1
|
Peluso P, Chankvetadze B. Recent developments in molecular modeling tools and applications related to pharmaceutical and biomedical research. J Pharm Biomed Anal 2024; 238:115836. [PMID: 37939549 DOI: 10.1016/j.jpba.2023.115836] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/21/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
In modern pharmaceutical and biomedical research, molecular modeling represents a useful tool to explore processes and their mechanistic bases at the molecular level. Integrating experimental and virtual analysis is a fruitful approach to study ligand-receptor interaction in chemical, biochemical and biological environments. In these fields, molecular docking and molecular dynamics are considered privileged techniques for modeling (bio)macromolecules and related complexes. This review aims to present the current landscape of molecular modeling in pharmaceutical and biomedical research by examining selected representative applications published in the last years and highlighting current topics and trends of this field. Thus, a systematic compilation of all published literature has not been attempted herein. After a brief overview of the main theoretical and computational tools used to investigate mechanisms at molecular level, recent applications of molecular modeling in drug discovery, ligand binding and for studying protein conformation and function will be discussed. Furthermore, specific sections will be devoted to the application of molecular modeling for unravelling enantioselective mechanisms underlying the enantioseparation of chiral compounds of pharmaceutical and biomedical interest as well as for studying new forms of noncovalent interactivity identified in biochemical and biological environments. The general aim of this review is to provide the reader with a modern overview of the topic, highlighting advancements and outlooks as well as drawbacks and pitfalls still affecting the applicability of theoretical and computational methods in the field of pharmaceutical and biomedical research.
Collapse
Affiliation(s)
- Paola Peluso
- Istituto di Chimica Biomolecolare ICB-CNR, Sede secondaria di Sassari, Traversa La Crucca 3, Regione Baldinca, Li Punti, 07100 Sassari, Italy.
| | - Bezhan Chankvetadze
- Institute of Physical and Analytical Chemistry, School of Exact and Natural Sciences, Tbilisi State University, Chavchavadze Ave 3, 0179 Tbilisi, Georgia
| |
Collapse
|
2
|
Yevsieieva LV, Lohachova KO, Kyrychenko A, Kovalenko SM, Ivanov VV, Kalugin ON. Main and papain-like proteases as prospective targets for pharmacological treatment of coronavirus SARS-CoV-2. RSC Adv 2023; 13:35500-35524. [PMID: 38077980 PMCID: PMC10698513 DOI: 10.1039/d3ra06479d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/23/2023] [Indexed: 10/16/2024] Open
Abstract
The pandemic caused by the coronavirus SARS-CoV-2 led to a global crisis in the world healthcare system. Despite some progress in the creation of antiviral vaccines and mass vaccination of the population, the number of patients continues to grow because of the spread of new SARS-CoV-2 mutations. There is an urgent need for direct-acting drugs capable of suppressing or stopping the main mechanisms of reproduction of the coronavirus SARS-CoV-2. Several studies have shown that the successful replication of the virus in the cell requires proteolytic cleavage of the protein structures of the virus. Two proteases are crucial in replicating SARS-CoV-2 and other coronaviruses: the main protease (Mpro) and the papain-like protease (PLpro). In this review, we summarize the essential viral proteins of SARS-CoV-2 required for its viral life cycle as targets for chemotherapy of coronavirus infection and provide a critical summary of the development of drugs against COVID-19 from the drug repurposing strategy up to the molecular design of novel covalent and non-covalent agents capable of inhibiting virus replication. We overview the main antiviral strategy and the choice of SARS-CoV-2 Mpro and PLpro proteases as promising targets for pharmacological impact on the coronavirus life cycle.
Collapse
Affiliation(s)
- Larysa V Yevsieieva
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| | - Kateryna O Lohachova
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| | - Alexander Kyrychenko
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| | - Sergiy M Kovalenko
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| | - Volodymyr V Ivanov
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| | - Oleg N Kalugin
- School of Chemistry, V. N. Karazin Kharkiv National University 4 Svobody sq. Kharkiv 61022 Ukraine
| |
Collapse
|
3
|
Manikanttha M, Deepti K, Tej MB, Tej MB, Gopi Reddy A, Kapavarapu R, Barange DK, V Basaveswara Rao M, Pal M. Ultrasound assisted Cu-catalyzed Ullmann-Goldberg type coupling-cyclization in a single pot: Synthesis and in silico evaluation of 11 H-pyrido[2,1- b]quinazolin-11-ones against SARS-CoV-2 RdRp. J Mol Struct 2023; 1280:135044. [PMID: 36743447 PMCID: PMC9884102 DOI: 10.1016/j.molstruc.2023.135044] [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: 12/07/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
The in silico evaluation of 11H-pyrido[2,1-b]quinazolin-11-one derivatives against SARS-CoV-2 RdRp was undertaken based on the reports on antiviral activities of this class of compounds in addition to the promising interactions of the antiviral drug penciclovir as well as quinazoline derivatives with SARS-CoV-2 RdRp in silico. The target compounds were prepared via an Ullmann-Goldberg type coupling followed by the subsequent cyclization (involving amidation) in a single pot. The methodology involved a CuI-catalyzed reaction of 2-iodobenzoate ester with 2-aminopyridine or quinolin-2-amine or thiazol-2-amine under ultrasound to give the expected products in acceptable (51-93%) yields. The molecular interactions of the synthesized 11H-pyrido[2,1-b]quinazolin-11-one derivatives with the SARS-CoV-2 RdRp (PDB: 7AAP) were evaluated in silico. The study suggested that though none of these compounds showed interactions better than penciclovir but the compound 3a and 3n appeared to be comparable along with 3b seemed to be nearly comparable to favipiravir and remdesivir. The compound 3n with the best binding energy (-79.85 Kcal/mol) participated in the H-bond interactions through its OMe group with THR556 as well as ARG624 and via the N-5 atom with the residue SER682. The in silico studies further suggested that majority of the compounds interacted with the main cavity of active site pocket whereas 3h and 3o that showed relatively lower binding energies (-66.06 and -66.28 Kcal/mol) interacted with the shallow cavity underneath the active site of SARS CoV-2 RdRp. The study also revealed that a OMe group was favourable for interaction with respect to its position in the order C-8 > C-1 > C-2. Further, the presence of a fused quinoline ring was tolerated whereas a fused thiazole ring decreased the interaction significantly. The in silico predictions of pharmacokinetic properties of 3a, 3b and 3n indicated that besides the BBB (Blood Brain Barrier) penetration potential these molecules may show a good overall ADME. Overall, the regioisomers 3a, 3b and 3n have emerged as molecules of possible interest in the context of targeting COVID-19.
Collapse
Affiliation(s)
- Matta Manikanttha
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Greenfields, Vaddeswaram, Guntur, Andhra Pradesh 522 502, India
| | - Kolli Deepti
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Greenfields, Vaddeswaram, Guntur, Andhra Pradesh 522 502, India
| | - Mandava Bhuvan Tej
- Department of Healthcare informatics, Sacred Heart University, 5151 Park Avenue, Fair fields, Connecticut CT06825, USA
| | - Mandava Bhagya Tej
- Department of MBBS, NRI Academi of Medical Sciences, Chinakakani, Guntur, Andhra Pradesh 522503, India
| | - A. Gopi Reddy
- Department of Pharmaceutical Chemistry, SANA College of Pharmacy, Kodad, Telangana, India
| | - Ravikumar Kapavarapu
- Department of Pharmaceutical Chemistry and Phytochemistry, Nirmala College of Pharmacy, Mangalagiri, Andhra Pradesh, India
| | | | - M. V Basaveswara Rao
- Department of Chemistry, Krishna University, Krishna District, Andhra Pradesh, India,Corresponding authors
| | - Manojit Pal
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad 500046, India,Corresponding authors
| |
Collapse
|
4
|
Kumari S, Chakraborty S, Ahmad M, Kumar V, Tailor PB, Biswal BK. Identification of probable inhibitors for the DNA polymerase of the Monkeypox virus through the virtual screening approach. Int J Biol Macromol 2023; 229:515-528. [PMID: 36584781 PMCID: PMC9794403 DOI: 10.1016/j.ijbiomac.2022.12.252] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Given the paucity of antiviral treatments for monkeypox disease, caused by the Monkeypox virus (MPXV), there is a pressing need for the development/identification of new drugs to treat the infection. MPXV possesses a linear dsDNA genome that is replicated by a DNA replication complex of which DNA polymerase (DPol) forms an important component. Owing to the importance of DPol in the viral life cycle, identifying/designing small molecules abolishing its function could yield new antivirals. In this study, we first used the AlphaFold artificial intelligence program to model the 3D structure of the MPXV DPol; like the fold of DPol from other organisms, the MPXV DPol structure has the characteristic exonuclease, thumb, palm, and fingers sub-domains arrangement. Subsequently, we have identified several inhibitors through virtual screening of ZINC and antiviral libraries. Molecules with phenyl scaffold along with alanine-based and tetrazole-based molecules showed the best docking score of -8 to -10 kcal/mol. These molecules bind in the palm and fingers sub-domains interface region, which partially overlaps with the DNA binding path. The delineation of DPol/inhibitor interactions showed that majorly active site residues ASP549, ASP753, TYR550, ASN551, SER552, and ASN665 interact with the inhibitors. These compounds exhibit good Absorption, Distribution, Metabolism and Excretion properties.
Collapse
Affiliation(s)
- Swati Kumari
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sayan Chakraborty
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Mohammed Ahmad
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Varun Kumar
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | - Bichitra K Biswal
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
5
|
Xu X, Chen Y, Lu X, Zhang W, Fang W, Yuan L, Wang X. An update on inhibitors targeting RNA-dependent RNA polymerase for COVID-19 treatment: Promises and challenges. Biochem Pharmacol 2022; 205:115279. [PMID: 36209840 PMCID: PMC9535928 DOI: 10.1016/j.bcp.2022.115279] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 01/18/2023]
Abstract
The highly transmissible variants of SARS-CoV-2, the causative pathogen of the COVID-19 pandemic, bring new waves of infection worldwide. Identification of effective therapeutic drugs to combat the COVID-19 pandemic is an urgent global need. RNA-dependent RNA polymerase (RdRp), an essential enzyme for viral RNA replication, is the most promising target for antiviral drug research since it has no counterpart in human cells and shows the highest conservation across coronaviruses. This review summarizes recent progress in studies of RdRp inhibitors, focusing on interactions between these inhibitors and the enzyme complex, based on structural analysis, and their effectiveness. In addition, we propose new possible strategies to address the shortcomings of current inhibitors, which may guide the development of novel efficient inhibitors to combat COVID-19.
Collapse
Affiliation(s)
- Xiaoying Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yuheng Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinyu Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Wanlin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Wenxiu Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Luping Yuan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Xiaoyan Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China.
| |
Collapse
|
6
|
Medhat MA, El-Kassas M, Karam-Allah H, Al Shafie A, Abd-Elsalam S, Moustafa E, Hassany SM, Salama M, Abd Elghafar MS, Sayed H, Badr M, Kamal DT, Shamseldeen A, Ossimi A, Moaz I, Esmael HED, Ezz Eldin AM, Ezzat S, Abdelghaffar H, Abdelghaffar K. Sofosbuvir/ledipasvir in combination or nitazoxanide alone are safe and efficient treatments for COVID-19 infection: A randomized controlled trial for repurposing antivirals. Arab J Gastroenterol 2022; 23:165-171. [PMID: 35690556 PMCID: PMC9116423 DOI: 10.1016/j.ajg.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/16/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND STUDY AIMS Currently, there is no therapy approved for COVID-19. We evaluated the efficacy and safety of sofosbuvir/ledipasvir and nitazoxanide for the treatment of patients with COVID-19 infection. PATIENTS AND METHODS A multicenter, open-label randomized controlled trial included one hundred and ninety patients with non-severe COVID-19 infection. Patients were randomized into three groups. All groups received standard care treatment (SCT). In addition, group 1 received sofosbuvir/ledipasvir, and group 2 received nitazoxanide. Follow-up by reverse-transcriptase polymerase chain reaction (RT-PCR) was done at intervals of 5, 8, 11, and 14 days. The primary endpoint was viral clearance. RESULTS Viral clearance was significantly higher in the sofosbuvir/ledipasvir and nitazoxanide groups compared to the SCT group in all follow-up intervals (p < 0.001). In the sofosbuvir/ledipasvir arm, 36.9% showed early viral clearance by day 5. By day 14, 83.1% of the sofosbuvir/ledipasvir group, 39.7% of the nitazoxanide group, and 19.4% of the SCT group tested negative for SARS-CoV-2. Sofosbuvir/ledipasvir and nitazoxanide treatment were the only significant factors in Cox regression of negative RT-PCR with the highest OR (17.88, 95% CI: 6.66-47.98 and 2.59, 95% CI: 1.11-6.07, respectively). No mortality or serious adverse events were recorded. CONCLUSION The addition of sofosbuvir/ledipasvir or nitazoxanide to the SCT results in an early and high viral clearance rate in mild and moderate patients with COVID-19. These drugs represent a safe and affordable treatment for COVID-19.
Collapse
Affiliation(s)
- Mohammed A Medhat
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt.
| | - Haidi Karam-Allah
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Al Shafie
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Sherief Abd-Elsalam
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ehab Moustafa
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sahar M Hassany
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwa Salama
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed S Abd Elghafar
- Anesthesia, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hamdy Sayed
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Mohamed Badr
- Critical Care Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Dalia T Kamal
- Clinical Pathology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Shamseldeen
- Critical Care Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Ashima'a Ossimi
- Critical Care Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Inas Moaz
- Epidemiology and Preventive Medicine Department, National Liver Institute, Menoufia University, Menoufia, Egypt
| | - Hossam El-Deen Esmael
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Azza M Ezz Eldin
- Clinical Pathology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sameera Ezzat
- Epidemiology and Preventive Medicine Department, National Liver Institute, Menoufia University, Menoufia, Egypt
| | | | | |
Collapse
|
7
|
IMTIAZ F, PASHA MK. A systematic review of RdRp of SARS-CoV-2 through artificial intelligence and machine learning utilizing structure-based drug design strategy. Turk J Chem 2021; 46:583-594. [PMID: 37720604 PMCID: PMC10503974 DOI: 10.55730/1300-0527.3355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 06/16/2022] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Since the coronavirus disease has been declared a global pandemic, it had posed a challenge among researchers and raised common awareness and collaborative efforts towards finding the solution. Caused by severe acute respiratory coronavirus syndrome-2 (SARS-CoV-2), coronavirus drug design strategy needs to be optimized. It is understandable that cognizance of the pathobiology of COVID-19 can help scientists in the development and discovery of therapeutically effective antiviral drugs by elucidating the unknown viral pathways and structures. Considering the role of artificial intelligence and machine learning with its advancements in the field of science, it is rational to use these methods which can aid in the discovery of new potent candidates in silico. Our review utilizes similar methodologies and focuses on RNA-dependent RNA polymerase (RdRp), based on its importance as an essential element for virus replication and also a promising target for COVID-19 therapeutics. Artificial neural network technique was used to shortlist articles with the support of PRISMA, from different research platforms including Scopus, PubMed, PubChem, and Web of Science, through a combination of keywords. "English language", from the year "2000" and "published articles in journals" were selected to carry out this research. We summarized that structural details of the RdRp reviewed in this analysis will have the potential to be taken into consideration when developing therapeutic solutions and if further multidisciplinary efforts are taken in this domain then potential clinical candidates for RdRp of SARS-CoV-2 could be successfully delivered for experimental validations.
Collapse
Affiliation(s)
- Fariha IMTIAZ
- Punjab University College of Pharmacy, University of the Punjab, Lahore,
Pakistan
| | | |
Collapse
|
8
|
Lochab A, Thareja R, Gadre SD, Saxena R. Potential Protein and Enzyme Targets for In‐silico Development and Repurposing of Drug Against Coronaviruses. ChemistrySelect 2021. [DOI: 10.1002/slct.202103350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Amit Lochab
- Department of Chemistry Kirori Mal College University of Delhi Delhi India
| | - Rakhi Thareja
- Department of Chemistry St. Stephens College University of Delhi Delhi India
| | - Sangeeta D. Gadre
- Department of Physics Kirori Mal College University of Delhi Delhi India
| | - Reena Saxena
- Department of Chemistry Kirori Mal College University of Delhi Delhi India
| |
Collapse
|
9
|
Radder SB, Melavanki R, Hiremath SM, Kusanur R, Khemalapure SS, Jeyaseelan SC. Synthesis, spectroscopic (FT-IR, FT-Raman, NMR & UV-Vis), reactive (ELF, LOL, Fukui), drug likeness and molecular docking insights on novel 4-[3-(3-methoxy-phenyl)-3-oxo-propenyl]-benzonitrile by experimental and computational methods. Heliyon 2021; 7:e08429. [PMID: 34877424 PMCID: PMC8632848 DOI: 10.1016/j.heliyon.2021.e08429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/21/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
The spectroscopic analysis such as FT-IR, FT-Raman, UV-Vis and NMR are conducted for the synthesized molecule by both experimental and theoretical approach. The theoretical computations were achieved by DFT method with B3LYP functional and 6-311 ++ G (d, P) basis set. Firstly the geometrical parameters obtained by DFT are compared with the related experimental parameters. Experimental FT-IR and FT-Raman spectra of the title molecule have been acquired. The vibrational analysis is conducted and the assignments concerned to the observed bands are mentioned through the potential energy distribution (PED). The GIAO method was employed for theoretical NMR analysis and the results are compared with experimental chemical shifts. In accumulation to these analyses NLO, NBO, FMO and MEP analysis have been conducted to understand the nature of the molecule. ELF and LOL were performed. The drug likeness and molecular docking studies also conducted. The potency of inhibition of molecule against MPRO and PLPRO receptors has been performed using molecular docking studies.
Collapse
Affiliation(s)
- Shivaraj B. Radder
- Department of Physics, M S Ramaiah Institute of Technology, Bangalore, 560054, Karnataka, India
- Affiliated to Visvesvaraya Technological University, Belgaum, 590018, Karnataka, India
| | - Raveendra Melavanki
- Department of Physics, M S Ramaiah Institute of Technology, Bangalore, 560054, Karnataka, India
- Affiliated to Visvesvaraya Technological University, Belgaum, 590018, Karnataka, India
| | - Sudhir M. Hiremath
- Department of P.G. Studies in Physics, KLE Society's J.T. College, Gadag, 582101, Karnataka, India
| | - Raviraj. Kusanur
- Department of Chemistry, R.V. College of Engineering, Bangalore, 560059, Karnataka, India
| | - Seema S. Khemalapure
- P.G. Department of Studies and Research in Physics, KLE Society's P.C. Jabin Science College, Hubballi, 580031, Karnataka, India
| | | |
Collapse
|
10
|
Sonkar C, Doharey PK, Rathore AS, Singh V, Kashyap D, Sahoo AK, Mittal N, Sharma B, Jha HC. Repurposing of gastric cancer drugs against COVID-19. Comput Biol Med 2021; 137:104826. [PMID: 34537409 PMCID: PMC8420180 DOI: 10.1016/j.compbiomed.2021.104826] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022]
Abstract
Corona Virus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has become a global pandemic. Additionally, the SARS-CoV-2 infection in the patients of Gastric Cancer (GC; the third leading cause of death in the world) pose a great challenge for the health management of the patients. Since there have been uncertainties to develop a new drug against COVID-19, there is an urgent need for repurposing drugs that can target key proteins of both SARS-CoV-2 and GC. The SARS-CoV-2-RdRp protein contains the NiRAN domain, which is known to have kinase-like folds. A docking study of the FDA approved drugs against GC was performed using AutoDock 4.2 and Glide Schrodinger suite 2019 against SARS-CoV-2-RdRp protein. MMGBSA and MD simulation studies were performed to investigate the binding and stability of the inhibitors with the target protein. In this study, we have found 12 kinase inhibitors with high binding energies namely Baricitinib, Brepocitinib, Decernotinib, Fasudil, Filgotinib, GSK2606414, Peficitinib, Ruxolitinib, Tofacitinib, Upadacitinib, Pamapimod and Ibrutinib. These FDA approved drugs against GC can play a key role in the treatment of COVID-19 patients along with GC as comorbidity. We also hypothesize that JAK, ITK, Rho-associated kinases, FGFR2, FYN, PERK, TYK2, p38-MAPK and SYK kinases can be considered as key therapeutic targets in COVID-19 treatment. Taken altogether, we have proposed the SARS-CoV-2-RdRp as a potential therapeutic target through in-silico studies. However, further in-vitro and in-vivo studies are required for the validation of the proposed targets and drugs for the treatment of COVID-19 patients already suffering from GC.
Collapse
Affiliation(s)
- Charu Sonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Pawan Kumar Doharey
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, U.P., India
| | - Anuranjan Singh Rathore
- SASTRA Deemed to Be University, Trichy-Tanjore Road, Thirumalaisamudram, Thanjavur, Tamil Nadu, 613401, India
| | - Vishal Singh
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, 211015, U.P., India
| | - Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, 211015, U.P., India
| | - Nitish Mittal
- Computational and Systems Biology, Biozentrum, University of Basel, Klingelbergstrasse 50-70, 4056, Basel, Switzerland
| | - Bechan Sharma
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, U.P., India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore, 453552, India.
| |
Collapse
|
11
|
Li J, McKay KT, Remington JM, Schneebeli ST. A computational study of cooperative binding to multiple SARS-CoV-2 proteins. Sci Rep 2021; 11:16307. [PMID: 34381116 PMCID: PMC8358031 DOI: 10.1038/s41598-021-95826-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/30/2021] [Indexed: 01/18/2023] Open
Abstract
Structure-based drug design targeting the SARS-CoV-2 virus has been greatly facilitated by available virus-related protein structures. However, there is an urgent need for effective, safe small-molecule drugs to control the spread of the virus and variants. While many efforts are devoted to searching for compounds that selectively target individual proteins, we investigated the potential interactions between eight proteins related to SARS-CoV-2 and more than 600 compounds from a traditional Chinese medicine which has proven effective at treating the viral infection. Our original ensemble docking and cooperative docking approaches, followed by a total of over 16-micorsecond molecular simulations, have identified at least 9 compounds that may generally bind to key SARS-CoV-2 proteins. Further, we found evidence that some of these compounds can simultaneously bind to the same target, potentially leading to cooperative inhibition to SARS-CoV-2 proteins like the Spike protein and the RNA-dependent RNA polymerase. These results not only present a useful computational methodology to systematically assess the anti-viral potential of small molecules, but also point out a new avenue to seek cooperative compounds toward cocktail therapeutics to target more SARS-CoV-2-related proteins.
Collapse
Affiliation(s)
- Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA.
| | - Kyle T McKay
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | - Jacob M Remington
- Department of Chemistry, University of Vermont, Burlington, VT, 05405, USA
| | | |
Collapse
|
12
|
Sun ZG, Yu FL, Qiu XT, Li S, Li XT, Zhu HL. The Promising Enzymes for Inhibitors Development against COVID-19. Mini Rev Med Chem 2021; 22:449-456. [PMID: 34353251 DOI: 10.2174/1389557521666210805104250] [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: 12/31/2020] [Revised: 02/11/2021] [Accepted: 04/12/2021] [Indexed: 01/18/2023]
Abstract
Since the outbreak of COVID-19, it has been an epidemic for nearly a year. COVID-19 has brought painful disasters to people all over the world. It not only threatens lives and health, but also induces economic crises. At present, promising methods to eradicate COVID-19 mainly include drugs and vaccines. Enzyme inhibitors have always been a reliable strategy for the treatment of related diseases. Scientists worldwide have worked together to study COVID-19, have obtained the structure of key SARS-CoV-2 associated enzymes, and reported the research of inhibitors of these enzymes. This article summarizes COVID-19-related enzyme inhibitors' recent development, mainly including 3CLpro, PLpro, TMPRSS2, and RdRp inhibitors, hoping to provide valuable weapons in the ensuing battle against COVID-19.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400. China
| | - Feng-Ling Yu
- Pharmacy Department, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400. China
| | - Xiang-Ting Qiu
- Clinical Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400. China
| | - Shuang Li
- Pharmacy Department, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400. China
| | - Xue-Tang Li
- Pharmacy Department, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400. China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing 210023. China
| |
Collapse
|
13
|
Pandey K, Lokhande KB, Swamy KV, Nagar S, Dake M. In Silico Exploration of Phytoconstituents From Phyllanthus emblica and Aegle marmelos as Potential Therapeutics Against SARS-CoV-2 RdRp. Bioinform Biol Insights 2021; 15:11779322211027403. [PMID: 34248355 PMCID: PMC8236766 DOI: 10.1177/11779322211027403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide has increased the importance of computational tools to design a drug or vaccine in reduced time with minimum risk. Earlier studies have emphasized the important role of RNA-dependent RNA polymerase (RdRp) in SARS-CoV-2 replication as a potential drug target. In our study, comprehensive computational approaches were applied to identify potential compounds targeting RdRp of SARS-CoV-2. To study the binding affinity and stability of the phytocompounds from Phyllanthus emblica and Aegel marmelos within the defined binding site of SARS-CoV-2 RdRp, they were subjected to molecular docking, 100 ns molecular dynamics (MD) simulation followed by post-simulation analysis. Furthermore, to assess the importance of features involved in the strong binding affinity, molecular field-based similarity analysis was performed. Based on comparative molecular docking and simulation studies of the selected phytocompounds with SARS-CoV-2 RdRp revealed that EBDGp possesses a stronger binding affinity (-23.32 kcal/mol) and stability than other phytocompounds and reference compound, Remdesivir (-19.36 kcal/mol). Molecular field-based similarity profiling has supported our study in the validation of the importance of the presence of hydroxyl groups in EBDGp, involved in increasing its binding affinity toward SARS-CoV-2 RdRp. Molecular docking and dynamic simulation results confirmed that EBDGp has better inhibitory potential than Remdesivir and can be an effective novel drug for SARS-CoV-2 RdRp. Furthermore, binding free energy calculations confirmed the higher stability of the SARS-CoV-2 RdRp-EBDGp complex. These results suggest that the EBDGp compound may emerge as a promising drug against SARS-CoV-2 and hence requires further experimental validation.
Collapse
Affiliation(s)
- Khushboo Pandey
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - K Venkateswara Swamy
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
- Bioinformatics and Drug Discovery Group, MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| | - Shuchi Nagar
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - Manjusha Dake
- Protein Biochemistry Laboratory, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| |
Collapse
|
14
|
Mariadasse R, Rajmichael R, Dwivedy A, Amala M, Ahmad M, Mutharasappan N, Biswal BK, Jeyakanthan J. Characterization of putative transcriptional regulator (PH0140) and its distal homologue. Cell Signal 2021; 84:110031. [PMID: 33932498 DOI: 10.1016/j.cellsig.2021.110031] [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: 02/18/2021] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 11/28/2022]
Abstract
In this study, a phylogenetic tree was constructed using 1854 sequences of various Lrp/AnsC (FFRPs) and ArsR proteins from pathogenic and non-pathogenic organisms. Despite having sequence similarities, FFRPs and ArsR proteins functioning differently as a transcriptional regulator and de-repressor in the presence of exogenous amino acids and metal ions, respectively. To understand these functional differences, the structures of various FFRPs and ArsR proteins (134 sequences) were modeled. Several ArsR proteins exhibited high similarity to the FFRPs while in few proteins, unusual structural folds were observed. However, the Helix-turn-Helix (HTH) domains are common among them and the ligand-binding domains are structurally dissimilar suggest the differences in their binding preferences. Despite low sequence conservation, most of these proteins revealed negatively charged surfaces in the active site pockets. Representative structures (PH0140 and TtArsR protein) from FFRPs and ArsR protein families were considered and evaluated for their functional differences using molecular modeling studies. Our earlier study has explained the binding preference of exogenous Tryptophan and the related transcriptional regulatory mechanism of PH0140 protein. In this study, a Cu2+ ion-induced de-repression mechanism of the TtArsR-DNA complex was characterized through docking and molecular dynamics. Further, the proteins were purified and their efficiency for sensing Tryptophan and Cu2+ ions were analyzed using cyclic voltammetry. Overall, the study explores the structural evolution and functional difference of FFRPs and ArsR proteins that present the possibilities of PH0140 and TtArsR as potential bio-sensory molecules.
Collapse
Affiliation(s)
- Richard Mariadasse
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi 630 004, India
| | - Raji Rajmichael
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi 630 004, India
| | | | - Mathimaran Amala
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi 630 004, India
| | | | - Nachiappan Mutharasappan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi 630 004, India
| | | | - Jeyaraman Jeyakanthan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi 630 004, India.
| |
Collapse
|
15
|
Agrawal N, Goyal A. Potential Candidates against COVID-19 Targeting RNA-Dependent RNA Polymerase: A Comprehensive Review. Curr Pharm Biotechnol 2021; 23:396-419. [PMID: 33882805 DOI: 10.2174/1389201022666210421102513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 11/22/2022]
Abstract
Due to the extremely contagious nature of SARS-COV-2, it presents a significant threat to humans worldwide. A plethora of studies are going on all over the world to discover the drug to fight SARS-COV-2. One of the most promising targets is RNA-dependent RNA polymerase (RdRp), responsible for viral RNA replication in host cells. Since RdRp is a viral enzyme with no host cell homologs, it allows the development of selective SARS-COV-2 RdRp inhibitors. A variety of studies used in silico approaches for virtual screening, molecular docking, and repurposing of already existing drugs and phytochemicals against SARS-COV-2 RdRp. This review focuses on collating compounds possessing the potential to inhibit SARS-COV-2 RdRp based on in silico studies to give medicinal chemists food for thought so that the existing drugs can be repurposed for the control and treatment of ongoing COVID-19 pandemic after performing in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura-281406 (U.P.), India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura-281406 (U.P.), India
| |
Collapse
|
16
|
Vicenti I, Zazzi M, Saladini F. SARS-CoV-2 RNA-dependent RNA polymerase as a therapeutic target for COVID-19. Expert Opin Ther Pat 2021; 31:325-337. [PMID: 33475441 PMCID: PMC7938656 DOI: 10.1080/13543776.2021.1880568] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Introduction: The current SARS-CoV-2 pandemic urgently demands for both prevention and treatment strategies. RNA-dependent RNA-polymerase (RdRp), which has no counterpart in human cells, is an excellent target for drug development. Given the time-consuming process of drug development, repurposing drugs approved for other indications or at least successfully tested in terms of safety and tolerability, is an attractive strategy to rapidly provide an effective medication for severe COVID-19 cases.Areas covered: The currently available data and upcominSg studies on RdRp which can be repurposed to halt SARS-CoV-2 replication, are reviewed.Expert opinion: Drug repurposing and design of novel compounds are proceeding in parallel to provide a quick response and new specific drugs, respectively. Notably, the proofreading SARS-CoV-2 exonuclease activity could limit the potential for drugs designed as immediate chain terminators and favor the development of compounds acting through delayed termination. While vaccination is awaited to curb the SARS-CoV-2 epidemic, even partially effective drugs from repurposing strategies can be of help to treat severe cases of disease. Considering the high conservation of RdRp among coronaviruses, an improved knowledge of its activity in vitro can provide useful information for drug development or drug repurposing to combat SARS-CoV-2 as well as future pandemics.
Collapse
Affiliation(s)
- Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Saladini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
17
|
Prussia AJ, Chennamadhavuni S. Biostructural Models for the Binding of Nucleoside Analogs to SARS-CoV-2 RNA-Dependent RNA Polymerase. J Chem Inf Model 2021; 61:1402-1411. [PMID: 33655751 PMCID: PMC7945588 DOI: 10.1021/acs.jcim.0c01277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 01/18/2023]
Abstract
SARS-CoV-2 is a positive-sense RNA virus that requires an RNA-dependent RNA polymerase (RdRp) for replication of its viral genome. Nucleoside analogs such as Remdesivir and β-d-N4-hydroxycytidine are antiviral candidates and may function as chain terminators or induce viral mutations, thus impairing RdRp function. Recently disclosed Cryo-EM structures of apo, RNA-bound, and inhibitor-bound SARS-CoV-2 RdRp provided insight into the inhibitor-bound structure by capturing the enzyme with its reaction product: Remdesivir covalently bound to the RNA primer strand. To gain a structural understanding of the binding of this and several other nucleoside analogs in the precatalytic state, molecular models were developed that predict the noncovalent interactions to a complex of SARS-CoV-2 RdRp, RNA, and catalytic metal cations. MM-GBSA evaluation of these interactions is consistent with resistance-conferring mutations and existing structure-activity relationship (SAR) data. Therefore, this approach may yield insights into antiviral mechanisms and guide the development of experimental drugs for COVID-19 treatment.
Collapse
|
18
|
M RK, Gideon DA, Mariadasse R, Nirusimhan V, A SR, Edward JC, Jeyaraman J, Dhayabaran V. In silico evaluation of isatin-based derivatives with RNA-dependent RNA polymerase of the novel coronavirus SARS-CoV-2. J Biomol Struct Dyn 2021; 40:6710-6724. [PMID: 33615998 DOI: 10.1080/07391102.2021.1890223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Isatin (1H-indole-2,3-dione)-containing compounds have been shown to possess several remarkable biological activities. We had previously explored a few isatin-based imidazole derivatives for their predicted dual activity against both inflammation and cancer. We explored 47 different isatin-based derivatives (IBDs) for other potential biological activities using in silico tools and found them to possess anti-viral activity. Using AutoDock tools, the binding site, binding energy, inhibitory constant/Ki and receptor-ligand interactions for each of the compounds were analyzed against SARS-CoV-2 RNA-dependent RNA polymerase (RdRp). The partition coefficient (logP) values were predicted using MedChem Designer tool. Based on the best Ki, binding energy and the ideal range of logP (between 1.0 and 3.0), 10 out of total 47 compounds were deemed to be prospective RdRp inhibitors. Some of these compounds gave better Ki, binding energy and logP values when compared to standard RdRp inhibitors, such as remdesivir (REM) (Ki = 15.61 μM, logP = 2.2; binding energy = -6.95), a clinically approved RdRp inhibitor and nine other RdRp inhibitors. The results showed that the 10 selected IBDs could be further explored. Molecular dynamics simulations (MDSs) showed that the selected RdRp-IBD complexes were highly stable compared to the native RdRp and RdRp-REM complex during 100 ns time periods. DFT studies were performed for the compounds 16a, 24a, 28a, 38a and 40a, to evaluate the charge transfer mechanism for the interactions between the IBDs and the RdRp residues. Among these, ADME profiling revealed that 28a is a possible lead compound which can be explored further for anti-RdRp activity in vitro. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rajesh Kumar M
- Department of Chemistry, Bishop Heber College (Autonomous), Tiruchirappalli, India
| | - Daniel A Gideon
- Department of Biotechnology & Bioinformatics, Bishop Heber College (Autonomous), Tiruchirappalli, India
| | | | - Vijay Nirusimhan
- Department of Biotechnology & Bioinformatics, Bishop Heber College (Autonomous), Tiruchirappalli, India
| | - Sherlin Rosita A
- Department of Biotechnology & Bioinformatics, Bishop Heber College (Autonomous), Tiruchirappalli, India
| | - Jesu Castin Edward
- Department of Biotechnology & Bioinformatics, Bishop Heber College (Autonomous), Tiruchirappalli, India
| | | | - Violet Dhayabaran
- Department of Chemistry, Bishop Heber College (Autonomous), Tiruchirappalli, India
| |
Collapse
|
19
|
Wakchaure PD, Ghosh S, Ganguly B. Revealing the Inhibition Mechanism of RNA-Dependent RNA Polymerase (RdRp) of SARS-CoV-2 by Remdesivir and Nucleotide Analogues: A Molecular Dynamics Simulation Study. J Phys Chem B 2020; 124:10641-10652. [PMID: 33190493 DOI: 10.1021/acs.jpcb.0c06747] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Antiviral drug therapy against SARS-CoV-2 is not yet established and posing a serious global health issue. Remdesivir is the first antiviral compound approved by the US FDA for the SARS-CoV-2 treatment for emergency use, targeting RNA-dependent RNA polymerase (RdRp) enzyme. In this work, we have examined the action of remdesivir and other two ligands screened from the library of nucleotide analogues using docking and molecular dynamics (MD) simulation studies. The MD simulations have been performed for all the ligand-bound RdRp complexes for the 30 ns time scale. This is one of the earlier reports to perform the MD simulations studies using the SARS-CoV-2 RdRp crystal structure (PDB ID 7BTF). The MD trajectories were analyzed and Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) calculations were performed to calculate the binding free energy. The binding energy data reveal that compound-17 (-59.6 kcal/mol) binds more strongly as compared to compound-8 (-46.3 kcal/mol) and remdesivir (-29.7 kcal/mol) with RdRp. The detailed analysis of trajectories shows that the remdesivir binds in the catalytic site and forms a hydrogen bond with the catalytic residues from 0 to 0.46 ns. Compound-8 binds in the catalytic site but does not form direct hydrogen bonds with catalytic residues. Compound-17 showed the formation of hydrogen bonds with catalytic residues throughout the simulation process. The MD simulation results such as hydrogen bonding, the center of mass distance analysis, snapshots at a different time interval, and binding energy suggest that compound-17 binds strongly with RdRp of SARS-CoV-2 and has the potential to develop as a new antiviral against COVID-19. Further, the frontier molecular orbital analysis and molecular electrostatic potential (MESP) iso-surface analysis using DFT calculations shed light on the superior binding of compound-17 with RdRp compared to remdesivir and compound-8. The computed as well as the experimentally reported pharmacokinetics and toxicity parameters of compound-17 is encouraging and therefore can be one of the potential candidates for the treatment of COVID-19.
Collapse
Affiliation(s)
- Padmaja D Wakchaure
- Computation and Simulation Unit (Analytical and Environmental Science Division and Centralized Instrument Facility), CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shibaji Ghosh
- Computation and Simulation Unit (Analytical and Environmental Science Division and Centralized Instrument Facility), CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bishwajit Ganguly
- Computation and Simulation Unit (Analytical and Environmental Science Division and Centralized Instrument Facility), CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
20
|
In Silico Identification of Potential Natural Product Inhibitors of Human Proteases Key to SARS-CoV-2 Infection. Molecules 2020; 25:molecules25173822. [PMID: 32842606 PMCID: PMC7504347 DOI: 10.3390/molecules25173822] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022] Open
Abstract
Presently, there are no approved drugs or vaccines to treat COVID-19, which has spread to over 200 countries and at the time of writing was responsible for over 650,000 deaths worldwide. Recent studies have shown that two human proteases, TMPRSS2 and cathepsin L, play a key role in host cell entry of SARS-CoV-2. Importantly, inhibitors of these proteases were shown to block SARS-CoV-2 infection. Here, we perform virtual screening of 14,011 phytochemicals produced by Indian medicinal plants to identify natural product inhibitors of TMPRSS2 and cathepsin L. AutoDock Vina was used to perform molecular docking of phytochemicals against TMPRSS2 and cathepsin L. Potential phytochemical inhibitors were filtered by comparing their docked binding energies with those of known inhibitors of TMPRSS2 and cathepsin L. Further, the ligand binding site residues and non-covalent interactions between protein and ligand were used as an additional filter to identify phytochemical inhibitors that either bind to or form interactions with residues important for the specificity of the target proteases. This led to the identification of 96 inhibitors of TMPRSS2 and 9 inhibitors of cathepsin L among phytochemicals of Indian medicinal plants. Further, we have performed molecular dynamics (MD) simulations to analyze the stability of the protein-ligand complexes for the three top inhibitors of TMPRSS2 namely, qingdainone, edgeworoside C and adlumidine, and of cathepsin L namely, ararobinol, (+)-oxoturkiyenine and 3α,17α-cinchophylline. Interestingly, several herbal sources of identified phytochemical inhibitors have antiviral or anti-inflammatory use in traditional medicine. Further in vitro and in vivo testing is needed before clinical trials of the promising phytochemical inhibitors identified here.
Collapse
|