1
|
Roney M, Uddin MN, Khan AA, Fatima S, Mohd Aluwi MFF, Hamim SMI, Ahmad A. Repurposing of dipeptidyl peptidase FDA-approved drugs in Alzheimer's disease using network pharmacology and in-silico approaches. Comput Biol Chem 2025; 116:108378. [PMID: 39938415 DOI: 10.1016/j.compbiolchem.2025.108378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/01/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) have similar clinical characteristics in the brain and islet, as well as an increased incidence with ageing and familial susceptibility. Therefore, in recent years there has been a great desire for research that elucidates how anti-diabetic drugs affect AD. This work attempts to first elucidate the possible mechanism of action of DPP-IV inhibitors in the treatment of AD by employing techniques from network pharmacology, molecular docking, molecular dynamic simulation, principal component analysis, and MM/PBSA. A total of 463 targets were identified from the SwissTargetPrediction and 784 targets were identified from the SuperPred databases. 79 common targets were screened using the PPI network. The GO and KEGG analyses indicated that the activity of DPP-IV against AD potentially involves the hsa04080 neuroactive ligand-receptor interaction signalling pathway, which contains 17 proteins, including CHRM2, CHRM3, CHRNB1, CHRNB4, CHRM1, PTGER2, CHRM4, CHRM5, TACR2, HTR2C, TACR1, F2, GABRG2, MC4R, HTR7, CHRNG, and DRD3. Molecular docking demonstrated that sitagliptin had the greatest binding affinity of -10.7 kcal/mol and established hydrogen bonds with the Asp103, Ser107, and Asn404 residues in the active site of the CHRM2 protein. Molecular dynamic simulation, PCA, and MM/PBSA were performed for the complex of sitagliptin with the above-mentioned proteins, which revealed a stable complex throughout the simulation. The work identifies the active component and possible molecular mechanism of sitagliptin in the treatment of AD and provides a theoretical foundation for future fundamental research and practical implementation.
Collapse
Affiliation(s)
- Miah Roney
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia; Centre for Bio-aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - Md Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka 1205, Bangladesh
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 12371, Saudi Arabia
| | - Mohd Fadhlizil Fasihi Mohd Aluwi
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia; Centre for Bio-aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - S M Istiaque Hamim
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - Asrar Ahmad
- Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC, USA
| |
Collapse
|
2
|
Kizhakkiniyakath SA, Choudhury T, Rajan MV, Rathee S, Meena B, Hariprasad G. In silico studies to understand the interactions of flavonoid inhibitor with nsp12-RNA dependent RNA polymerase of SARS-CoV-2 and its homologs. Biochem Biophys Rep 2025; 42:101975. [PMID: 40160515 PMCID: PMC11952025 DOI: 10.1016/j.bbrep.2025.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Aim COVID 19 continues to be a major health concern. RNA dependent RNA polymerase of SARS-CoV-2 which is crucial for replication is therefore a potential drug target. Methodology Based on experimental structures of RdRp from SARS-CoV-2, computational models were generated of its homologs from SARS-C o V-1, MERS and Bat. SARS CoV-2 RdRp was used for virtual screening at nucleotide binding site with molecule from COCONUT Natural Products database using Glide. Complexes with the top inhibitor molecule were modelled using Discovery Studio and Desmond suite of programs. Results SARS-CoV-2 RdRp has a minimum of 80 % sequence similarity with its homologs, with the secondary structural elements, catalytic residues and metal binding residues being conserved. Certain residue variations in SARS-CoV-2 RdRp seems to be responsible for the stability of the enzyme. Docking and simulation studies showed that a flavonoid molecule with Coconut ID: CNP0127177.0 (HHF318) has binding affinity in low nano-molar range against RdRp from SARS-CoV-2 which was comparable or better than currently used inhibitors. This affinity stems from cationic-π with Arg555, and π-stacking interactions with a nucleobase of RNA. Molecule also engages with other residues that are crucial for its functions. This flavonoid molecule has similar physio-chemical properties like ATP towards SARS-CoV-2 RdRp, and has low potency for human ATP binding proteins. Conclusion HHF318 is a potential inhibitor of SARS-CoV-2 RdRp with good potency, specificity and pharmacokinetic properties for it to be developed as a drug candidate for COVID19.
Collapse
Affiliation(s)
| | - Tejaswini Choudhury
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Madhan Vishal Rajan
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sagar Rathee
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | | | | |
Collapse
|
3
|
Shen R, Zhu Q, Hu L, Ma J, Wang W, Dong H. Bridging Dimensionality Reduction and Stochastic Sampling: The DA2-MC Algorithm for Protein Dynamics. J Phys Chem Lett 2025:4788-4795. [PMID: 40335286 DOI: 10.1021/acs.jpclett.5c00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Elucidating protein dynamics and conformational changes is crucial for understanding their biological functions. This work introduces a data-driven accelerated conformational searching algorithm incorporating a Monte Carlo strategy, termed the DA2-MC method, which integrates dimensionality reduction techniques with Monte Carlo strategies to efficiently explore unknown protein conformations. The DA2-MC method was applied to investigate the folding mechanisms of two miniproteins, chignolin and WW domain, revealing their dynamic behavior in different conformational states at a reasonable computational cost. A Markov state model-based analysis of chignolin's folding pathway corroborated the dynamic insights obtained from the DA2-MC method. Moreover, free energy calculations initiated with the intermediate structures identified by DA2-MC yielded results consistent with published literature, affirming the method's reliability in accelerating conformational searches and reconstructing equilibrium properties. Collectively, the DA2-MC method emerges as an effective tool for efficiently exploring protein conformations, facilitating the identification of potential functional conformations on complex energy landscapes.
Collapse
Affiliation(s)
- Ruizhe Shen
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, & School of Physics, Nanjing University, Nanjing 210093, China
| | - Qiang Zhu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Key Laboratory of Mesoscopic Chemistry of Ministry of Education, Institute of Theoretical and Computational Chemistry, & School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Limu Hu
- Key Laboratory of Mesoscopic Chemistry of Ministry of Education, Institute of Theoretical and Computational Chemistry, & School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jing Ma
- Key Laboratory of Mesoscopic Chemistry of Ministry of Education, Institute of Theoretical and Computational Chemistry, & School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, & School of Physics, Nanjing University, Nanjing 210093, China
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China
| | - Hao Dong
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Centre (ChemBIC), ChemBioMed Interdisciplinary Research Centre at Nanjing University, Nanjing University, Nanjing 210023, China
| |
Collapse
|
4
|
Vural BE, Yandım C. Effects of F53 hotspot mutations on the molecular dynamics of MEK1 protein and the binding of its FDA-approved inhibitors. Int J Biol Macromol 2025; 306:141329. [PMID: 39986525 DOI: 10.1016/j.ijbiomac.2025.141329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 01/25/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
MEK1 (MAP2K1) could emerge as an oncogenic protein in the presence of certain mutations, and could be inhibited by FDA-approved drugs (trametinib, cobimetinib, binimetinib and selumetinib). However, how the mutations on MEK1's hotspot residue F53 affect the bindings of these therapeutic molecules remained largely unexplored. We performed molecular dynamics (MD) simulations with wild-type and mutated (F53L/V/C/I/Y) MEK1 structures in the presence and absence of these drugs and observed changes on the motion of MEK1. A longer duration in the lowest energy state conformation was observed during the simulations in the presence of F53 mutations. This was complemented by cross-correlated motions of amino acids of MEK1. More importantly, the binding affinities of inhibitors were affected. There was a marked reduction in the calculated binding affinity of trametinib in the presence of F53C mutation. On the other hand, the binding affinities of cobimetinib and selumetinib could overcome F53 mutations on MEK1. Binimetinib interestingly exhibited an increased binding affinity when F53C/I mutations were present. Taken together, our results provide a comprehensive perspective on the structural and drug-binding effects of observed mutations on the hotspot residue F53 within MEK1; warranting further research in stratifying F53 hotspot mutations for effective drug binding.
Collapse
Affiliation(s)
- Berkin Ersin Vural
- İzmir University of Economics, Faculty of Engineering, Department of Genetics and Bioengineering, 35330 Balçova, İzmir, Turkey
| | - Cihangir Yandım
- İzmir University of Economics, Faculty of Engineering, Department of Genetics and Bioengineering, 35330 Balçova, İzmir, Turkey; İzmir Biomedicine and Genome Center (IBG), Dokuz Eylül University Health Campus, 35340 İnciraltı, İzmir, Turkey.
| |
Collapse
|
5
|
Kumar A, Purohit N, Singh PP, Jangid K, Kumar V, Bharat JS, Chakraborty S, Kumar V, Jaitak V. Identification of phytochemicals from genus Potentilla as estrogen receptor-α inhibitors through molecular docking, molecular dynamic simulation and DFT calculations. J Biomol Struct Dyn 2025:1-17. [PMID: 40307235 DOI: 10.1080/07391102.2025.2498622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/29/2024] [Indexed: 05/02/2025]
Abstract
Breast cancer is among the most prevalent causes of death in women worldwide. About 70-75% of these cancers are hormone-dependent, expressing estrogen receptors (ERs), mainly ER-α, making it an essential target for managing breast cancer. Potentilla genus has been traditionally used worldwide for its diverse biological activities, including antidiabetic, anti-inflammatory, antioxidant, etc. In the present study, phytochemicals isolated from various species of the Potentilla species were evaluated for their in silico ER-α inhibitory activity through molecular docking, molecular dynamic simulation, Density Functional Theory calculations and free energy calculations. Four hundred seventy-one molecules were used through ligand preparation and docked inside the generated grid on ER-α protein cavity and the standard drug tamoxifen. Fourteen molecules have shown better dock (-14.42 to -12.57 kcal/mol) scores than tamoxifen (-10.71 kcal/mol). Most of the molecules belong to the category of flavonoid glycosides. Molecules with good binding free energy (-78.81 to -12.94 kcal/mol) indicate stability inside the binding pocket. Further, based on dock score, pharmacokinetic parameters, and binding free energy, two hit molecules, 1 and 2, were selected for their molecular dynamic simulation, MM/PBSA and DFT calculations for assessing their stability and structural dynamics inside the binding cavity as well as their reactivity. Through MD simulation analysis, it was evaluated that Compound 1 could distort the protein to a greater extent. In contrast, compound 2 was stable throughout the simulation time of 150 ns and can be further explored in vitro and in vivo studies as ER-α inhibitors in breast cancer.
Collapse
Affiliation(s)
- Amit Kumar
- Natural Product Chemistry Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Nehal Purohit
- Natural Product Chemistry Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Praval Pratap Singh
- Department of Computational Sciences, Central University of Punjab, Bathinda, India
| | - Kailash Jangid
- Department of Chemistry, Central University of Punjab, Bathinda, India
| | - Vijay Kumar
- Department of Chemistry, Central University of Punjab, Bathinda, India
| | - Jare Shrikrushna Bharat
- Natural Product Chemistry Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Sudip Chakraborty
- Department of Computational Sciences, Central University of Punjab, Bathinda, India
| | - Vinod Kumar
- Department of Chemistry, Central University of Punjab, Bathinda, India
| | - Vikas Jaitak
- Natural Product Chemistry Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| |
Collapse
|
6
|
Shahrior R, Tamkin S, Khan MB, Meraj AJ, Bhuiyan H. In-silico investigation integrated with machine learning to identify potential inhibitors targeting AKT2: Key driver of cancer cell progression and metastasis. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 267:108793. [PMID: 40305999 DOI: 10.1016/j.cmpb.2025.108793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND AND OBJECTIVE In search of a key driver for the invasive growth of cancer metastasis, AKT2 is found to be exceptionally expressed in colorectal cancer and its metastasis. Again, exceeding genomic arrangements of AKT2 can be held responsible for HGSC (High-grade serous ovarian cancer) and breast cancer cell metastasis. FDA-approved capivasertib, a potential drug targeting the AKT signaling pathway, has a few side effects such as plausible alterations of liver function and gastrointestinal issues. Hence, this research aims to detect compounds with higher drug potency for selective AKT2 inhibition to encounter the incidence of different types of cancer cell metastasis. METHODS Eight machine-learning models were engaged to classify active and inactive drug candidates among 1148 collected compounds from the CHEMBL database. Potential drug candidates with greater IC50 value and no Lipinski violations were then addressed to molecular docking and molecular dynamics simulation using PyRx, AutoDock Vina and Desmond package. RESULTS From docking studies, three of the initial drug candidates provided greater binding affinities within a range from -10.9 to -9.8 kcal/mol, comparable to that of Capivasertib and backed up by post-docking MM/GBSA analysis. Again, the prediction of pharmacokinetic properties and bioactivity scores of drug candidates revealed their drug-likeliness and safer ADMET profiles for future clinical trials. Finally, 100 ns MD simulation computation for these lead compounds exhibited greater stability and drug potency during interactions with AKT2 protein, followed by PCA and DCCM analysis. CONCLUSION However, future in-vivo research can ascertain whether our proposed drug candidates can pass the standard clinical trials as publicly accessible novel drug targets.
Collapse
Affiliation(s)
- Rahat Shahrior
- Department of Biomedical Engineering, Khulna University of Engineering and Technology, Khulna 9203, Bangladesh
| | - Salwa Tamkin
- Department of Computer Science and Engineering, BRAC University, Kha 224, Bir Uttam Rafiqul Islam Avenue, Merul Badda, Dhaka, 1212, Bangladesh
| | - Mohammad Badhruddouza Khan
- Department of Biomedical Engineering, Khulna University of Engineering and Technology, Khulna 9203, Bangladesh
| | - Ahmed Jebail Meraj
- Department of Biomedical Engineering, Khulna University of Engineering and Technology, Khulna 9203, Bangladesh
| | - Hanif Bhuiyan
- Data Scientist, Performance & Analytics Group, City of Gold Coast, Bundall, QLD 4217, Australia.
| |
Collapse
|
7
|
McElhenney SJ, Yu J. Collective Variables and Facilitated Conformational Opening during Translocation of Human Mitochondrial RNA Polymerase (POLRMT) from Atomic Simulations. J Chem Theory Comput 2025. [PMID: 40238747 DOI: 10.1021/acs.jctc.4c01568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Collective variable (CV) identification is challenging in complex dynamical systems. To study the translocation of a single-subunit RNA polymerase (RNAP) during human mitochondrial transcription, we employed all-atom molecular dynamics (MD) as a vehicle to illustrate CV refinement in conformational samplings and dimension reduction analyses. RNAP translocation is an essential mechanical step of transcription elongation that dictates gene expression. The translocation generally follows from polymerization product release and proceeds to initial binding or preinsertion of incoming nucleotides. The human mitochondrial DNA-dependent RNAP (or POLRMT) plays a critical role in cellular metabolism and can be a key molecular off-target in the design of nucleotide analogue antiviral and antitumor drugs due to its structural similarities with many viral RNAPs or RNA-dependent RNA polymerases (RdRps). While POLRMT shares particularly high structural similarity with bacteriophage T7 RNAP, previous experimental studies and our current simulations suggest that POLRMT's mechanochemical coupling mechanisms may be distinct. In the current work, we modeled POLRMT elongation complexes and performed equilibrium MD simulations on the pre- and post-translocation models, with extensive samplings around two potential translocation paths (with or without coupling to the fingers subdomain conformational change). We then compared time-lagged independent component analysis (tICA) and the neural network implementation of the variational approach for Markov processes (VAMPnets) as dimensional reduction methods on selected atomic coordinate sets to best represent the sampled features from the MD simulations. Our results indicate that POLRMT translocation is likely coupled with NTP binding to enable fingers subdomain opening at post-translocation which would otherwise be nonstabilized, or the translocations may proceed futilely without the fingers opening for incoming NTP initial binding or incorporation. The time scale of the coupled translocation reaches over hundreds of microseconds, as predicted by the VAMPnets analyses. Such a time scale seems to match a last postcatalytic kinetic step suggested for the POLRMT elongation cycle by previous experimental measurements. Our MD simulation studies combining atomic coordinate refinements and dimension reduction analyses on top of extensive conformational samplings thus suggest a variation of Brownian ratcheting in POLRMT translocation, as if the Brownian motions of translocation are coupled with NTP binding, which captures transient fingers subdomain opening to couple the translocation with a sustained fingers opening.
Collapse
Affiliation(s)
- Shannon J McElhenney
- Department of Chemistry, University of California-Irvine, Irvine, California 92697, United States
| | - Jin Yu
- Department of Chemistry, University of California-Irvine, Irvine, California 92697, United States
- Department of Physics and Astronomy, University of California-Irvine, Irvine, California 92697, United States
| |
Collapse
|
8
|
Ahmad I, Tassawer A, Khan MU, Shehroz M, Nishan U, Sheheryar S, Dib H, Abdelfattah MAO, Shah M. Identification of novel amides and alkaloids as putative inhibitors of dopamine transporter for schizophrenia using computer-aided virtual screening. Front Pharmacol 2025; 16:1509263. [PMID: 40303924 PMCID: PMC12039762 DOI: 10.3389/fphar.2025.1509263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/27/2025] [Indexed: 05/02/2025] Open
Abstract
Schizophrenia is a complex psychiatric disorder marked by delusions, memory impairments, hallucinations, disorganized behavior, and severe cognitive deficits. Targeting the dopamine transporter (DAT) protein is promising for treating cognitive symptoms, especially in patients resistant to antipsychotic treatments. In this study, phytochemicals from six medicinal plants underwent virtual screening, and molecular dynamics simulation to identify potential agents targeting DAT. Key drug-like properties, safety, and biological activity were evaluated for identified hits. Pharmacokinetic simulation and pharmacophoric analysis were also performed. Among 990 screened phytochemicals, three alkaloids and six amides, predominantly from Piper retrofractum, and one diterpene were identified as potential antischizophrenic agents based on their stronger binding affinities and favorable docking scores compared to the standard (Lumateperone). Amides showed more potential for DAT than alkaloids. The dynamic behavior and stability of the top three amides, namely, Chenoalbicin, Dipiperamide G, and Lyciumamide C, were evaluated using various molecular dynamics analyses. RMSD (Root Mean Square Deviation), RMSF (Root Mean Square Fluctuation), Rg (Radius of Gyration), and SASA (Solvent Accessible Surface Area) analyses demonstrated favorable characteristics for all three ligands. However, binding free energy, cross-correlation, PCA (Principal Component Analysis) and FEL (Free Energy Landscape) analyses indicated that Lyciumamide C exhibited the highest stability and binding affinity in dynamic environments, Pharmacophoric features highlighted the distinct interacting components for the top three amides. Pharmacokinetic simulations revealed significant peak concentrations and sustained levels can be indicated as Lyciumamide C > Chenoalbicin > Dipiperamide G. The higher and more sustained brain concentrations of Lyciumamide C suggest its most promising pharmacokinetic profile for targeting DAT. Overall, our screened metabolites followed drug-ability criteria and require further experimental validation.
Collapse
Affiliation(s)
- Iqra Ahmad
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Anam Tassawer
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Muhammad Umer Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Shehroz
- Department of Bioinformatics, Kohsar University Murree, Murree, Pakistan
| | - Umar Nishan
- Hainan International Joint Research Center of Marine Advanced Photoelectric Functional Materials, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, China
- Department of Chemistry, Kohat University of Science and Technology, Kohat, Pakistan
| | - Sheheryar Sheheryar
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| | - Hanna Dib
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait
| | | | - Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| |
Collapse
|
9
|
Ibrahim MJ, Nangia A, Das S, Verma T, Rajeswari VD, Venkatraman G, Gnanasambandan R. Exploring Holy Basil's Bioactive Compounds for T2DM Treatment: Docking and Molecular Dynamics Simulations with Human Omentin-1. Cell Biochem Biophys 2025; 83:793-810. [PMID: 39259407 DOI: 10.1007/s12013-024-01511-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2024] [Indexed: 09/13/2024]
Abstract
Type 2 Diabetes Mellitus (T2DM) presents a substantial health concern on a global scale, driving the search for innovative therapeutic strategies. Phytochemicals from medicinal plants, particularly Ocimum tenuiflorum (Holy Basil), have garnered attention for their potential in T2DM management. The increased focus on plant-based treatments stems from their perceived safety profile, lower risk of adverse effects, and the diverse range of bioactive molecules they offer, which can target multiple pathways involved in T2DM. Computational techniques explored the binding interactions between O. tenuiflorum phytochemicals and Human Omentin-1, a potential T2DM target. ADMET evaluation and targeted docking identified lead compounds: Luteolin (-4.84 kcal/mol), Madecassic acid (-4.12 kcal/mol), Ursolic acid (-5.91 kcal/mol), Stenocereol (-5.59 kcal/mol), and Apigenin (-4.64 kcal/mol), to have a better binding affinity to target protein compared to the control drug, Metformin (-2.01 kcal/mol). Subsequent molecular dynamics simulations evaluated the stability of Stenocereol, Luteolin, and Metformin complexes for 200 nanoseconds, analysing RMSD, RMSF, RG, SASA, PCA, FEL, and MM-PBSA parameters. Results indicated Stenocereol's strong binding affinity with Omentin-1, suggesting its potential as a potent therapeutic agent for T2DM management. These findings lay the groundwork for further experimental validation and drug discovery endeavours.
Collapse
Affiliation(s)
- Mohammad Jasim Ibrahim
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Aayushi Nangia
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Soumik Das
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Tanishque Verma
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Ganesh Venkatraman
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - R Gnanasambandan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
10
|
Saha S, Sharma A, Bhowmik D, Kumar D. Investigation into in silico and in vitro approaches for inhibitors targeting MCM10 in Leishmania donovani: a comprehensive study. Mol Divers 2025; 29:575-590. [PMID: 38722455 DOI: 10.1007/s11030-024-10876-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/10/2024] [Indexed: 02/02/2025]
Abstract
Visceral Leishmaniasis (VL), the second neglected tropical disease caused by various Leishmania species, presents a significant public health challenge due to limited treatment options and the absence of vaccines. The agent responsible for visceral leishmaniasis, also referred to as "black fever" in India, is Leishmania donovani. This study focuses on L. donovani Minichromosome maintenance 10 (LdMcm10), a crucial protein in the DNA replication machinery, as a potential therapeutic target in Leishmania therapy using in silico and in vitro approaches. We employed bioinformatics tools, molecular docking, and molecular dynamics simulations to predict potential inhibitors against the target protein. The research revealed that the target protein lacks homologues in the host, emphasizing its potential as a drug target. Ligands from the DrugBank database were screened against LdMcm10 using PyRx software. The top three compounds, namely suramin, vapreotide, and pasireotide, exhibiting the best docking scores, underwent further investigation through molecular dynamic simulation and in vitro analysis. The observed structural dynamics suggested that LdMcm10-ligand complexes maintained consistent binding throughout the 300 ns simulation period, with minimal variations in their backbone. These findings suggest that these three compounds hold promise as potential lead compounds for developing new drugs against leishmaniasis. In vitro experiments also demonstrated a dose-dependent reduction in L. donovani viability for suramin, vapreotide, and pasireotide, with computed IC50 values providing quantitative metrics of their anti-leishmanial efficacy. The research offers a comprehensive understanding of LdMcm10 as a drug target and provides a foundation for further investigations and clinical exploration, ultimately advancing drug discovery strategies for leishmaniasis treatment.
Collapse
Affiliation(s)
- Satabdi Saha
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| | - Anupama Sharma
- Department of Computational Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Deep Bhowmik
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
11
|
Cordeiro Josino LP, da Penha Valente RP, de Souza da Silva ML, Alves CN, Lima AH. Molecular dynamics of transferrin receptor binder peptides: unlocking blood-brain barrier for enhanced CNS drug delivery. J Biomol Struct Dyn 2025:1-10. [PMID: 39743789 DOI: 10.1080/07391102.2024.2446676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/27/2024] [Indexed: 01/04/2025]
Abstract
A cystine-dense peptide (CDP) named TfRB1 was identified for its ability to bind to the transferrin receptor (TfR). CDPs are stabilized by their disulfide bonds, and variants of TfRB1 - specifically TfRB1G1, TfRB1G2, and TfRB1G3 - are explored for their potential to transport molecules across the blood-brain barrier (BBB) into the central nervous system (CNS). This study employed molecular modeling and dynamics simulations to characterize the interactions between these TfRB1 variants and TfR. Binding free energy calculations showed a strong correlation with experimental binding affinities of -10.99 kcal/mol for TfRB1G2 and -13.18 kcal/mol for TfRB1G3, with a relative error of 1.98%. The key forces driving these interactions include electrostatic and van der Waals forces, with mutations in TfRB1G3 (T9M and A13D) enhancing its binding affinity through improved interactions with residues such as Arg633. The free energy landscape analysis revealed that TfRB1G3 maintains the N-terminal residues of TfR in an α-helical conformation, unlike TfRB1G2. Per-residue free energy decomposition identified key residues - Leu619, Arg629, Tyr643, and Phe650 - as crucial for TfR binding, underscoring their competitive nature with transferrin. Additionally, Glu612, which is favorable for binding in TfRB1G2, becomes unfavorable in TfRB1G3. Conversely, Arg633 shifts from unfavorable in TfRB1G2 to favorable in TfRB1G3, compensating for the loss of favorable interaction with Glu612. These findings provide valuable molecular insights into the TfRB1 peptides' potential as drug carriers, highlighting their capability to deliver molecules to the CNS and compete with transferrin for BBB transport.
Collapse
Affiliation(s)
- Luiz Patrick Cordeiro Josino
- Programa de Pós-Graduação em Química Medicinal e Modelagem Molecular, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belem, Brazil
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Universidade Federal do Pará, Belem, Brazil
| | - Renan Patrick da Penha Valente
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Universidade Federal do Pará, Belem, Brazil
- Programa de Pós-Graduação em Química, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Belem, Brazil
| | - Maria Luane de Souza da Silva
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Universidade Federal do Pará, Belem, Brazil
- Programa de Pós-Graduação em Química, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Belem, Brazil
| | - Cláudio Nahum Alves
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Universidade Federal do Pará, Belem, Brazil
- Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Belem, Brazil
| | - Anderson H Lima
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Universidade Federal do Pará, Belem, Brazil
- Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Belem, Brazil
| |
Collapse
|
12
|
Moradi S, Nowroozi A, Aryaei Nezhad M, Jalali P, Khosravi R, Shahlaei M. A review on description dynamics and conformational changes of proteins using combination of principal component analysis and molecular dynamics simulation. Comput Biol Med 2024; 183:109245. [PMID: 39388840 DOI: 10.1016/j.compbiomed.2024.109245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Understanding how proteins behave dynamically and undergo conformational changes is essential to comprehending their biological roles. This review article examines the potent tool of using Molecular Dynamics simulations in conjunction with Principal Component Analysis (PCA) to explore protein dynamics. Molecular dynamics data can be made easier to read by removing prominent patterns through the use of PCA, a sophisticated dimensionality reduction approach. Researchers can obtain critical insights into the fundamental principles governing protein function by using PCA on MD simulation data. We provide a systematic approach to PCA that includes data collection, input coordinate selection, and result interpretation. Protein collective movements and fundamental dynamics are made visible by PCA, which makes it possible to identify conformational substates that are crucial to function. By means of principal component analysis, scientists are able to observe and measure large-scale movements, like hinge bending and domain motions, as well as pinpoint areas of protein structural stiffness and flexibility. Moreover, PCA allows temporal separation, distinguishing slower global motions from faster local changes. A strong foundation for researching protein dynamics is provided by the combination of PCA and Molecular Dynamics simulations, which have applications in drug development and enhance our comprehension of intricate biological systems.
Collapse
Affiliation(s)
- Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Nowroozi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Aryaei Nezhad
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parvin Jalali
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rasool Khosravi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
13
|
Zhuang L, Ali A, Yang L, Ye Z, Li L, Ni R, An Y, Ali SL, Gong W. Leveraging computer-aided design and artificial intelligence to develop a next-generation multi-epitope tuberculosis vaccine candidate. INFECTIOUS MEDICINE 2024; 3:100148. [DOI: https:/doi.org/10.1016/j.imj.2024.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Zhuang L, Ali A, Yang L, Ye Z, Li L, Ni R, An Y, Ali SL, Gong W. Leveraging computer-aided design and artificial intelligence to develop a next-generation multi-epitope tuberculosis vaccine candidate. INFECTIOUS MEDICINE 2024; 3:100148. [PMID: 39687693 PMCID: PMC11647498 DOI: 10.1016/j.imj.2024.100148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Tuberculosis (TB) remains a global public health challenge. The existing Bacillus Calmette-Guérin vaccine has limited efficacy in preventing adult pulmonary TB, necessitating the development of new vaccines with improved protective effects. METHODS Computer-aided design and artificial intelligence technologies, combined with bioinformatics and immunoinformatics approaches, were used to design a multi-epitope vaccine (MEV) against TB. Comprehensive bioinformatics analyses were conducted to evaluate the physicochemical properties, spatial structure, immunogenicity, molecular dynamics (MD), and immunological characteristics of the MEV. RESULTS We constructed a MEV, designated ZL12138L, containing 13 helper T lymphocyte epitopes, 12 cytotoxic T lymphocyte epitopes, 8 B-cell epitopes, as well as Toll-like receptor (TLR) agonists and helper peptides. Bioinformatics analyses revealed that ZL12138L should exhibit excellent immunogenicity and antigenicity, with no toxicity or allergenicity, and had potential to induce robust immune responses and high solubility, the immunogenicity score was 4.14449, the antigenicity score was 0.8843, and the immunological score was 0.470. Moreover, ZL12138L showed high population coverage for human leukocyte antigen class I and II alleles, reaching 92.41% and 90.17%, respectively, globally. Molecular docking analysis indicated favorable binding affinity of ZL12138L with TLR-2 and TLR-4, with binding energies of -1173.4 and -1360.5 kcal/mol, respectively. Normal mode analysis and MD simulations indicated the stability and dynamic properties of the vaccine construct. Immune simulation predictions suggested that ZL12138L could effectively activate innate and adaptive immune cells, inducing high levels of Type 1 T helper cell cytokines. CONCLUSIONS This study provides compelling evidence for ZL12138L as a promising TB vaccine candidate. Future research will focus on experimental validation and further optimization of the vaccine design.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Awais Ali
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Ling Yang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Zhaoyang Ye
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Linsheng Li
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Ruizi Ni
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Yajing An
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Syed Luqman Ali
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| |
Collapse
|
15
|
Alghamdi A, A Awadh Ali N, Alafnan A, Zainal Abidin SA, Alamri A, Hussein W, Khalifa NE, Awaji AYM, Al Rashah K, Ahmed Younes KM, Anwar S. Deciphering the toxicological and computational assessment of Verbascum yemenese.: Integrating phytochemistry and bioinformatics tools. Food Chem Toxicol 2024; 193:115028. [PMID: 39368542 DOI: 10.1016/j.fct.2024.115028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/17/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
This study explores the phytochemical composition and biological activities of Verbascum yemenense, a plant known for its medicinal properties. The plant extract revealed a rich presence of bioactive compounds that exhibited significant antioxidant properties against free radicals. The enzyme inhibition potential was particularly notable against cholinesterases (AChE: 2.56 mg GALAE/g; BChE: 1.98 mg GALAE/g), and tyrosinase (87.94 mg KAE/g), α-glucosidase suggesting potential therapeutic applications in neurodegenerative diseases, skin disorders and diabetes. Molecular docking studies and Molecular Dynamics simulations, providing insights into the interaction mechanisms of the identified compounds with the target proteins. Molecular docking studies revealed high binding affinities of the phytoconstituents, with compounds like VY4 and phyllanthusol-A (VY15) showing substantial docking scores against AChE (-9.840 kcal/mol) and BChE (-9.853 kcal/mol), respectively. For instance, the RMSD values during the MD simulations for compound VY17 in the AML complex showed a stable conformation, fluctuating within a range of 0.75 Å to 1.75 Å, indicating a strong and consistent interaction with the enzyme. MESP studies highlighted VY17's distinctive electrostatic features, notably a pronounced electronegative region, which might contribute to its binding efficiency. These findings suggest that V. yemenense is a promising candidate for developing novel therapeutic agents.
Collapse
Affiliation(s)
- Adel Alghamdi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Baha University, P.O. Box 1988, Al-Baha, Saudi Arabia
| | - Nasser A Awadh Ali
- Department of Pharmacognosy and Medicinal Herbs, Faculty of Pharmacy, Al-Baha University, P.O. Box 1988, Al-Baha, Saudi Arabia
| | - Ahmed Alafnan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Malaysia
| | - Abdulwahab Alamri
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Weiam Hussein
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Nasrin E Khalifa
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | | | - Khaled Al Rashah
- Medical Services, Ministry of Interior, Comprehensive Specialized Clinic for Security Forces, Najran, Saudi Arabia
| | - Kareem Mahmoud Ahmed Younes
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia; Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia.
| |
Collapse
|
16
|
Siddiquee NH, Talukder MEK, Ahmed E, Zeba LT, Aivy FS, Rahman MH, Barua D, Rumman R, Hossain MI, Shimul MEK, Rama AR, Chowdhury S, Hossain I. Cheminformatics-based analysis identified (Z)-2-(2,5-dimethoxy benzylidene)-6-(2-(4-methoxyphenyl)-2-oxoethoxy) benzofuran-3(2H)-one as an inhibitor of Marburg replication by interacting with NP. Microb Pathog 2024; 195:106892. [PMID: 39216611 DOI: 10.1016/j.micpath.2024.106892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The highly pathogenic Marburg virus (MARV) is a member of the Filoviridae family, a non-segmented negative-strand RNA virus. This article represents the computer-aided drug design (CADD) approach for identifying drug-like compounds that prevent the MARV virus disease by inhibiting nucleoprotein, which is responsible for their replication. This study used a wide range of in silico drug design techniques to identify potential drugs. Out of 368 natural compounds, 202 compounds passed ADMET, and molecular docking identified the top two molecules (CID: 1804018 and 5280520) with a high binding affinity of -6.77 and -6.672 kcal/mol, respectively. Both compounds showed interactions with the common amino acid residues SER_216, ARG_215, TYR_135, CYS_195, and ILE_108, which indicates that lead compounds and control ligands interact in the common active site/catalytic site of the protein. The negative binding free energies of CID: 1804018 and 5280520 were -66.01 and -31.29 kcal/mol, respectively. Two lead compounds were re-evaluated using MD modeling techniques, which confirmed CID: 1804018 as the most stable when complexed with the target protein. PC3 of the (Z)-2-(2,5-dimethoxybenzylidene)-6-(2-(4-methoxyphenyl)-2-oxoethoxy) benzofuran-3(2H)-one (CID: 1804018) was 8.74 %, whereas PC3 of the 2'-Hydroxydaidzein (CID: 5280520) was 11.25 %. In this study, (Z)-2-(2,5-dimethoxybenzylidene)-6-(2-(4-methoxyphenyl)-2-oxoethoxy) benzofuran-3(2H)-one (CID: 1804018) unveiled the significant stability of the proteins' binding site in ADMET, Molecular docking, MM-GBSA and MD simulation analysis studies, which also showed a high negative binding free energy value, confirming as the best drug candidate which is found in Angelica archangelica which may potentially inhibit the replication of MARV nucleoprotein.
Collapse
Affiliation(s)
- Noimul Hasan Siddiquee
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh; Bioinformatics Laboratory (BioLab), Bangladesh
| | - Md Enamul Kabir Talukder
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Bangladesh
| | - Ezaz Ahmed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh; Bioinformatics Laboratory (BioLab), Bangladesh
| | - Labiba Tasnim Zeba
- Bioinformatics Laboratory (BioLab), Bangladesh; Department of Mathematics & Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Farjana Sultana Aivy
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh; Bioinformatics Laboratory (BioLab), Bangladesh
| | - Md Hasibur Rahman
- Bioinformatics Laboratory (BioLab), Bangladesh; Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Durjoy Barua
- Bioinformatics Laboratory (BioLab), Bangladesh; Department of Pharmacy, BGC Trust University, Bangladesh
| | - Rahnumazzaman Rumman
- Bioinformatics Laboratory (BioLab), Bangladesh; Department Of Environmental Science and Disaster Management, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Ifteker Hossain
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh; Bioinformatics Laboratory (BioLab), Bangladesh
| | - Md Ebrahim Khalil Shimul
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Bangladesh
| | - Anika Rahman Rama
- Bioinformatics Laboratory (BioLab), Bangladesh; Department of Genetic Engineering and Biotechnology, East West University, Dhaka, Bangladesh
| | - Sristi Chowdhury
- Bioinformatics Laboratory (BioLab), Bangladesh; Department of Biochemistry and Molecular Biology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Imam Hossain
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh; Bioinformatics Laboratory (BioLab), Bangladesh.
| |
Collapse
|
17
|
Khan MU, Sakhawat A, Rehman R, Wali AH, Ghani MU, Akram A, Javed MA, Ali Q, Yu-Ming Z, Ali D, Yu-Ming Z. Identification of novel natural compounds against CFTR p.Gly628Arg pathogenic variant. AMB Express 2024; 14:99. [PMID: 39249658 PMCID: PMC11383896 DOI: 10.1186/s13568-024-01762-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) protein is an ion channel found in numerous epithelia and controls the flow of water and salt across the epithelium. The aim of our study to find natural compounds that can improve lung function for people with cystic fibrosis (CF) caused by the p.Gly628Arg (rs397508316) mutation of CFTR protein. The sequence of CFTR protein as a target structure was retrieved from UniProt and PDB database. The ligands that included Armepavine, Osthole, Curcumin, Plumbagine, Quercetin, and one Trikafta (R*) reference drug were screened out from PubChem database. Autodock vina software carried out docking, and binding energies between the drug and the target were included using docking-score. The following tools examined binding energy, interaction, stability, toxicity, and visualize protein-ligand complexes. The compounds having binding energies of -6.4, -5.1, -6.6, -5.1, and - 6.5 kcal/mol for Armepavine, Osthole, Curcumin, Plumbagine, Quercetin, and R*-drug, respectively with mutated CFTR (Gly628Arg) structure were chosen as the most promising ligands. The ligands bind to the mutated CFTR protein structure active sites in hydrophobic bonds, hydrogen bonds, and electrostatic interactions. According to ADMET analyses, the ligands Armepavine and Quercetin also displayed good pharmacokinetic and toxicity characteristics. An MD simulation for 200 ns was also established to ensure that Armepavine and Quercetin ligands attached to the target protein favorably and dynamically, and that protein-ligand complex stability was maintained. It is concluded that Armepavine and Quercetin have stronger capacity to inhibit the effect of mutated CFTR protein through improved trafficking and restoration of original function.
Collapse
Affiliation(s)
- Muhammad Umer Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan.
| | - Azra Sakhawat
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Raima Rehman
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Abbas Haider Wali
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Usman Ghani
- Precision Genomics Research Lab, Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Areeba Akram
- Precision Genomics Research Lab, Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Arshad Javed
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan
| | - Qurban Ali
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan.
| | - Zhou Yu-Ming
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, 341000, Jiangxi Province, P.R. China
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh, 11451, Saudi Arabia
| | - Zhou Yu-Ming
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, 341000, Jiangxi Province, P.R. China
| |
Collapse
|
18
|
Rigobello L, Lugli F, Caporali L, Bartocci A, Fadanni J, Zerbetto F, Iommarini L, Carelli V, Ghelli AM, Musiani F. A computational study to assess the pathogenicity of single or combinations of missense variants on respiratory complex I. Int J Biol Macromol 2024; 273:133086. [PMID: 38871105 DOI: 10.1016/j.ijbiomac.2024.133086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Variants found in the respiratory complex I (CI) subunit genes encoded by mitochondrial DNA can cause severe genetic diseases. However, it is difficult to establish a priori whether a single or a combination of CI variants may impact oxidative phosphorylation. Here we propose a computational approach based on coarse-grained molecular dynamics simulations aimed at investigating new CI variants. One of the primary CI variants associated with the Leber hereditary optic neuropathy (m.14484T>C/MT-ND6) was used as a test case and was investigated alone or in combination with two additional rare CI variants whose role remains uncertain. We found that the primary variant positioned in the E-channel region, which is fundamental for CI function, stiffens the enzyme dynamics. Moreover, a new mechanism for the transition between π- and α-conformation in the helix carrying the primary variant is proposed. This may have implications for the E-channel opening/closing mechanism. Finally, our findings show that one of the rare variants, located next to the primary one, further worsens the stiffening, while the other rare variant does not affect CI function. This approach may be extended to other variants candidate to exert a pathogenic impact on CI dynamics, or to investigate the interaction of multiple variants.
Collapse
Affiliation(s)
- Laura Rigobello
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna I-40127, Italy
| | - Francesca Lugli
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Bologna I-40126, Italy.
| | - Leonardo Caporali
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna I-40124, Italy
| | - Alessio Bartocci
- Department of Physics, University of Trento, Trento I-38123, Italy; INFN-TIFPA, Trento Institute for Fundamental Physics and Applications, Trento I-38123, Italy
| | - Jacopo Fadanni
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Bologna I-40126, Italy
| | - Francesco Zerbetto
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Bologna I-40126, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna I-40127, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna I-40124, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna I-40123, Italy
| | - Anna Maria Ghelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna I-40127, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna I-40124, Italy
| | - Francesco Musiani
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna I-40127, Italy.
| |
Collapse
|
19
|
Palma J, Pierdominici-Sottile G. Fortuitous Correlations in Molecular Dynamics Simulations: Their Harmful Influence on the Probability Distributions of the Main Principal Components. ACS OMEGA 2024; 9:20488-20501. [PMID: 38737025 PMCID: PMC11080035 DOI: 10.1021/acsomega.4c01515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 05/14/2024]
Abstract
Nonsense correlations frequently develop between independent random variables that evolve with time. Therefore, it is not surprising that they appear between the components of vectors carrying out multidimensional random walks, such as those describing the trajectories of biomolecules in molecular dynamics simulations. The existence of these correlations does not imply in itself a problem. Still, it can present a problem when the trajectories are analyzed with an algorithm such as the Principal Component Analysis (PCA) because it seeks to maximize correlations without discriminating whether they have physical origin or not. In this Article, we employ random walks occurring on multidimensional harmonic potentials to evaluate the influence of fortuitous correlations in PCA. We demonstrate that, because of them, this algorithm affords misleading results when applied to a single trajectory. The errors do not only affect the directions of the first eigenvectors and their eigenvalues, but the very definition of the molecule's "essential space" may be wrong. Additionally, the main principal component's probability distributions present artificial structures which do not correspond with the shape of the potential energy surface. Finally, we show that the PCA of two realistic protein models, human serum albumin and lysozyme, behave similarly to the simple harmonic models. In all cases, the problems can be mitigated and eventually eliminated by doing PCA on concatenated trajectories formed from a large enough number of individual simulations.
Collapse
Affiliation(s)
- Juliana Palma
- Departamento
de Ciencia y Tecnología, Universidad
Nacional de Quilmes, Sáenz Peña 352 Bernal, Buenos
Aires B1876BXD, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires C1425FQB, Argentina
| | - Gustavo Pierdominici-Sottile
- Departamento
de Ciencia y Tecnología, Universidad
Nacional de Quilmes, Sáenz Peña 352 Bernal, Buenos
Aires B1876BXD, Argentina
- Consejo
Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires C1425FQB, Argentina
| |
Collapse
|
20
|
Nath M, Bhowmik D, Saha S, Nandi R, Kumar D. Identification of potential inhibitor against Leishmania donovani mitochondrial DNA primase through in-silico and in vitro drug repurposing approaches. Sci Rep 2024; 14:3246. [PMID: 38332162 PMCID: PMC10853515 DOI: 10.1038/s41598-024-53316-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Leishmania donovani is the causal organism of leishmaniasis with critical health implications affecting about 12 million people around the globe. Due to less efficacy, adverse side effects, and resistance, the available therapeutic molecules fail to control leishmaniasis. The mitochondrial primase of Leishmania donovani (LdmtPRI1) is a vital cog in the DNA replication mechanism, as the enzyme initiates the replication of the mitochondrial genome of Leishmania donovani. Hence, we target this protein as a probable drug target against leishmaniasis. The de-novo approach enabled computational prediction of the three-dimensional structure of LdmtPRI1, and its active sites were identified. Ligands from commercially available drug compounds were selected and docked against LdmtPRI1. The compounds were chosen for pharmacokinetic study and molecular dynamics simulation based on their binding energies and protein interactions. The LdmtPRI1 gene was cloned, overexpressed, and purified, and a primase activity assay was performed. The selected compounds were verified experimentally by the parasite and primase inhibition assay. Capecitabine was observed to be effective against the promastigote form of Leishmania donovani, as well as inhibiting primase activity. This study's findings suggest capecitabine might be a potential anti-leishmanial drug candidate after adequate further studies.
Collapse
Affiliation(s)
- Mitul Nath
- Department of Microbiology, Assam University, Silchar, Assam, 788011, India
| | - Deep Bhowmik
- Department of Microbiology, Assam University, Silchar, Assam, 788011, India
| | - Satabdi Saha
- Department of Microbiology, Assam University, Silchar, Assam, 788011, India
| | - Rajat Nandi
- Department of Microbiology, Assam University, Silchar, Assam, 788011, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, Assam, 788011, India.
| |
Collapse
|
21
|
Oladipo SD, Luckay RC, Olofinsan KA, Obakachi VA, Zamisa SJ, Adeleke AA, Badeji AA, Ogundare SA, George BP. Antidiabetes and antioxidant potential of Schiff bases derived from 2-naphthaldehye and substituted aromatic amines: Synthesis, crystal structure, Hirshfeld surface analysis, computational, and invitro studies. Heliyon 2024; 10:e23174. [PMID: 38163168 PMCID: PMC10756989 DOI: 10.1016/j.heliyon.2023.e23174] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Three Schiff bases were synthesised by the condensation reaction between 2-napthaldehyde and aromatic amines to afford (E)-N-mesityl-1-(naphthalen-2-yl)methanimine (L1), (E)-N-(2,6-dimethylphenyl)-1-(naphthalen-2-yl)methanimine (L2) and (E)-N-(2,6-diisopropylphenyl)-1-(naphthalen-2-yl)methanimine (L3). The synthesised compounds were characterised using UV-visible, NMR (13C & 1H), and Fourier transform infrared spectroscopic methods while their purity was ascertained by elemental analysis. Structural analysis revealed that the naphthalene ring is almost coplanar with the imine functional group as evident by C1-C10-C11-N1 torsion angles of 176.4(2)° and 179.4(1)° in L2 and L3, respectively. Of all the various intermolecular contacts, H⋯H interactions contributed mostly towards the Hirshfeld surfaces of both L2 (58.7 %) and L3 (69.7 %). Quantum chemical descriptors of L1 - L3 were determined using Density Functional Theory (DFT) and the results obtained showed that the energy band gap (ΔE) for L1, L2 and L3 are 3.872, 4.023 and 4.004 eV respectively. The antidiabetic potential of the three compounds were studied using α-amylase and α-glucosidase assay. Compound L1 showed very promising antidiabetic activities with IC50 values of 58.85 μg/mL and 57.60 μg/mL while the reference drug (Acarbose) had 405.84 μg/mL and 35.69 μg/mL for α-amylase and α-glucosidase respectively. In-silico studies showed that L1 docking score as well as binding energies are higher than that of acarbose, which are recognized inhibitors of α-amylase together with α-glucosidase. Further insight from the RMSF, RMSD and RoG analysis predicted that, throughout the simulation L1 showcased evident influence on the structural stability of α-amylase. The antioxidant potential of the compounds was carried out using nitric oxide (NO), ferric reducing ability power (FRAP) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) assays. The compounds exhibited good to fairly antioxidant properties with L1 as well as L3 having IC50 values of 70.91 and 91.21 μg/mL respectively for NO scavenging activities assay, which comparatively outshined acarbose (reference drug) with IC50 value of 109.95 μg/mL. Pharmacology and pharmacokinetics approximations of L1 - L3 showed minimal violation of Lipinski's Ro5 and this projects them to be less toxic and orally bioavailable as potential templates for the design of therapeutics with antioxidant and antidiabetic activities.
Collapse
Affiliation(s)
- Segun D. Oladipo
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
- Department of Chemical Sciences, Olabisi Onabanjo University, P.M.B 2002, Ago-Iwoye, Nigeria
| | - Robert C. Luckay
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Kolawole A. Olofinsan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O Box 17011, Doornfontein, 2028, South Africa
| | - Vincent A. Obakachi
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| | - Sizwe J. Zamisa
- School of Chemistry and Physics, Westville Campus, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000, South Africa
| | - Adesola A. Adeleke
- Department of Chemical Sciences, Olabisi Onabanjo University, P.M.B 2002, Ago-Iwoye, Nigeria
| | - Abosede A. Badeji
- Department of Chemical Sciences, Tai Solarin University of Education, Ijagun, Ogun State, Nigeria
| | - Segun A. Ogundare
- Department of Chemical Sciences, Olabisi Onabanjo University, P.M.B 2002, Ago-Iwoye, Nigeria
| | - Blassan P. George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O Box 17011, Doornfontein, 2028, South Africa
| |
Collapse
|
22
|
Zhuang L, Zhao Y, Yang L, Li L, Ye Z, Ali A, An Y, Ni R, Ali SL, Gong W. Harnessing bioinformatics for the development of a promising multi-epitope vaccine against tuberculosis: The ZL9810L vaccine. DECODING INFECTION AND TRANSMISSION 2024; 2:100026. [DOI: https:/doi.org/10.1016/j.dcit.2024.100026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Ormazábal A, Palma J, Pierdominici-Sottile G. Dynamics and Function of sRNA/mRNAs Under the Scrutiny of Computational Simulation Methods. Methods Mol Biol 2024; 2741:207-238. [PMID: 38217656 DOI: 10.1007/978-1-0716-3565-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
Molecular dynamics simulations have proved extremely useful in investigating the functioning of proteins with atomic-scale resolution. Many applications to the study of RNA also exist, and their number increases by the day. However, implementing MD simulations for RNA molecules in solution faces challenges that the MD practitioner must be aware of for the appropriate use of this tool. In this chapter, we present the fundamentals of MD simulations, in general, and the peculiarities of RNA simulations, in particular. We discuss the strengths and limitations of the technique and provide examples of its application to elucidate small RNA's performance.
Collapse
Affiliation(s)
- Agustín Ormazábal
- Departmento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Godoy Cruz, CABA, Argentina
| | - Juliana Palma
- Departmento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Godoy Cruz, CABA, Argentina
| | - Gustavo Pierdominici-Sottile
- Departmento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Godoy Cruz, CABA, Argentina.
| |
Collapse
|
24
|
Verkhivker G, Alshahrani M, Gupta G. Exploring Conformational Landscapes and Cryptic Binding Pockets in Distinct Functional States of the SARS-CoV-2 Omicron BA.1 and BA.2 Trimers: Mutation-Induced Modulation of Protein Dynamics and Network-Guided Prediction of Variant-Specific Allosteric Binding Sites. Viruses 2023; 15:2009. [PMID: 37896786 PMCID: PMC10610873 DOI: 10.3390/v15102009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
A significant body of experimental structures of SARS-CoV-2 spike trimers for the BA.1 and BA.2 variants revealed a considerable plasticity of the spike protein and the emergence of druggable binding pockets. Understanding the interplay of conformational dynamics changes induced by the Omicron variants and the identification of cryptic dynamic binding pockets in the S protein is of paramount importance as exploring broad-spectrum antiviral agents to combat the emerging variants is imperative. In the current study, we explore conformational landscapes and characterize the universe of binding pockets in multiple open and closed functional spike states of the BA.1 and BA.2 Omicron variants. By using a combination of atomistic simulations, a dynamics network analysis, and an allostery-guided network screening of binding pockets in the conformational ensembles of the BA.1 and BA.2 spike conformations, we identified all experimentally known allosteric sites and discovered significant variant-specific differences in the distribution of binding sites in the BA.1 and BA.2 trimers. This study provided a structural characterization of the predicted cryptic pockets and captured the experimentally known allosteric sites, revealing the critical role of conformational plasticity in modulating the distribution and cross-talk between functional binding sites. We found that mutational and dynamic changes in the BA.1 variant can induce the remodeling and stabilization of a known druggable pocket in the N-terminal domain, while this pocket is drastically altered and may no longer be available for ligand binding in the BA.2 variant. Our results predicted the experimentally known allosteric site in the receptor-binding domain that remains stable and ranks as the most favorable site in the conformational ensembles of the BA.2 variant but could become fragmented and less probable in BA.1 conformations. We also uncovered several cryptic pockets formed at the inter-domain and inter-protomer interface, including functional regions of the S2 subunit and stem helix region, which are consistent with the known role of pocket residues in modulating conformational transitions and antibody recognition. The results of this study are particularly significant for understanding the dynamic and network features of the universe of available binding pockets in spike proteins, as well as the effects of the Omicron-variant-specific modulation of preferential druggable pockets. The exploration of predicted druggable sites can present a new and previously underappreciated opportunity for therapeutic interventions for Omicron variants through the conformation-selective and variant-specific targeting of functional sites involved in allosteric changes.
Collapse
Affiliation(s)
- Gennady Verkhivker
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Mohammed Alshahrani
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
| | - Grace Gupta
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
| |
Collapse
|