Cargando…

Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation

BACKGROUND: Hyperuricemia (HUA) is a major public health issue with a high prevalence worldwide. Wuling San (WLS) is an effective treatment for HUA. However, the active compounds and the related mechanism are unclear. In this study, we aimed to explore the active compounds and the underlying pharmac...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Jing, Lin, Zhijian, Wang, Yu, Ding, Xueli, Zhang, Bing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9994669/
https://www.ncbi.nlm.nih.gov/pubmed/36911073
http://dx.doi.org/10.2147/DDDT.S398625
_version_ 1784902662962020352
author Huang, Jing
Lin, Zhijian
Wang, Yu
Ding, Xueli
Zhang, Bing
author_facet Huang, Jing
Lin, Zhijian
Wang, Yu
Ding, Xueli
Zhang, Bing
author_sort Huang, Jing
collection PubMed
description BACKGROUND: Hyperuricemia (HUA) is a major public health issue with a high prevalence worldwide. Wuling San (WLS) is an effective treatment for HUA. However, the active compounds and the related mechanism are unclear. In this study, we aimed to explore the active compounds and the underlying pharmacological mechanisms of WLS against HUA. METHODS: First, a network pharmacology approach was used to detect active compounds of WLS, and potential targets and signaling pathways involved in the treatment of HUA were predicted. Then, a molecular docking strategy was used to predict the affinity between active compounds and key targets. Finally, to verify the prediction, the HUA rat model was established. RESULTS: 49 active compounds with 108 common targets were obtained. Besides, cerevisterol, luteolin, ergosterol peroxide, beta-sitosterol, and sitosterol were identified as key active compounds. In PPI analysis, TNF, IL6, CASP3, PPARG, STAT3, and other 12 core targets were obtained. GO enrichment analysis indicated that WLS was likely to interfere with oxidative stress in the treatment of HUA, and KEGG enrichment analysis indicated multiple inflammation-related signaling pathways possibly involved in the treatment of HUA by WLS, including TNF, and NOD-like receptor, HIF-1, PI3K-Akt, and IL-17 signaling pathways. The results of molecular docking indicated that the active compounds had good binding properties to their key targets. In the validation experiments, WLS significantly reduced the levels of serum uric acid (SUA) and serum malondialdehyde (MDA). Moreover, WLS not only significantly increased the levels of total antioxidant capacity (T-AOC) and superoxide dismutase (SOD), but also inhibited the expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). CONCLUSION: In the present study, we demonstrate that WLS has multicomponent, multitarget, and multi-pathway properties in the treatment of HUA. Its potential capability to reduce SUA could be ascribed to oxidative stress improvement and inflammation inhibition.
format Online
Article
Text
id pubmed-9994669
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-99946692023-03-09 Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation Huang, Jing Lin, Zhijian Wang, Yu Ding, Xueli Zhang, Bing Drug Des Devel Ther Original Research BACKGROUND: Hyperuricemia (HUA) is a major public health issue with a high prevalence worldwide. Wuling San (WLS) is an effective treatment for HUA. However, the active compounds and the related mechanism are unclear. In this study, we aimed to explore the active compounds and the underlying pharmacological mechanisms of WLS against HUA. METHODS: First, a network pharmacology approach was used to detect active compounds of WLS, and potential targets and signaling pathways involved in the treatment of HUA were predicted. Then, a molecular docking strategy was used to predict the affinity between active compounds and key targets. Finally, to verify the prediction, the HUA rat model was established. RESULTS: 49 active compounds with 108 common targets were obtained. Besides, cerevisterol, luteolin, ergosterol peroxide, beta-sitosterol, and sitosterol were identified as key active compounds. In PPI analysis, TNF, IL6, CASP3, PPARG, STAT3, and other 12 core targets were obtained. GO enrichment analysis indicated that WLS was likely to interfere with oxidative stress in the treatment of HUA, and KEGG enrichment analysis indicated multiple inflammation-related signaling pathways possibly involved in the treatment of HUA by WLS, including TNF, and NOD-like receptor, HIF-1, PI3K-Akt, and IL-17 signaling pathways. The results of molecular docking indicated that the active compounds had good binding properties to their key targets. In the validation experiments, WLS significantly reduced the levels of serum uric acid (SUA) and serum malondialdehyde (MDA). Moreover, WLS not only significantly increased the levels of total antioxidant capacity (T-AOC) and superoxide dismutase (SOD), but also inhibited the expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). CONCLUSION: In the present study, we demonstrate that WLS has multicomponent, multitarget, and multi-pathway properties in the treatment of HUA. Its potential capability to reduce SUA could be ascribed to oxidative stress improvement and inflammation inhibition. Dove 2023-03-04 /pmc/articles/PMC9994669/ /pubmed/36911073 http://dx.doi.org/10.2147/DDDT.S398625 Text en © 2023 Huang et al. https://creativecommons.org/licenses/by-nc/3.0/This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/ (https://creativecommons.org/licenses/by-nc/3.0/) ). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Huang, Jing
Lin, Zhijian
Wang, Yu
Ding, Xueli
Zhang, Bing
Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title_full Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title_fullStr Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title_full_unstemmed Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title_short Wuling San Based on Network Pharmacology and in vivo Evidence Against Hyperuricemia via Improving Oxidative Stress and Inhibiting Inflammation
title_sort wuling san based on network pharmacology and in vivo evidence against hyperuricemia via improving oxidative stress and inhibiting inflammation
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9994669/
https://www.ncbi.nlm.nih.gov/pubmed/36911073
http://dx.doi.org/10.2147/DDDT.S398625
work_keys_str_mv AT huangjing wulingsanbasedonnetworkpharmacologyandinvivoevidenceagainsthyperuricemiaviaimprovingoxidativestressandinhibitinginflammation
AT linzhijian wulingsanbasedonnetworkpharmacologyandinvivoevidenceagainsthyperuricemiaviaimprovingoxidativestressandinhibitinginflammation
AT wangyu wulingsanbasedonnetworkpharmacologyandinvivoevidenceagainsthyperuricemiaviaimprovingoxidativestressandinhibitinginflammation
AT dingxueli wulingsanbasedonnetworkpharmacologyandinvivoevidenceagainsthyperuricemiaviaimprovingoxidativestressandinhibitinginflammation
AT zhangbing wulingsanbasedonnetworkpharmacologyandinvivoevidenceagainsthyperuricemiaviaimprovingoxidativestressandinhibitinginflammation