1
|
Golivi Y, Behera SK, Alam A, Peela S. Interaction of Heat Shock Protein 90 (HSP90), Ganetespib, and 5-Fluorouracil by Computational Approach for Colorectal Cancer Therapy. Crit Rev Oncog 2025; 30:31-42. [PMID: 39819433 DOI: 10.1615/critrevoncog.2024056394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The heat shock protein 90 kDa (HSP90) is highly conserved across diverse species, including humans, and upregulated in various cancers. As a result, it has been identified as a promising target for advancing anticancer medicine. The introduction of combinatorial chemistry in drug discovery has emphasized the need to develop new technologies in screening, designing, decoding, synthesizing, and screening combinatorial drug libraries. The current investigation was carried out to report improved inhibition efficacy of ganetespib, fluorouracil (5-FU), and its combinatorial drug treatment (ganetespib + 5-FU) against the HSP90 molecular chaperone through an in silico approach. Both drugs and their combination are ATP-competitive inhibitors; they inhibit the HSP90α N-terminal and block the ATP binding site. The structural and functional basis and their combination were confirmed through molecular docking interaction with HSP90α. The inhibitors' conformational effects and their combination against the HSP90α protein were studied using powerful MD simulations. The key interacting residues of HSP90α with ganetespib, 5-FU, and ganetespib + 5-FU were identified via energy binding calculations and molecular dynamics. This study is the first to offer atomistic insights into the interaction between ganetespib, 5-FU, and ganetespib + 5-FU with the HSP90α protein N-terminal domain. The results of our in silico study will open better avenues for developing potential cancer inhibitors in the near future.
Collapse
Affiliation(s)
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, (NIPER), Ahmedabad, Gujarat 382355, India
| | - Afroz Alam
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sujatha Peela
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam 532410, Andhra Pradesh, India
| |
Collapse
|
2
|
Behera S, Gupta N. In Silico Analysis of Anti-Cancer Activity of Exopolysaccharide Isolated from Novel Pseudolagarobasidium acaciicola through Mass Production, Gel Permeation Separation, and Compositional Analysis. Crit Rev Oncog 2025; 30:119-135. [PMID: 39819439 DOI: 10.1615/critrevoncog.2024056496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Bacteria, fungi, and algae are examples of microorganisms that synthesize polysaccharides, which are macromolecules that belong to the carbohydrate class. Production of polysaccharides represents an alternative to chemical and plant-derived compounds that could be used for human well-being which requires implementation of different methods standardized during the extraction and purification process. In the current investigation, Pseudolagarobasidium acaciicola, a novel fungal source of exopolysaccharide (EPS) was used which produced 2773.23 ± 100.39 mg/L when cultured under pre-optimized composed medium for 7 days under submerged culture conditions. Biochemical estimation of crude polysaccharides revealed the presence of carbohydrates, protein, reducing sugar, least phenolics and no flavonoids. Partially purified EPS (ppEPS) was subjected to monosaccharide analysis, molecular weight determination and structural confirmation using FTIR and LCMS analysis. The presence of maltose, fructose, xylose, galactose, glucose, raffinose and sorbose was evident in the ppEPS using HPTLC at 285 nm, with molecular weight of dextran 70 (tentative). Characterization revealed the presence of functional groups including -OH, -COO, C-O-C and C-O with compounds like cellulose, phosphate and 3'-Sialyl-N-acetyllactosamine with glycan as the main structural form. Hence, our hypothesis is: the fungal strain may be used as a novel source of glycan and explore more possibilities for enhanced recovery of EPS important for further drug discovery and formulation programs. Based on existing research on the anti-cancerous characteristics of β-Glycans, an in silico study was carried out, which suggested that β-Glycans may operate more potent against its receptor CLEC7A than the oral chemotherapy drug imatinib.
Collapse
Affiliation(s)
- Smita Behera
- Plant Pathology and Microbiology Laboratory, Regional Plant Resource Centre, Bhubaneswar, India
| | - Nibha Gupta
- Plant Pathology and Microbiology Laboratory, Regional Plant Resource Centre, Bhubaneswar, India
| |
Collapse
|
3
|
Das RP, Behera SK, Sahoo B, Arakha M, Pradhan AK. Comparative analysis of backbone atom cross-correlation matrices and folding dynamics of amyloid fibril and its complexes with novel biosurfactants isolated from Bacillus strain: a binding free energy calculation (mM-PBSA) and MD simulation approach. J Biomol Struct Dyn 2024:1-16. [PMID: 39731748 DOI: 10.1080/07391102.2024.2446677] [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: 03/06/2024] [Accepted: 09/25/2024] [Indexed: 12/30/2024]
Abstract
In the relentless pursuit of unraveling the intricate pathophysiology of Alzheimer's disease (AD), amyloid β (Aβ) proteins emerge as focal points due to their pivotal role in disease progression. The pathological hallmark of AD involves the aberrant aggregation of Aβ peptides into amyloid fibrils, precipitating a cascade of neurodegenerative events culminating in cognitive decline and neuronal loss. This study adopts a computational framework to investigate the potential therapeutic efficacy of novel biosurfactants (BS) in mitigating Aβ fibril formation. Initial analyses encompassing sequence alignment, structural elucidation, and functional characterization reveal distinctive attributes of the Aβ peptide and the identified BS candidates. Quantum chemical calculations, using the ORCA Program (v4.0) employed Density Functional Theory (DFT), specifically the Becke 3-parameter Lee-Yang-Parr (B3LYP) method, to investigate the electronic structure and energetics of novel isolates. Molecular docking through AutoDock Vina (version 1.1.2) employing advanced algorithms elucidates the binding affinities and interaction energies between Aβ fibrils and BS molecules. The observed binding energy of -7.0 kcal/mol for BG2A and -6.6 kcal/mol for BG2B, underscoring the robustness and stability of the formed complexes. The binding mechanism of docked complexes was predicted through molecular dynamics (MD) simulations using GROMACS 2021.3 and Charmm36 force field, capture complex dynamics over 100 nanoseconds. Analysis via RMSD, RMSF, Rg, PCA, and SASA offers insights into Aβ-BS complex stability and dynamics. These promising results highlight the potential of BG2A and BG2B as therapeutic candidates against AD. However, rigorous preclinical and clinical validation is crucial to ascertain their safety, efficacy, and translational relevance.
Collapse
Affiliation(s)
- Rohit Pritam Das
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Banishree Sahoo
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Manoranjan Arakha
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Arun Kumar Pradhan
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| |
Collapse
|
4
|
Labidi A, Bensghaier R, Jebali S, Latrous L. Validated LC/MS method for simultaneous determination of elbasvir and grazoprevir in human plasma. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:1034-1045. [PMID: 38823440 DOI: 10.1016/j.pharma.2024.05.006] [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: 02/27/2024] [Revised: 04/14/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
A sensitive and accurate LC/MS method for the determination of elbasvir (ELB) and grazoprevir (GZP) in human plasma was established using daclatasvir (DCT) as an internal standard. The analytes were separated on a Waters Spherisorb phenyl column (150mm×4.6mm ID, 5μm particle size) maintained at 40°C±2°C. Gradient elution, at a flow rate of 0.8mLmin-1, was used. The mobile phase consists of 90% of acetonitrile mixed to 10% of a 5mM ammonium formate buffer (+0.1% v/v of trimethylamine, pH was adjusted to 3.2 by formic acid) as phase A and 10% of acetonitrile mixed to 90% of the same buffer as phase B. Liquid-liquid extraction with ethyl acetate solvent was used to recuperate compounds from plasma. The method was validated over a concentration range of 2 and 100ng/mL for GZP and between 1 and 50ng/mL for ELB. The intra- and inter-day precision and accuracy of the quality control samples at low, medium, and high concentration levels exhibited relative standard deviations (RSD)<15%, and the accuracy values ranged from 94.2 to 107.8%. The robustness of the method was established using a two-level full factorial design.
Collapse
Affiliation(s)
- Aymen Labidi
- Laboratoire National de Contrôle Des Médicaments, 11 bis, Rue Jebel Lakhdar Bab Saadoun, 1006 Tunis, Tunisia
| | - Rafika Bensghaier
- Laboratoire de Chimie Minérale Appliquée (LR19ES02), Faculté Des Sciences de Tunis, Université de Tunis El Manar, Campus Universitaire Farhat HACHED, 2092 Tunis, Tunisia; Laboratoire des Matériaux Composites et des Minéraux Argileux, Centre National de Recherches en Sciences des Matériaux Technopôle Borj Cedria, BP 73, 8027 Soliman, Tunisia
| | - Sami Jebali
- Institut National de Recherche et d'Analyse Physico-chimique, Laboratoire Matériaux Traitement et Analyse (LMTA), Biotechpole Sidi-Thabet, Ariana, Tunisia
| | - Latifa Latrous
- Laboratoire de Chimie Minérale Appliquée (LR19ES02), Faculté Des Sciences de Tunis, Université de Tunis El Manar, Campus Universitaire Farhat HACHED, 2092 Tunis, Tunisia; Institut Préparatoire Aux Études D'Ingénieurs El Manar, Université de Tunis El Manar, B.P. 244 El Manar II, 2092 Tunis, Tunisia.
| |
Collapse
|
5
|
Molaakbari E, Aallae MR, Golestanifar F, Garakani-Nejad Z, Khosravi A, Rezapour M, Eshaghi Malekshah R, Ghomi M, Ren G. In silico assessment of hesperidin on SARS-CoV-2 main protease and RNA polymerase: Molecular docking and dynamics simulation approach. Biochem Biophys Rep 2024; 39:101804. [PMID: 39193225 PMCID: PMC11347860 DOI: 10.1016/j.bbrep.2024.101804] [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: 05/06/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
The present study uses molecular docking and dynamic simulations to evaluate the inhibitory effect of flavonoid glycosides-based compounds on coronavirus Main protease (Mpro) and RNA polymerase. The Molegro Virtual Docker (MVD) software is utilized to simulate and calculate the binding parameters of compounds with coronavirus. The docking results show that the selected herbal compounds are more effective than those of chemical compounds. It is also revealed that five herbal ligands and two chemical ligands have the best docking scores. Furthermore, a Molecular Dynamics (MD) simulation was conducted for Hesperidin, confirming docking results. Analysis based on different parameters such as Root-mean-square deviation (RMSD), Root mean square fluctuation (RMSF), Radius of gyration (Rg), Solvent accessibility surface area (SASA), and the total number of hydrogen bonds suggests that Hesperidin formed a stable complex with Mpro. Absorption, Distribution, Metabolism, Excretion, And Toxicity (ADMET) analysis was performed to compare Hesperidin and Grazoprevir as potential antiviral medicines, evaluating both herbal and chemical ligand results. According to the study, herbal compounds could be effective on coronavirus and are admissible candidates for developing potential operative anti-viral medicines. Hesperidin was found to be the most acceptable interaction. Grazoprevir is an encouraging candidate for drug development and clinical trials, with the potential to become a highly effective Mpro inhibitor. Compared to RNA polymerase, Mpro showed a greater affinity for bonding with Hesperidin. van der Waals and electrostatic energies dominated, creating a stable Hesperidin-Mpro and Hesperidin-RNA polymerase complex.
Collapse
Affiliation(s)
- Elaheh Molaakbari
- Leishmaniasis Research Center, Kerman University of Medical Science, Kerman, Iran
| | | | | | | | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Science, Kerman, Iran
| | - Mohsen Rezapour
- Department of Biostatics and Data Science, University of Texas, Health Science Center at Houston, Texas, USA
| | | | - Mahsa Ghomi
- Students Research Committee, Faculty of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Guogang Ren
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, AL10 9AB, UK
| |
Collapse
|
6
|
Shelake G, Baviskar S, Panda AK, Solankure S, Pandey K, Chauthe S, Behera SK. Exploring the rare variants associated with Type 2 Diabetes Mellitus in Indian population and its disease-drug association studies: an in-silico approach. J Biomol Struct Dyn 2024; 42:6307-6322. [PMID: 37440426 DOI: 10.1080/07391102.2023.2233634] [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: 02/06/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023]
Abstract
The diversified eating habits and religious culture of Indian population may be one of the reasons they largely contribute to the global diabetes burden. In the present investigation, an in-silico approach was carried out to explore hub genes in the Indian population with Type 2 Diabetes Mellitus (T2DM) that are scantily reported in the GWAS catalogue and probable potential anti-diabetic drugs from plants. This computational approach unwrapped LEP (leptin) as the hub gene among 170 genes analyzed with 14 non-synonymous single nucleotide polymorphisms (nsSNPs) with MAF < 0.01. The mutation of the LEP gene leads to a decrease in leptin concentration, which increases the risk of obesity and T2DM. According to the DUET webserver, 11 of 14 mutations examined were found to destabilize the LEP protein. Among 14, four barely reported LEP variants rs781301976 (I45N), rs776443424 (S52F), rs200915360 (D76Y), and rs1191666811 (D162N) were unzipped to be associated with T2DM, which may be the probable potential drug targets. The virtual screening revealed Vescalagin as having the highest binding energy among 336 natural compounds. Molecular docking of Vescalagin depicted higher binding energy (-9.0 kcal/mol) against mutated LEP [rs200915360 (D76Y)] compared to wild (-8.9 kcal/mol) and LEP-Metformin complexes. The trajectory analysis of MD simulations revealed that Vescalagin was more effective than Metformin in stabilizing the system. The present study suggests that the associations of the investigated nsSNPs in LEP [rs200915360 (D76Y)] and others can be key factors in the predominant role of T2DM morbidity in the Indian population that can be used as potential markers and drug targets for T2DM therapeutics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ganesh Shelake
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Shraddha Baviskar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Amrita Kumari Panda
- Department of Biotechnology, Sant Gahira Guru Vishwavidyalaya, Sarguja, Ambikapur, Chhattisgarh, India
| | - Sunetra Solankure
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Komal Pandey
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Siddheshwar Chauthe
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
7
|
Saha D, Kothari S, Kulkarni SD, Thambiraja M, Yennamalli RM, Das DK. Genetic heterogeneity and respiratory chain enzyme analysis in pediatric Indian patients with mitochondrial disorder: Report of novel variants in POLG1 gene and their functional implication using molecular dynamic simulation. Mitochondrion 2024; 76:101870. [PMID: 38471579 DOI: 10.1016/j.mito.2024.101870] [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: 12/05/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Mitochondrial disorders are a heterogeneous group of disorders caused by mutations in the mitochondrial DNA or in nuclear genes encoding the mitochondrial proteins and subunits. Polymerase Gamma (POLG) is a nuclear gene and mutation in the POLG gene are one of the major causes of inherited mitochondrial disorders. In this study, 15 pediatric patients, with a wide spectrum of clinical phenotypes were screened using blood samples (n = 15) and muscle samples (n = 4). Respiratory chain enzyme analysis in the muscle samples revealed multi-complex deficiencies with Complex I deficiency present in (1/4) patients, Complex II (2/4), Complex III (3/4) and Complex IV (2/4) patients. Multiple large deletions were observed in 4/15 patients using LR-PCR. Whole exome sequencing (WES) revealed a compound heterozygous mutation consisting of a POLG1 novel variant (NP_002684.1:p.Trp261X) and a missense variant (NP_002684.1:p. Leu304Arg) in one patient and another patient harboring a novel homozygous POLG1 variant (NP_002684.1:p. Phe750Val). These variants (NP_002684.1:p. Leu304Arg) and (NP_002684.1:p. Phe750Val) and their interactions with DNA were modelled using molecular docking and molecular dynamics (MD) simulation studies. The protein conformation was analyzed as root mean square deviation (RMSD), root mean square fluctuation (RMSF) which showed local fluctuations in the mutants compared to the wildtype. However, Solvent Accessible Surface Area (SASA) significantly increased for NP_002684.1:p.Leu304Arg and decreased in NP_002684.1:p.Phe750Val mutants. Further, Contact Order analysis indicated that the Aromatic-sulfur interactions were destabilizing in the mutants. Overall, these in-silico analysis has revealed a destabilizing mutations suggesting pathogenic variants in POLG1 gene.
Collapse
Affiliation(s)
- Debolina Saha
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive and Child Health, JM Street, Parel, Mumbai 400012, India
| | - Sonam Kothari
- Department of Pediatric Neurology, Bai Jerbai Wadia Hospital for Children, Acharya Donde Marg, Parel, Mumbai, Maharashtra 400012, India
| | - Shilpa Duttaprasanna Kulkarni
- Department of Pediatric Neurology, Bai Jerbai Wadia Hospital for Children, Acharya Donde Marg, Parel, Mumbai, Maharashtra 400012, India
| | - Menaka Thambiraja
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamilnadu 613401, India
| | - Ragothaman M Yennamalli
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamilnadu 613401, India.
| | - Dhanjit K Das
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive and Child Health, JM Street, Parel, Mumbai 400012, India.
| |
Collapse
|
8
|
Bhavsar D, Kutre S, Shikhare P, Kumar S, Behera SK, Chauthe SK. Pharmacoinformatics approach for type 2 diabetes mellitus therapeutics using phytocompounds from Costus genus: an in-silico investigation. J Biomol Struct Dyn 2024:1-17. [PMID: 38511497 DOI: 10.1080/07391102.2024.2330712] [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: 09/22/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Type 2 Diabetes Mellitus (T2DM), as a significant health concern globally, particularly in India, underscoring the vital need for effective therapeutics. Current drug therapies for T2DM may have limitations, leading researchers to explore natural products as alternatives. In this study. We have investigated the anti-diabetic compounds from the Costus genus, known as the insulin plant, which is abundant in southern India. The bioinformatics tools and software used for in-silico analysis to identify potential therapeutic compounds and hub genes associated with T2DM in the Indian population that could cut short the in-vitro and in-vivo experimental approaches in near future. The systematic review and combinatorial in-silico analysis revealed IGF2BP2, INS and TCF as the key targets that are associated with T2DM. The compounds stigmasterol, cycloartenol, and diosgenone were explored to be potent among all the 38 phytocompounds from genus Costus with binding energies -8.48, -10.07, and -10.31 kcal/mol against IGF2BP2, INS and TCF. The molecular dynamics (MD) simulation studies of these complexes demonstrated stable and consistent dynamic behavior, particularly in the INS-cycloartenol, IGF2BP2-stigmasterol and TCF7L2-diosgenone complexes. The identified compounds and associated targets represent potential candidates for T2DM therapeutics in the Indian population. The pharmacoinformatics approach presented in the study could streamline the drug discovery process by prioritizing compounds for further experimental validation.
Collapse
Affiliation(s)
- Drashti Bhavsar
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Suraj Kutre
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Priti Shikhare
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Sunil Kumar
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute (IASRI), New Delhi, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Siddheshwar Kisan Chauthe
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| |
Collapse
|
9
|
Vijay A, Sreyas Adury VS, Mukherjee A. Targeting RdRp of SARS-CoV-2 with De Novo Molecule Generation. ACS APPLIED BIO MATERIALS 2024; 7:609-616. [PMID: 37566736 DOI: 10.1021/acsabm.3c00339] [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: 08/13/2023]
Abstract
Viruses are known for their extremely high mutation rates, allowing them to evade both the human immune system and many forms of standard medicine. Despite this, the RNA dependent RNA polymerase (RdRp) of the RNA viruses has been largely conserved, and any significant mutation of this protein is unlikely. The recent COVID-19 pandemic presents a need for therapeutics. We have designed a de novo drug design algorithm that generates strong binding ligands from scratch, based on only the structure of the target protein's receptor. In this paper, we applied our method to target SARS-CoV-2 RdRp and generated several de novo molecules. We then chose some drug molecules based on the structural similarity to some of our strongest binding de novo molecules. Subsequently, we showed, using rigorous all-atom explicit-water free energy calculations in near-microsecond time scales using state-of-the-art well-tempered metadynamics simulations, that some of our de novo generated ligands bind more strongly to RdRp than the recent FDA approved drug remdesivir in its active form, remdesivir triphosphate (RTP). We elucidated the binding mechanism for some of the top binders and compared it with RTP. We believe that this work will be useful both by presenting lead structures for RdRp inhibition and by delivering key insights into the residues of the protein potentially involved in the binding/unbinding of these small molecule drugs, leading to more targeted studies in the future.
Collapse
Affiliation(s)
- Amal Vijay
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411008, India
| | | | - Arnab Mukherjee
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411008, India
| |
Collapse
|
10
|
Lambring CB, Fiadjoe H, Behera SK, Basha R. Docking and molecular dynamic simulations of Mithramycin-A and Tolfenamic acid against Sp1 and survivin. Process Biochem 2024; 137:207-216. [PMID: 38912413 PMCID: PMC11192519 DOI: 10.1016/j.procbio.2023.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Therapeutic targeting of Sp1 transcription factor and survivin, are studied in various cancers due to their consistent overexpression. These markers result in poorer cancer prognoses and their downregulation has been investigated as an effective treatment approach. Mithramycin-A and Tolfenamic acid are two drugs with innate anti-cancer properties and are suggested to be able to target Sp1 through GC/GT DNA binding interference, however in-depth binding and mechanistic studies are lacking. Through docking analysis, we investigated Mithramycin-A and Tolfenamic acid in terms of their specific binding interactions with Sp1 and survivin. Through further molecular dynamics simulations including Root Mean Square (RMS) Fluctuation and RMS Deviation, rGYr, and H-bond analysis, we identified critical residues involved in drug interactions with each protein in question. We show Mithramycin-A as the superior binding candidate to each protein and found that it exhibited stronger binding with Sp1, and then survivin. Subsequent molecular dynamics simulations followed the same trend as initial binding energy calculations and showed crucial amino acids involved in each Mithramycin-A-protein complex. Our findings warrant further investigation into Mithramycin-A and its specific interaction with Sp1 and their downstream targets giving a better understanding of Mithramycin-A and its potential as an effective cancer treatment.
Collapse
Affiliation(s)
| | - Hope Fiadjoe
- UNT Health Science Center at Fort Worth, Fort Worth, TX 76107, USA
| | | | - Riyaz Basha
- UNT Health Science Center at Fort Worth, Fort Worth, TX 76107, USA
| |
Collapse
|
11
|
Rohilla A, Rohilla S. Drug Repositioning: A Monetary Stratagem to Discover a New Application of Drugs. Curr Drug Discov Technol 2024; 21:e101023222023. [PMID: 38629171 DOI: 10.2174/0115701638253929230922115127] [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: 03/19/2023] [Revised: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 04/19/2024]
Abstract
Drug repurposing, also referred to as drug repositioning or drug reprofiling, is a scientific approach to the detection of any new application for an already approved or investigational drug. It is a useful policy for the invention and development of new pharmacological or therapeutic applications of different drugs. The strategy has been known to offer numerous advantages over developing a completely novel drug for certain problems. Drug repurposing has numerous methodologies that can be categorized as target-oriented, drug-oriented, and problem-oriented. The choice of the methodology of drug repurposing relies on the accessible information about the drug molecule and like pharmacokinetic, pharmacological, physicochemical, and toxicological profile of the drug. In addition, molecular docking studies and other computer-aided methods have been known to show application in drug repurposing. The variation in dosage for original target diseases and novel diseases presents a challenge for researchers of drug repurposing in present times. The present review critically discusses the drugs repurposed for cancer, covid-19, Alzheimer's, and other diseases, strategies, and challenges of drug repurposing. Moreover, regulatory perspectives related to different countries like the United States (US), Europe, and India have been delineated in the present review.
Collapse
Affiliation(s)
- Ankur Rohilla
- Department of Pharmacology, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, 140413, Mohali, India
| | - Seema Rohilla
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, Haryana, India
| |
Collapse
|
12
|
Behera SK, Lambring CB, Hashmi A, Gottipolu S, Basha R. In Silico Analysis Determining the Binding Interactions of NAD(P)H: Quinone Oxidoreductase 1 and Resveratrol via Docking and Molecular Dynamic Simulations. EUROPEAN JOURNAL OF BIOLOGY 2023; 82:280-288. [PMID: 38264080 PMCID: PMC10805530 DOI: 10.26650/eurjbiol.2023.1352396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Objective NAD(P)H: Quinone oxidoreductase1 (NQO1) plays a crucial role in cellular defense against oxidative stress. Overexpression of NQO1 is linked to various cancer pathways. Despite its potential, the actual mechanisms to inhibit NQO1 and increase the efficacy of standard therapeutic options are not yet established. Resveratrol is an anti-cancer polyphenol found in dietary products and red wine. The objective of this investigation is to employ in silico methods to explore how resveratrol interacts with NQO1. Materials and Methods Docking analysis of resveratrol against NQO1 was performed using Glide. The most efficiently docked complex was characterized and analyzed by measuring intermolecular (IM) hydrogen (H)-bonds and binding energy values, additional hydrophobic, and electrostatic interactions. IM interaction between complexed protein and compound was demonstrated using LigPlot+ and the Schrödinger ligand interaction module. Molecular dynamics tools were employed to examine the physical movement of molecules to evaluate how macromolecular structures relate to their functions. Results The results of this investigation depicted a strong affinity of resveratrol against NQO1 followed by MD simulations (NQO1-resveratrol complex-binding energy: -2.847kcal/mol). Resveratrol's robust binding affinity through docking and molecular dynamic simulations highlights a significant change around 90 ns. The H-bonds number was inversely linked with the resveratrol-NQO1 complex stability. The NQO1-Resveratrol complex displayed dynamic motion, as revealed by porcupine projections, indicating alterations in its movement and flexibility. Conclusion The present in silico analysis suggests a possible alteration in resveratrol's orientation in the protein binding pocket. The findings encourage further investigation, including validation using in vitro and in vivo assays.
Collapse
Affiliation(s)
| | | | | | | | - Riyaz Basha
- University of North Texas Health Science Center at Fort Worth, Texas, USA
| |
Collapse
|
13
|
Pamarthy D, Behera SK, Swain S, Yadav S, Suresh S, Jain N, Bhadra MP. Diaryl ether derivative inhibits GPX4 expression levels to induce ferroptosis in thyroid cancer cells. Drug Dev Res 2023; 84:861-887. [PMID: 37070554 DOI: 10.1002/ddr.22059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 04/19/2023]
Abstract
Papillary thyroid carcinoma contributes to about 80% of the total thyroid cancer cases. BRAFV600E is a frequently occurring mutation in PTCs. Although several BRAF inhibitors are available, many thyroid cancer patients acquire resistance to BRAF inhibitors. Therefore, new targets and drugs need to be identified as therapies. Ferroptosis is a recently discovered type of cell death, and inhibiting glutathione peroxidase 4 (GPX4) using small molecules was found to trigger ferroptosis. But it is unknown whether inhibiting GPX4 renders thyroid cancer cells susceptible to ferroptosis. To identify novel GPX4 inhibitors, we focused on our previously reported cohort of diaryl ether and dibenzoxepine molecules. In this study, we asked whether diaryl ether and dibenzoxepine derivatives trigger ferroptosis in thyroid cancer cells. To answer this question, we screened diaryl ether and dibenzoxepine derivatives in cell-based assays and performed mechanism of action studies. We found that a diaryl ether derivative, 16 decreased thyroid cell proliferation and triggered ferroptosis by inhibiting GPX4 expression levels. Molecular modeling and dynamics simulations showed that 16 binds to the active site of GPX4. Upon deciphering the mode of 16-induced ferroptosis, we found that 16 treatments decrease mitochondrial polarization and reduce mitochondrial respiration similar to a ferroptosis inducer, RSL3. We conclude that the diaryl ether derivative, 16 inhibits GPX4 expression levels to induce ferroptosis in thyroid cancer cells. Based on our observations, we suggest that 16 can be lead-optimized and developed as a ferroptosis-inducing agent to treat thyroid cancers.
Collapse
Affiliation(s)
- Deepika Pamarthy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, (NIPER), Ahmedabad, Gujarat, India
| | - Sonam Swain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanjay Yadav
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
| | - Surisetti Suresh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
| | - Nishant Jain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manika Pal Bhadra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
14
|
Singh R, Kumar P, Sindhu J, Devi M, Kumar A, Lal S, Singh D, Kumar H. Thiazolidinedione-triazole conjugates: design, synthesis and probing of the α-amylase inhibitory potential. Future Med Chem 2023; 15:1273-1294. [PMID: 37551699 DOI: 10.4155/fmc-2023-0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Aim: The primary objective of this investigation was the synthesis, spectral interpretation and evaluation of the α-amylase inhibition of rationally designed thiazolidinedione-triazole conjugates (7a-7aa). Materials & methods: The designed compounds were synthesized by stirring a mixture of thiazolidine-2,4-dione, propargyl bromide, cinnamaldehyde and azide derivatives in polyethylene glycol-400. The α-amylase inhibitory activity of the synthesized conjugates was examined by integrating in vitro and in silico studies. Results: The investigated derivatives exhibited promising α-amylase inhibitory activity, with IC50 values ranging between 0.028 and 0.088 μmol ml-1. Various computational approaches were employed to get detailed information about the inhibition mechanism. Conclusion: The thiazolidinedione-triazole conjugate 7p, with IC50 = 0.028 μmol ml-1, was identified as the best hit for inhibiting α-amylase.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar, 125004, India
| | - Meena Devi
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Ashwani Kumar
- Department of Pharmaceutical Sciences, GJUS&T, Hisar, 125001, India
| | - Sohan Lal
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak, 124001, India
| | - Harish Kumar
- Department of Chemistry, School of Basic Sciences, Central University Haryana, Mahendergarh, 123029, India
| |
Collapse
|
15
|
Patel U, Desai K, Dabhi RC, Maru JJ, Shrivastav PS. Bioprospecting phytochemicals of Rosmarinus officinalis L. for targeting SARS-CoV-2 main protease (M pro): a computational study. J Mol Model 2023; 29:161. [PMID: 37115321 PMCID: PMC10141822 DOI: 10.1007/s00894-023-05569-6] [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: 09/24/2022] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
CONTEXT The persistent spread of highly contagious COVID-19 disease is one of the deadliest occurrences in the history of mankind. Despite the distribution of numerous efficacious vaccines and their extensive usage, the perpetual effectiveness of immunization is being catechized. Therefore, discovering an alternative therapy to control and prevent COVID-19 infections has become a top priority. The main protease (Mpro) plays a key role in viral replication, making it an intriguing pharmacological target for SARS-CoV-2. METHODS In this context, virtual screening of thirteen bioactive polyphenols and terpenoids of Rosmarinus officinalis L. was performed using several computational modules including molecular docking, ADMET, drug-likeness characteristics, and molecular dynamic simulation to predict the potential inhibitors against SARS-CoV-2 Mpro (PDB: 6LU7). The results suggest that apigenin, betulinic acid, luteolin, carnosol, and rosmarinic acid may emerge as potential inhibitors of SARS-CoV-2 with acceptable drug-likeness, pharmacokinetics, ADMET characteristics, and binding interactions comparable with remdesivir and favipiravir. These findings imply that some of the active components of Rosmarinus officinalis L. can serve as an effective antiviral source for the development of therapeutics for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Unnati Patel
- Department of Chemistry, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Krishna Desai
- Department of Botany, Bioinformatics and Climate Change Impacts Management, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Ranjitsinh C Dabhi
- Department of Chemistry, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Jayesh J Maru
- Department of Chemistry, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Pranav S Shrivastav
- Department of Chemistry, University School of Sciences, Gujarat University, Ahmedabad, 380009, India.
| |
Collapse
|
16
|
Pradhan S, Behera SK, Samal SK, Panda I, Sahu PK, Priyadarshini S. Interaction Between 2‐Methylimidazole and 1‐Butanol/1‐Octanol: Thermophysical and Computational Studies. ChemistrySelect 2023. [DOI: 10.1002/slct.202204931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Sanghamitra Pradhan
- Department of Chemistry Institute of Technical Education and Research (FET) Siksha ‘O' Anusandhan Deemed to be University Khandagiri square Bhubaneswar 751030 Odisha India
| | - Santosh Kumar Behera
- Department of Biotechnology National Institute of Pharmaceutical Education and Research Ahmedabad Gujarat 382355 India
| | - Sangram Keshari Samal
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies ICMR-Regional Medical Research Center Bhubaneswar 751 023 Odisha India
| | - Itishree Panda
- Department of Chemistry Institute of Technical Education and Research (FET) Siksha ‘O' Anusandhan Deemed to be University Khandagiri square Bhubaneswar 751030 Odisha India
| | - Pratyush Kumar Sahu
- Department of Chemistry Institute of Technical Education and Research (FET) Siksha ‘O' Anusandhan Deemed to be University Khandagiri square Bhubaneswar 751030 Odisha India
| | - Swayamprabha Priyadarshini
- Department of Chemistry Institute of Technical Education and Research (FET) Siksha ‘O' Anusandhan Deemed to be University Khandagiri square Bhubaneswar 751030 Odisha India
| |
Collapse
|
17
|
Varshney N, Kashyap D, Behra SK, Saini V, Chaurasia A, Kumar S, Jha HC. Predictive profiling of gram-negative antibiotics in CagA oncoprotein inactivation: a molecular dynamics simulation approach. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:501-521. [PMID: 37462112 DOI: 10.1080/1062936x.2023.2230876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/24/2023] [Indexed: 07/20/2023]
Abstract
Gastric cancer (GC) is the fifth most prevalent form of cancer worldwide. CagA - positive Helicobacter pylori infects more than 60% of the human population. Moreover, chronic infection of CagA-positive H. pylori can directly affect GC incidence. In the current study, we have repurposed FDA-approved antibiotics that are viable alternatives to current regimens and can potentially be used as combination therapy against the CagA of H. pylori. The 100 FDA-approved gram negative antibiotics were screened against CagA protein using the AutoDock 4.2 tool. Further, top nine compounds were selected based on higher binding affinity with CagA. The trajectory analysis of MD simulations reflected that binding of these drugs with CagA stabilizes the system. Nonetheless, atomic density map and principal component analysis also support the notion of stable binding of antibiotics to the protein. The residues ASP96, GLN100, PRO184, and THR185 of compound cefpiramide, doxycycline, delafloxacin, metacycline, oxytetracycline, and ertapenem were involved in the binding with CagA protein. These residues are crucial for the CagA that aids in entry or pathogenesis of the bacterium. The screened FDA-approved antibiotics have a potential druggability to inhibit CagA and reduce the progression of H. pylori borne diseases.
Collapse
Affiliation(s)
- N Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - D Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - S K Behra
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, India
| | - V Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - A Chaurasia
- Division of Crop Protection, ICAR -Indian Institute of Vegetable Research, Varanasi, India
| | - S Kumar
- Division of Agricultural Bioinformatics (CABIN), ICAR-Indian Agricultural Statistics Research Institute (IASRI), Delhi, India
| | - H C Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
18
|
Singh R, Kumar P, Sindhu J, Devi M, Kumar A, Lal S, Singh D. Parsing structural fragments of thiazolidin-4-one based α-amylase inhibitors: A combined approach employing in vitro colorimetric screening and GA-MLR based QSAR modelling supported by molecular docking, molecular dynamics simulation and ADMET studies. Comput Biol Med 2023; 157:106776. [PMID: 36947906 DOI: 10.1016/j.compbiomed.2023.106776] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/20/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
α-Amylase (EC.3.2.1.1) is a ubiquitous digestive endoamylase. The abrupt rise in blood glucose levels due to the hydrolysis of carbohydrates by α-amylase at a faster rate is one of the main reasons for type 2 diabetes. The inhibitors prevent the action of digestive enzymes, slowing the digestion of carbs and eventually assisting in the management of postprandial hyperglycemia. In the course of developing α-amylase inhibitors, we have screened 2-aryliminothiazolidin-4-one based analogs for their in vitro α-amylase inhibitory potential and employed various in silico approaches for the detailed exploration of the bioactivity. The DNSA bioassay revealed that compounds 5c, 5e, 5h, 5j, 5m, 5o and 5t were more potent than the reference drug (IC60 value = 22.94 ± 0.24 μg mL-1). The derivative 5o with -NO2 group at both the rings was the most potent analog with an IC60 value of 19.67 ± 0.20 μg mL-1 whereas derivative 5a with unsubstituted aromatic rings showed poor inhibitory potential with an IC60 value of 33.40 ± 0.15 μg mL-1. The reliable QSAR models were developed using the QSARINS software. The high value of R2ext = 0.9632 for model IM-9 showed that the built model can be applied to predict the α-amylase inhibitory activity of the untested molecules. A consensus modelling approach was also employed to test the reliability and robustness of the developed QSAR models. Molecular docking and molecular dynamics were employed to validate the bioassay results by studying the conformational changes and interaction mechanisms. A step further, these compounds also exhibited good ADMET characteristics and bioavailability when tested for in silico pharmacokinetics prediction parameters.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India.
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar, 125004, India
| | - Meena Devi
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Ashwani Kumar
- Department of Pharmaceutical Sciences, GJUS&T, Hisar, 125001, India
| | - Sohan Lal
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak, 124001, India
| |
Collapse
|
19
|
Dhanalakshmi M, Das K, Pandya M, Shah S, Gadnayak A, Dave S, Das J. Artificial Neural Network-Based Study Predicts GS-441524 as a Potential Inhibitor of SARS-CoV-2 Activator Protein Furin: a Polypharmacology Approach. Appl Biochem Biotechnol 2022; 194:4511-4529. [PMID: 35507249 PMCID: PMC9066385 DOI: 10.1007/s12010-022-03928-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Furin, a pro-protein convertase, plays a significant role as a biological scissor in bacterial, viral, and even mammalian substrates which in turn decides the fate of many viral and bacterial infections along with the numerous ailments caused by cancer, diabetes, inflammations, and neurological disorders. In the wake of the current pandemic caused by the virus SARS-CoV-2, furin has become the center of attraction for researchers as the spike protein contains a polybasic furin cleavage site. In the present work, we have searched for novel inhibitors against this interesting human target from FDA-approved antiviral. To enhance the selection of new inhibitors, we employed Kohonen's artificial neural network-based self-organizing maps for ligand-based virtual screening. Promising results were obtained which can help in drug repurposing and network pharmacology studies can address the errors generated due to promiscuity/polypharmacology. We found 15 existing FDA antiviral drugs having the potential to inhibit furin. Among these, six compounds have targets on important human proteins (LDLR, FCGR1A, PCK1, TLR7, DNA, and PNP). The role of these 15 drugs inhibiting furin can be established by studying further on patients infected with number of viruses including SARS-CoV-2. Here we propose two promising candidate FDA drugs GS-441524 and Grazoprevir (MK-5172) for repurposing as inhibitors of furin. The best results were observed with GS-441524.
Collapse
Affiliation(s)
- M Dhanalakshmi
- Research and Development Centre, Bharathiar University, Marudhamalai Rd, Coimbatore, Tamil Nadu, India
| | - Kajari Das
- Department of Biotechnology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Medha Pandya
- The KPES Science College, Maharaja krishnakumarsinhji Bhavnagar University, Bhavnagar, Gujarat, India
| | - Sejal Shah
- Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - Ayushman Gadnayak
- Centre for Genomics & Biomedical Informatics, IMS and SUM Hospital, Siksha "O" Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sushma Dave
- Department of Applied Sciences, JIET, Jodhpur, Rajasthan, India.
| | | |
Collapse
|
20
|
Contractor D, Globisch C, Swaroop S, Jain A. Structural basis of Omicron immune evasion: A comparative computational study. Comput Biol Med 2022; 147:105758. [PMID: 35763933 PMCID: PMC9212419 DOI: 10.1016/j.compbiomed.2022.105758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/10/2022] [Accepted: 06/18/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The vaccines used against SARS-CoV-2 by now have been able to develop some neutralising antibodies in the vaccinated population and their effectiveness has been challenged by the emergence of the new strains with numerous mutations in the spike protein of SARS-CoV-2. Since S protein is the major immunogenic protein of the virus which contains Receptor Binding Domain (RBD) that interacts with the human Angiotensin-Converting Enzyme 2 (ACE2) receptors, any mutations in this region should affect the neutralisation potential of the antibodies leading to the immune evasion. Several variants of concern of the virus have emerged so far, amongst which the most critical are Delta and recently reported Omicron. In this study, we have mapped and reported mutations on the modelled RBD and evaluated binding affinities of various human antibodies with it. METHOD Docking and molecular dynamics simulation studies have been used to explore the effect of mutations on the structure of RBD and RBD-antibody interaction. RESULTS These analyses show that the mutations mostly at the interface of a nearby region lower the binding affinity of the antibody by ten to forty percent, with a downfall in the number of interactions formed as a whole. It implies the generation of immune escape variants. CONCLUSIONS Notable mutations and their effect was characterised that explain the structural basis of antibody efficacy in Delta and a compromised neutralisation effect for the Omicron variant. Thus, our results pave the way for robust vaccine design that can be effective for many variants.
Collapse
Affiliation(s)
- Darshan Contractor
- Department of Bioengineering and Biotechnology, Birla Institute of Technology (BIT), Mesra, Ranchi, 835215, Jharkhand, India; Department of Biotechnology, Sun Pharmaceutical Industries Ltd., Tandalja, Vadodara, 390012, Gujarat, India
| | | | - Shiv Swaroop
- Department of Biochemistry, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, 305817, Rajasthan, India.
| | - Alok Jain
- Department of Bioengineering and Biotechnology, Birla Institute of Technology (BIT), Mesra, Ranchi, 835215, Jharkhand, India.
| |
Collapse
|
21
|
Tambe V, Patel S, Shard A, Behera SK, Polaka S, Anup N, Gadeval A, Kalia K, Tekade RK. Dendronized Polymeric Biomaterial for Loading, Stabilization, and Targeted Cytosolic Delivery of microRNA in Cancer Cells. ACS APPLIED BIO MATERIALS 2022. [DOI: 10.1021/acsabm.2c00179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vishakha Tambe
- Department of Pharmaceutics, Department of Pharmaceuticals, Ministry of Chem-icals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Sagarkumar Patel
- Department of Medicinal Chemistry, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Amit Shard
- Department of Medicinal Chemistry, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Santosh Kumar Behera
- Central Instrumentation Facility, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Suryanarayana Polaka
- Department of Pharmaceutics, Department of Pharmaceuticals, Ministry of Chem-icals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Neelima Anup
- Department of Pharmaceutics, Department of Pharmaceuticals, Ministry of Chem-icals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Anuradha Gadeval
- Department of Pharmaceutics, Department of Pharmaceuticals, Ministry of Chem-icals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Kiran Kalia
- Department of Biotechnology, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- Department of Pharmaceutics, Department of Pharmaceuticals, Ministry of Chem-icals and Fertilizers, National Institute of Pharmaceutical Education and Research (NIPER)─Ahmedabad, An Institute of National Importance, Government of India, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
22
|
Debnath SK, Debnath M, Srivastava R, Omri A. Drugs repurposing for SARS-CoV-2: new insight of COVID-19 druggability. Expert Rev Anti Infect Ther 2022; 20:1187-1204. [PMID: 35615888 DOI: 10.1080/14787210.2022.2082944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The ongoing epidemic of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) creates a massive panic worldwide due to the absence of effective medicines. Developing a new drug or vaccine is time-consuming to pass safety and efficacy testing. Therefore, repurposing drugs have been introduced to treat COVID-19 until effective drugs are developed. AREA COVERED A detailed search of repurposing drugs against SARS-CoV-2 was carried out using the PubMed database, focusing on articles published 2020 years onward. A different class of drugs has been described in this article to target hosts and viruses. Based on the previous pandemic experience of SARS-CoV and MERS, several antiviral and antimalarial drugs are discussed here. This review covers the failure of some repurposed drugs that showed promising activity in the earlier CoV-pandemic but were found ineffective against SARS-CoV-2. All these discussions demand a successful drug development strategy for screening and identifying an effective drug for better management of COVID-19. The drug development strategies described here will serve a new scope of research for academicians and researchers. EXPERT OPINION Repurposed drugs have been used since COVID-19 to eradicate disease propagation. Drugs found effective for MERS and SARS may not be effective against SARS-CoV-2. Drug libraries and artificial intelligence are helpful tools to screen and identify different molecules targeting viruses or hosts.
Collapse
Affiliation(s)
- Sujit Kumar Debnath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Monalisha Debnath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug & Vaccine Delivery Systems Facility, Laurentian University, Sudbury, Canada
| |
Collapse
|
23
|
Pramanik D, Pawar AB, Roy S, Singh JK. Mechanistic insights of key host proteins and potential repurposed inhibitors regulating SARS-CoV-2 pathway. J Comput Chem 2022; 43:1237-1250. [PMID: 35535951 PMCID: PMC9348233 DOI: 10.1002/jcc.26888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/03/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022]
Abstract
The emergence of pandemic situations originated from severe acute respiratory syndrome (SARS)‐CoV‐2 and its new variants created worldwide medical emergencies. Due to the non‐availability of efficient drugs and vaccines at these emergency hours, repurposing existing drugs can effectively treat patients critically infected by SARS‐CoV‐2. Finding a suitable repurposing drug with inhibitory efficacy to a host‐protein is challenging. A detailed mechanistic understanding of the kinetics, (dis)association pathways, key protein residues facilitating the entry–exit of the drugs with targets are fundamental in selecting these repurposed drugs. Keeping this target as the goal of the paper, the potential repurposing drugs, Nafamostat, Camostat, Silmitasertib, Valproic acid, and Zotatifin with host‐proteins HDAC2, CSK22, eIF4E2 are studied to elucidate energetics, kinetics, and dissociation pathways. From an ensemble of independent simulations, we observed the presence of single or multiple dissociation pathways with varying host‐proteins‐drug systems and quantitatively estimated the probability of unbinding through these specific pathways. We also explored the crucial gateway residues facilitating these dissociation mechanisms. Interestingly, the residues we obtained for HDAC2 and CSK22 are also involved in the catalytic activity. Our results demonstrate how these potential drugs interact with the host machinery and the specific target residues, showing involvement in the mechanism. Most of these drugs are in the preclinical phase, and some are already being used to treat severe COVID‐19 patients. Hence, the mechanistic insight presented in this study is envisaged to support further findings of clinical studies and eventually develop efficient inhibitors to treat SARS‐CoV‐2.
Collapse
Affiliation(s)
- Debabrata Pramanik
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, India
| | | | - Sudip Roy
- Prescience Insilico Private Limited, Bangalore, India
| | - Jayant Kumar Singh
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, India.,Prescience Insilico Private Limited, Bangalore, India
| |
Collapse
|
24
|
Probing the Immune System Dynamics of the COVID-19 Disease for Vaccine Designing and Drug Repurposing Using Bioinformatics Tools. IMMUNO 2022. [DOI: 10.3390/immuno2020022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of COVID-19 is complicated by immune dysfunction. The impact of immune-based therapy in COVID-19 patients has been well documented, with some notable studies on the use of anti-cytokine medicines. However, the complexity of disease phenotypes, patient heterogeneity and the varying quality of evidence from immunotherapy studies provide problems in clinical decision-making. This review seeks to aid therapeutic decision-making by giving an overview of the immunological responses against COVID-19 disease that may contribute to the severity of the disease. We have extensively discussed theranostic methods for COVID-19 detection. With advancements in technology, bioinformatics has taken studies to a higher level. The paper also discusses the application of bioinformatics and machine learning tools for the diagnosis, vaccine design and drug repurposing against SARS-CoV-2.
Collapse
|
25
|
Nguyen QV, Chong LC, Hor YY, Lew LC, Rather IA, Choi SB. Role of Probiotics in the Management of COVID-19: A Computational Perspective. Nutrients 2022; 14:274. [PMID: 35057455 PMCID: PMC8781206 DOI: 10.3390/nu14020274] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/01/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) was declared a pandemic at the beginning of 2020, causing millions of deaths worldwide. Millions of vaccine doses have been administered worldwide; however, outbreaks continue. Probiotics are known to restore a stable gut microbiota by regulating innate and adaptive immunity within the gut, demonstrating the possibility that they may be used to combat COVID-19 because of several pieces of evidence suggesting that COVID-19 has an adverse impact on gut microbiota dysbiosis. Thus, probiotics and their metabolites with known antiviral properties may be used as an adjunctive treatment to combat COVID-19. Several clinical trials have revealed the efficacy of probiotics and their metabolites in treating patients with SARS-CoV-2. However, its molecular mechanism has not been unraveled. The availability of abundant data resources and computational methods has significantly changed research finding molecular insights between probiotics and COVID-19. This review highlights computational approaches involving microbiome-based approaches and ensemble-driven docking approaches, as well as a case study proving the effects of probiotic metabolites on SARS-CoV-2.
Collapse
Affiliation(s)
- Quang Vo Nguyen
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Wilayah Persekutuan, Kuala Lumpur 50490, Malaysia;
| | - Li Chuin Chong
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul 34820, Turkey;
| | - Yan-Yan Hor
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Gyeongbuk, Korea;
| | - Lee-Ching Lew
- Probionic Corporation, Jeonbuk Institute for Food-Bioindustry, Jeonju 54810, Korea;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Sy-Bing Choi
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Wilayah Persekutuan, Kuala Lumpur 50490, Malaysia;
| |
Collapse
|
26
|
Verma R, Pradhan D, Nayek A, Singh H, Jain AK, Khan LA. Target-based drug repurposing against Candida albicans-A computational modeling, docking, and molecular dynamic simulations study. J Cell Biochem 2021; 123:289-305. [PMID: 34672012 DOI: 10.1002/jcb.30163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/21/2022]
Abstract
The emergence of multidrug-resistant strains of Candida albicans has become a global threat mostly due to co-infection with immune-compromised patients leading to invasive candidiasis. The life-threatening form of the disease can be managed quickly and effectively by drug repurposing. Thus, the study used in silico approaches to evaluate Food and Drug Administration (FDA) approved drugs against three drug targets-TRR1, TOM40, and YHB1. The tertiary structures of three drug targets were modeled, refined, and evaluated for their structural integrity based on PROCHECK, ERRAT, and PROSA. High-throughput virtual screening of FDA-approved drugs (8815), interaction analysis, and energy profiles had revealed that DB01102 (Arbutamine), DB01611 (Hydroxychloroquine), and DB09319 (Carindacillin) exhibited better binding affinity with TRR1, TOM40, and YHB1, respectively. Notably, the molecular dynamic simulation explored that Gln45, Thr119, and Asp288 of TRR1; Thr107 and Ser121 of TOM40; Arg193, Glu213, and Ser228 of YHB1 are crucial residues for stable drug-target interaction. Additionally, it also prioritized Arbutamine-TRR1 as the best drug-target complex based on MM-PBSA (-52.72 kcal/mol), RMSD (2.43 Å), and radius of gyration (-21.49 Å) analysis. In-depth, PCA results supported the findings of molecular dynamic simulations. Interestingly, the conserved region (>70%) among the TRR1 sequences from pathogenic Candida species indicated the effectiveness of Arbutamine against multiple species of Candida as well. Thus, the study dispenses new insight and enriches the understanding of developing an advanced technique to consider potential antifungals against C. albicans. Nonetheless, a detailed experimental validation is needed to investigate the efficacy of Arbutamin against life-threatening candidiasis.
Collapse
Affiliation(s)
- Rashi Verma
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India.,Biomedical Informatics Centre, Indian Council of Medical Research, New Delhi, India
| | - Dibyabhaba Pradhan
- Computational Genomics Centre, All India Institute of Medical Sciences, Indian Council of Medical Research, New Delhi, India
| | - Arnab Nayek
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Harpreet Singh
- Computational Genomics Centre, All India Institute of Medical Sciences, Indian Council of Medical Research, New Delhi, India
| | - Arun Kumar Jain
- Biomedical Informatics Centre, Indian Council of Medical Research, New Delhi, India
| | | |
Collapse
|
27
|
Dhote AM, Patil VR, Lokwani DK, Amnerkar ND, Ugale VG, Charbe NB, Bhongade BA, Khadse SC. Strategic analyses to identify key structural features of antiviral/antimalarial compounds for their binding interactions with 3CLpro, PLpro and RdRp of SARS-CoV-2: in silico molecular docking and dynamic simulation studies. J Biomol Struct Dyn 2021; 40:11914-11931. [PMID: 34431452 DOI: 10.1080/07391102.2021.1965912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/05/2021] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV-2), a novel member of the betacoronavirus family is a single-stranded RNA virus that has spread worldwide prompting the World Health Organization to declare a global pandemic. This creates an alarming situation and generates an urgent need to develop innovative therapeutic agents. In this context, an in silico molecular docking and molecular dynamics (MD) simulation study on the existing 58 antiviral and antimalarial compounds was performed on 3CLpro, PLpro and RdRp SARS-CoV-2 proteins. The antiviral compounds are best fitted in the binding pockets and interact more profoundly with the amino acid residues compared to antimalarial compounds. An HIV protease inhibitor, saquinavir showed a good dock score and binding free energy with varied binding interactions against 3CLpro and PLpro. While, adefovir, a nucleotide HBV DNA polymerase inhibitor exhibited good dock score and binding interactions against RdRp. Although, the antimalarial compounds showed relatively less dock score but were found to be crucial in displaying essential binding interactions with these proteins. The MD simulation runs for 100 ns on 3CLpro-saquinavir, PLpro-saquinavir and RdRp-adefovir complexes using Desmond revealed fairly stable nature of interactions. This study helped in understanding the key interactions of the vital functionalities that provide a concrete base to develop lead molecules effective against SARS-CoV-2.
Collapse
Affiliation(s)
- Ashish M Dhote
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Vikas R Patil
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Deepak K Lokwani
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Nikhil D Amnerkar
- Department of Pharmaceutical Chemistry, Adv. V. R. Manohar Institute of Diploma in Pharmacy, Nagpur, India
| | - Vinod G Ugale
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| | - Nitin B Charbe
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX, USA
| | - Bhoomendra A Bhongade
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah, UAE
| | - Saurabh C Khadse
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Maharashtra, India
| |
Collapse
|
28
|
Goswami D. Comparative assessment of RNA-dependent RNA polymerase (RdRp) inhibitors under clinical trials to control SARS-CoV2 using rigorous computational workflow. RSC Adv 2021; 11:29015-29028. [PMID: 35478553 PMCID: PMC9038185 DOI: 10.1039/d1ra04460e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
The devastating effect of SARS-CoV2 continues and the scientific community is pursuing to find the strategy to combat the spread of the virus. The approach is adapted to target this virus with medicine in combination with existing vaccines. For this, the medications that can specifically inhibit an enzyme essential for viral replication 'RNA-dependant-RNA polymerase (RdRp)' of SARS-CoV2 are being developed. RdRp is the enzyme commonly found in all RNA viruses but is absent in humans. There are in total 60 different RdRp inhibitors already under clinical trials for combating other RNA viruses, which are sought to even work for SARS-CoV2. These inhibitors are classified as nucleoside/nucleotide analogues and nonnucleoside/nonnucleotide analogues. In this study, all the known RdRp inhibitors were computationally targeted in the native form and their active form making the use of molecular docking, MM-GBSA and molecular dynamics (MD) simulations to find the top two of each nucleoside/nucleotide analogues and nonnucleoside/nonnucleotide analogues. The results showed ribavirin 5'-triphosphate and favipiravir ribonucleoside triphosphate (favipiravir-RTP) to be the top two nucleotide analogues while pimodivir and dihydropyrazolopyridinone analogue 8d were the top two nonnucleosides/non-nucleotide analogues.
Collapse
Affiliation(s)
- Dweipayan Goswami
- Department of Microbiology & Biotechnology, University School of Sciences, Gujarat University Ahmedabad 380009 Gujarat India
| |
Collapse
|