CC BY 4.0 · Chinese medicine and natural products 2022; 02(01): e32-e43
DOI: 10.1055/s-0042-1747918
Original Article

Potential Targets and Molecular Mechanism of Quercetin Against Knee Osteoarthritis

Lingling Li
1   College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Shandong, China
,
Hailiang Huang
1   College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Shandong, China
› Author Affiliations
Funding This work was funded by the Project of Chinese Medicine Science and Technology Development Planning in Shandong Province (2017-018), the Project of Scientific Research and Development Plan of Colleges and Universities in Shandong Province (J18KB130), and the Project of the First Batch of Excellent Scientific Research and Innovation Team in Shandong University of Chinese Medicine (220316).

Abstract

Objective The objective of this study was to clarify the potential mechanism of quercetin against knee osteoarthritis (KOA) based on network pharmacology and molecular docking.

Methods The targets of quercetin were predicted by PubChem and Swiss Target Prediction databases, and the targets of KOA were obtained by DisGeNET, OMIM, and GeneCards databases. Then, the targets of quercetin and KOA were intersected to find the potential targets of quercetin against KOA. The protein–protein interaction network was constructed through the STRING database, and the core targets were screened. Gene ontology (GO) functions enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed using DAVID database. The drug–target–pathway–disease network was constructed by Cytoscape software, and the molecular docking verification was performed by Vina.

Results There were 49 potential targets for quercetin against KOA, including 10 core targets. GO functions enrichment analysis showed that the biological process of quercetin against KOA mainly involved the negative regulation of apoptotic process, collagen catabolic process, and extracellular matrix disassembly. KEGG pathway enrichment analysis showed that quercetin against KOA was closely related to PI3K-Akt signaling pathway, Rap 1 signaling pathway, FoxO signaling pathway, Ras signaling pathway, TNF signaling pathway, and ErbB signaling pathway. The results of molecular docking showed that the binding energies between ligand and receptors were less than −5 kcal • mol−1.

Conclusions The molecular mechanism of quercetin against KOA involves many targets and pathways, which can regulate the proliferation and apoptosis of chondrocytes, degradation of extracellular matrix, and inflammatory reaction. Quercetin can stably bind to the active pockets of core target proteins, thereby exerting the effect against KOA.

Credit Autorship Contribution Statement

Lingling Li: Conceptualization, writing-original draft. Hailiang Huang: Methodology, supervision, and writing - review & editing.




Publication History

Received: 23 October 2021

Accepted: 23 January 2022

Article published online:
07 July 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Hügle T, Geurts J. What drives osteoarthritis?Synovial versus subchondral bone pathology. Rheumatology (Oxford) 2017; 56 (09) 1461-1471
  • 2 Madry H, Kon E, Condello V. et al. Early osteoarthritis of the knee. Knee Surg Sports Traumatol Arthrosc 2016; 24 (06) 1753-1762
  • 3 Lee BJ, Yang S, Kwon S, Choi KH, Kim W. Association between metabolic syndrome and knee osteoarthritis: a cross-sectional nationwide survey study. J Rehabil Med 2019; 51 (06) 464-470
  • 4 Roos EM, Arden NK. Strategies for the prevention of knee osteoarthritis. Nat Rev Rheumatol 2016; 12 (02) 92-101
  • 5 Kolasinski SL, Neogi T, Hochberg MC. et al. 2019 American college of rheumatology/arthritis foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis Care Res (Hoboken) 2020; 72 (02) 149-162
  • 6 Basu A, Schell J, Scofield RH. Dietary fruits and arthritis. Food Funct 2018; 9 (01) 70-77
  • 7 Wei B, Zhang Y, Tang L, Ji Y, Yan C, Zhang X. Protective effects of quercetin against inflammation and oxidative stress in a rabbit model of knee osteoarthritis. Drug Dev Res 2019; 80 (03) 360-367
  • 8 Zhang J, Yin J, Zhao D. et al. Therapeutic effect and mechanism of action of quercetin in a rat model of osteoarthritis. J Int Med Res 2020; 48 (03) 300060519873461
  • 9 Hu Y, Gui Z, Zhou Y, Xia L, Lin K, Xu Y. Quercetin alleviates rat osteoarthritis by inhibiting inflammation and apoptosis of chondrocytes, modulating synovial macrophages polarization to M2 macrophages. Free Radic Biol Med 2019; 145: 146-160
  • 10 Luo TT, Lu Y, Yan SK, Xiao X, Rong XL, Guo J. Network pharmacology in research of Chinese medicine formula: methodology, application and prospective. Chin J Integr Med 2020; 26 (01) 72-80
  • 11 Vakser IA. Protein-protein docking: from interaction to interactome. Biophys J 2014; 107 (08) 1785-1793
  • 12 Xue LC, Dobbs D, Bonvin AM, Honavar V. Computational prediction of protein interfaces: a review of data driven methods. FEBS Lett 2015; 589 (23) 3516-3526
  • 13 Kim S, Chen J, Cheng T. et al. PubChem in 2021: new data content and improved web interfaces. Nucleic Acids Res 2021; 49 (D1): D1388-D1395
  • 14 Daina A, Michielin O, Zoete V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res 2019; 47 (W1): W357-W364
  • 15 Wishart DS, Feunang YD, Guo AC. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res 2018; 46 (D1): D1074-D1082
  • 16 Uniprot C. UniProt Consortium. UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res 2019; 47 (D1): D506-D515
  • 17 Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J. et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res 2020; 48 (D1): D845-D855
  • 18 Amberger JS, Bocchini CA, Scott AF, Hamosh A. OMIM.org: leveraging knowledge across phenotype-gene relationships. Nucleic Acids Res 2019; 47 (D1): D1038-D1043
  • 19 Stelzer G, Rosen N, Plaschkes I. et al. The GeneCards suite: from gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics 2016; 54: 1 , 33
  • 20 Szklarczyk D, Gable AL, Lyon D. et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 2019; 47 (D1): D607-D613
  • 21 Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009; 4 (01) 44-57
  • 22 Burley SK, Bhikadiya C, Bi C. et al. RCSB Protein Data Bank: powerful new tools for exploring 3D structures of biological macromolecules for basic and applied research and education in fundamental biology, biomedicine, biotechnology, bioengineering and energy sciences. Nucleic Acids Res 2021; 49 (D1): D437-D451
  • 23 Mooers BHM. Shortcuts for faster image creation in PyMOL. Protein Sci 2020; 29 (01) 268-276
  • 24 El-Hachem N, Haibe-Kains B, Khalil A, Kobeissy FH, Nemer G. AutoDock and AutoDockTools for protein-ligand docking: beta-site amyloid precursor protein cleaving enzyme 1(BACE1) as a case study. Methods Mol Biol 2017; 1598: 391-403
  • 25 Tang X, Wang S, Zhan S. et al. The prevalence of symptomatic knee osteoarthritis in China: results from the China health and retirement longitudinal study. Arthritis Rheumatol 2016; 68 (03) 648-653
  • 26 Liu Y, Zhang H, Liang N. et al. Prevalence and associated factors of knee osteoarthritis in a rural Chinese adult population: an epidemiological survey. BMC Public Health 2016; 16: 94
  • 27 Cross M, Smith E, Hoy D. et al. The global burden of hip and knee osteoarthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis 2014; 73 (07) 1323-1330
  • 28 Hunter DJ, Schofield D, Callander E. The individual and socioeconomic impact of osteoarthritis. Nat Rev Rheumatol 2014; 10 (07) 437-441
  • 29 Zhang W, Bai Y, Wang Y, Xiao W. Polypharmacology in drug discovery: a review from systems pharmacology perspective. Curr Pharm Des 2016; 22 (21) 3171-3181
  • 30 Rose BJ, Kooyman DL. A tale of two joints: the role of matrix metalloproteases in cartilage biology. Dis Markers 2016; 2016: 4895050
  • 31 Cui N, Hu M, Khalil RA. Biochemical and biological attributes of matrix metalloproteinases. Prog Mol Biol Transl Sci 2017; 147: 1-73
  • 32 Lipari L, Gerbino A. Expression of gelatinases (MMP-2, MMP-9) in human articular cartilage. Int J Immunopathol Pharmacol 2013; 26 (03) 817-823
  • 33 Watanabe H, Ishii H, Takahashi K, Takai S, Ozawa H. Suitable reference gene selection for gene expression studies in knee osteoarthritis synovium using quantitative PCR analysis. Connect Tissue Res 2018; 59 (04) 356-368
  • 34 Cuesta C, Arévalo-Alameda C, Castellano E. The importance of being PI3K in the RAS signaling network. Genes (Basel) 2021; 12 (07) 1094
  • 35 Hao Q, Wang L, Tang H. Vascular endothelial growth factor induces protein kinase D-dependent production of proinflammatory cytokines in endothelial cells. Am J Physiol Cell Physiol 2009; 296 (04) C821-C827
  • 36 Pufe T, Harde V, Petersen W, Goldring MB, Tillmann B, Mentlein R. Vascular endothelial growth factor (VEGF) induces matrix metalloproteinase expression in immortalized chondrocytes. J Pathol 2004; 202 (03) 367-374
  • 37 Wu H, Xu T, Chen Z. et al. Specific inhibition of FAK signaling attenuates subchondral bone deterioration and articular cartilage degeneration during osteoarthritis pathogenesis. J Cell Physiol 2020; 235 (11) 8653-8666
  • 38 Zhang X, Zhu J, Liu F. et al. Reduced EGFR signaling enhances cartilage destruction in a mouse osteoarthritis model. Bone Res 2014; 2: 14015
  • 39 Ke H, Mou X, Xia Q. Remifentanil repairs cartilage damage and reduces the degradation of cartilage matrix in post-traumatic osteoarthritis, and inhibits IL-1β-induced apoptosis of articular chondrocytes via inhibition of PI3K/AKT/NF-κB phosphorylation. Ann Transl Med 2020; 8 (22) 1487
  • 40 Sun K, Luo J, Guo J, Yao X, Jing X, Guo F. The PI3K/AKT/mTOR signaling pathway in osteoarthritis: a narrative review. Osteoarthritis Cartilage 2020; 28 (04) 400-409
  • 41 Matsuzaki T, Alvarez-Garcia O, Mokuda S. et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci Transl Med 2018; 10 (428) eaan0746
  • 42 Akasaki Y, Alvarez-Garcia O, Saito M, Caramés B, Iwamoto Y, Lotz MK. FoxO transcription factors support oxidative stress resistance in human chondrocytes. Arthritis Rheumatol 2014; 66 (12) 3349-3358
  • 43 Xue JF, Shi ZM, Zou J, Li XL. Inhibition of PI3K/AKT/mTOR signaling pathway promotes autophagy of articular chondrocytes and attenuates inflammatory response in rats with osteoarthritis. Biomed Pharmacother 2017; 89: 1252-1261
  • 44 Choi MC, Jo J, Park J, Kang HK, Park Y. NF-κB signaling pathways in osteoarthritic cartilage destruction. Cells 2019; 8 (07) 734
  • 45 Looi CK, Hii LW, Ngai SC, Leong CO, Mai CW. The role of Ras-associated protein 1 (Rap1) in cancer: bad actor or good player?. Biomedicines 2020; 8 (09) E334
  • 46 Krygowska AA, Castellano E. PI3K: a crucial piece in the RAS signaling puzzle. Cold Spring Harb Perspect Med 2018; 8 (06) a031450
  • 47 Soliman AM, Alqahtani AS, Ghorab M. Novel sulphonamide benzoquinazolinones as dual EGFR/HER2 inhibitors, apoptosis inducers and radiosensitizers. J Enzyme Inhib Med Chem 2019; 34 (01) 1030-1040