Preprint Article Version 1 Preserved in Portico This version is not peer-reviewed

Comparative Molecular Docking of Apigenin and Luteolin Versus Conventional Ligands for TP-53, pRb, APOBEC3H, and HPV-16 E6: Potential Clinical Applications in Preventing Gy-necological Malignancies

Version 1 : Received: 15 August 2024 / Approved: 23 August 2024 / Online: 26 August 2024 (11:33:11 CEST)

How to cite: Dunjic, M.; Turini, S.; Nejkovic, L.; Sulovic, N.; Cvetkovic, S.; Dunjic, M.; Dunjic, K.; Dolovac, D. Comparative Molecular Docking of Apigenin and Luteolin Versus Conventional Ligands for TP-53, pRb, APOBEC3H, and HPV-16 E6: Potential Clinical Applications in Preventing Gy-necological Malignancies. Preprints 2024, 2024081765. https://doi.org/10.20944/preprints202408.1765.v1 Dunjic, M.; Turini, S.; Nejkovic, L.; Sulovic, N.; Cvetkovic, S.; Dunjic, M.; Dunjic, K.; Dolovac, D. Comparative Molecular Docking of Apigenin and Luteolin Versus Conventional Ligands for TP-53, pRb, APOBEC3H, and HPV-16 E6: Potential Clinical Applications in Preventing Gy-necological Malignancies. Preprints 2024, 2024081765. https://doi.org/10.20944/preprints202408.1765.v1

Abstract

This study presents a detailed comparative analysis of Molecular Docking data, focusing on the binding interactions of conventional ligands and natural compounds, Apigenin and Luteolin, with TP-53, pRb, and APOBEC. Utilizing advanced bioinformatics techniques, coupled with Ar-tificial Intelligence software and High-Performance Computing (HPC), we measured and con-trasted the binding energies of these interactions. Additionally, we investigated protein-protein interactions between HPV-16 oncoprotein E6 and tumor suppressors TP-53 and pRb. Our findings demonstrate that the natural compounds Apigenin and Luteolin exhibit significantly higher binding affinities to TP-53, pRb, and APOBEC compared to conventional pharmacological ligands. The binding energies for Apigenin and Luteolin were approximately -6.9 kcal/mol and -6.6 kcal/mol, respectively, indicating their strong potential as therapeutic agents in inhibiting the oncogenic functions of HPV-16. In contrast, conventional ligands showed lower binding affinities, with energies around -4.5 to -5.5 kcal/mol. The study further revealed that the protein-protein interactions between E6 and TP-53, and E6 and pRb, exhibited considerably higher binding en-ergies, approximately -976.7 kcal/mol, due to the multiple interaction sites and the complex nature of protein interfaces. A conversion formula was developed to translate these high-energy inter-actions to a comparable scale for non-protein-protein interactions, highlighting the superior binding potential of natural compounds, which, through the same conversion formula, was shown to be higher than that of interaction between E6 with TP-53 and E6 with pRb. These results underscore the therapeutic promise of Apigenin and Luteolin in preventing HPV-16-related on-cogenesis. By demonstrating their higher binding affinities to crucial tumor suppressors, this study supports the development of natural compound-based therapies. The findings also em-phasize the necessity for further experimental validation to explore these compounds' efficacy in clinical settings. This comprehensive analysis provides a robust framework for understanding the comparative binding interactions and lays the groundwork for innovative therapeutic strategies against HPV-16.

Keywords

Molecular Docking; HPV-16; TP-53; pRb; APOBEC; Apigenin; Luteolin; Protein-Protein Interac-tion; High-Performance Computing (HPC); Oncoprotein E6

Subject

Medicine and Pharmacology, Medicine and Pharmacology

Comments (0)

We encourage comments and feedback from a broad range of readers. See criteria for comments and our Diversity statement.

Leave a public comment
Send a private comment to the author(s)
* All users must log in before leaving a comment
Views 0
Downloads 0
Comments 0


×
Alerts
Notify me about updates to this article or when a peer-reviewed version is published.
We use cookies on our website to ensure you get the best experience.
Read more about our cookies here.