Screening fructosamine-3-kinase (FN3K) inhibitors, a deglycating enzyme of oncogenic Nrf2: Human FN3K homology modelling, docking and molecular dynamics simulations

dc.contributor.authorBeeraka, Narasimha M.
dc.contributor.authorZhang, Jin
dc.contributor.authorMandal, Subhankar
dc.contributor.authorVikram P. R., Hemanth
dc.contributor.authorLiu, Junqi
dc.contributor.authorB. M., Namitha
dc.contributor.authorZhao, Di
dc.contributor.authorVishwanath, Prashanth
dc.contributor.authorGurupadayya, B. M.
dc.contributor.authorFan, Ruitai
dc.contributor.departmentPediatrics, School of Medicine
dc.date.accessioned2024-04-18T12:34:20Z
dc.date.available2024-04-18T12:34:20Z
dc.date.issued2023-11-01
dc.description.abstractFructosamine-3-kinase (FN3K) is involved in the deglycation of Nrf2, a significant regulator of oxidative stress in cancer cells. However, the intricate functional aspects of FN3K and Nrf2 in breast cancers have not been explored vividly. The objectives of this study are to design the human FN3K protein using homology modeling followed by the screening of several anticancer molecules and examining their efficacy to modulate FN3K activity, Nrf2-mediated antioxidant signalling. Methods pertinent to homology modeling, virtual screening, molecular docking, molecular dynamics simulations, assessment of ADME properties, cytotoxicity assays for anticancer molecules of natural/synthetic origin in breast cancer cells (BT-474, T-47D), and Western blotting were used in this study. The screened anticancer molecules including kinase inhibitors of natural and synthetic origin interacted with the 3-dimensional structure of the catalytic domain in human FN3K protein designed through homology modeling by significant CDOCKER interaction energies. Subsequently, gefitinib, sorafenib, neratinib, tamoxifen citrate, and cyclosporine A enhanced the expression of FN3K in BT-474 cell lines with simultaneous alteration in Nrf2-driven antioxidant signalling. Oxaliplatin significantly downregulated FN3K expression and modulated Nrf2-driven antioxidant signalling when compared to cisplatin and other anticancer drugs. Hence, the study concluded the potential implications of existing anticancer drugs to modulate FN3K activity in breast cancers.
dc.eprint.versionFinal published version
dc.identifier.citationBeeraka NM, Zhang J, Mandal S, et al. Screening fructosamine-3-kinase (FN3K) inhibitors, a deglycating enzyme of oncogenic Nrf2: Human FN3K homology modelling, docking and molecular dynamics simulations. PLoS One. 2023;18(11):e0283705. Published 2023 Nov 1. doi:10.1371/journal.pone.0283705
dc.identifier.urihttps://hdl.handle.net/1805/40110
dc.language.isoen_US
dc.publisherPublic Library of Science
dc.relation.isversionof10.1371/journal.pone.0283705
dc.relation.journalPLoS One
dc.rightsAttribution 4.0 Internationalen
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/
dc.sourcePMC
dc.subjectAntineoplastic agents
dc.subjectAntioxidants
dc.subjectMolecular docking simulation
dc.titleScreening fructosamine-3-kinase (FN3K) inhibitors, a deglycating enzyme of oncogenic Nrf2: Human FN3K homology modelling, docking and molecular dynamics simulations
dc.typeArticle
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
pone.0283705.pdf
Size:
5.48 MB
Format:
Adobe Portable Document Format
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: