1
|
Falbo E, Delre P, Lavecchia A. From Apo to Ligand-Bound: Unraveling PPARγ-LBD Conformational Shifts via Advanced Molecular Dynamics. ACS OMEGA 2025; 10:13303-13318. [PMID: 40224459 PMCID: PMC11983173 DOI: 10.1021/acsomega.4c11128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor whose ligand-induced conformational changes, primarily driven by helix 12 (H12) repositioning, regulate transcriptional activity. However, the precise mechanism remains elusive. In this study, we performed classical molecular dynamics (cMD) simulations of the PPARγ ligand binding domain (LBD) in complex with two agonists (BRL, 3EA), a partial agonist (GW0072), and an antagonist (EKP), generating 3 μs trajectories for each system. To gain deeper insights, we integrated machine learning-assisted clustering with MD simulations, revealing a favorable trend in binding free energy (ΔG b), suggesting enhanced complex stability. A case study on EKP demonstrated that, despite fitting within the binding site, it failed to induce rapid LBD or H12 rearrangements in the apo agonist-induced conformation. Additionally, we investigated the apo-state conformations of PPARγ-LBD influenced by agonist and antagonist ligands, utilizing cMD and Gaussian accelerated molecular dynamics (GaMD) over a cumulative 6 μs (3 μs cMD + 3 μs GaMD). Key residues known to modulate PPARγ function upon mutation were analyzed, and simulations confirmed the high stability of both apo and ligand-bound conformations. Notably, in the apo state, specific H12 residues interacted with other PPARγ-LBD regions, preventing disorder and abrupt transitions. These findings guided the selection of collective variables (CVs) for well-tempered metadynamics (WT-MetaD) simulations, which-in the apo-agonist state-captured the H12 shift from agonist- to antagonist-like conformations, consistent with resolved X-ray structures. Overall, this computational framework provides novel insights into PPARγ-LBD conformational dynamics and establishes a valuable approach for rationally assessing the effects of modulators on PPARγ activity.
Collapse
Affiliation(s)
| | | | - Antonio Lavecchia
- Department of Pharmacy, “Drug
Discovery Laboratory”, University
of Naples Federico II, via Domenico Montesano 49, I-80131 Naples, Italy
| |
Collapse
|
2
|
Salvi V, Khodke P, Patil P, Kumbhar BV. Identification of a potential anti-viral drug targeting allosteric site of papain-like protease against rubella using a molecular modeling approach. J Biomol Struct Dyn 2024:1-17. [PMID: 39714197 DOI: 10.1080/07391102.2024.2443132] [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: 12/12/2023] [Accepted: 06/27/2024] [Indexed: 12/24/2024]
Abstract
Rubella virus (RUBV) is responsible for causing rashes, lymphadenopathy, and fever which are the hallmarks of an acute viral illness called Rubella. For RUBV replication, the non-structural polyprotein p200 must be cleaved by the rubella papain-like protease (RubPro) into the multifunctional proteins p150 and p90. Hence, RubPro is an attractive target for anti-viral drug discovery. Moreover, the binding of host Calmodulin 1 (CaM) to RubPro modulates the protease activity and infectivity of RUBV. However, the binding mode of CaM and RubPro remain uncertain. Therefore, our investigation not only delves into understanding the interaction between CaM and the RubPro but also aims to recognize the allosteric site for the development of antiviral protease inhibitors. In this study, we interestingly identified the allosteric site in close vicinity with the CaM binding domain of RubPro. Considering the allosteric site of RubPro, we employed a computational modelling approach to identify the potential antiviral compounds. Leveraging ChemDiv protease inhibitors database, we employed structure-based virtual screening, ADME, pass prediction, and docking studies, unveiling three potent compounds: C073-2897, C073-3328, and C073-3368. Moreover, molecular dynamics simulation analysis revealed that these compounds affect the RubPro structure and dynamics and may also influence the binding of CaM with RubPro. Notably, binding energy calculation showed that the compound C073-3328 exhibits higher binding affinity, while C073-3368 displays a lower binding affinity with RubPro. These compounds signify potential for managing RUBV infections and pioneering effective antiviral treatments. This computational study could pave the way for improved methods of managing or controlling rubella infections.
Collapse
Affiliation(s)
- Vrinda Salvi
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, Maharashtra, India
| | - Purva Khodke
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, Maharashtra, India
| | - Pruthanka Patil
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, Maharashtra, India
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Azarkar S, Abedi M, Lavasani ASO, Ammameh AH, Goharipanah F, Baloochi K, Bakhshi H, Jafari A. Curcumin as a natural potential drug candidate against important zoonotic viruses and prions: A narrative review. Phytother Res 2024; 38:3080-3121. [PMID: 38613154 DOI: 10.1002/ptr.8119] [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: 01/27/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 04/14/2024]
Abstract
Zoonotic diseases are major public health concerns and undeniable threats to human health. Among Zoonotic diseases, zoonotic viruses and prions are much more difficult to eradicate, as they result in higher infections and mortality rates. Several investigations have shown curcumin, the active ingredient of turmeric, to have wide spectrum properties such as anti-microbial, anti-vascular, anti-inflammatory, anti-tumor, anti-neoplastic, anti-oxidant, and immune system modulator properties. In the present study, we performed a comprehensive review of existing in silico, in vitro, and in vivo evidence on the antiviral (54 important zoonotic viruses) and anti-prion properties of curcumin and curcuminoids in PubMed, Google Scholar, Science Direct, Scopus, and Web of Science databases. Database searches yielded 13,380 results, out of which 216 studies were eligible according to inclusion criteria. Of 216 studies, 135 (62.5%), 24 (11.1%), and 19 (8.8%) were conducted on the effect of curcumin and curcuminoids against SARS-CoV-2, Influenza A virus, and dengue virus, respectively. This review suggests curcumin and curcuminoids as promising therapeutic agents against a wide range of viral zoonoses by targeting different proteins and signaling pathways.
Collapse
Affiliation(s)
- Setareh Azarkar
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Masoud Abedi
- Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | | | | | - Fatemeh Goharipanah
- Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Kimiya Baloochi
- Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Hasan Bakhshi
- Vector-Borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirsajad Jafari
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Zheng Y, Schupp JC, Adams T, Clair G, Justet A, Ahangari F, Yan X, Hansen P, Carlon M, Cortesi E, Vermant M, Vos R, De Sadeleer LJ, Rosas IO, Pineda R, Sembrat J, Königshoff M, McDonough JE, Vanaudenaerde BM, Wuyts WA, Kaminski N, Ding J. Unagi: Deep Generative Model for Deciphering Cellular Dynamics and In-Silico Drug Discovery in Complex Diseases. RESEARCH SQUARE 2023:rs.3.rs-3676579. [PMID: 38196613 PMCID: PMC10775382 DOI: 10.21203/rs.3.rs-3676579/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Human diseases are characterized by intricate cellular dynamics. Single-cell sequencing provides critical insights, yet a persistent gap remains in computational tools for detailed disease progression analysis and targeted in-silico drug interventions. Here, we introduce UNAGI, a deep generative neural network tailored to analyze time-series single-cell transcriptomic data. This tool captures the complex cellular dynamics underlying disease progression, enhancing drug perturbation modeling and discovery. When applied to a dataset from patients with Idiopathic Pulmonary Fibrosis (IPF), UNAGI learns disease-informed cell embeddings that sharpen our understanding of disease progression, leading to the identification of potential therapeutic drug candidates. Validation via proteomics reveals the accuracy of UNAGI's cellular dynamics analyses, and the use of the Fibrotic Cocktail treated human Precision-cut Lung Slices confirms UNAGI's predictions that Nifedipine, an antihypertensive drug, may have antifibrotic effects on human tissues. UNAGI's versatility extends to other diseases, including a COVID dataset, demonstrating adaptability and confirming its broader applicability in decoding complex cellular dynamics beyond IPF, amplifying its utility in the quest for therapeutic solutions across diverse pathological landscapes.
Collapse
Affiliation(s)
- Yumin Zheng
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, Canada
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jonas C. Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Taylor Adams
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Aurelien Justet
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Farida Ahangari
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Xiting Yan
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Paul Hansen
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Marianne Carlon
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Emanuela Cortesi
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Laurens J. De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Ivan O Rosas
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ricardo Pineda
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John E. McDonough
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Wim A. Wuyts
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Jun Ding
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, Canada
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| |
Collapse
|
5
|
Milani G, Budriesi R, Tavazzani E, Cavalluzzi MM, Mattioli LB, Miniero DV, Delre P, Belviso BD, Denegri M, Cuocci C, Rotondo NP, De Palma A, Gualdani R, Caliandro R, Mangiatordi GF, Kumawat A, Camilloni C, Priori S, Lentini G. hERG stereoselective modulation by mexiletine-derived ureas: Molecular docking study, synthesis, and biological evaluation. Arch Pharm (Weinheim) 2023; 356:e2300116. [PMID: 37460390 DOI: 10.1002/ardp.202300116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 10/06/2023]
Abstract
Long QT syndrome (LQTS) is a disorder of cardiac electrophysiology resulting in life-threatening arrhythmias; nowadays, only a few drugs are available for the management of LQTS. Focusing our attention on LQT2, one of the most common subtypes of LQTS caused by mutations in the human ether-à-go-go-related gene (hERG), in the present work, the stereoselectivity of the recently discovered mexiletine-derived urea 8 was investigated on the hERG potassium channel. According to preliminary in silico predictions, in vitro studies revealed a stereoselective behavior, with the meso form showing the greatest hERG opening activity. In addition, functional studies on guinea pig isolated left atria, aorta, and ileum demonstrated that 8 does not present any cardiac or intestinal liability in our ex vivo studies. Due to its overall profile, (R,S)-8 paves the way for the design and development of a new series of compounds potentially useful in the treatment of both congenital and drug-induced forms of LQTS.
Collapse
Affiliation(s)
- Gualtiero Milani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Budriesi
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | | | | | - Laura Beatrice Mattioli
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Pietro Delre
- Chemistry Department, University of Bari Aldo Moro, Bari, Italy
- CNR-Institute of Crystallography, Bari, Italy
| | | | | | | | - Natalie Paola Rotondo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa De Palma
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Roberta Gualdani
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Amit Kumawat
- Department of Biosciences, University of Milan, Milano, Italy
| | - Carlo Camilloni
- Department of Biosciences, University of Milan, Milano, Italy
| | - Silvia Priori
- ICS-Maugeri IRCCS, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
6
|
Abatematteo FS, Delre P, Mercurio I, Rezelj VV, Siliqi D, Beaucourt S, Lattanzi G, Colabufo NA, Leopoldo M, Saviano M, Vignuzzi M, Mangiatordi GF, Abate C. A conformational rearrangement of the SARS-CoV-2 host protein sigma-1 is required for antiviral activity: insights from a combined in-silico/in-vitro approach. Sci Rep 2023; 13:12798. [PMID: 37550340 PMCID: PMC10406941 DOI: 10.1038/s41598-023-39662-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023] Open
Abstract
The development of effective drugs to treat coronavirus infections remains a significant challenge for the scientific community. Recent evidence reports on the sigma-1 receptor (S1R) as a key druggable host protein in the SARS-CoV-1 and SARS-CoV-2 interactomes and shows a potent antiviral activity against SARS-CoV-2 for the S1R antagonist PB28. To improve PB28 activity, we designed and tested a series of its analogues and identified a compound that is fourfold more potent against SARS-CoV-2 than PB28 itself. Interestingly, we found no direct correlation between S1R affinity and SARS-CoV-2 antiviral activity. Building on this, we employed comparative induced fit docking and molecular dynamics simulations to gain insights into the possible mechanism that occurs when specific ligand-protein interactions take place and that may be responsible for the observed antiviral activity. Our findings offer a possible explanation for the experimental observations, provide insights into the S1R conformational changes upon ligand binding and lay the foundation for the rational design of new S1R ligands with potent antiviral activity against SARS-CoV-2 and likely other viruses.
Collapse
Affiliation(s)
- Francesca Serena Abatematteo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona, 4, 70125, Bari, Italy
| | - Pietro Delre
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola 122/O, 70126, Bari, Italy
| | - Ivan Mercurio
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola 122/O, 70126, Bari, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Antonio Vivaldi 43, 81100, Caserta, Italy
| | - Veronica V Rezelj
- Viral Populations and Pathogenesis Unit, UMR 3569, CNRS, Institut Pasteur, Paris, France
| | - Dritan Siliqi
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola 122/O, 70126, Bari, Italy
| | - Stephanie Beaucourt
- Viral Populations and Pathogenesis Unit, UMR 3569, CNRS, Institut Pasteur, Paris, France
| | - Gianluca Lattanzi
- Department of Physics, University of Trento, Via Sommarive 9, 38123, Povo-Trento, Italy
- TIFPA Trento Institute for Fundamental Physics and Applications, Via Sommarive 9, 38123, Povo-Trento, Italy
| | - Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona, 4, 70125, Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona, 4, 70125, Bari, Italy
| | - Michele Saviano
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Vivaldi 43, 81100, Caserta, Italy
| | - Marco Vignuzzi
- Viral Populations and Pathogenesis Unit, UMR 3569, CNRS, Institut Pasteur, Paris, France
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
| | - Giuseppe Felice Mangiatordi
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola 122/O, 70126, Bari, Italy.
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona, 4, 70125, Bari, Italy.
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola 122/O, 70126, Bari, Italy.
| |
Collapse
|
7
|
Farkaš B, Minneci M, Misevicius M, Rozas I. A Tale of Two Proteases: M Pro and TMPRSS2 as Targets for COVID-19 Therapies. Pharmaceuticals (Basel) 2023; 16:834. [PMID: 37375781 PMCID: PMC10301481 DOI: 10.3390/ph16060834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Considering the importance of the 2019 outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) resulting in the coronavirus disease 2019 (COVID-19) pandemic, an overview of two proteases that play an important role in the infection by SARS-CoV-2, the main protease of SARS-CoV-2 (MPro) and the host transmembrane protease serine 2 (TMPRSS2), is presented in this review. After summarising the viral replication cycle to identify the relevance of these proteases, the therapeutic agents already approved are presented. Then, this review discusses some of the most recently reported inhibitors first for the viral MPro and next for the host TMPRSS2 explaining the mechanism of action of each protease. Afterward, some computational approaches to design novel MPro and TMPRSS2 inhibitors are presented, also describing the corresponding crystallographic structures reported so far. Finally, a brief discussion on a few reports found some dual-action inhibitors for both proteases is given. This review provides an overview of two proteases of different origins (viral and human host) that have become important targets for the development of antiviral agents to treat COVID-19.
Collapse
Affiliation(s)
| | | | | | - Isabel Rozas
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, D02 R590 Dublin, Ireland; (B.F.); (M.M.); (M.M.)
| |
Collapse
|
8
|
Rullo M, La Spada G, Miniero DV, Gottinger A, Catto M, Delre P, Mastromarino M, Latronico T, Marchese S, Mangiatordi GF, Binda C, Linusson A, Liuzzi GM, Pisani L. Bioisosteric replacement based on 1,2,4-oxadiazoles in the discovery of 1H-indazole-bearing neuroprotective MAO B inhibitors. Eur J Med Chem 2023; 255:115352. [PMID: 37178666 DOI: 10.1016/j.ejmech.2023.115352] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
Following a hybridization strategy, a series of 5-substituted-1H-indazoles were designed and evaluated in vitro as inhibitors of human monoamine oxidase (hMAO) A and B. Among structural modifications, the bioisostere-based introduction of 1,2,4-oxadiazole ring returned the most potent and selective human MAO B inhibitor (compound 20, IC50 = 52 nM, SI > 192). The most promising inhibitors were studied in cell-based neuroprotection models of SH-SY5Y and astrocytes line against H2O2. Moreover, preliminary drug-like features (aqueous solubility at pH 7.4; hydrolytic stability at acidic and neutral pH) were assessed for selected 1,2,4-oxadiazoles and compared to amide analogues through RP-HPLC methods. Molecular docking simulations highlighted the crucial role of molecular flexibility in providing a better shape complementarity for compound 20 within MAO B enzymatic cleft than rigid analogue 18. Enzymatic kinetics analysis along with thermal stability curves (Tm shift = +2.9 °C) provided clues of a tight-binding mechanism for hMAO B inhibition by 20.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Gabriella La Spada
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Daniela Valeria Miniero
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andrea Gottinger
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Marco Catto
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Pietro Delre
- CNR, Institute of Crystallography, 70126, Bari, Italy
| | - Margherita Mastromarino
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Tiziana Latronico
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Sara Marchese
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | | | - Claudia Binda
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Anna Linusson
- Department of Chemistry, Umeå University, 90187, Umeå, Sweden
| | - Grazia Maria Liuzzi
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Leonardo Pisani
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
9
|
Abatematteo FS, Majellaro M, Montsch B, Prieto-Díaz R, Niso M, Contino M, Stefanachi A, Riganti C, Mangiatordi GF, Delre P, Heffeter P, Sotelo E, Abate C. Development of Fluorescent 4-[4-(3 H-Spiro[isobenzofuran-1,4'-piperidin]-1'-yl)butyl]indolyl Derivatives as High-Affinity Probes to Enable the Study of σ Receptors via Fluorescence-Based Techniques. J Med Chem 2023; 66:3798-3817. [PMID: 36919956 PMCID: PMC10041534 DOI: 10.1021/acs.jmedchem.2c01227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Sigma (σ) receptor subtypes, σ1 and σ2, are targets of wide pharmaceutical interest. The σ2 receptor holds promise for the development of diagnostics and therapeutics against cancer and Alzheimer's disease. Nevertheless, little is known about the mechanisms activated by the σ2 receptor. To contribute to the exploitation of its therapeutic potential, we developed novel specific fluorescent ligands. Indole derivatives bearing the N-butyl-3H-spiro[isobenzofuran-1,4'-piperidine] portion were functionalized with fluorescent tags. Nanomolar-affinity fluorescent σ ligands, spanning from green to red to near-infrared emission, were obtained. Compounds 19 (σ pan affinity) and 29 (σ2 selective), which displayed the best compromise between pharmacodynamic and photophysical properties, were investigated in flow cytometry, confocal, and live cell microscopy, demonstrating their specificity for the σ2 receptor. To the best of our knowledge, these are the first red-emitting fluorescent σ2 ligands, validated as powerful tools for the study of σ2 receptors via fluorescence-based techniques.
Collapse
Affiliation(s)
| | - Maria Majellaro
- Centro Singular Investigación Quimica Biologica e Materiales Moleculares (CIQUS), Departamento de Quimica Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Bianca Montsch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Rubén Prieto-Díaz
- Centro Singular Investigación Quimica Biologica e Materiales Moleculares (CIQUS), Departamento de Quimica Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze del Farmaco, Via Orabona, 4, 79125 Bari, Italy
| | | | - Angela Stefanachi
- Dipartimento di Farmacia-Scienze del Farmaco, Via Orabona, 4, 79125 Bari, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | | | - Pietro Delre
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola, 70126 Bari, Italy
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Eddy Sotelo
- Centro Singular Investigación Quimica Biologica e Materiales Moleculares (CIQUS), Departamento de Quimica Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Via Orabona, 4, 79125 Bari, Italy
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia, Via Amendola, 70126 Bari, Italy
| |
Collapse
|
10
|
Ershov PV, Yablokov EO, Mezentsev YV, Chuev GN, Fedotova MV, Kruchinin SE, Ivanov AS. SARS-COV-2 Coronavirus Papain-like Protease PLpro as an Antiviral Target for Inhibitors of Active Site and Protein-Protein Interactions. Biophysics (Nagoya-shi) 2023; 67:902-912. [PMID: 36883182 PMCID: PMC9984130 DOI: 10.1134/s0006350922060082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 03/06/2023] Open
Abstract
The papain-like protease PLpro of the SARS-CoV-2 coronavirus is a multifunctional enzyme that catalyzes the proteolytic processing of two viral polyproteins, pp1a and pp1ab. PLpro also cleaves peptide bonds between host cell proteins and ubiquitin (or ubiquitin-like proteins), which is associated with a violation of immune processes. Nine structures of the most effective inhibitors of the PLpro active center were prioritized according to the parameters of biochemical (IC 50) and cellular tests to assess the suppression of viral replication (EC 50) and cytotoxicity (CC 50). A literature search has shown that PLpro can interact with at least 60 potential protein partners in cells, 23 of which are targets for other viral proteins (human papillomavirus and Epstein-Barr virus). The analysis of protein-protein interactions showed that the proteins USP3, UBE2J1, RCHY1, and FAF2 involved in deubiquitinylation and ubiquitinylation processes contain the largest number of bonds with other proteins; the interaction of viral proteins with them can affect the architecture of the entire network of protein-protein interactions. Using the example of a spatial model of the PLpro/ubiquitin complex and a set of 154 naturally occurring compounds with known antiviral activity, 13 compounds (molecular masses in the range of 454-954 Da) were predicted as potential PLpro inhibitors. These compounds bind to the "hot" amino acid residues of the protease at the positions Gly163, Asp164, Arg166, Glu167, and Tyr264 involved in the interaction with ubiquitin. Thus, pharmacological effects on peripheral PLpro sites, which play important roles in binding protein substrates, may be an additional target-oriented antiviral strategy.
Collapse
Affiliation(s)
- P. V. Ershov
- Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - E. O. Yablokov
- Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | | | - G. N. Chuev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow oblast Russia
| | - M. V. Fedotova
- Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia
| | - S. E. Kruchinin
- Krestov Institute of Solution Chemistry, Russian Academy of Sciences, 153045 Ivanovo, Russia
| | - A. S. Ivanov
- Institute of Biomedical Chemistry, 119121 Moscow, Russia
| |
Collapse
|
11
|
Graziano G, Delre P, Carofiglio F, Brea J, Ligresti A, Kostrzewa M, Riganti C, Gioè-Gallo C, Majellaro M, Nicolotti O, Colabufo NA, Abate C, Loza MI, Sotelo E, Mangiatordi GF, Contino M, Stefanachi A, Leonetti F. N-adamantyl-anthranil amide derivatives: New selective ligands for the cannabinoid receptor subtype 2 (CB2R). Eur J Med Chem 2023; 248:115109. [PMID: 36657299 DOI: 10.1016/j.ejmech.2023.115109] [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: 11/30/2022] [Revised: 12/28/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Cannabinoid type 2 receptor (CB2R) is a G-protein-coupled receptor that, together with Cannabinoid type 1 receptor (CB1R), endogenous cannabinoids and enzymes responsible for their synthesis and degradation, forms the EndoCannabinoid System (ECS). In the last decade, several studies have shown that CB2R is overexpressed in activated central nervous system (CNS) microglia cells, in disorders based on an inflammatory state, such as neurodegenerative diseases, neuropathic pain, and cancer. For this reason, the anti-inflammatory and immune-modulatory potentials of CB2R ligands are emerging as a novel therapeutic approach. The design of selective ligands is however hampered by the high sequence homology of transmembrane domains of CB1R and CB2R. Based on a recent three-arm pharmacophore hypothesis and latest CB2R crystal structures, we designed, synthesized, and evaluated a series of new N-adamantyl-anthranil amide derivatives as CB2R selective ligands. Interestingly, this new class of compounds displayed a high affinity for human CB2R along with an excellent selectivity respect to CB1R. In this respect, compounds exhibiting the best pharmacodynamic profile in terms of CB2R affinity were also evaluated for the functional behavior and molecular docking simulations provided a sound rationale by highlighting the relevance of the arm 1 substitution to prompt CB2R action. Moreover, the modulation of the pro- and anti-inflammatory cytokines production was also investigated to exert the ability of the best compounds to modulate the inflammatory cascade.
Collapse
Affiliation(s)
- Giovanni Graziano
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy
| | - Pietro Delre
- CNR - Institute of Crystallography, Via Giovanni Amendola, 122/O, 70126, Bari, Italy
| | - Francesca Carofiglio
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy
| | - Josè Brea
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Av. Barcelona, 15782, Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Turin, Italy
| | - Claudia Gioè-Gallo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Orazio Nicolotti
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy
| | - Nicola Antonio Colabufo
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy
| | - Carmen Abate
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy; CNR - Institute of Crystallography, Via Giovanni Amendola, 122/O, 70126, Bari, Italy
| | - Maria Isabel Loza
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Av. Barcelona, 15782, Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | | | - Marialessandra Contino
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy.
| | - Angela Stefanachi
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy.
| | - Francesco Leonetti
- Department of Pharmacy-Pharmaceutical Sciences, University of the Studies of Bari "Aldo Moro", Via E.Orabona 4, 70125, Bari, Italy
| |
Collapse
|
12
|
Intranuovo F, Brunetti L, DelRe P, Mangiatordi GF, Stefanachi A, Laghezza A, Niso M, Leonetti F, Loiodice F, Ligresti A, Kostrzewa M, Brea J, Loza MI, Sotelo E, Saviano M, Colabufo NA, Riganti C, Abate C, Contino M. Development of N-(1-Adamantyl)benzamides as Novel Anti-Inflammatory Multitarget Agents Acting as Dual Modulators of the Cannabinoid CB2 Receptor and Fatty Acid Amide Hydrolase. J Med Chem 2023; 66:235-250. [PMID: 36542836 DOI: 10.1021/acs.jmedchem.2c01084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cannabinoid type 2 receptor (CB2R), belonging to the endocannabinoid system, is overexpressed in pathologies characterized by inflammation, and its activation counteracts inflammatory states. Fatty acid amide hydrolase (FAAH) is an enzyme responsible for the degradation of the main endocannabinoid anandamide; thus, the simultaneous CB2R activation and FAAH inhibition may be a synergistic anti-inflammatory strategy. Encouraged by principal component analysis (PCA) data identifying a wide chemical space shared by CB2R and FAAH ligands, we designed a small library of adamantyl-benzamides, as potential dual agents, CB2R agonists, and FAAH inhibitors. The new compounds were tested for their CB2R affinity/selectivity and CB2R and FAAH activity. Derivatives 13, 26, and 27, displaying the best pharmacodynamic profile as CB2R full agonists and FAAH inhibitors, decreased pro-inflammatory and increased anti-inflammatory cytokines production. Molecular docking simulations complemented the experimental findings by providing a molecular rationale behind the observed activities. These multitarget ligands constitute promising anti-inflammatory agents.
Collapse
Affiliation(s)
- Francesca Intranuovo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Leonardo Brunetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Pietro DelRe
- Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | | | - Angela Stefanachi
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Jose Brea
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Maria Isabel Loza
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Eddy Sotelo
- ComBioMed Research Group, Centro de Química Biológica y Materiales Moleculares (CIQUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Michele Saviano
- Institute of Crystallography, National Research Council of Italy, Via Vivaldi, 43, Caserta 81100, Italy
| | - Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università Degli Studi di Torino, Torino 10126, Italy
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy.,Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| |
Collapse
|
13
|
Schuurs ZP, McDonald JP, Croft LV, Richard DJ, Woodgate R, Gandhi NS. Integration of molecular modelling and in vitro studies to inhibit LexA proteolysis. Front Cell Infect Microbiol 2023; 13:1051602. [PMID: 36936756 PMCID: PMC10020695 DOI: 10.3389/fcimb.2023.1051602] [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: 09/23/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction As antibiotic resistance has become more prevalent, the social and economic impacts are increasingly pressing. Indeed, bacteria have developed the SOS response which facilitates the evolution of resistance under genotoxic stress. The transcriptional repressor, LexA, plays a key role in this response. Mutation of LexA to a non-cleavable form that prevents the induction of the SOS response sensitizes bacteria to antibiotics. Achieving the same inhibition of proteolysis with small molecules also increases antibiotic susceptibility and reduces drug resistance acquisition. The availability of multiple LexA crystal structures, and the unique Ser-119 and Lys-156 catalytic dyad in the protein enables the rational design of inhibitors. Methods We pursued a binary approach to inhibit proteolysis; we first investigated β-turn mimetics, and in the second approach we tested covalent warheads targeting the Ser-119 residue. We found that the cleavage site region (CSR) of the LexA protein is a classical Type II β-turn, and that published 1,2,3-triazole compounds mimic the β-turn. Generic covalent molecule libraries and a β-turn mimetic library were docked to the LexA C-terminal domain using molecular modelling methods in FlexX and CovDock respectively. The 133 highest-scoring molecules were screened for their ability to inhibit LexA cleavage under alkaline conditions. The top molecules were then tested using a RecA-mediated cleavage assay. Results The β-turn library screen did not produce any hit compounds that inhibited RecA-mediated cleavage. The covalent screen discovered an electrophilic serine warhead that can inhibit LexA proteolysis, reacting with Ser-119 via a nitrile moiety. Discussion This research presents a starting point for hit-to-lead optimisation, which could lead to inhibition of the SOS response and prevent the acquisition of antibiotic resistance.
Collapse
Affiliation(s)
- Zachariah P. Schuurs
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Translational Research Institute (TRI), Brisbane, QLD, Australia
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - John P. McDonald
- Laboratory of Genomic Integrity, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Laura V. Croft
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Translational Research Institute (TRI), Brisbane, QLD, Australia
| | - Derek J. Richard
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Translational Research Institute (TRI), Brisbane, QLD, Australia
| | - Roger Woodgate
- Laboratory of Genomic Integrity, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Neha S. Gandhi, ; Roger Woodgate,
| | - Neha S. Gandhi
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Translational Research Institute (TRI), Brisbane, QLD, Australia
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- *Correspondence: Neha S. Gandhi, ; Roger Woodgate,
| |
Collapse
|
14
|
Kim H, Hauner D, Laureanti JA, Agustin K, Raugei S, Kumar N. Mechanistic investigation of SARS-CoV-2 main protease to accelerate design of covalent inhibitors. Sci Rep 2022; 12:21037. [PMID: 36470873 PMCID: PMC9722715 DOI: 10.1038/s41598-022-23570-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/02/2022] [Indexed: 12/12/2022] Open
Abstract
Targeted covalent inhibition represents one possible strategy to block the function of SARS-CoV-2 Main Protease (MPRO), an enzyme that plays a critical role in the replication of the novel SARS-CoV-2. Toward the design of covalent inhibitors, we built a covalent inhibitor dataset using deep learning models followed by high throughput virtual screening of these candidates against MPRO. Two top-ranking inhibitors were selected for mechanistic investigations-one with an activated ester warhead that has a piperazine core and the other with an acrylamide warhead. Specifically, we performed a detailed analysis of the free energetics of covalent inhibition by hybrid quantum mechanics/molecular mechanics simulations. Cleavage of a fragment of the non-structured protein (NSP) from the SARS-CoV-2 genome was also simulated for reference. Simulations show that both candidates form more stable enzyme-inhibitor (E-I) complexes than the chosen NSP. It was found that both the NSP fragment and the activated ester inhibitor react with CYS145 of MPRO in a concerted manner, whereas the acrylamide inhibitor follows a stepwise mechanism. Most importantly, the reversible reaction and the subsequent hydrolysis reaction from E-I complexes are less probable when compared to the reactions with an NSP fragment, showing promise for these candidates to be the base for efficient MPRO inhibitors.
Collapse
Affiliation(s)
- Hoshin Kim
- grid.451303.00000 0001 2218 3491Physical and Computational Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Darin Hauner
- grid.451303.00000 0001 2218 3491Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Joseph A. Laureanti
- grid.451303.00000 0001 2218 3491Physical and Computational Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Kruel Agustin
- grid.451303.00000 0001 2218 3491Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Simone Raugei
- grid.451303.00000 0001 2218 3491Physical and Computational Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Neeraj Kumar
- grid.451303.00000 0001 2218 3491Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| |
Collapse
|
15
|
Rieder AS, Deniz BF, Netto CA, Wyse ATS. A Review of In Silico Research, SARS-CoV-2, and Neurodegeneration: Focus on Papain-Like Protease. Neurotox Res 2022; 40:1553-1569. [PMID: 35917086 PMCID: PMC9343570 DOI: 10.1007/s12640-022-00542-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 01/18/2023]
Abstract
Since the appearance of SARS-CoV-2 and the COVID-19 pandemic, the search for new approaches to treat this disease took place in the scientific community. The in silico approach has gained importance at this moment, once the methodologies used in this kind of study allow for the identification of specific protein-ligand interactions, which may serve as a filter step for molecules that can act as specific inhibitors. In addition, it is a low-cost and high-speed technology. Molecular docking has been widely used to find potential viral protein inhibitors for structural and non-structural proteins of the SARS-CoV-2, aiming to block the infection and the virus multiplication. The papain-like protease (PLpro) participates in the proteolytic processing of SARS-CoV-2 and composes one of the main targets studied for pharmacological intervention by in silico methodologies. Based on that, we performed a systematic review about PLpro inhibitors from the perspective of in silico research, including possible therapeutic molecules in relation to this viral protein. The neurological problems triggered by COVID-19 were also briefly discussed, especially relative to the similarities of neuroinflammation present in Alzheimer's disease. In this context, we focused on two molecules, curcumin and glycyrrhizinic acid, given their PLpro inhibitory actions and neuroprotective properties and potential therapeutic effects on COVID-19.
Collapse
Affiliation(s)
- Alessandra S Rieder
- Laboratory of Neuroprotection and Neurometabolic Diseases, Wyse's Lab, Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Bruna F Deniz
- Laboratory of Neuroprotection and Neurometabolic Diseases, Wyse's Lab, Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Carlos Alexandre Netto
- Laboratory of Neuroprotection and Neurometabolic Diseases, Wyse's Lab, Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Neurometabolic Diseases, Wyse's Lab, Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
16
|
Negru PA, Miculas DC, Behl T, Bungau AF, Marin RC, Bungau SG. Virtual screening of substances used in the treatment of SARS-CoV-2 infection and analysis of compounds with known action on structurally similar proteins from other viruses. Biomed Pharmacother 2022; 153:113432. [PMID: 36076487 PMCID: PMC9289048 DOI: 10.1016/j.biopha.2022.113432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is considered the etiological agent of the disease that caused the COVID-19 pandemic, and for which there is currently no effective treatment. This pandemic has shown that the rapid identification of therapeutic compounds is critical (when a new virus with high transmissibility occurs) to prevent or reduce as much as possible the loss of human lives. To meet the urgent need for drugs, many strategies were applied for the discovery, respectively the identification of potential therapies / drugs for SARS-CoV-2. Molecular docking and virtual screening are two of the in silico tools/techniques that provided the identification of few SARS-CoV-2 inhibitors, removing ineffective or less effective drugs and thus preventing the loss of resources such as time and additional costs. The main target of this review is to provide a comprehensive overview of how in-silico tools have been used in the crisis management of anti-SARS-CoV-2 drugs, especially in virtual screening of substances used in the treatment of SARS-CoV-2 infection and analysis of compounds with known action on structurally similar proteins from other viruses; also, completions were added to the way in which these methods came to meet the requirements of biomedical research in the field. Moreover, the importance and impact of the topic approached for researchers was highlighted by conducting an extensive bibliometric analysis.
Collapse
Affiliation(s)
- Paul Andrei Negru
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Denisa Claudia Miculas
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Alexa Florina Bungau
- Medicine Programm of Study, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Ruxandra-Cristina Marin
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
17
|
Alvarado YJ, Olivarez Y, Lossada C, Vera-Villalobos J, Paz JL, Vera E, Loroño M, Vivas A, Torres FJ, Jeffreys LN, Hurtado-León ML, González-Paz L. Interaction of the new inhibitor paxlovid (PF-07321332) and ivermectin with the monomer of the main protease SARS-CoV-2: A volumetric study based on molecular dynamics, elastic networks, classical thermodynamics and SPT. Comput Biol Chem 2022; 99:107692. [PMID: 35640480 PMCID: PMC9107165 DOI: 10.1016/j.compbiolchem.2022.107692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023]
Abstract
The COVID-19 pandemic has accelerated the study of drugs, most notably ivermectin and more recently Paxlovid (PF-07321332) which is in phase III clinical trials with experimental data showing covalent binding to the viral protease Mpro. Theoretical developments of catalytic site-directed docking support thermodynamically feasible non-covalent binding to Mpro. Here we show that Paxlovid binds non-covalently at regions other than the catalytic sites with energies stronger than reported and at the same binding site as the ivermectin B1a homologue, all through theoretical methodologies, including blind docking. We volumetrically characterize the non-covalent interaction of the ivermectin homologues (avermectins B1a and B1b) and Paxlovid with the mMpro monomer, through molecular dynamics and scaled particle theory (SPT). Using the fluctuation-dissipation theorem (FDT), we estimated the electric dipole moment fluctuations at the surface of each of complex involved in this study, with similar trends to that observed in the interaction volume. Using fluctuations of the intrinsic volume and the number of flexible fragments of proteins using anisotropic and Gaussian elastic networks (ANM+GNM) suggests the complexes with ivermectin are more dynamic and flexible than the unbound monomer. In contrast, the binding of Paxlovid to mMpro shows that the mMpro-PF complex is the least structurally dynamic of all the species measured in this investigation. The results support a differential molecular mechanism of the ivermectin and PF homologues in the mMpro monomer. Finally, the results showed that Paxlovid despite beingbound in different sites through covalent or non-covalent forms behaves similarly in terms of its structural flexibility and volumetric behaviour.
Collapse
Affiliation(s)
- Ysaias José Alvarado
- Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Investigación y Tecnología de Materiales (CITeMA), Laboratorio de Caracterización Molecular y Biomolecular, 4001 Maracaibo, Bolivarian Republic of Venezuela.
| | - Yosmari Olivarez
- Universidad del Zulia (LUZ). Facultad Experimental de Ciencias (FEC), Departamento de Quimica, Laboratorio de Electronica Molecular, 4001 Maracaibo, Bolivarian Republic of Venezuela
| | - Carla Lossada
- Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Investigación y Tecnología de Materiales (CITeMA), Laboratorio de Caracterización Molecular y Biomolecular, 4001 Maracaibo, Bolivarian Republic of Venezuela
| | - Joan Vera-Villalobos
- Facultad de Ciencias Naturales y Matemáticas, Departamento de Química y Ciencias Ambientales, Laboratorio de Análisis Químico Instrumental (LAQUINS), Escuela Superior Politécnica del Litoral, Guayaquil, Ecuador
| | - José Luis Paz
- Departamento Académico de Química Inorgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Eddy Vera
- Universidad del Zulia (LUZ). Facultad Experimental de Ciencias (FEC), Departamento de Quimica, Laboratorio de Electronica Molecular, 4001 Maracaibo, Bolivarian Republic of Venezuela
| | - Marcos Loroño
- Departamento Académico de Química Analítica e Instrumental, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Alejandro Vivas
- Universidad del Zulia (LUZ). Facultad Experimental de Ciencias (FEC), Departamento de Quimica, Laboratorio de Electronica Molecular, 4001 Maracaibo, Bolivarian Republic of Venezuela
| | - Fernando Javier Torres
- Grupo de Química Computacional y Teórica (QCT-UR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia; Grupo de Química Computacional y Teórica (QCT-USFQ), Instituto de Simulación Computacional (ISC-USFQ), Departamento de Ingeniería Química, Universidad San Francisco de Quito (USFQ), Quito, Ecuador
| | - Laura N Jeffreys
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - María Laura Hurtado-León
- Universidad del Zulia (LUZ), Facultad Experimental de Ciencias (FEC), Departamento de Biología, Laboratorio de Genética y Biología Molecular (LGBM), Maracaibo 4001, Zulia, Bolivarian Republic of Venezuela
| | - Lenin González-Paz
- Universidad del Zulia (LUZ), Facultad Experimental de Ciencias (FEC), Departamento de Biología, Laboratorio de Genética y Biología Molecular (LGBM), Maracaibo 4001, Zulia, Bolivarian Republic of Venezuela; Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Estudios Botanicos y Agroforestales, (CEBA), Laboratorio de Proteccion Vegetal, 4001 Maracaibo, Bolivarian Republic of Venezuela.
| |
Collapse
|
18
|
Ton AT, Pandey M, Smith JR, Ban F, Fernandez M, Cherkasov A. Targeting SARS-CoV-2 Papain-Like Protease in the Post-Vaccine Era. Trends Pharmacol Sci 2022; 43:906-919. [PMID: 36114026 PMCID: PMC9399131 DOI: 10.1016/j.tips.2022.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022]
Abstract
While vaccines remain at the forefront of global healthcare responses, pioneering therapeutics against SARS-CoV-2 are expected to fill the gaps for waning immunity. Rapid development and approval of orally available direct-acting antivirals targeting crucial SARS-CoV-2 proteins marked the beginning of the era of small-molecule drugs for COVID-19. In that regard, the papain-like protease (PLpro) can be considered a major SARS-CoV-2 therapeutic target due to its dual biological role in suppressing host innate immune responses and in ensuring viral replication. Here, we summarize the challenges of targeting PLpro and innovative early-stage PLpro-specific small molecules. We propose that state-of-the-art computer-aided drug design (CADD) methodologies will play a critical role in the discovery of PLpro compounds as a novel class of COVID-19 drugs.
Collapse
Affiliation(s)
- Anh-Tien Ton
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Mohit Pandey
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Jason R Smith
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada; Department of Chemistry, Simon Fraser University, Burnaby, Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Michael Fernandez
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
19
|
Daoui O, Elkhattabi S, Chtita S. Rational identification of small molecules derived from 9,10-dihydrophenanthrene as potential inhibitors of 3CL pro enzyme for COVID-19 therapy: a computer-aided drug design approach. Struct Chem 2022; 33:1667-1690. [PMID: 35818588 PMCID: PMC9261181 DOI: 10.1007/s11224-022-02004-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/23/2022] [Indexed: 01/11/2023]
Abstract
Small molecules such as 9,10-dihydrophenanthrene derivatives have remarkable activity toward inhibition of SARS-CoV-2 3CLpro and COVID-19 proliferation, which show a strong correlation between their structures and bioactivity. Therefore, these small compounds could be suitable for clinical pharmaceutical use against COVID-19. The objective of this study was to remodel the structures of 9,10-dihydrophenanthrene derivatives to achieve a powerful biological activity against 3CLpro and favorable pharmacokinetic properties for drug design and discovery. Therefore, by the use of bioinformatics techniques, we developed robust 3D-QSAR models that are capable of describing the structure-activity relationship for 46 molecules based on 9,10-dihydrophenanthrene derivatives using CoMFA/SE (R 2 = 0.97, Q 2 = 0.81, R 2 pred = 0.95, c R 2 p = 0.71) and CoMSIA/SEHDA (R 2 = 0.94, Q 2 = 0.76, R 2 pred = 0.91, c R 2 p = 0.65) techniques. Accordingly, 96 lead compounds were generated based on a template molecule that showed the highest observed activity in vitro (T40, pIC50 = 5.81) and predicted their activities and bioavailability in silico. The rational screening outputs of 3D-QSAR, Molecular docking, ADMET, and MM-GBSA led to the identification of 9 novel modeled molecules as potent noncovalent drugs against SARS-CoV-2-3CLpro. Finally, by molecular dynamics simulations, the stability and structural dynamics of 3CLpro free and complex (PDB code: 6LU7) were discussed in the presence of samples of 9,10-dihydrophenanthrene derivative in an aqueous environment. Overall, the retrosynthesis of the proposed drug compounds in this study and the evaluation of their bioactivity in vitro and in vivo may be interesting for designing and discovering a new drug effective against COVID-19. Supplementary Information The online version contains supplementary material available at 10.1007/s11224-022-02004-z.
Collapse
Affiliation(s)
- Ossama Daoui
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, BP Box 72, Fez, Morocco
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, BP Box 72, Fez, Morocco
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M’Sik, Hassan II University of Casablanca, B.P 7955 Casablanca, Morocco
| |
Collapse
|
20
|
Development of a database of RNA helicase inhibitors (VHIMDB) of pathogenic viruses and in silico screening for the potential drug molecules. THE EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The pathogenic RNA virus that infects human beings contains the RNA helicase enzyme, responsible for the replication of the viral genome. The enzyme is used as a suitable target against which the drug molecule acts. Therefore, the identification and proposal the novel compounds that can be targeted toward the helicase enzymes to stop the functioning of the enzyme is desirable. Although many viral helicase inhibitor molecules have been identified, still yet no unique database is available for these compounds. This research work envisages developing a curated database of RNA helicase inhibitors. The database contains in total of 353 entries that are computationally predicted and experimentally verified RNA helicase inhibitors. The database contains information like compound name, chemical properties, chemical format, and name of the target virus to which it acts against it with a user-friendly menu-driven search engine. Presently, the database is freely available at: https://vhimdb.rsatpathy.in/. Further, in silico screening of the whole database by drug-likeness and toxicity resulted in 14 potential drug molecules. The selected molecules were analyzed for their effectiveness in binding by using molecular docking score and interaction with the helicase enzymes of three categories of pathogenic viruses (SARS-CoV-2, SARS-CoV, and MERS-CoV).
Collapse
|
21
|
Bhowmick S, AlFaris NA, Zaidan ALTamimi J, ALOthman ZA, Patil PC, Aldayel TS, Wabaidur SM, Saha A. Identification of bio-active food compounds as potential SARS-CoV-2 PLpro inhibitors-modulators via negative image-based screening and computational simulations. Comput Biol Med 2022; 145:105474. [PMID: 35395517 PMCID: PMC8973019 DOI: 10.1016/j.compbiomed.2022.105474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022]
Abstract
Despite significant studies on the COVID-19 pandemic, scientists around the world are still battling to find a definitive therapy against the ongoing severe global health crisis. In this study, advanced computational approaches have been employed to identify bioactive food constituents as potential SARS-CoV-2 PLpro inhibitors-modulators. As a validated antiviral drug target, PLpro has gained tremendous attention for therapeutics developments. Therefore, targeting the multifunctional SARS-CoV-2 PLpro protein, ∼1039 bioactive dietary compounds have been screened extensively through novel techniques like negative image-based (NIB) screening and molecular docking approaches. In particular, the three different models of NIB screening have been generated and used to re-score the dietary compounds based on the negative image which is created by reversing the shape and electrostatics features of PLpro protein's ligand-binding cavity. Further, 100 ns molecular dynamics simulation has been performed and MM-GBSA based binding free energies have been estimated for the final proposed four dietary compounds (PC000550, PC000361, PC000558, and PC000573) as potential inhibitors/modulators of SARS-CoV-2 PLpro protein. Employed computational study outcome also has been compared with respect to the earlier experimentally investigated compound GRL0617 against SARS-CoV-2 PLpro protein, which suggests much greater interaction potential in terms of binding affinity and other energetic contributions for the proposed dietary compounds. Hence, the present study suggests that proposed dietary compounds can be suitable chemical entities for modulating the activity of PLpro protein or can be further utilized for optimizing or screening of novel chemical surrogates, however also needs experimental evaluation for entry in clinical studies for better assessment.
Collapse
Affiliation(s)
- Shovonlal Bhowmick
- Department of Chemical Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, India
| | - Nora Abdullah AlFaris
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia,Corresponding author
| | - Jozaa Zaidan ALTamimi
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Zeid A. ALOthman
- Chemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Pritee Chunarkar Patil
- Department of Bioinformatics, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth Deemed University, Pune-Satara Road, Pune, India
| | - Tahany Saleh Aldayel
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, India,Corresponding author
| |
Collapse
|
22
|
Kulandaisamy R, Kushwaha T, Dalal A, Kumar V, Singh D, Baswal K, Sharma P, Praneeth K, Jorwal P, Kayampeta SR, Sharma T, Maddur S, Kumar M, Kumar S, Polamarasetty A, Singh A, Sehgal D, Gholap SL, Appaiahgari MB, Katika MR, Inampudi KK. Repurposing of FDA Approved Drugs Against SARS-CoV-2 Papain-Like Protease: Computational, Biochemical, and in vitro Studies. Front Microbiol 2022; 13:877813. [PMID: 35620103 PMCID: PMC9127501 DOI: 10.3389/fmicb.2022.877813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
The pandemic caused by SARS-CoV-2 (SCoV-2) has impacted the world in many ways and the virus continues to evolve and produce novel variants with the ability to cause frequent global outbreaks. Although the advent of the vaccines abated the global burden, they were not effective against all the variants of SCoV-2. This trend warrants shifting the focus on the development of small molecules targeting the crucial proteins of the viral replication machinery as effective therapeutic solutions. The PLpro is a crucial enzyme having multiple roles during the viral life cycle and is a well-established drug target. In this study, we identified 12 potential inhibitors of PLpro through virtual screening of the FDA-approved drug library. Docking and molecular dynamics simulation studies suggested that these molecules bind to the PLpro through multiple interactions. Further, IC50 values obtained from enzyme-inhibition assays affirm the stronger affinities of the identified molecules for the PLpro. Also, we demonstrated high structural conservation in the catalytic site of PLpro between SCoV-2 and Human Coronavirus 229E (HCoV-229E) through molecular modelling studies. Based on these similarities in PLpro structures and the resemblance in various signalling pathways for the two viruses, we propose that HCoV-229E is a suitable surrogate for SCoV-2 in drug-discovery studies. Validating our hypothesis, Mefloquine, which was effective against HCoV-229E, was found to be effective against SCoV-2 as well in cell-based assays. Overall, the present study demonstrated Mefloquine as a potential inhibitor of SCoV-2 PLpro and its antiviral activity against SCoV-2. Corroborating our findings, based on the in vitro virus inhibition assays, a recent study reported a prophylactic role for Mefloquine against SCoV-2. Accordingly, Mefloquine may further be investigated for its potential as a drug candidate for the treatment of COVID.
Collapse
Affiliation(s)
| | - Tushar Kushwaha
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Anu Dalal
- Department of Chemistry, Indian Institute of Technology-Delhi, New Delhi, India
| | - Vikas Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Deepa Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Kamal Baswal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Pratibha Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Kokkula Praneeth
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Pankaj Jorwal
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sarala R Kayampeta
- Research and Development Division, Srikara Biologicals Private Limited, Tirupati, India
| | - Tamanna Sharma
- Central Research Laboratory Mobile Virology Research and Development BSL3 Lab, Employees' State Insurance Corporation Medical College and Hospital, Hyderabad, India
| | - Srinivas Maddur
- Central Research Laboratory Mobile Virology Research and Development BSL3 Lab, Employees' State Insurance Corporation Medical College and Hospital, Hyderabad, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Aparoy Polamarasetty
- Faculty of Biology, Indian Institute of Petroleum and Energy, Visakhapatnam, India
| | - Aekagra Singh
- Virology Lab, Department of Life Sciences, Shiv Nadar University, Greater Noida, India
| | - Deepak Sehgal
- Virology Lab, Department of Life Sciences, Shiv Nadar University, Greater Noida, India
| | - Shivajirao L Gholap
- Department of Chemistry, Indian Institute of Technology-Delhi, New Delhi, India
| | - Mohan B Appaiahgari
- Research and Development Division, Srikara Biologicals Private Limited, Tirupati, India
| | - Madhumohan R Katika
- Central Research Laboratory Mobile Virology Research and Development BSL3 Lab, Employees' State Insurance Corporation Medical College and Hospital, Hyderabad, India.,Stem Cell Facility and Regenerative Medicine, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - Krishna K Inampudi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
23
|
Hognon C, Marazzi M, García-Iriepa C. Atomistic-Level Description of the Covalent Inhibition of SARS-CoV-2 Papain-like Protease. Int J Mol Sci 2022; 23:5855. [PMID: 35628665 PMCID: PMC9143025 DOI: 10.3390/ijms23105855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/13/2022] [Accepted: 05/21/2022] [Indexed: 12/25/2022] Open
Abstract
Inhibition of the papain-like protease (PLpro) of SARS-CoV-2 has been demonstrated to be a successful target to prevent the spreading of the coronavirus in the infected body. In this regard, covalent inhibitors, such as the recently proposed VIR251 ligand, can irreversibly inactivate PLpro by forming a covalent bond with a specific residue of the catalytic site (Cys111), through a Michael addition reaction. An inhibition mechanism can therefore be proposed, including four steps: (i) ligand entry into the protease pocket; (ii) Cys111 deprotonation of the thiol group by a Brønsted-Lowry base; (iii) Cys111-S- addition to the ligand; and (iv) proton transfer from the protonated base to the covalently bound ligand. Evaluating the energetics and PLpro conformational changes at each of these steps could aid the design of more efficient and selective covalent inhibitors. For this aim, we have studied by means of MD simulations and QM/MM calculations the whole mechanism. Regarding the first step, we show that the inhibitor entry in the PLpro pocket is thermodynamically favorable only when considering the neutral Cys111, that is, prior to the Cys111 deprotonation. For the second step, MD simulations revealed that His272 would deprotonate Cys111 after overcoming an energy barrier of ca. 32 kcal/mol (at the QM/MM level), but implying a decrease of the inhibitor stability inside the protease pocket. This information points to a reversible Cys111 deprotonation, whose equilibrium is largely shifted toward the neutral Cys111 form. Although thermodynamically disfavored, if Cys111 is deprotonated in close proximity to the vinylic carbon of the ligand, then covalent binding takes place in an irreversible way (third step) to form the enolate intermediate. Finally, due to Cys111-S- negative charge redistribution over the bound ligand, proton transfer from the initially protonated His272 is favored, finally leading to an irreversibly modified Cys111 and a restored His272. These results elucidate the selectivity of Cys111 to enable formation of a covalent bond, even if a weak proton acceptor is available, as His272.
Collapse
Affiliation(s)
- Cécilia Hognon
- Grupo de Reactividad y Estructura Molecular (RESMOL), Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Alcalá de Henares, 28801 Madrid, Spain;
| | - Marco Marazzi
- Grupo de Reactividad y Estructura Molecular (RESMOL), Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Alcalá de Henares, 28801 Madrid, Spain;
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, Alcalá de Henares, 28801 Madrid, Spain
| | - Cristina García-Iriepa
- Grupo de Reactividad y Estructura Molecular (RESMOL), Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Alcalá de Henares, 28801 Madrid, Spain;
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, Alcalá de Henares, 28801 Madrid, Spain
| |
Collapse
|
24
|
Mahmoudi S, Dehkordi MM, Asgarshamsi MH. The effect of various compounds on the COVID mechanisms, from chemical to molecular aspects. Biophys Chem 2022; 288:106824. [PMID: 35728510 PMCID: PMC9095071 DOI: 10.1016/j.bpc.2022.106824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/13/2022] [Accepted: 05/07/2022] [Indexed: 02/07/2023]
Abstract
The novel coronavirus that caused COVID-19 pandemic is SARS-CoV-2. Although various vaccines are currently being used to prevent the disease's severe consequences, there is still a need for medications for those who become infected. The SARS-CoV-2 has a variety of proteins that have been studied extensively since the virus's advent. In this review article, we looked at chemical to molecular aspects of the various structures studied that have pharmaceutical activity and attempted to find a link between drug activity and compound structure. For example, designing of the compounds which bind to the allosteric site and modify hydrogen bonds or the salt bridges can disrupt SARS-CoV2 RBD–ACE2 complex. It seems that quaternary ammonium moiety and quinolin-1-ium structure could act as a negative allosteric modulator to reduce the tendency between spike-ACE2. Pharmaceutical structures with amino heads and hydrophobic tails can block envelope protein to prevent making mature SARS-CoV-2. Also, structures based on naphthalene pharmacophores or isosteres can form a strong bond with the PLpro and form a π-π and the Mpro's active site can be occupied by octapeptide compounds or linear compounds with a similar fitting ability to octapeptide compounds. And for protein RdRp, it is critical to consider pH and pKa so that pKa regulation of compounds to comply with patients is very effective, thus, the presence of tetrazole, phenylpyrazole groups, and analogs of pyrophosphate in the designed drugs increase the likelihood of the RdRp active site inhibition. Finally, it can be deduced that designing hybrid drug molecules along with considering the aforementioned characteristics would be a suitable approach for developing medicines in order to accurate targeting and complete inhibition this virus.
Collapse
Affiliation(s)
- Samira Mahmoudi
- Department of Microbial Biotechnology, School of Biological Sciences, Islamic Azad University Tehran North Branch, Tehran, Iran.
| | - Mehrdad Mohammadpour Dehkordi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Hossein Asgarshamsi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
25
|
Tinivella A, Pinzi L, Gambacorta G, Baxendale I, Rastelli G. Identification of potential biological targets of oxindole scaffolds via in silico repositioning strategies. F1000Res 2022; 11:Chem Inf Sci-217. [PMID: 37767081 PMCID: PMC10521104 DOI: 10.12688/f1000research.109017.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 09/29/2023] Open
Abstract
Background: Drug repurposing is an alternative strategy to traditional drug discovery that aims at predicting new uses for already existing drugs or clinical candidates. Drug repurposing has many advantages over traditional drug development, such as reduced attrition rates, time and costs. This is especially the case considering that most drugs investigated for repurposing have already been assessed for their safety in clinical trials. Repurposing campaigns can also be designed for libraries of already synthesized molecules at different levels of biological experimentation, from null to in vitro and in vivo. Such an extension of the "repurposing" concept is expected to provide significant advantages for the identification of novel drugs, as the synthetic accessibility of the desired compounds is often one of the limiting factors in the traditional drug discovery pipeline. Methods: In this work, we performed a computational repurposing campaign on a library of previously synthesized oxindole-based compounds, in order to identify potential new targets for this versatile scaffold. To this aim, ligand-based approaches were firstly applied to evaluate the similarity degree of the investigated compound library, with respect to ligands extracted from the DrugBank, Protein Data Bank (PDB) and ChEMBL databases. In particular, the 2D fingerprint-based and 3D shape-based similarity profiles were evaluated and compared for the oxindole derivates. Results: The analyses predicted a set of potential candidate targets for repurposing, some of them emerging by consensus of different computational analyses. One of the identified targets, i.e., the vascular endothelial growth factor receptor 2 (VEGFR-2) kinase, was further investigated by means of docking calculations, followed by biological testing of one candidate. Conclusions: While the compound did not show potent inhibitory activity towards VEGFR-2, the study highlighted several other possibilities of therapeutically relevant targets that may be worth of consideration for drug repurposing.
Collapse
Affiliation(s)
- Annachiara Tinivella
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Ian Baxendale
- Department of Chemistry, University of Durham, Durham, UK
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
26
|
Jiang H, Yang P, Zhang J. Potential Inhibitors Targeting Papain-Like Protease of SARS-CoV-2: Two Birds With One Stone. Front Chem 2022; 10:822785. [PMID: 35281561 PMCID: PMC8905519 DOI: 10.3389/fchem.2022.822785] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/28/2022] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2), the pathogen of the Coronavirus disease-19 (COVID-19), is still devastating the world causing significant chaos to the international community and posing a significant threat to global health. Since the first outbreak in late 2019, several lines of intervention have been developed to prevent the spread of this virus. Nowadays, some vaccines have been approved and extensively administered. However, the fact that SARS-CoV-2 rapidly mutates makes the efficacy and safety of this approach constantly under debate. Therefore, antivirals are still needed to combat the infection of SARS-CoV-2. Papain-like protease (PLpro) of SARS-CoV-2 supports viral reproduction and suppresses the innate immune response of the host, which makes PLpro an attractive pharmaceutical target. Inhibition of PLpro could not only prevent viral replication but also restore the antiviral immunity of the host, resulting in the speedy recovery of the patient. In this review, we describe structural and functional features on PLpro of SARS-CoV-2 and the latest development in searching for PLpro inhibitors. Currently available inhibitors targeting PLpro as well as their structural basis are also summarized.
Collapse
Affiliation(s)
- Haihai Jiang
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
- *Correspondence: Haihai Jiang, ; Jin Zhang,
| | - Peiyao Yang
- Queen Mary School, Nanchang University, Nanchang, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
- *Correspondence: Haihai Jiang, ; Jin Zhang,
| |
Collapse
|
27
|
Liang JJ, Pitsillou E, Ververis K, Guallar V, Hung A, Karagiannis TC. Investigation of small molecule inhibitors of the SARS-CoV-2 papain-like protease by all-atom microsecond modelling, PELE Monte Carlo simulations, and in vitro activity inhibition. Chem Phys Lett 2022; 788:139294. [PMID: 34961797 PMCID: PMC8693950 DOI: 10.1016/j.cplett.2021.139294] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 12/16/2022]
Abstract
The SARS-CoV-2 papain-like (PLpro) protease is essential for viral replication. We investigated potential antiviral effects of hypericin relative to the well-known noncovalent PLpro inhibitor GRL-0617. Molecular dynamics and PELE Monte Carlo simulations highlight favourable binding of hypericin and GRL-0617 to the naphthalene binding pocket of PLpro. Although not potent as GRL-0617 (45.8 vs 1.6 µM for protease activity, respectively), in vitro fluorogenic enzymatic assays with hypericin show concentration-dependent inhibition of both PLpro protease and deubiquitinating activities. Given its use in supplementations and the FDA conditional approval of a synthetic version, further evaluation of hypericin as a potential SARS-CoV-2 antiviral is warranted.
Collapse
Affiliation(s)
- Julia J. Liang
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia,School of Science, STEM College, RMIT University, VIC 3001, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia,School of Science, STEM College, RMIT University, VIC 3001, Australia
| | - Katherine Ververis
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Victor Guallar
- Barcelona Supercomputing Center, Jordi Girona 29, E-08034 Barcelona, Spain,ICREA, Passeig Lluís Companys 23, E-08010 Barcelona, Spain
| | - Andrew Hung
- School of Science, STEM College, RMIT University, VIC 3001, Australia
| | - Tom C. Karagiannis
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3052, Australia,Corresponding author at: Head Epigenomic Medicine Program, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
28
|
Kandeel M, Kim J, Fayez M, Kitade Y, Kwon HJ. Antiviral drug discovery by targeting the SARS-CoV-2 polyprotein processing by inhibition of the main protease. PeerJ 2022; 10:e12929. [PMID: 35186496 PMCID: PMC8833224 DOI: 10.7717/peerj.12929] [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: 09/16/2021] [Accepted: 01/21/2022] [Indexed: 01/11/2023] Open
Abstract
The spread of SARS-CoV-2, the causative agent for COVID-19, has led to a global and deadly pandemic. To date, few drugs have been approved for treating SARS-CoV-2 infections. In this study, a structure-based approach was adopted using the SARS-CoV-2 main protease (Mpro) and a carefully selected dataset of 37,060 compounds comprising Mpro and antiviral protein-specific libraries. The compounds passed two-step docking filtration, starting with standard precision (SP) followed by extra precision (XP) runs. Fourteen compounds with the highest XP docking scores were examined by 20 ns molecular dynamics simulations (MDs). Based on backbone route mean square deviations (RMSD) and molecular mechanics/generalized Born surface area (MM/GBSA) binding energy, four drugs were selected for comprehensive MDs analysis at 100 ns. Results indicated that birinapant, atazanavir, and ritonavir potently bound and stabilized SARS-CoV-2 Mpro structure. Binding energies higher than -102 kcal/mol, RMSD values <0.22 nm, formation of several hydrogen bonds with Mpro, favourable electrostatic contributions, and low radii of gyration were among the estimated factors contributing to the strength of the binding of these three compounds with Mpro. The top two compounds, atazanavir and birinapant, were tested for their ability to prevent SARS-CoV-2 plaque formation. At 10 µM of birinapant concentration, antiviral tests against SARS-CoV-2 demonstrated a 37% reduction of virus multiplication. Antiviral assays demonstrated that birinapant has high anti-SARS-CoV-2 activity in the low micromolar range, with an IC50 value of 18 ± 3.6 µM. Therefore, birinapant is a candidate for further investigation to determine whether it is a feasible therapy option.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-hofuf, Al-ahsa, Saudi Arabia,Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh, Egypt
| | - Jinsoo Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Mahmoud Fayez
- Al-Ahsa Veterinary Diagnostic Laboratory, Ministry of Agriculture, Al-Ahsa, Saudi Arabia,Veterinary Serum and Vaccine Institute, Cairo, Dokki, Egypt
| | - Yukio Kitade
- Department of Applied Chemistry, Faculty of Engineering, Aichi Institute of Technology, Toyota, Japan
| | - Hyung-Joo Kwon
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
29
|
Sencanski M, Perovic V, Milicevic J, Todorovic T, Prodanovic R, Veljkovic V, Paessler S, Glisic S. Identification of SARS-CoV-2 Papain-like Protease (PLpro) Inhibitors Using Combined Computational Approach. ChemistryOpen 2022; 11:e202100248. [PMID: 35103413 PMCID: PMC8805381 DOI: 10.1002/open.202100248] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/13/2022] [Indexed: 11/25/2022] Open
Abstract
In the current pandemic, finding an effective drug to prevent or treat the infection is the highest priority. A rapid and safe approach to counteract COVID-19 is in silico drug repurposing. The SARS-CoV-2 PLpro promotes viral replication and modulates the host immune system, resulting in inhibition of the host antiviral innate immune response, and therefore is an attractive drug target. In this study, we used a combined in silico virtual screening for candidates for SARS-CoV-2 PLpro protease inhibitors. We used the Informational spectrum method applied for Small Molecules for searching the Drugbank database followed by molecular docking. After in silico screening of drug space, we identified 44 drugs as potential SARS-CoV-2 PLpro inhibitors that we propose for further experimental testing.
Collapse
Affiliation(s)
- Milan Sencanski
- National Institute of the Republic of SerbiaUniversity of BelgradeMike Petrovica Alasa 12–1411000BelgradeSerbia
| | - Vladimir Perovic
- National Institute of the Republic of SerbiaUniversity of BelgradeMike Petrovica Alasa 12–1411000BelgradeSerbia
| | - Jelena Milicevic
- National Institute of the Republic of SerbiaUniversity of BelgradeMike Petrovica Alasa 12–1411000BelgradeSerbia
| | - Tamara Todorovic
- Faculty of ChemistryUniversity of BelgradeStudentski Trg 12–1611000BelgradeSerbia
| | - Radivoje Prodanovic
- Faculty of ChemistryUniversity of BelgradeStudentski Trg 12–1611000BelgradeSerbia
| | | | - Slobodan Paessler
- Department of PathologyUniversity of Texas Medical BranchGalvestonTX 77550USA
- Institute for Human Infections and ImmunityUniversity of Texas Medical BranchGalvestonTX 77555USA
| | - Sanja Glisic
- National Institute of the Republic of SerbiaUniversity of BelgradeMike Petrovica Alasa 12–1411000BelgradeSerbia
| |
Collapse
|
30
|
Bhowmick S, Saha A, AlFaris NA, ALTamimi JZ, ALOthman ZA, Aldayel TS, Wabaidur SM, Islam MA. Identification of potent food constituents as SARS-CoV-2 papain-like protease modulators through advanced pharmacoinformatics approaches. J Mol Graph Model 2021; 111:108113. [PMID: 34959151 PMCID: PMC8688376 DOI: 10.1016/j.jmgm.2021.108113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/05/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022]
Abstract
The current ongoing pandemic of COVID-19 urges immediate treatment measures for controlling the highly contagious SARS-CoV-2 infections. The papain-like protease (PLpro), which is released from nsp3, is presently being evaluated as a significant anti-viral drug target for COVID-19 therapy development. Particularly, PLpro is implicated in the cleavage of viral polyproteins and antagonizes the host innate immune response through its deubiquitinating and deISGylating actions, thus making it a high-profile antiviral therapeutic target. The present study reports a few specific food compounds that can bind tightly with the SARS-CoV-2 PLpro protein identified through extensive computational screening techniques. Precisely, extensive advanced computational approaches combining target-based virtual screening, particularly employing sub-structure based similarity search, molecular docking, molecular dynamics (MD) simulations, and MM-GBSA based binding free energy calculations have been employed for the identification of the most promising food compounds with substantial functional implications as SARS-CoV-2 PLpro protein inhibitors/modulators. Observations from the present research investigation also provide a deeper understanding of the binding modes of the proposed four food compounds with SARS-CoV-2 PLpro protein. In docking analyses, all compounds have established essential inter-molecular interaction profiles at the active site cavity of the SARS-CoV-2 PLpro protein. Similarly, the long-range 100 ns conventional MD simulation studies also provided an in-depth understanding of probable interactions and dynamic behaviour of the SARS-CoV-2 PLpro protein-food compound complexes. Binding free energies of all molecular systems revealed a strong interaction affinity of food compounds towards the SARS-CoV-2 PLpro protein. Moreover, clear-cut comparative analyses against the known standard inhibitor also suggest that proposed food compounds may act as potential active chemical entities for modulating the action of the SARS-CoV-2 PLpro protein.
Collapse
Affiliation(s)
- Shovonlal Bhowmick
- Department of Chemical Technology, University of Calcutta, 92, A.P.C. Road, Kolkata, 700009, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92, A.P.C. Road, Kolkata, 700009, India.
| | - Nora Abdullah AlFaris
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, P. O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Jozaa Zaidan ALTamimi
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, P. O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Zeid A ALOthman
- Department of Chemistry, P.O. Box 2455, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Tahany Saleh Aldayel
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah bint Abdulrahman University, P. O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Saikh Mohammad Wabaidur
- Department of Chemistry, P.O. Box 2455, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Md Ataul Islam
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom.
| |
Collapse
|
31
|
Szabó PB, Sabanés Zariquiey F, Nogueira JJ. Cosolvent and Dynamic Effects in Binding Pocket Search by Docking Simulations. J Chem Inf Model 2021; 61:5508-5523. [PMID: 34730967 PMCID: PMC8659376 DOI: 10.1021/acs.jcim.1c00924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Indexed: 11/30/2022]
Abstract
The lack of conformational sampling in virtual screening projects can lead to inefficient results because many of the potential drugs may not be able to bind to the target protein during the static docking simulations. Here, we performed ensemble docking for around 2000 United States Food and Drug Administration (FDA)-approved drugs with the RNA-dependent RNA polymerase (RdRp) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a target. The representative protein structures were generated by clustering classical molecular dynamics trajectories, which were evolved using three solvent scenarios, namely, pure water, benzene/water and phenol/water mixtures. The introduction of dynamic effects in the theoretical model showed improvement in docking results in terms of the number of strong binders and binding sites in the protein. Some of the discovered pockets were found only for the cosolvent simulations, where the nonpolar probes induced local conformational changes in the protein that lead to the opening of transient pockets. In addition, the selection of the ligands based on a combination of the binding free energy and binding free energy gap between the best two poses for each ligand provided more suitable binders than the selection of ligands based solely on one of the criteria. The application of cosolvent molecular dynamics to enhance the sampling of the configurational space is expected to improve the efficacy of virtual screening campaigns of future drug discovery projects.
Collapse
Affiliation(s)
- P. Bernát Szabó
- Department
of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
- Department
of Chemistry, Universidad Autónoma
de Madrid, Calle Francisco Tomás y Valiente, 7, 28049 Madrid, Spain
| | | | - Juan J. Nogueira
- Department
of Chemistry, Universidad Autónoma
de Madrid, Calle Francisco Tomás y Valiente, 7, 28049 Madrid, Spain
- IADCHEM,
Institute for Advanced Research in Chemistry, Universidad Autónoma de Madrid, Calle Francisco Tomás y Valiente, 7, 28049 Madrid, Spain
| |
Collapse
|
32
|
Gligorijevic N, Radomirovic M, Nedic O, Stojadinovic M, Khulal U, Stanic-Vucinic D, Cirkovic Velickovic T. Molecular Mechanisms of Possible Action of Phenolic Compounds in COVID-19 Protection and Prevention. Int J Mol Sci 2021; 22:12385. [PMID: 34830267 PMCID: PMC8625847 DOI: 10.3390/ijms222212385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022] Open
Abstract
The worldwide outbreak of COVID-19 was caused by a pathogenic virus called Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Therapies against SARS-CoV-2 target the virus or human cells or the immune system. However, therapies based on specific antibodies, such as vaccines and monoclonal antibodies, may become inefficient enough when the virus changes its antigenicity due to mutations. Polyphenols are the major class of bioactive compounds in nature, exerting diverse health effects based on their direct antioxidant activity and their effects in the modulation of intracellular signaling. There are currently numerous clinical trials investigating the effects of polyphenols in prophylaxis and the treatment of COVID-19, from symptomatic, via moderate and severe COVID-19 treatment, to anti-fibrotic treatment in discharged COVID-19 patients. Antiviral activities of polyphenols and their impact on immune system modulation could serve as a solid basis for developing polyphenol-based natural approaches for preventing and treating COVID-19.
Collapse
Affiliation(s)
- Nikola Gligorijevic
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia; (N.G.); (O.N.)
| | - Mirjana Radomirovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia; (M.R.); (M.S.); (D.S.-V.)
| | - Olgica Nedic
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia; (N.G.); (O.N.)
| | - Marija Stojadinovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia; (M.R.); (M.S.); (D.S.-V.)
| | - Urmila Khulal
- Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium;
- Global Campus, Ghent University, Yeonsu-gu, Incheon 21985, Korea
| | - Dragana Stanic-Vucinic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia; (M.R.); (M.S.); (D.S.-V.)
| | - Tanja Cirkovic Velickovic
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia; (M.R.); (M.S.); (D.S.-V.)
- Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium;
- Global Campus, Ghent University, Yeonsu-gu, Incheon 21985, Korea
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| |
Collapse
|
33
|
Bioeconomy during the COVID-19 and perspectives for the post-pandemic world: Example from EU. EFB BIOECONOMY JOURNAL 2021. [PMCID: PMC8683579 DOI: 10.1016/j.bioeco.2021.100013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Creanza TM, Delre P, Ancona N, Lentini G, Saviano M, Mangiatordi GF. Structure-Based Prediction of hERG-Related Cardiotoxicity: A Benchmark Study. J Chem Inf Model 2021; 61:4758-4770. [PMID: 34506150 PMCID: PMC9282647 DOI: 10.1021/acs.jcim.1c00744] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
![]()
Drug-induced blockade of the human
ether-à-go-go-related
gene (hERG) channel is today considered the main
cause of cardiotoxicity in postmarketing surveillance. Hence, several
ligand-based approaches were developed in the last years and are currently
employed in the early stages of a drug discovery process for in silico cardiac safety assessment of drug candidates.
Herein, we present the first structure-based classifiers able to discern hERG binders from nonbinders. LASSO regularized support
vector machines were applied to integrate docking scores and protein–ligand
interaction fingerprints. A total of 396 models were trained and validated
based on: (i) high-quality experimental bioactivity information returned
by 8337 curated compounds extracted from ChEMBL (version 25) and (ii)
structural predictor data. Molecular docking simulations were performed
using GLIDE and GOLD software programs and four different hERG structural models, namely, the recently published structures
obtained by cryoelectron microscopy (PDB codes: 5VA1 and 7CN1) and
two published homology models selected for comparison. Interestingly,
some classifiers return performances comparable to ligand-based models
in terms of area under the ROC curve (AUCMAX = 0.86 ±
0.01) and negative predictive values (NPVMAX = 0.81 ±
0.01), thus putting forward the herein proposed computational workflow
as a valuable tool for predicting hERG-related cardiotoxicity
without the limitations of ligand-based models, typically affected
by low interpretability and a limited applicability domain. From a
methodological point of view, our study represents the first example
of a successful integration of docking scores and protein–ligand
interaction fingerprints (IFs) through a support vector machine (SVM)
LASSO regularized strategy. Finally, the study highlights the importance
of using hERG structural models accounting for ligand-induced
fit effects and allowed us to select the best-performing protein conformation
(made available in the Supporting Information, SI) to be employed
for a reliable structure-based prediction of hERG-related cardiotoxicity.
Collapse
Affiliation(s)
- Teresa Maria Creanza
- CNR-Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Via Amendola 122/o, 70126 Bari, Italy
| | - Pietro Delre
- Chemistry Department, University of Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.,CNR-Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | - Nicola Ancona
- CNR-Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Via Amendola 122/o, 70126 Bari, Italy
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy
| | - Michele Saviano
- CNR-Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | | |
Collapse
|
35
|
Yan F, Gao F. An overview of potential inhibitors targeting non-structural proteins 3 (PL pro and Mac1) and 5 (3CL pro/M pro) of SARS-CoV-2. Comput Struct Biotechnol J 2021; 19:4868-4883. [PMID: 34457214 PMCID: PMC8382591 DOI: 10.1016/j.csbj.2021.08.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/02/2021] [Accepted: 08/21/2021] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need to develop effective treatments for coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The rapid spread of SARS-CoV-2 has resulted in a global pandemic that has not only affected the daily lives of individuals but also had a significant impact on the global economy and public health. Although extensive research has been conducted to identify inhibitors targeting SARS-CoV-2, there are still no effective treatment strategies to combat COVID-19. SARS-CoV-2 comprises two important proteolytic enzymes, namely, the papain-like proteinase, located within non-structural protein 3 (nsp3), and nsp5, both of which cleave large replicase polypeptides into multiple fragments that are required for viral replication. Moreover, a domain within nsp3, known as the macrodomain (Mac1), also plays an important role in viral replication. Inhibition of their functions should be able to significantly interfere with the replication cycle of the virus, and therefore these key proteins may serve as potential therapeutic targets. The functions of the above viral targets and their corresponding inhibitors have been summarized in the current review. This review provides comprehensive updates of nsp3 and nsp5 inhibitor development and would help advance the discovery of novel anti-viral therapeutics against SARS-CoV-2.
Collapse
Affiliation(s)
- Fangfang Yan
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Feng Gao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| |
Collapse
|
36
|
Rehman MFU, Akhter S, Batool AI, Selamoglu Z, Sevindik M, Eman R, Mustaqeem M, Akram MS, Kanwal F, Lu C, Aslam M. Effectiveness of Natural Antioxidants against SARS-CoV-2? Insights from the In-Silico World. Antibiotics (Basel) 2021; 10:1011. [PMID: 34439061 PMCID: PMC8388999 DOI: 10.3390/antibiotics10081011] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
The SARS CoV-2 pandemic has affected millions of people around the globe. Despite many efforts to find some effective medicines against SARS CoV-2, no established therapeutics are available yet. The use of phytochemicals as antiviral agents provides hope against the proliferation of SARS-CoV-2. Several natural compounds were analyzed by virtual screening against six SARS CoV-2 protein targets using molecular docking simulations in the present study. More than a hundred plant-derived secondary metabolites have been docked, including alkaloids, flavonoids, coumarins, and steroids. SARS CoV-2 protein targets include Main protease (MPro), Papain-like protease (PLpro), RNA-dependent RNA polymerase (RdRp), Spike glycoprotein (S), Helicase (Nsp13), and E-Channel protein. Phytochemicals were evaluated by molecular docking, and MD simulations were performed using the YASARA structure using a modified genetic algorithm and AMBER03 force field. Binding energies and dissociation constants allowed the identification of potentially active compounds. Ligand-protein interactions provide an insight into the mechanism and potential of identified compounds. Glycyrrhizin and its metabolite 18-β-glycyrrhetinic acid have shown a strong binding affinity for MPro, helicase, RdRp, spike, and E-channel proteins, while a flavonoid Baicalin also strongly binds against PLpro and RdRp. The use of identified phytochemicals may help to speed up the drug development and provide natural protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Fayyaz ur Rehman
- Department of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Institute of Chemistry, University of Sargodha, Sargodha 41600, Pakistan; (S.A.); (R.E.)
| | - Shahzaib Akhter
- Institute of Chemistry, University of Sargodha, Sargodha 41600, Pakistan; (S.A.); (R.E.)
| | - Aima Iram Batool
- Department of Zoology, University of Sargodha, Sargodha 41600, Pakistan;
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Omer Halisdemir University, Nigde 51240, Turkey;
| | - Mustafa Sevindik
- Department of Food Processing, Bahçe Vocational School, Osmaniye Korkut Ata University, Osmaniye 80000, Turkey;
| | - Rida Eman
- Institute of Chemistry, University of Sargodha, Sargodha 41600, Pakistan; (S.A.); (R.E.)
| | - Muhammad Mustaqeem
- Department of Chemistry, University of Sargodha, Bhakkar Campus, Bhakkar 30000, Pakistan;
| | - Muhammad Safwan Akram
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK;
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Fariha Kanwal
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 201620, China;
| | - Changrui Lu
- Department of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Mehwish Aslam
- School of Biological Sciences, University of the Punjab, Lahore 54600, Pakistan
| |
Collapse
|
37
|
Di Gaetano S, Capasso D, Delre P, Pirone L, Saviano M, Pedone E, Mangiatordi GF. More Is Always Better Than One: The N-Terminal Domain of the Spike Protein as Another Emerging Target for Hampering the SARS-CoV-2 Attachment to Host Cells. Int J Mol Sci 2021; 22:6462. [PMID: 34208755 PMCID: PMC8235207 DOI: 10.3390/ijms22126462] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 12/31/2022] Open
Abstract
Although the approved vaccines are proving to be of utmost importance in containing the Coronavirus disease 2019 (COVID-19) threat, they will hardly be resolutive as new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, a single-stranded RNA virus) variants might be insensitive to the immune response they induce. In this scenario, developing an effective therapy is still a dire need. Different targets for therapeutic antibodies and diagnostics have been identified, among which the SARS-CoV-2 spike (S) glycoprotein, particularly its receptor-binding domain, has been defined as crucial. In this context, we aim to focus attention also on the role played by the S N-terminal domain (S1-NTD) in the virus attachment, already recognized as a valuable target for neutralizing antibodies, in particular, building on a cavity mapping indicating the presence of two druggable pockets and on the recent literature hypothesizing the presence of a ganglioside-binding domain. In this perspective, we aim at proposing S1-NTD as a putative target for designing small molecules hopefully able to hamper the SARS-CoV-2 attachment to host cells.
Collapse
Affiliation(s)
- Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
| | - Domenica Capasso
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
- CESTEV, University of Naples “Federico II”, 80145 Naples, Italy
| | - Pietro Delre
- Institute of Crystallography, CNR, 70126 Bari, Italy; (P.D.); (G.F.M.)
- Chemistry Department, University of Bari, 70121 Bari, Italy
| | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
| | - Michele Saviano
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
- Institute of Crystallography, CNR, 70126 Bari, Italy; (P.D.); (G.F.M.)
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
| | | |
Collapse
|
38
|
Mahmoudvand S, Shokri S. Interactions between SARS coronavirus 2 papain-like protease and immune system: A potential drug target for the treatment of COVID-19. Scand J Immunol 2021; 94:e13044. [PMID: 33872387 PMCID: PMC8250271 DOI: 10.1111/sji.13044] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/24/2021] [Accepted: 04/11/2021] [Indexed: 12/01/2022]
Abstract
Coronaviruses (CoVs) are a large family of respiratory viruses which can cause mild to moderate upper respiratory tract infections. Recently, new coronavirus named as Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) has been identified which is a major threat to public health. Innate immune responses play a vital role in a host's defence against viruses. Interestingly, CoVs have evolved elaborate strategies to evade the complex system of sensors and signalling molecules to suppress host immunity. SARS‐CoV‐2 papain‐like protease (PLpro), as an important coronavirus enzyme, regulates viral spread and innate immune responses. SCoV‐2 PLpro is multifunctional enzyme with deubiquitinating (DUB) and deISGylating activity. The PLpro can interact with key regulators in signalling pathways such as STING, NF‐κB, cytokine production, MAPK and TGF‐β and hijack those to block the immune responses. Therefore, the PLpro can be as an important target for the treatment of COVID‐19. Until now, several drugs or compounds have been identified that can inhibit PLpro activity. Here we discuss about the dysregulation effects of PLpro on immune system and drugs that have potential inhibitors for SCoV‐2 PLpro.
Collapse
Affiliation(s)
- Shahab Mahmoudvand
- Student Research CommitteeAhvaz Jundishapur University of Medical SciencesAhvazIran
- Department of VirologySchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Somayeh Shokri
- Student Research CommitteeAhvaz Jundishapur University of Medical SciencesAhvazIran
- Department of VirologySchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
39
|
Ismail MI, Ragab HM, Bekhit AA, Ibrahim TM. Targeting multiple conformations of SARS-CoV2 Papain-Like Protease for drug repositioning: An in-silico study. Comput Biol Med 2021; 131:104295. [PMID: 33662683 PMCID: PMC7902231 DOI: 10.1016/j.compbiomed.2021.104295] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 12/16/2022]
Abstract
Papain-Like Protease (PLpro) is a key protein for SARS-CoV-2 viral replication which is the cause of the emerging COVID-19 pandemic. Targeting PLpro can suppress viral replication and provide treatment options for COVID-19. Due to the dynamic nature of its binding site loop, PLpro multiple conformations were generated through a long-range 1 micro-second molecular dynamics (MD) simulation. Clustering the MD trajectory enabled us to extract representative structures for the conformational space generated. Adding to the MD representative structures, X-ray structures were involved in an ensemble docking approach to screen the FDA approved drugs for a drug repositioning endeavor. Guided by our recent benchmarking study of SARS-CoV-2 PLpro, FRED docking software was selected for such a virtual screening task. The results highlighted potential consensus binders to many of the MD clusters as well as the newly introduced X-ray structure of PLpro complexed with a small molecule. For instance, three drugs Benserazide, Dobutamine and Masoprocol showed a superior consensus enrichment against the PLpro conformations. Further MD simulations for these drugs complexed with PLpro suggested the superior stability and binding of dobutamine and masoprocol inside the binding site compared to Benserazide. Generally, this approach can facilitate identifying drugs for repositioning via targeting multiple conformations of a crucial target for the rapidly emerging COVID-19 pandemic.
Collapse
Affiliation(s)
- Muhammad I Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Al-Sherouk City, Cairo-Suez Desert Road, 11837, Cairo, Egypt
| | - Hanan M Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Adnan A Bekhit
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Pharmacy Program, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, P.O. Box 32038, Bahrain
| | - Tamer M Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| |
Collapse
|
40
|
Huynh T, Cornell W, Luan B. In silico Exploration of Inhibitors for SARS-CoV-2's Papain-Like Protease. Front Chem 2021; 8:624163. [PMID: 33614597 PMCID: PMC7889802 DOI: 10.3389/fchem.2020.624163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/08/2020] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an ongoing global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with very limited treatments so far. Demonstrated with good druggability, two major proteases of SARS-CoV-2, namely main protease (Mpro) and papain-like protease (PLpro) that are essential for viral maturation, have become the targets for many newly designed inhibitors. Unlike Mpro that has been heavily investigated, PLpro is not well-studied so far. Here, we carried out the in silico high-throughput screening of all FDA-approved drugs via the flexible docking simulation for potential inhibitors of PLpro and explored the molecular mechanism of binding between a known inhibitor rac5c and PLpro. Our results, from molecular dynamics simulation, show that the chances of drug repurposing for PLpro might be low. On the other hand, our long (about 450 ns) MD simulation confirms that rac5c can be bound stably inside the substrate-binding site of PLpro and unveils the molecular mechanism of binding for the rac5c-PLpro complex. The latter may help perform further structural optimization and design potent leads for inhibiting PLpro.
Collapse
Affiliation(s)
- Tien Huynh
- Computational Biological Center, IBM Thomas J. Watson Research, New York, NY, United States
| | - Wendy Cornell
- Computational Biological Center, IBM Thomas J. Watson Research, New York, NY, United States
| | - Binquan Luan
- Computational Biological Center, IBM Thomas J. Watson Research, New York, NY, United States
| |
Collapse
|
41
|
El Hassab MA, Ibrahim TM, Shoun AA, Al-Rashood ST, Alkahtani HM, Alharbi A, Eskandrani RO, Eldehna WM. In silico identification of potential SARS COV-2 2′-O-methyltransferase inhibitor: fragment-based screening approach and MM-PBSA calculations. RSC Adv 2021; 11:16026-16033. [PMID: 35481212 PMCID: PMC9029993 DOI: 10.1039/d1ra01809d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
In the present era, there are many efforts trying to face the emerging and successive waves of the COVID-19 pandemic. This has led to considering new and unusual targets for SARS CoV-2. 2′-O-Methyltransferase (nsp16) is a key and attractive target in the SARS CoV-2 life cycle since it is responsible for the viral RNA protection via a cap formation process. In this study, we propose a new potential inhibitor for SARS COV-2 2′-O-methyltransferase (nsp16). A fragment library was screened against the co-crystal structure of the SARS COV-2 2′-O-methyltransferase complexed with Sinefungin (nsp16 – PDB ID: 6WKQ), and consequently the best proposed fragments were linked via a de novo approach to build molecule AP-20. Molecule AP-20 displayed a superior docking score to Sinefungin and reproduced the key interactions in the binding site of 2′-O-methyltransferase. Three molecular dynamic simulations of the 2′-O-methyltransferase apo structure and its complexed forms with AP-20 and Sinefungin were performed for 150 nano-seconds to provide insights on the dynamic nature of such setups and to assess the stability of the proposed AP-20/enzyme complex. AP-20/enzyme complex demonstrated better stability for the ligand–enzyme complex compared to Sinefungin in a respective setup. Furthermore, MM-PBSA binding free energy calculations showed a better profile for AP-20/enzyme complex compared to Sinefungin/enzyme complex emphasizing the potential inhibitory effect of AP-20 on SARS COV-2 2′-O-methyltransferase. We endorse our designed molecule AP-20 to be further explored via experimental evaluations to confront the spread of the emerging COVID-19. Also, in silico ADME profiling has ascribed to AP-20 an excellent safety and metabolic stability profile. The identification of AP-20 as a potential SARS COV-2 2′-O-methyltransferase inhibitor: fragment-based screening approach and MM-PBSA calculations.![]()
Collapse
Affiliation(s)
- Mahmoud A. El Hassab
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- Badr University in Cairo (BUC)
- Cairo
- Egypt
| | - Tamer M. Ibrahim
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Kafrelsheikh University
- Kafrelsheikh
- Egypt
| | - Aly A. Shoun
- Department of Microbiology & Immunology
- Faculty of Pharmacy
- Sinai University
- North Sinai
- Egypt
| | - Sara T. Al-Rashood
- Department of Pharmaceutical Chemistry
- College of Pharmacy
- King Saud University
- Riyadh
- Saudi Arabia
| | - Hamad M. Alkahtani
- Department of Pharmaceutical Chemistry
- College of Pharmacy
- King Saud University
- Riyadh
- Saudi Arabia
| | - Amal Alharbi
- Department of Pharmaceutical Chemistry
- College of Pharmacy
- King Saud University
- Riyadh
- Saudi Arabia
| | - Razan O. Eskandrani
- Department of Pharmaceutical Chemistry
- College of Pharmacy
- King Saud University
- Riyadh
- Saudi Arabia
| | - Wagdy M. Eldehna
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Kafrelsheikh University
- Kafrelsheikh
- Egypt
| |
Collapse
|