EXPLORING INNOVATIVE INHIBITOR CANDIDATES TARGETING THE RHOF PROTEIN: AN IN-SILICO INVESTIGATION.

Main Article Content

Madhavi Latha Bingi
Vani Kondaparthi
Thirupathi Damera
Priyadarshini Gangidi
Hareesh Reddy Badepally
Mounika Badineni
Kiran Kumar Mustyala
Vasavi Malkhed

Keywords

.

Abstract

Cancer is a broad category of diseases characterized by untamed cell division and proliferation that damages nearby tissues and progresses to other parts of the body. The Rhof family, which is a subgroup of the broader Rhof family of tiny signaling G proteins, controls several physiological functions such as actin cytoskeleton, cell motility, and gene expression. Dysregulation or mutations in Rho GTPases can result in disorders such as cancer, as they are essential for cellular motility and proliferation. The present investigation assesses and validates the three-dimensional arrangement of the Rhof protein utilizing In-silico approaches. Virtual screening is underway to create potent inhibitors that selectively target the GTP interacting domain of the Rhof protein. The results of the research indicate that the amino acid residues ARG122, GLY123, ILE124 and PRO125, ARG166, ALA195, LEU196, LYS198, GLU200 ARG201, and LYS204 have strong interactions with ligands and significant influence on complex formation. Computational approaches reveal that the ADME features of screened ligand molecules suggest the optimum level of permissibility of drug-like properties. The utilization of Rhof structural information, active site details, and selected ligand molecules greatly aid in identifying new therapeutic structures for Rhof protein.

Abstract 93 | pdf Downloads 28

References

[1]. Ferlay J, Colombet M, Soerjomataram I, et.al. Global and Regional Estimates of the Incidence and Mortality for 38 Cancers: GLOBOCAN 2018. International Agency for Research on Cancer/World Health Organization; 2018.
[2]. World Health Organization (WHO). Global Health Estimates 2020: Deaths by Cause, Age, Sex, by Country and by Region, 2000-2019. WHO; 2020.
[3]. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002 Dec 12;420(6916):629-35.
[4]. Jung, H., Yoon, S. R., Lim, J., Cho, H. J., & Lee, H. G. (2020). Dysregulation of Rho GTPases in Human Cancers. Cancers.
[5] Senior, A.W., Evans, R., Jumper, J. et al. Improved protein structure prediction using potentials from deep learning. Nature 577, 706–710 (2020).
[6]. Ellenbroek SI, Collard JG. Rho GTPases: functions and association with cancer. Clin. Exp Metastasis. 2007;24(8):657-72.
[7]. Xu,H., Niu,M., Yuan,X. etal. CD44 as a tumor biomarker and therapeutic target. Exp. Hematol Oncol 9,36 (2020)
[8]. Hamidi, H., Ivaska, J. every step of the way: integrins in cancer progression and metastasis. Nat Rev Cancer 18, 533-548 (2018).
[9]. Koledova Z. 3D Cell Culture: An Introduction. Methods Mol Biol. 2017;1612:1-11.
[10]. Li Q, Shah S. Structure-Based Virtual Screening. Methods Mol Biol. 2017;1558:111-124.
[11]. Lin, X., Li, X., & Lin, X. (2020). A Review on Applications of Computational Methods in Drug Screening and Design.
[12]. Davidavičienė, V. (2018). Research Methodology: An Introduction. In: Marx Gómez, J., Mouselli, S. (eds) Modernizing the Academic Teaching and Research Environment. Progress in IS. Springer, Cham.
[13]. The UniProt Consortium, UniProt: a worldwide hub of protein knowledge, Nucleic Acids Research, Volume 47, Issue D1, 08 January 2019, Pages D506–D515.
[14]. Drozdetskiy, A., Cole, C., Procter, J., & Barton, G. J. (2015). JPred4: a protein secondary structure prediction server. Nucleic Acids Research.
[15] Anderson, Nicole M.et.al, The tumor microenvironment, Current Biology, Volume 30, Issue 16, R921 – R925
[16]. Webb, B. & S. (2016). Comparative Protein Structure Modeling Using MODELLER. Current Protocols in Bioinformatics,. . . Current Protocols in Bioinformatics, 54(1).
[17]. Andrew Waterhouse, Martino Bertoni, Stefan Bienert, Gabriel Studer, Gerardo Tauriello, Rafal Gumienny, Florian T Heer, Tjaart A P de Beer, Christine Rempfer, Lorenza Bordoli, Rosalba Lepore, Torsten Schwede, SWISS-MODEL: homology modelling of protein structures and complexes, Nucleic Acids Research, Volume 46, Issue W1, 2 July 2018, Pages W296–W303.
[18]. Byrne, B. M. (2005). Factor Analytic Models: Viewing the Structure of an Assessment Instrument From Three Perspectives. Journal of Personality Assessment, 85(1), 17–32. https://doi.org/10.1207/s15327752jpa8501_02
[19]. Brocke, S. A., Degen, A., MacKerell, A. D., Jr, Dutagaci, B., & Feig, M.. (2018). Prediction of Membrane Permeation of Drug Molecules by Combining an Implicit Membrane Model with Machine Learning. . Journal of Chemical Information and Modeling, 59(3), 1147–1162.
[20]. Valckenaers, P. (2018). RTI Reference Architecture – PROSA Revisited. SpringerLink. [21]. Al-Refaei, M. A., Makki, R. M., & Ali, H. M. (2020). Structure prediction of transferrin receptor protein 1 (TfR1) by homology modelling, docking, and molecular dynamics simulation studies. 6(1(e03221).
[22]. Banach M, Konieczny L, Roterman I. Secondary and Supersecondary Structure of Proteins in Light of the Structure of Hydrophobic Cores. Methods Mol Biol. 2019;1958:347-378.
[23]. Thomas Simonson, Georgios Archontis, and Martin Karplus. Free Energy simulations Come of Age: Protein-Ligand Recognition. Acc. Chem. Res. 2002, 35, 6, 430–437.
[24] Monticelli, L., Tieleman, D.P. (2013). Force Fields for Classical Molecular Dynamics. In: Monticelli, L., Salonen, E. (eds) Biomolecular Simulations. Methods in Molecular Biology, vol 924. Humana Press, Totowa, NJ.
[25]. Simone Brogi, Teodorico Castro Ramalho, Kamil Kuca, José L. Medina-Franco, Marian Valko. In silico Methods for Drug Design and Discovery. Frontiers in Chemistry, August 2020
[26]. Tian, W., Chen, C., Lei, X., Zhao, J., & Liang, J (2018). CASTp 3.0: computed atlas of surface topography of proteins. Nucleic Acids Research, 46(W1), W363–W367.
[27]. Pavlo.Polishchuk. Interpretation of Quantitative structure- Activity Relationship Models: Past, Present, and future. J. Chem. Inf. Model. 2017, 57, 11, 2618–2639
[28]. Lu, H., Zhou, Q., He, J. et al. Recent advances in the development of protein–protein interactions modulators: mechanisms and clinical trials. Sig Transduct Target Ther 5, 213 (2020).
[29]. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010 Jan 30;31(2):455-61.
[30]. Pattar, S.V., Adhoni, S.A., Kamanavalli, C.M. et al. In silico molecular docking studies and MM/GBSA analysis of coumarin-carbonodithioate hybrid derivatives divulge the anticancer potential against breast cancer. Beni-Suef Univ J Basic Appl Sci 9, 36 (2020).
[31]. Martí-Renom, M. A., Stuart, A. C., Fiser, A., Sánchez, R., Melo, F., & Sali, A. (2000). Comparative Protein Structure Modeling of Genes and Genomes. Annual Review of Biophysics and Biomolecular Structure,. 29(1), 295–325.
[32]. Prajapat, M., Shekhar, N., Sarma, P., Avti, P., Singh, S., Kaur, H., Bhattacharyya, A., Kumar, S., Sharma, S., Prakash, A., & Medhi, B. (2020). Virtual screening and molecular dynamics study of approved drugs as inhibitors of spike protein S1 domain and ACE2 interaction in SARS-CoV-2. Journal of Molecular Graphics and Modelling,. Journal of Molecular Graphics and Modelling, 101(107716).
[33]. Forli, S., Huey, R., Pique, M. et al. Computational protein–ligand docking and virtual drug screening with the AutoDock suite. Nat Protoc 11, 905–919 (2016).
[34] Wang E, Liu H, Wang J, Weng G, Sun H, Wang Z, Kang Y, Hou T. Development and Evaluation of MM/GBSA Based on a Variable Dielectric GB Model for Predicting Protein-Ligand Binding Affinities. J Chem Inf Model. 2020 Nov 23; 60(11):5353-5365.
[35]. Yim DS, Choi S, Bae SH. Predicting human pharmacokinetics from preclinical data: absorption. Transl Clin Pharmacol. 2020 Sep; 28(3):126-135.
[36]. Lucas AJ, Sproston JL, Barton P, Riley RJ. Estimating human ADME properties, pharmacokinetic parameters and likely clinical dose in drug discovery. Expert Opin Drug Discov. 2019 Dec; 14(12):1313-1327.
[37]. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017 Mar 3;7: 42717.
[38]. Gimeno A, Ojeda-Montes MJ, Tomás-Hernández S, Cereto-Massagué A, Beltrán-Debón R, Mulero M, Pujadas G, Garcia-Vallvé S. The Light and Dark Sides of Virtual Screening: What Is There to Know? Int J Mol Sci. 2019 Mar 19; 20(6):1375.
[39]. Pundir S, Martin MJ, O'Donovan C; UniProt Consortium. UniProt Tools. Curr Protoc Bioinformatics. 2016 Mar 24; 53: 1.29.1-1.29.15.
[40]. Wright GJ. Signal initiation in biological systems: the properties and detection of transient extracellular protein interactions. Mol Biosyst. 2009 Dec; 5(12):1405-12.
[41]. Vadija, R., Mustyala, K. K., Nambigari, N., Dulapalli, R., Dumpati, R. K., Ramatenki, V., Vellanki, S. P., & Vuruputuri, U. (2016). Homology modeling and virtual screening studies of FGF-7 protein—a structure-based approach to design new molecules against tumor angiogenesis. . 9(3), 69–78. T.
[42] Chen, D., Oezguen, N., Urvil, P., Ferguson, C., Dann, S.M., & Savidge, T.C. (2016). Regulation of protein-ligand binding affinity by hydrogen bond pairing. Science Advances, 2. (2016). Regulation of protein-ligand binding affinity by hydrogen bond pairing. Science Advances, 2(3).
[43]. Perumalsamy, H., Sankarapandian, K., Veerappan, K., Natarajan, S., Kandaswamy, N., Thangavelu, L., & Balusamy, S. R. (2020). In silico molecular docking studies and MM/GBSA analysis of coumarin-carbonodithioate hybrid derivatives divulge the anticancer potential against breast cancer. Beni-Suef University Journal of Basic and Applied Sciences.
[44]. Zoe Cournia, Bryce Allen, and Woody Sherman.Relative Binding Free Energy Calculations in Drug Discovery: Recent Advances and Practical Considerations.Journal of Chemical Information and Modeling 2017 57 (12), 2911-2937.

Most read articles by the same author(s)