pericarpium citri reticulatae and Fritillaria thunbergii in the treatment of adenoid hypertrophy based on network pharmacology and molecular docking. Journal of Contemporary Medical Practice, 2022, 4: 27. https://doi.org/10.53469/JCMP.2022.04(03).27

[28]

Jiang, S. S., Huang, C. M., Wang, S. L., et al. Network pharmacology-based strategy for predicting therapy targets of Citri reticulatae pericarpium on myocardial hypertrophy. BioMed Research International, 2022, 2022: 4293265. https://doi.org/10.1155/2022/4293265

[29]

Yu, Z. Y., Wu, Y. L., Ma, Y. J., et al. Systematic analysis of the mechanism of aged citrus peel (Chenpi) in oral squamous cell carcinoma treatment via network pharmacology, molecular docking and experimental validation. Journal of Functional Foods, 2022, 91: 105012. https://doi.org/10.1016/J.JFF.2022.105012

[30]

Zhou, L., Gu, W. W., Kui, F. G., et al. The mechanism and candidate compounds of aged citrus peel (chenpi) preventing chronic obstructive pulmonary disease and its progression to lung cancer. Food & Nutrition Research, 2021, 65: 7526. https://doi.org/10.29219/ FNR.V65.7526

[31]

Zhao, C., Zhi, C. Y., Zhou, J. H. Mechanism of Jiawei Zhengqi Powder in the treatment of ulcerative colitis based on network pharmacology and molecular docking. BioMed Research International, 2023: 8397111. https://doi.org/10.1155/2023/8397111

[32]

Zeng, Q., Li, L. F., Siu, W. S., et al. A combined molecular biology and network pharmacology approach to investigate the multi-target mechanisms of Chaihu Shugan San on Alzheimer’s disease. Biomedicine & Pharmacotherapy, 2019, 120: 109370. https://doi.org/ 10.1016/j.biopha.2019.109370

[33]

Guo, D. H., Hao, J. P., Li, X. J., et al. Exploration in the mechanism of Zhisou San for the treatment of cough variant asthma based on network pharmacology. Evidence-Based Complementary and Alternative Medicine, 2022: 1698571. https://doi.org/10.1155/2022/1698571

[34]

Wang, M. Q., Gu, W. W., Kui, F. G., et al. The clinical efficiency and the mechanism of Sanzi Yangqin Decoction for chronic obstructive pulmonary disease. Evidence-Based Complementary and Alternative Medicine, 2021: 5565562. https://doi.org/10.1155/2021/5565562

[35]

Xie, F. Q., Zheng, W. J., Yan, Q., et al. Network pharmacology-deciphering the molecular mechanism of San-Zi-Yang-Qin decoction for the treatment of chronic obstructive pulmonary disease. European Journal of Integrative Medicine, 2019, 31: 100962. https://doi.org/10.1016/j.eujim.2019.100962

[36]

Zhong, Y., Chen, R. F., Fang, Y. F. Network pharmacology-based strategy to reveal the mechanism of Cassiae semen against cataracts. Computational and Mathematical Methods in Medicine, 2022: 5654120. https://doi.org/10.1155/2022/5654120

[37]

Huang, L. L., Zhu, H. Y., Tang, Y. Q., et al. Exploring the mechanism of Cassiae semen in regulating lipid metabolism through network pharmacology and experimental validation. Bioscience Reports, 2023, 43: 1375. https://doi.org/10.1042/BSR20221375

[38]

Chen, C., Zhou, S., Meng, Q. G. A molecular docking study of Rhizoma Atractylodis and Rhizoma Atractylodis Macrocephalae herbal pair with respect to type 2 diabetes mellitus. Journal of Traditional Chinese Medical Sciences, 2018, 5: 185–198. https://doi.org/10.1016/j.jtcms.2018.05.004

[39]

Nguyen, L. T. H., Nguyen, N. P. K., Tran, K. N., et al. Network pharmacology and experimental validation to investigate the antidepressant potential of Atractylodes lancea (Thunb.) DC. Life (Basel, Switzerland), 2022, 12: 1925. https://doi.org/10.3390/LIFE12111925

[40]

Ye, J. H., Li, L., Hu, Z. X. Exploring the molecular mechanism of action of Yinchen Wuling Powder for the treatment of hyperlipidemia, using network pharmacology, molecular docking, and molecular dynamics simulation. BioMed Research International, 2021: 9965906. https://doi.org/10.1155/2021/9965906

[41]

Huang, Z. Q., Shi, X. Q., Li, X. C., et al. Network pharmacology approach to uncover the mechanism governing the effect of Simiao powder on knee osteoarthritis. BioMed Research International, 2020: 1503. https://doi.org/10.1155/2020/6971503

[42]

Wang, Y., Sun, Y., Wang, R. R., et al. Network pharmacology and molecular docking analysis on the pharmacological mechanisms of modified Sanmiaosan in treating ulcerative colitis. Computational and Mathematical Methods in Medicine, 2022: 2556521. https://doi.org/10.1155/2022/2556521

[43]

Zheng, L., Jiang, H. N., Li, R. Q., et al. The pharmacological mechanisms of Xiaochaihutang in treating breast cancer based on network pharmacology. Contrast Media & Molecular Imaging, 2022: 3900636.

[44]

The UniProt Consortium. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Research, 2020, 51: 1100. https://doi.org/10.1093/NAR/GKAA1100

[45]

Piñero, J., Saüch, J., Sanz, F., et al. The disGeNET cytoscape app: exploring and visualizing disease genomics data. Computational and Structural Biotechnology Journal, 2021, 19: 2960–2967. https://doi.org/10.1016/J.CSBJ.2021.05.015

[46]
Chin, C. H., Chen, S. H., Wu, H. H., et al. cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Systems Biology, 2014 , 8: S11. https://doi.org/10.1186/1752-0509-8-S4-S11
[47]

Giovanni, S., Gabriele, T., Mohammed, F., et al. Biological network analysis with centiscape: centralities and experimental dataset integration. F1000Research, 2014, 3: 139. https://doi.org/10.12688/f1000research.4477.2

[48]

Chen, R. Y., Guan, Z. Y., Zhong, X. X., et al. Network pharmacology prediction: the possible mechanisms of cinobufotalin against osteosarcoma. Computational and Mathematical Methods in Medicine, 2022: 3197402. https://doi.org/10.1155/2022/3197402

[49]

Bindea, G., Mlecnik, B., Hackl, H., et al. ClueGO: a cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics (Oxford, England), 2009, 25: 1091–1093. https://doi.org/10.1093/bioinformatics/btp101

[50]

Kanehisa, M., Goto, S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Research, 2000, 28: 27–30. https://doi.org/10.1093/nar/28.1.27

[51]

Li, S. A., Du, G. G., Shi, J. J., et al. Preparation of antihypertensive peptides from quinoa via fermentation with Lactobacillus paracasei. eFood, 2022, 3: 20. https://doi.org/10.1002/EFD2.20

[52]

Hou, C. L., Shi, C. C., Ren, J. Y. Xanthine oxidase targeted model setup and its application for antihyperuricemic compounds prediction by in silico methods. eFood, 2022, 2: 296–306. https://doi.org/10.53365/EFOOD.K/147019

[53]

Feng, S. M., Dai, Z. Q., Liu, A. B., et al. Intake of stigmasterol and β-sitosterol alters lipid metabolism and alleviates NAFLD in mice fed a high-fat Western-style diet. BBA-Molecular and Cell Biology of Lipids, 2018, 1863: 1274–1284. https://doi.org/10.1016/j.bbalip.2018.08.004

[54]

Chen, J., Liu, J., Wang, Y., et al. Wogonin mitigates nonalcoholic fatty liver disease via enhancing PPARα/AdipoR2, in vivo and in vitro. Biomedicine & Pharmacotherapy, 2017, 91: 621–631. https://doi.org/10.1016/j.biopha.2017.04.125

[55]

Deng, C. R., Gao, C. Y., Tian, X. H., et al. Pharmacokinetics, tissue distribution and excretion of luteolin and its major metabolites in rats: metabolites predominate in blood, tissues and are mainly excreted via bile. Journal of Functional Foods, 2017, 35: 332–340. https://doi.org/10.1016/j.jff.2017.05.056

[56]

Tu, Y. F., Zhou, L., Li, L., et al. Development and validation of an LC-MS/MS method for the quantification of flavonoid glucuronides (wogonoside, baicalin, and apigenin-glucuronide) in the bile and blood samples: application to a portal vein infusion study. Analytical Biochemistry, 2020, 601: 113723. https://doi.org/10.1016/j.ab.2020.113723

[57]

Batta, A. K., Xu, G., Honda, A., et al. Stigmasterol reduces plasma cholesterol levels and inhibits hepatic synthesis and intestinal absorption in the rat. Metabolism, 2006, 55: 292–299. https://doi.org/10.1016/j.metabol.2005.08.024

[58]

Kotani, A., Nagami, K., Mino, C., et al. Determination of nobiletin in rat plasma after ingestion of citrus depressa juice by capillary liquid chromatography with electrochemical detection using boron-doped diamond electrode. Electrochemistry, 2015, 83: 363–367. https://doi.org/10.5796/electrochemistry.83.363

[59]

Yu, C. P., Shia, C. S., Lin, H. J., et al. Analysis of the pharmacokinetics and metabolism of aloe-emodin following intravenous and oral administrations in rats. Biomedical Chromatography, 2016, 30: 1641–1647. https://doi.org/10.1002/bmc.3735

[60]

Ito, N., Hakamata, H., Kusu, F. Simultaneous determination of β-sitosterol, campesterol, stigmasterol, and brassicasterol in serum by high-performance liquid chromatography with electrochemical detection. Analytical Methods, 2010, 2: 174–179. https://doi.org/10.1039/B9AY00195F

[61]

Su, H., Liu, R. J., Chang, M., et al. Effect of dietary alpha-linolenic acid on blood inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. European Journal of Nutrition, 2018, 57: 877–891. https://doi.org/10.1007/s00394-017-1386-2

[62]

Xu, S. Z., Qi, X. J., Liu, Y. Q., et al. UPLC-MS/MS of atractylenolide I, atractylenolide II, atractylenolide III, and atractyloside A in rat plasma after oral administration of raw and wheat bran-processed atractylodis rhizoma. Molecules, 2018, 23: 3234. https://doi.org/10.3390/molecules23123234

[63]

Xie, X., Yan, D., Li, H. B., et al. Enhancement of adiponectin ameliorates nonalcoholic fatty liver disease via inhibition of FoxO1 in type I diabetic rats. Journal of Diabetes Research, 2018: 6254340.

[64]

Hao, W. T., Biao, L. J., Gang, T. Y., et al. Association of TNF-α, IGF-1, and IGFBP-1 levels with the severity of osteopenia in mice with nonalcoholic fatty liver disease. Journal of Orthopaedic Surgery and Research, 2023, 18: 915. https://doi.org/10.1186/S13018-023-04385-1

[65]

Mei, D. Y., Feng, P. X., You, L. J., et al. Association of inflammatory cytokines with non-alcoholic fatty liver disease. Frontiers in Immunology, 2022, 13: 880298. https://doi.org/10.3389/FIMMU.2022.880298

[66]

Nelson J. E., Handa P., Aouizerat B., et al. Increased parenchymal damage and steatohepatitis in Caucasian non-alcoholic fatty liver disease patients with common IL1B and IL6 polymorphisms. Alimentary Pharmacology & Therapeutics, 2016, 44: 1253–1264. https://doi.org/10.1111/apt.13824

[67]

Fang, C. Y., Cai, X. B., Hayashi, S., et al. Caffeine-stimulated muscle IL-6 mediates alleviation of non-alcoholic fatty liver disease. BBA-Molecular and Cell Biology of Lipids, 2018, 1864: 271–280. https://doi.org/10.1016/j.bbalip.2018.12.003

[68]

Liu, X. L., Pan, Q., Cao, H. X., et al. Lipotoxic hepatocyte-derived exosomal microRNA 192-5p activates macrophages through Rictor/Akt/Forkhead Box transcription factor O1 signaling in nonalcoholic fatty liver disease. Hepatology, 2020, 72: 454–469. https://doi.org/10.1002/hep.31050

[69]

NasiriAnsari, N., Nikolopoulou, C., Papoutsi, K., et al. Empagliflozin attenuates non-alcoholic fatty liver disease (NAFLD) in high fat diet fed ApoE−/− mice by activating autophagy and reducing ER stress and apoptosis. International Journal of Molecular Sciences, 2021, 22: 818. https://doi.org/10.3390/IJMS22020818

[70]

Clémence, R., Anne, F., Sandrine, E., et al. Oxidative stress in NAFLD: role of nutrients and food contaminants. Biomolecules, 2020, 10: 1702. https://doi.org/10.3390/BIOM10121702

[71]
Duan, Y. L., Yang, Y., Zhao, S. Q., et al. Crosstalk in extrahepatic and hepatic system in NAFLD/NASH. Liver International: Official Journal of the International Association for the Study of the Liver, 2024 : 15967. https://doi.org/10.1111/LIV.15967
[72]
Michael, S., Jennifer, N., Shirihai, O. S., et al. Mitochondrial oxidative function in NAFLD: friend or foe? Molecular Metabolism, 2020 , 50: 101134. https://doi.org/10.1016/J.MOLMET.2020.101134
[73]

Hernández, A., Arab, J. P., Reyes, D., et al. Extracellular vesicles in NAFLD/ALD: from pathobiology to therapy. Cells, 2020, 9: 817. https://doi.org/10.3390/cells9040817

[74]

Ferreira, D. F., Fiamoncini, J., Prist, I. H., et al. Novel role of TLR4 in NAFLD development: modulation of metabolic enzymes expression. BBA-Molecular and Cell Biology of Lipids, 2015, 1851: 1353–1359. https://doi.org/10.1016/j.bbalip.2015.07.002

[75]

Ahmed, M. H., Byrne, C. D. Modulation of sterol regulatory element binding proteins (SREBPs) as potential treatments for non-alcoholic fatty liver disease (NAFLD). Drug Discovery Today, 2007, 12: 740–747. https://doi.org/10.1016/j.drudis.2007.07.009

[76]

Asadipooya, K., Lankarani, K. B., Raj, R., et al. RAGE is a potential cause of onset and progression of nonalcoholic fatty liver disease. International Journal of Endocrinology, 2019: 2151302. https://doi.org/10.1155/2019/2151302

[77]

Deprince, A., Haas, J. T., Staels, B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Molecular Metabolism, 2020, 42: 101092. https://doi.org/10.1016/j.molmet.2020.101092

[78]
Zhang, J., Liang, Y., Ren, L., et al. In vitro anti-inflammatory potency of sanguinarine and chelerythrine via interaction with glucocorticoid receptor. eFood, 2021 , 40: 496-508. https://doi.org/10.2991/EFOOD.K.210118.001
[79]
Li, T. H., Shi, L. L., Liu, W. Q., et al. Aloe-emodin induces mitochondrial dysfunction and pyroptosis by activation of the Caspase-9/3/Gasdermin E axis in HeLa cells. Frontiers in Pharmacology, 2022 : 13854526. https://doi.org/10.3389/fphar.2022.854526
[80]

Sampath, S. J. P., Rath, S. N., Kotikalapudi, N., et al. Beneficial effects of secretome derived from mesenchymal stem cells with stigmasterol to negate IL-1β-induced inflammation in-vitro using rat chondrocytes-OA management. Inflammopharmacology, 2021, 29: 1701–1717. https://doi.org/10.1007/s10787-021-00874-z

[81]

Eun, S. H., Woo, J. T., Kim, D. H. Tangeretin inhibits IL-12 expression and NF-κB activation in dendritic cells and attenuates colitis in mice. Planta Medica, 2016, 83: 527–533. https://doi.org/10.1055/s-0042-119074

Food & Medicine Homology
Article number: 9420015

{{item.num}}

Comments on this article

Go to comment

< Back to all reports

Review Status: {{reviewData.commendedNum}} Commended , {{reviewData.revisionRequiredNum}} Revision Required , {{reviewData.notCommendedNum}} Not Commended Under Peer Review

Review Comment

Close
Close
Cite this article:
Ren C-X, Gao M-Y, Li N, et al. Identification and mechanism elucidation of medicative diet for food therapy XQCSY in NAFLD prevention: an integrative in silico study. Food & Medicine Homology, 2024, 1(2): 9420015. https://doi.org/10.26599/FMH.2024.9420015

1619

Views

402

Downloads

8

Crossref

Altmetrics

Received: 09 June 2024
Revised: 02 July 2024
Accepted: 04 July 2024
Published: 04 August 2024
© National R & D Center for Edible Fungus Processing Technology 2024. Published by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).