Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter December 14, 2020

Molecular docking study of flavonoid compounds for possible matrix metalloproteinase-13 inhibition

  • Amir Taherkhani , Shirin Moradkhani , Athena Orangi , Alireza Jalalvand and Zahra Khamverdi EMAIL logo

Abstract

Objectives

Matrix metalloproteinase-13 (MMP-13) has been reported to be involved in different biological processes such as degradation of extracellular matrix proteins, activating or degrading some significant regulatory proteins, wound healing, tissue remodeling, cartilage degradation, bone development, bone mineralization, ossification, cell migration, and tumor cell invasion. Further, MMP-13 participates in many oral diseases such as tooth decay, gingivitis, and degradation of enamel and tissue around the implant. In addition, inhibition of MMP-13 has shown therapeutic properties for Alzheimer’s disease (AD). We performed molecular docking to assess the binding affinity of 29 flavonoid compounds with the MMP-13. Additionally, pharmacokinetic and toxicity characteristics of the top-ranked flavonoids were studied. The current study also intended to identify the most important amino acids involved in the inhibition of MMP-13 based on topological feature (degree) in the ligand-amino acid network for MMP-13.

Methods

Molecular docking and network analysis were studied using AutoDock and Cytoscape software, respectively. Pharmacokinetic and toxicity characteristics of compounds were predicted using bioinformatics web tools.

Results

The results revealed that nine of the studied flavonoids had considerable estimated free energy of binding and inhibition constant: Rutin, nicotiflorin, orientin, vitexin, apigenin-7-glucoside, quercitrin, isoquercitrin, quercitrin-3-rhamnoside, and vicenin-2. Proline-242 was found to be the most important amino acid inhibiting the enzyme.

Conclusions

The results of the current study may be helpful in the prevention and therapeutic procedures of many disorders such as cancer, tooth caries, and AD. Nevertheless, validation tests are required in the future.


Corresponding author: Zahra Khamverdi, Department of Restorative Dentistry, Dental Research Center, Dental School, Hamadan University of Medical Sciences, Hamadan, Iran, Phone: +98-9183122095, Fax: +98-8138381085, E-mail:

Acknowledgments

The authors would like to appreciate the Deputy of Research and Technology, Dental Research Center and Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan – Iran, for their supports.

  1. Research funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

  2. Author contributions: AT, ZKH, and SHM designed the study. Energy minimization of the protein and small molecules was performed by AT and AO, respectively. Docking operations were conducted by AT. Molecular dynamics simulations were performed by AJ and AT. The ADME and toxicity studies were conducted by AT, ZKH, AO, SHM, and AJ. The network analysis was conducted and interpreted by AT. The results were analyzed and discussed by AT, ZKH, AO, SHM, and AJ. AT and SHM wrote the manuscript. All authors read and approved the final version of the manuscript.

  3. Competing interests: The authors declare that they have no competing interests.

  4. Availability of data and materials: The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

  5. Ethics approval and consent to participate: The present study was approved by the Ethics Committee of Hamadan University of Medical Sciences, Hamadan, Iran (ethics no. IR.UMSHA.REC.1398.576).

  6. Consent for publication: Not applicable.

References

1. Consortium, U. UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res 2018;47:D506–15.10.1093/nar/gky1049Search in Google Scholar

2. Freije, JM, Diez-Itza, I, Balbín, M, Sánchez, LM, Blasco, R, Tolivia, J, et al.. Molecular cloning and expression of collagenase-3, a novel human matrix metalloproteinase produced by breast carcinomas. J Biol Chem 1994;269:16766–73.10.1016/S0021-9258(19)89457-7Search in Google Scholar

3. Knäuper, V, López-Otin, C, Smith, B, Knight, G, Murphy, G. Biochemical characterization of human collagenase-3. J Biol Chem 1996;271:1544–50. https://doi.org/10.1074/jbc.271.3.1544.Search in Google Scholar

4. Knäuper, V, Will, H, López-Otin, C, Smith, B, Atkinson, SJ, Stanton, H, et al.. Cellular mechanisms for human procollagenase-3 (MMP-13) activation Evidence that MT1-MMP (MMP-14) and gelatinase a (MMP-2) are able to generate active enzyme. J Biol Chem 1996;271:17124–31. https://doi.org/10.1074/jbc.271.29.17124.Search in Google Scholar

5. Fosang, AJ, Last, K, Knäuper, V, Murphy, G, Neame, PJ. Degradation of cartilage aggrecan by collagenase‐3 (MMP‐13). FEBS Lett 1996;380:17–20. https://doi.org/10.1016/0014-5793(95)01539-6.Search in Google Scholar

6. Knäuper, V, Cowell, S, Smith, B, López-Otin, C, O’Shea, M, Morris, H, et al.. The role of the C-terminal domain of human collagenase-3 (MMP-13) in the activation of procollagenase-3, substrate specificity, and tissue inhibitor of metalloproteinase interaction. J Biol Chem 1997;272:7608–16. https://doi.org/10.1074/jbc.272.12.7608.Search in Google Scholar PubMed

7. Johnson, AR, Pavlovsky, AG, Ortwine, DF, Prior, F, Man, C-F, Bornemeier, DA, et al.. Discovery and characterization of a novel inhibitor of matrix metalloprotease-13 that reduces cartilage damage in vivo without joint fibroplasia side effects. J Biol Chem 2007;282:27781–91. https://doi.org/10.1074/jbc.m703286200.Search in Google Scholar

8. Becker, DP, Barta, TE, Bedell, LJ, Boehm, TL, Bond, BR, Carroll, J, et al.. Orally active MMP-1 sparing α-tetrahydropyranyl and α-piperidinyl sulfone matrix metalloproteinase (MMP) inhibitors with efficacy in cancer, arthritis, and cardiovascular disease. J Med Chem 2010;53:6653–80. https://doi.org/10.1021/jm100669j.Search in Google Scholar PubMed

9. Devel, L, Beau, F, Amoura, M, Vera, L, Cassar-Lajeunesse, E, Garcia, S, et al.. Simple pseudo-dipeptides with a P2′ glutamate a novel inhibitor family of matrix metalloproteases and other metzincins. J Biol Chem 2012;287:26647–56. https://doi.org/10.1074/jbc.m112.380782.Search in Google Scholar PubMed PubMed Central

10. De Savi, C, Waterson, D, Pape, A, Lamont, S, Hadley, E, Mills, M, et al.. Hydantoin based inhibitors of MMP13—discovery of AZD6605. Bioorg Med Chem Lett 2013;23:4705–12. https://doi.org/10.1016/j.bmcl.2013.05.089.Search in Google Scholar PubMed

11. Kennedy, AM, Inada, M, Krane, SM, Christie, PT, Harding, B, López-Otín, C, et al.. MMP13 mutation causes spondyloepimetaphyseal dysplasia, Missouri type (SEMD MO). J Clin Invest 2005;115:2832–42. https://doi.org/10.1172/jci22900.Search in Google Scholar

12. Lausch, E, Keppler, R, Hilbert, K, Cormier-Daire, V, Nikkel, S, Nishimura, G, et al.. Mutations in MMP9 and MMP13 determine the mode of inheritance and the clinical spectrum of metaphyseal anadysplasia. Am J Hum Genet 2009;85:168–78. https://doi.org/10.1016/j.ajhg.2009.06.014.Search in Google Scholar

13. Zhu, BL, Long, Y, Luo, W, Yan, Z, Lai, YJ, Zhao, LG, et al.. MMP13 inhibition rescues cognitive decline in Alzheimer transgenic mice via BACE1 regulation. Brain: J Neurol 2019;142:176–92. https://doi.org/10.1093/brain/awy305.Search in Google Scholar

14. Vos, T, Abajobir, AA, Abate, KH, Abbafati, C, Abbas, KM, Abd-Allah, F, et al.. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017;390:1211–59.10.1016/S0140-6736(17)32154-2Search in Google Scholar

15. Hedenbjörk-Lager, A, Bjørndal, L, Gustafsson, A, Sorsa, T, Tjäderhane, L, Åkerman, S, et al.. Caries correlates strongly with salivary levels of matrix metalloproteinase-8. Caries Res 2015;49:1–8. https://doi.org/10.1159/000360625.Search in Google Scholar PubMed

16. Loreto, C, Galanti, C, Musumeci, G, Rusu, M, Leonardi, R. Immunohistochemical analysis of matrix metalloproteinase-13 in human caries dentin. Eur J Histochem 2014;58:47–51. doi:https://doi.org/10.4081/ejh.2014.2318.Search in Google Scholar PubMed PubMed Central

17. de Oliveira Carrilho, MR, Tay, FR, Pashley, DH, Tjäderhane, L, Carvalho, RM. Mechanical stability of resin–dentin bond components. Dent Mater 2005;21:232–41. https://doi.org/10.1016/j.dental.2004.06.001.Search in Google Scholar PubMed

18. Pashley, DH, Tay, F, Yiu, C, Hashimoto, M, Breschi, L, Carvalho, R, et al.. Collagen degradation by host-derived enzymes during aging. J Dent Res 2004;83:216–21. https://doi.org/10.1177/154405910408300306.Search in Google Scholar PubMed

19. Sorsa, T, Tjäderhane, L, Salo, T. Matrix metalloproteinases (MMPs) in oral diseases. Oral Dis 2004;10:311–8. https://doi.org/10.1111/j.1601-0825.2004.01038.x.Search in Google Scholar PubMed

20. Reddy, MS, Geurs, NC, Gunsolley, JC. Periodontal host modulation with antiproteinase, anti‐inflammatory, and bone‐sparing agents. A systematic review. Ann Periodontol 2003;8:12–37. https://doi.org/10.1902/annals.2003.8.1.12.Search in Google Scholar PubMed

21. Gomes-Silva, W, Prado-Ribeiro, AC, Brandao, TB, Morais-Faria, K, de Castro Junior, G, Mak, MP, et al.. Postradiation matrix metalloproteinase-20 expression and its impact on dental micromorphology and radiation-related caries. Caries Res 2017;51:216–24. https://doi.org/10.1159/000457806.Search in Google Scholar PubMed

22. Xi, L, Li, S, Yao, X, Wei, Y, Li, J, Liu, H, et al.. In silico study combining docking and QSAR methods on a series of matrix metalloproteinase 13 inhibitors. Arch Pharmazie 2014;347:825–33. https://doi.org/10.1002/ardp.201400200.Search in Google Scholar PubMed

23. Hidalgo, M, Eckhardt, SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst 2001;93:178–93. https://doi.org/10.1093/jnci/93.3.178.Search in Google Scholar PubMed

24. Coussens, LM, Fingleton, B, Matrisian, LM. Matrix metalloproteinase inhibitors and cancer—trials and tribulations. Science 2002;295:2387–92. https://doi.org/10.1126/science.1067100.Search in Google Scholar PubMed

25. Huang, S-Y, Zou, X. Advances and challenges in protein-ligand docking. Int J Mol Sci 2010;11:3016–34. https://doi.org/10.3390/ijms11083016.Search in Google Scholar PubMed PubMed Central

26. Chen, C, Huang, S, Chen, CL, Su, SB, Fang, DD. Isoliquiritigenin inhibits ovarian cancer metastasis by reversing epithelial-to-mesenchymal transition. Molecules 2019;24:1–13. doi:https://doi.org/10.3390/molecules24203725.Search in Google Scholar PubMed PubMed Central

27. Hong, SH, Cha, HJ. Anti-proliferative and pro-apoptotic effects of licochalcone A through ROS-mediated cell cycle arrest and apoptosis in human bladder cancer cells. Int J Mol Sci 2019;20. https://doi.org/10.3390/ijms20153820.Search in Google Scholar PubMed PubMed Central

28. Iida, K, Naiki, T, Naiki-Ito, A. Luteolin suppresses bladder cancer growth via regulation of mechanistic target of rapamycin (mTOR) pathway. John Wiley & Sons Australia, Ltd.; 2020.10.1111/cas.14334Search in Google Scholar PubMed PubMed Central

29. Li, LJ, Li, GW, Xie, Y. Regulatory effects of glabridin and quercetin on energy metabolism of breast cancer cells. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China J Chin Mater Med 2019;44:3786–91. https://doi.org/10.19540/j.cnki.cjcmm.20190505.401.Search in Google Scholar PubMed

30. Hyuga, S, Hyuga, M, Yoshimura, M, Amakura, Y, Goda, Y, Hanawa, T. Herbacetin, a constituent of ephedrae herba, suppresses the HGF-induced motility of human breast cancer MDA-MB-231 cells by inhibiting c-Met and Akt phosphorylation. Planta Med 2013;79:1525–30. https://doi.org/10.1055/s-0033-1350899.Search in Google Scholar PubMed

31. Wood, N. The effects of selected dietary bioflavonoid supplementation on dental caries in young rats fed a high-sucrose diet. J Med Food 2007;10:694–701. https://doi.org/10.1089/jmf.2007.412.Search in Google Scholar PubMed

32. Koo, H, Schobel, B, Scott-Anne, K, Watson, G, Bowen, W, Cury, J, et al.. Apigenin and tt-farnesol with fluoride effects on S. mutans biofilms and dental caries. J Dent Res 2005;84:1016–20. https://doi.org/10.1177/154405910508401109.Search in Google Scholar PubMed PubMed Central

33. Zhao, B, Zhang, Y, Xiong, Y, Xu, X. Rutin promotes the formation and osteogenic differentiation of human periodontal ligament stem cell sheets in vitro. Int J Mol Med 2019;44:2289–97. https://doi.org/10.3892/ijmm.2019.4384.Search in Google Scholar PubMed PubMed Central

34. Gomez-Florit, M, Monjo, M, Ramis, JM. Identification of quercitrin as a potential therapeutic agent for periodontal applications. J Periodontol 2014;85:966–74. https://doi.org/10.1902/jop.2014.130438.Search in Google Scholar PubMed

35. Grenier, D, Chen, H, Ben Lagha, A, Fournier-Larente, J, Morin, MP. Dual action of myricetin on porphyromonas gingivalis and the inflammatory response of host cells: a promising therapeutic molecule for periodontal diseases. PloS One 2015;10:e0131758. https://doi.org/10.1371/journal.pone.0131758.Search in Google Scholar PubMed PubMed Central

36. Sabogal-Guáqueta, AM, Muñoz-Manco, JI, Ramírez-Pineda, JR, Lamprea-Rodriguez, M, Osorio, E, Cardona-Gómez, GP. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015;93:134–45. https://doi.org/10.1016/j.neuropharm.2015.01.027.Search in Google Scholar PubMed PubMed Central

37. Dourado, NS, Souza, CDS, de Almeida, MMA, Bispo da Silva, A, Dos Santos, BL, Silva, VDA, et al.. Neuroimmunomodulatory and neuroprotective effects of the flavonoid apigenin in in vitro models of neuroinflammation associated with Alzheimer’s disease. Front Aging Neurosci 2020;12:119. https://doi.org/10.3389/fnagi.2020.00119.Search in Google Scholar PubMed PubMed Central

38. Markham, KR. Techniques of flavonoid identification. London: Academic Press; 1982.Search in Google Scholar

39. Yonekura-Sakakibara, K, Higashi, Y, Nakabayashi, R. The origin and evolution of plant flavonoid metabolism. Front Plant Sci 2019;10:943. https://doi.org/10.3389/fpls.2019.00943.Search in Google Scholar PubMed PubMed Central

40. Bylka, W, Matlawska, I, Pilewski, N. Natural flavonoids as antimicrobial agents. Jana 2004;7:9–16.Search in Google Scholar

41. Berman, HM, Battistuz, T, Bhat, TN, Bluhm, WF, Bourne, PE, Burkhardt, K, et al.. The protein data bank. Acta Crystallogr Sect D Biol Crystallogr 2002;58:899–907. https://doi.org/10.1107/s0907444902003451.Search in Google Scholar PubMed

42. Singh, KD, Kirubakaran, P, Manikandaprabhu, S, Nagamani, S, Srinivasan, P, Karthikeyan, M. Docking studies of adenosine analogues with NS5 methyltransferase of yellow fever virus. Indian J Microbiol 2012;52:28–34.10.1007/s12088-011-0201-7Search in Google Scholar PubMed PubMed Central

43. Artimo, P, Jonnalagedda, M, Arnold, K, Baratin, D, Csardi, G, De Castro, E, et al.. ExPASy: SIB bioinformatics resource portal. Nucleic Acids Res 2012;40:W597–603. https://doi.org/10.1093/nar/gks400.Search in Google Scholar

44. Nara, H, Kaieda, A, Sato, K, Naito, T, Mototani, H, Oki, H, et al.. Discovery of novel, highly potent, and selective matrix metalloproteinase (MMP)-13 inhibitors with a 1,2,4-triazol-3-yl moiety as a zinc binding group using a structure-based design approach. J Med Chem 2017;60:608–26. https://doi.org/10.1021/acs.jmedchem.6b01007.Search in Google Scholar

45. Kim, S, Chen, J, Cheng, T, Gindulyte, A, He, J, He, S, et al.. PubChem 2019 update: improved access to chemical data. Nucleic Acids Res 2018;47:D1102–D9. https://doi.org/10.1093/nar/gky1033.Search in Google Scholar

46. Xu, Y, Lai, L, Gabrilove, J, Scheinberg, D. Antitumor activity of actinonin in vitro and in vivo. Clin Canc Res 1998;4:171–6.Search in Google Scholar

47. Paolocci, N, Tavazzi, B, Biondi, R, Gluzband, YA, Amorini, AM, Tocchetti, CG, et al.. Metalloproteinase inhibitor counters high-energy phosphate depletion and AMP deaminase activity enhancing ventricular diastolic compliance in subacute heart failure. J Pharmacol Exp Therapeut 2006;317:506–13. https://doi.org/10.1124/jpet.105.099168.Search in Google Scholar

48. Prato, M, Giribaldi, G, Polimeni, M, Gallo, V, Arese, P. Phagocytosis of hemozoin enhances matrix metalloproteinase-9 activity and TNF-α production in human monocytes: role of matrix metalloproteinases in the pathogenesis of falciparum malaria. J Immunol 2005;175:6436–42. https://doi.org/10.4049/jimmunol.175.10.6436.Search in Google Scholar

49. Nishimura, R, Wakabayashi, M, Hata, K, Matsubara, T, Honma, S, Wakisaka, S, et al.. Osterix regulates calcification and degradation of chondrogenic matrices through matrix metalloproteinase 13 (MMP13) expression in association with transcription factor Runx2 during endochondral ossification. J Biol Chem 2012;287:33179–90. https://doi.org/10.1074/jbc.m111.337063.Search in Google Scholar

50. Lipinski, CA, Lombardo, F, Dominy, BW, Feeney, PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 1997;23:3–25. https://doi.org/10.1016/s0169-409x(96)00423-1.Search in Google Scholar

51. Muthusamy, K, Prasad, S, Nagamani, S. Role of hydrophobic patch in LRP6: a promising drug target for Alzheimer’s disease. Indian J Pharmaceut Sci 2016;78:240–51. https://doi.org/10.4172/pharmaceutical-sciences.1000109.Search in Google Scholar

52. Morris, GM, Huey, R, Lindstrom, W, Sanner, MF, Belew, RK, Goodsell, DS, et al.. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J Comput Chem 2009;30:2785–91. https://doi.org/10.1002/jcc.21256.Search in Google Scholar

53. Morris, GM, Goodsell, DS, Halliday, RS, Huey, R, Hart, WE, Belew, RK, et al.. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J Comput Chem 1998;19:1639–62. https://doi.org/10.1002/(sici)1096-987x(19981115)19:14<1639::aid-jcc10>3.0.co;2-b.10.1002/(SICI)1096-987X(19981115)19:14<1639::AID-JCC10>3.0.CO;2-BSearch in Google Scholar

54. Liu, Z, Zhao, J, Li, W, Shen, L, Huang, S, Tang, J, et al.. Computational screen and experimental validation of anti-influenza effects of quercetin and chlorogenic acid from traditional Chinese medicine. Sci Rep 2016;6:19095. https://doi.org/10.1038/srep19095.Search in Google Scholar PubMed PubMed Central

55. Huey, R, Morris, GM. Using AutoDock 4 with AutoDocktools: a tutorial. USA: The Scripps Research Institute; 2008:54–6 pp.Search in Google Scholar

56. Huey, R, Morris, GM, Olson, AJ, Goodsell, DS. A semiempirical free energy force field with charge‐based desolvation. J Comput Chem 2007;28:1145–52. https://doi.org/10.1002/jcc.20634.Search in Google Scholar PubMed

57. Jayaraj, JM, Reteti, E, Kesavan, C, Muthusamy, K. Structural insights on Vitamin D receptor and screening of new potent agonist molecules: structure and ligand-based approach. J Biomol Struct Dyn 2020:1–12. https://doi.org/10.1080/07391102.2020.1775122.Search in Google Scholar PubMed

58. de Amorim, AL, de Lima, AVM, do Rosário, AC, dos Santos Souza, ÉT, Ferreira, JV, da Silva Hage-Melim, LI. Molecular modeling of inhibitors against fructose bisphosphate aldolase from Candida albicans. In Silico pharmacology; 2018, vol 6:2 p.10.1007/s40203-018-0040-xSearch in Google Scholar PubMed PubMed Central

59. Afonso, IF. Modelagem molecular e avaliação da relação estrutura-atividade acoplados a estudos farmacocinéticos e toxicológicos in silico de derivados heterocíclicos com atividade antimicrobiana: dissertation. Rio de Janeiro, Brazil: Universidade Federal do Rio de Janeiro; 2008.Search in Google Scholar

60. Buzzi, FdC. Síntese de novas moléculas com potencial terapêutico: imidas cíclicas, chalconas e compostos relacionados. Santa Catarina: Federal University of Santa Catarina; 2007.Search in Google Scholar

61. Silva, Vbd. Estudos de modelagem molecular e relação estrutura atividade da oncoproteína hnRNP K e ligantes. São Paulo, Brazil: Universidade de São Paulo; 2008.10.11606/D.60.2008.tde-02102008-164529Search in Google Scholar

62. Yamashita, F, Hashida, M. In silico approaches for predicting ADME properties of drugs. Drug Metabol Pharmacokinet 2004;19:327–38. https://doi.org/10.2133/dmpk.19.327.Search in Google Scholar PubMed

63. Moda, TL. Desenvolvimento de modelos in silico de propriedades de ADME para a triagem de novos candidatos a fármacos. São Paulo, Brazil: Universidade de São Paulo; 2007.10.11606/D.76.2007.tde-22032007-112055Search in Google Scholar

64. Ganeshpurkar, A, Saluja, AK. The pharmacological potential of rutin. Saudi Pharmaceut J: SPJ: Off Publ Saudi Pharmaceut Soc 2017;25:149–64. https://doi.org/10.1016/j.jsps.2016.04.025.Search in Google Scholar PubMed PubMed Central

65. Habtemariam, S. Rutin as a natural therapy for Alzheimer’s disease: insights into its mechanisms of action. Curr Med Chem 2016;23:860–73. https://doi.org/10.2174/0929867323666160217124333.Search in Google Scholar PubMed

66. Khan, F, Pandey, P, Upadhyay, TK, Jafri, A, Jha, NK, Mishra, R, et al.. Anti-cancerous effect of rutin against HPV-C33A cervical cancer cells via G0/G1 cell cycle arrest and apoptotic induction. Endocrine, metabolic & immune disorders drug targets. Sharjah, U.A.E.: Bentham Science Publishers; 2019.10.2174/1871530319666190806122257Search in Google Scholar PubMed

67. Lal Shyaula, S, Abbas, G, Siddiqui, H, Sattar, SA, Choudhary, MI, Basha, FZ. Synthesis and antiglycation activity of kaempferol-3-O-rutinoside (nicotiflorin). Med Chem 2012;8:415–20. https://doi.org/10.2174/1573406411208030415.Search in Google Scholar PubMed

68. Huang, JL, Fu, ST, Jiang, YY, Cao, YB, Guo, ML, Wang, Y, et al.. Protective effects of Nicotiflorin on reducing memory dysfunction, energy metabolism failure and oxidative stress in multi-infarct dementia model rats. Pharmacol Biochem Behav 2007;86:741–8. https://doi.org/10.1016/j.pbb.2007.03.003.Search in Google Scholar PubMed

69. Zhao, J, Zhang, S, You, S, Liu, T, Xu, F, Ji, T, et al.. Hepatoprotective effects of nicotiflorin from nymphaea candida against concanavalin A-induced and D-galactosamine-induced liver injury in mice. Int J Mol Sci 2017;18:1–12. doi:https://doi.org/10.3390/ijms18030587.Search in Google Scholar PubMed PubMed Central

70. Ben Bakrim, W, El Bouzidi, L, Nuzillard, JM. Bioactive metabolites from the leaves of Withania adpressa. Pharmaceut Biol 2018;56:505–10. https://doi.org/10.1080/13880209.2018.1499781.Search in Google Scholar PubMed PubMed Central

71. Yadav, M, Song, F, Huang, J, Chakravarti, A, Jacob, NK. Ocimum flavone Orientin as a countermeasure for thrombocytopenia. Sci Rep 2018;8:5075. https://doi.org/10.1038/s41598-018-23419-x.Search in Google Scholar PubMed PubMed Central

72. Pastene, E, Bocaz, G, Peric, I, Montes, M, Silva, V, Riffo, E. Separation by capillary electrophoresis of C-glycosylflavonoids in Passiflora sp. extracts. Bol Soc Chil Quim 2000;45:461–7. https://doi.org/10.4067/s0366-16442000000300017.Search in Google Scholar

73. Dykes, L, Rooney, LW. Sorghum and millet phenols and antioxidants. Review. J Cereal Sci 2006;44:236–51.10.1016/j.jcs.2006.06.007Search in Google Scholar

74. Lam, KY, Ling, APK, Koh, RY, Wong, YP, Say, YH. A review on medicinal properties of orientin. Adv Pharmacol Sci 2016;2016:1–9. doi:https://doi.org/10.1155/2016/4104595.Search in Google Scholar PubMed PubMed Central

75. Praveena, R, Sadasivam, K, Deepha, V, Sivakumar, R. Antioxidant potential of orientin: a combined experimental and DFT approach. J Mol Struct 2014;1061:114–23. https://doi.org/10.1016/j.molstruc.2014.01.002.Search in Google Scholar

76. Feghali, CA, Wright, TM. Cytokines in acute and chronic inflammation. Front Biosci 1997;2:d12–26. https://doi.org/10.2741/a171.Search in Google Scholar PubMed

77. Gabay, C. Interleukin-6 and chronic inflammation. Arthritis Res Ther 2006;8:S3. https://doi.org/10.1186/ar1917.Search in Google Scholar PubMed PubMed Central

78. Skovronsky, DM, Lee, VM-Y, Trojanowski, JQ. Neurodegenerative diseases: new concepts of pathogenesis and their therapeutic implications. Annu Rev Pathol Mech Dis 2006;1:151–70. https://doi.org/10.1146/annurev.pathol.1.110304.100113.Search in Google Scholar PubMed

79. Kilic, FS, Sirmagul, B, Yildirim, E, Oner, S, Erol, K. Antinociceptive effects of gabapentin & its mechanism of action in experimental animal studies. Indian J Med Res 2012;135:630.Search in Google Scholar

80. Thangaraj, K, Natesan, K, Palani, M, Vaiyapuri, M. Orientin, a flavanoid, mitigates 1, 2 dimethylhydrazine-induced colorectal lesions in Wistar rats fed a high-fat diet. Toxicol Rep 2018;5:977–87. https://doi.org/10.1016/j.toxrep.2018.09.004.Search in Google Scholar PubMed PubMed Central

81. Zhong, Y, Zheng, QY, Sun, CY, Zhang, Z, Han, K. Orientin improves cognition by enhancing autophagosome clearance in an Alzheimer’s mouse model. J Mol Neurosci. 2019;69:246–53. https://doi.org/10.1007/s12031-019-01353-5.Search in Google Scholar PubMed

82. Gamboa, JL, Billings, FTIV, Bojanowski, MT, Gilliam, LA, Yu, C, Roshanravan, B, et al.. Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease. Physiol Rep 2016;4:e12780. https://doi.org/10.14814/phy2.12780.Search in Google Scholar PubMed PubMed Central

83. Choo, C, Sulong, N, Man, F, Wong, T. Vitexin and isovitexin from the leaves of Ficus deltoidea with in-vivo α-glucosidase inhibition. J Ethnopharmacol 2012;142:776–81. https://doi.org/10.1016/j.jep.2012.05.062.Search in Google Scholar PubMed

84. Kim, J, Lee, I, Seo, J, Jung, M, Kim, Y, Yim, N, et al.. Vitexin, orientin and other flavonoids from Spirodela polyrhiza inhibit adipogenesis in 3T3‐L1 cells. Phytother Res 2010;24:1543–8. https://doi.org/10.1002/ptr.3186.Search in Google Scholar PubMed

85. Kim, JH, Lee, BC, Kim, JH, Sim, GS, Lee, DH, Lee, KE, et al.. The isolation and antioxidative effects of vitexin from Acer palmatum. Arch Pharm Res 2005;28:195. https://doi.org/10.1007/bf02977715.Search in Google Scholar PubMed

86. An, F, Yang, G, Tian, J, Wang, S. Antioxidant effects of the orientin and vitexin in Trollius chinensis Bunge in D-galactose-aged mice. Neural Regen Res 2012;7:2565. https://doi.org/10.3969/j.issn.1673-5374.2012.33.001.Search in Google Scholar PubMed PubMed Central

87. Borghi, SM, Carvalho, TT, Staurengo-Ferrari, L, Hohmann, MS, Pinge-Filho, P, Casagrande, R, et al.. Vitexin inhibits inflammatory pain in mice by targeting TRPV1, oxidative stress, and cytokines. J Nat Prod 2013;76:1141–9. https://doi.org/10.1021/np400222v.Search in Google Scholar PubMed

88. Choi, JS, Islam, MN, Ali, MY, Kim, EJ, Kim, YM, Jung, HA. Effects of C-glycosylation on anti-diabetic, anti-Alzheimer’s disease and anti-inflammatory potential of apigenin. Food Chem Toxicol 2014;64:27–33. https://doi.org/10.1016/j.fct.2013.11.020.Search in Google Scholar PubMed

89. Liu, X, Jiang, Q, Liu, H, Luo, S. Vitexin induces apoptosis through mitochondrial pathway and PI3K/Akt/mTOR signaling in human non-small cell lung cancer A549 cells. Biol Res 2019;52:7. https://doi.org/10.1186/s40659-019-0214-y.Search in Google Scholar PubMed PubMed Central

90. Nasr Bouzaiene, N, Chaabane, F, Sassi, A, Chekir-Ghedira, L, Ghedira, K. Effect of apigenin-7-glucoside, genkwanin and naringenin on tyrosinase activity and melanin synthesis in B16F10 melanoma cells. Life Sci 2016;144:80–5. https://doi.org/10.1016/j.lfs.2015.11.030.Search in Google Scholar PubMed

91. Nakazaki, E, Tsolmon, S, Han, J, Isoda, H. Proteomic study of granulocytic differentiation induced by apigenin 7-glucoside in human promyelocytic leukemia HL-60 cells. Eur J Nutr 2013;52:25–35. https://doi.org/10.1007/s00394-011-0282-4.Search in Google Scholar PubMed

92. Fang, Y, Lu, Y, Zang, X, Wu, T, Qi, X, Pan, S, et al.. 3D-QSAR and docking studies of flavonoids as potent Escherichia coli inhibitors. Sci Rep 2016;6:23634. https://doi.org/10.1038/srep23634.Search in Google Scholar PubMed PubMed Central

93. Shen, L, Yang, W. Molecular dynamics simulations with quantum mechanics/molecular mechanics and adaptive neural networks. J Chem Theor Comput. 2018;14:1442–55. https://doi.org/10.1021/acs.jctc.7b01195.Search in Google Scholar PubMed PubMed Central


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/jbcpp-2020-0036).


Received: 2020-02-12
Accepted: 2020-09-10
Published Online: 2020-12-14

© 2020 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 11.5.2024 from https://www.degruyter.com/document/doi/10.1515/jbcpp-2020-0036/html
Scroll to top button