1
|
Schuhmann F, Bordallo HN, Pezeshkian W. Physics-Based Protein Networks Might Recover Effectful Mutations─a Case Study on Cathepsin G. J Phys Chem B 2024; 128:10043-10050. [PMID: 39357873 PMCID: PMC11492240 DOI: 10.1021/acs.jpcb.4c04140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024]
Abstract
Molecular dynamics simulations have been remarkably effective for observing and analyzing structures and dynamics of proteins, with longer trajectories being computed every day. Still, often, relevant time scales are not observed. Adequately analyzing the generated trajectories can highlight the interesting areas within a protein such as mutation sites or allosteric hotspots, which might foreshadow dynamics untouched by the simulations. We employ a physics-based protein network and propose that such a network can adequately analyze the protein dynamics. The analysis is conducted on simulations of cathepsin G and neutrophil elastase, which are remarkably similar but with different specificities. However, a single mutation in cathepsin G recovers the specificity of neutrophil elastase. The physics-based network built on the interactions between residues instead of the distances can pinpoint the active triad in the proteins studied. Overall, the network seems to capture the structural behavior better than purely distance-based networks.
Collapse
Affiliation(s)
- Fabian Schuhmann
- Niels
Bohr International Academy, Niels Bohr Institute,
University of Copenhagen, Blegdamsvej 17, 2100 Copenhagen, Denmark
| | - Heloisa N. Bordallo
- Niels
Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Weria Pezeshkian
- Niels
Bohr International Academy, Niels Bohr Institute,
University of Copenhagen, Blegdamsvej 17, 2100 Copenhagen, Denmark
| |
Collapse
|
2
|
Sladek V, Artiushenko PV, Fedorov DG. Effect of Direct and Water-Mediated Interactions on the Identification of Hotspots in Biomolecular Complexes with Multiple Subsystems. J Chem Inf Model 2024; 64:7602-7615. [PMID: 39283296 DOI: 10.1021/acs.jcim.4c00973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Identification of important residues in biochemical complexes is often a crucial step for many problems in molecular biology and biochemistry. A method is proposed to identify hotspots in biomolecular complexes based on a new metric, derived from networks representing molecular subunits (residues, bridging solvent molecules, ligands etc.) connected by interactions. A singular value decomposition of the weighted adjacency matrix is used to construct a scalar rank for each subunit that reflects its importance in the residue interaction network. This metric is called the singular value centrality. In addition, a new formalism is proposed to account for water-mediated interactions in the ranking of residues. Interactions for a residue network can be provided by various computational methods. In this work interactions are obtained from full quantum-mechanical calculations of protein-protein complexes using the fragment molecular orbital method. The ranking results are shown to be in good agreement with earlier computational and experimental studies. The developed method can be used to gain a deeper insight into the role of subunits in complex biomolecular systems.
Collapse
Affiliation(s)
- Vladimir Sladek
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 845 38 Bratislava, Slovakia
| | - Polina V Artiushenko
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 845 38 Bratislava, Slovakia
| | - Dmitri G Fedorov
- Research Center for Computational Design of Advanced Functional Materials (CD-FMat) National Institute of Advanced Industrial Science and Technology (AIST), Central 2 Umezono 1-1-1, Tsukuba 305-8568, Japan
| |
Collapse
|
3
|
Shahu M, Schuhmann F, Wong SY, Solov’yov IA, Koch KW. Allosteric Communication of the Dimerization and the Catalytic Domain in Photoreceptor Guanylate Cyclase. Biochemistry 2024; 63:2131-2140. [PMID: 39175413 PMCID: PMC11375764 DOI: 10.1021/acs.biochem.4c00170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Phototransduction in vertebrate photoreceptor cells is controlled by Ca2+-dependent feedback loops involving the membrane-bound guanylate cyclase GC-E that synthesizes the second messenger guanosine-3',5'-cyclic monophosphate. Intracellular Ca2+-sensor proteins named guanylate cyclase-activating proteins (GCAPs) regulate the activity of GC-E by switching from a Ca2+-bound inhibiting state to a Ca2+-free/Mg2+-bound activating state. The gene GUCY2D encodes for human GC-E, and mutations in GUCY2D are often associated with an imbalance of Ca2+ and cGMP homeostasis causing retinal disorders. Here, we investigate the Ca2+-dependent inhibition of the constitutively active GC-E mutant V902L. The inhibition is not mediated by GCAP variants but by Ca2+ replacing Mg2+ in the catalytic center. Distant from the cyclase catalytic domain is an α-helical domain containing a highly conserved helix-turn-helix motif. Mutating the critical amino acid position 804 from leucine to proline left the principal activation mechanism intact but resulted in a lower level of catalytic efficiency. Our experimental analysis of amino acid positions in two distant GC-E domains implied an allosteric communication pathway connecting the α-helical and the cyclase catalytic domains. A computational connectivity analysis unveiled critical differences between wildtype GC-E and the mutant V902L in the allosteric network of critical amino acid positions.
Collapse
Affiliation(s)
- Manisha
Kumari Shahu
- Department
of Neuroscience, Carl von Ossietzky Universität
Oldenburg, Carl-von-Ossietzky-Str.
9-11, 26129 Oldenburg ,Germany
| | - Fabian Schuhmann
- Niels
Bohr International Academy, Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 Copenhagen, Denmark
- Institute
of Physics, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129 Oldenburg, Germany
| | - Siu Ying Wong
- Institute
of Physics, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129 Oldenburg, Germany
| | - Ilia A. Solov’yov
- Institute
of Physics, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129 Oldenburg, Germany
- Research
Centre for Neurosensory Science, Carl von
Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129 Oldenburg ,Germany
- Center
for Nanoscale Dynamics (CENAD), Institute of Physics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstr. 114-118, 26129 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Department
of Neuroscience, Carl von Ossietzky Universität
Oldenburg, Carl-von-Ossietzky-Str.
9-11, 26129 Oldenburg ,Germany
- Research
Centre for Neurosensory Science, Carl von
Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129 Oldenburg ,Germany
| |
Collapse
|
4
|
Kenward C, Vuckovic M, Paetzel M, Strynadka NCJ. Kinetic comparison of all eleven viral polyprotein cleavage site processing events by SARS-CoV-2 main protease using a linked protein FRET platform. J Biol Chem 2024; 300:107367. [PMID: 38750796 PMCID: PMC11209022 DOI: 10.1016/j.jbc.2024.107367] [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: 02/22/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
The main protease (Mpro) remains an essential therapeutic target for COVID-19 post infection intervention given its critical role in processing the majority of viral proteins encoded by the genome of severe acute respiratory syndrome related coronavirus 2 (SARS-CoV-2). Upon viral entry, the +ssRNA genome is translated into two long polyproteins (pp1a or the frameshift-dependent pp1ab) containing all the nonstructural proteins (nsps) required by the virus for immune modulation, replication, and ultimately, virion assembly. Included among these nsps is the cysteine protease Mpro (nsp5) which self-excises from the polyprotein, dimerizes, then sequentially cleaves 11 of the 15 cut-site junctions found between each nsp within the polyprotein. Many structures of Mpro (often bound to various small molecule inhibitors or peptides) have been detailed recently, including structures of Mpro bound to each of the polyprotein cleavage sequences, showing that Mpro can accommodate a wide range of targets within its active site. However, to date, kinetic characterization of the interaction of Mpro with each of its native cleavage sequences remains incomplete. Here, we present a robust and cost-effective FRET based system that benefits from a more consistent presentation of the substrate that is also closer in organization to the native polyprotein environment compared to previously reported FRET systems that use chemically modified peptides. Using this system, we were able to show that while each site maintains a similar Michaelis constant, the catalytic efficiency of Mpro varies greatly between cut-site sequences, suggesting a clear preference for the order of nsp processing.
Collapse
Affiliation(s)
- Calem Kenward
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Marija Vuckovic
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark Paetzel
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
Sawang N, Phongphanphanee S, Wong-ekkabut J, Sutthibutpong T. Biophysical Interpretation of Evolutionary Consequences on the SARS-CoV2 Main Protease through Molecular Dynamics Simulations and Network Topology Analysis. J Phys Chem B 2023; 127:2331-2343. [PMID: 36913683 PMCID: PMC10022058 DOI: 10.1021/acs.jpcb.2c08312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/25/2023] [Indexed: 03/14/2023]
Abstract
In this study, we present a combined analysis procedure between atomistic molecular dynamics (MD) simulations and network topology to obtain more understanding on the evolutionary consequences on protein stability and substrate binding of the main protease enzyme of SARS-CoV2. Communicability matrices of the protein residue networks (PRNs) were extracted from MD trajectories of both Mpro enzymes in complex with the nsp8/9 peptide substrate to compare the local communicability within both proteases that would affect the enzyme function, along with biophysical details on global protein conformation, flexibility, and contribution of amino acid side chains to both intramolecular and intermolecular interactions. The analysis displayed the significance of the mutated residue 46 with the highest communicability gain to the binding pocket closure. Interestingly, the mutated residue 134 with the highest communicability loss corresponded to a local structural disruption of the adjacent peptide loop. The enhanced flexibility of the disrupted loop connecting to the catalytic residue Cys145 introduced an extra binding mode that brought the substrate in proximity and could facilitate the reaction. This understanding might provide further help in the drug development strategy against SARS-CoV2 and prove the capability of the combined techniques of MD simulations and network topology analysis as a "reverse" protein engineering tool.
Collapse
Affiliation(s)
- Nuttawat Sawang
- Theoretical
and Computational Physics Group, Department of Physics, King Mongkut’s University of Technology Thonburi
(KMUTT), 126 Pracha-Uthit Road, Bang Mod, Thrung Khru, Bangkok 10140, Thailand
- Center
of Excellence in Theoretical and Computational Science (TaCS-CoE),
Faculty of Science, King Mongkut’s
University of Technology Thonburi (KMUTT), 126 Pracha Uthit Rd., Bang Mod, Thung Khru, Bangkok 10140, Thailand
| | - Saree Phongphanphanee
- Computational
Biomodelling Laboratory for Agricultural Science and Technology (CBLAST),
Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Thailand
Center of Excellence in Physics (ThEP Center), Ministry of Higher Education, Science, Research and Innovation, Bangkok 10400, Thailand
- Department
of Materials Science, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Jirasak Wong-ekkabut
- Computational
Biomodelling Laboratory for Agricultural Science and Technology (CBLAST),
Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Thailand
Center of Excellence in Physics (ThEP Center), Ministry of Higher Education, Science, Research and Innovation, Bangkok 10400, Thailand
- Department
of Physics, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Thana Sutthibutpong
- Theoretical
and Computational Physics Group, Department of Physics, King Mongkut’s University of Technology Thonburi
(KMUTT), 126 Pracha-Uthit Road, Bang Mod, Thrung Khru, Bangkok 10140, Thailand
- Center
of Excellence in Theoretical and Computational Science (TaCS-CoE),
Faculty of Science, King Mongkut’s
University of Technology Thonburi (KMUTT), 126 Pracha Uthit Rd., Bang Mod, Thung Khru, Bangkok 10140, Thailand
- Computational
Biomodelling Laboratory for Agricultural Science and Technology (CBLAST),
Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
6
|
Charles S, Edgar MP, Mahapatra RK. Artificial intelligence based virtual screening study for competitive and allosteric inhibitors of the SARS-CoV-2 main protease. J Biomol Struct Dyn 2023; 41:15286-15304. [PMID: 36943715 DOI: 10.1080/07391102.2023.2188419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023]
Abstract
SARS-CoV-2 is a highly contagious and dangerous coronavirus that first appeared in late 2019 causing COVID-19, a pandemic of acute respiratory illnesses that is still a threat to health and the general public safety. We performed deep docking studies of 800 M unique compounds in both the active and allosteric sites of the SARS-COV-2 Main Protease (Mpro) and the 15 M and 13 M virtual hits obtained were further taken for conventional docking and molecular dynamic (MD) studies. The best XP Glide docking scores obtained were -14.242 and -12.059 kcal/mol by CHEMBL591669 and the highest binding affinities were -10.5 kcal/mol (from 444215) and -11.2 kcal/mol (from NPC95421) for active and allosteric sites, respectively. Some hits can bind both sites making them a great area of concern. Re-docking of 8 random allosteric complexes in the active site shows a significant reduction in docking scores with a t-test P value of 2.532 × 10-11 at 95% confidence. Some specific interactions have higher elevations in docking scores. MD studies on 15 complexes show that single-ligand systems are stable as compared to double-ligand systems, and the allosteric binders identified are shown to modulate the active site binding as evidenced by the changes in the interaction patterns and stability of ligands and active site residues. When an allosteric complex was docked to the second monomer to check for homodimer formation, the validated homodimer could not be re-established, further supporting the potential of the identified allosteric binders. These findings could be important in developing novel therapeutics against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ssemuyiga Charles
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
- Department of Microbiology, Biotechnology and Plant Sciences, School of Biological Sciences, Makerere University, Kampala, Uganda
| | - Mulumba Pius Edgar
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
| | | |
Collapse
|
7
|
Banerjee A, Gosavi S. Potential Self-Peptide Inhibitors of the SARS-CoV-2 Main Protease. J Phys Chem B 2023; 127:855-865. [PMID: 36689738 PMCID: PMC9883841 DOI: 10.1021/acs.jpcb.2c05917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/23/2022] [Indexed: 01/24/2023]
Abstract
The SARS-CoV-2 main protease (Mpro) plays an essential role in viral replication, cleaving viral polyproteins into functional proteins. This makes Mpro an important drug target. Mpro consists of an N-terminal catalytic domain and a C-terminal α-helical domain (MproC). Previous studies have shown that peptides derived from a given protein sequence (self-peptides) can affect the folding and, in turn, the function of that protein. Since the SARS-CoV-1 MproC is known to stabilize its Mpro and regulate its function, we hypothesized that SARS-CoV-2 MproC-derived self-peptides may modulate the folding and the function of SARS-CoV-2 Mpro. To test this, we studied the folding of MproC in the presence of various self-peptides using coarse-grained structure-based models and molecular dynamics simulations. In these simulations of MproC and one self-peptide, we found that two self-peptides, the α1-helix and the loop between α4 and α5 (loop4), could replace the equivalent native sequences in the MproC structure. Replacement of either sequence in full-length Mpro should, in principle, be able to perturb Mpro function albeit through different mechanisms. Some general principles for the rational design of self-peptide inhibitors emerge: The simulations show that prefolded self-peptides are more likely to replace native sequences than those which do not possess structure. Additionally, the α1-helix self-peptide is kinetically stable and once inserted rarely exchanges with the native α1-helix, while the loop4 self-peptide is easily replaced by the native loop4, making it less useful for modulating function. In summary, a prefolded α1-derived peptide should be able to inhibit SARS-CoV-2 Mpro function.
Collapse
Affiliation(s)
- Arkadeep Banerjee
- Simons Centre for the Study
of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065, India
| | - Shachi Gosavi
- Simons Centre for the Study
of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065, India
| |
Collapse
|
8
|
Sanjeev BS, Chitara D, Madhumalar A. Physiological models to study the effect of molecular crowding on multi-drug bound proteins: insights from SARS-CoV-2 main protease. J Biomol Struct Dyn 2022; 40:13564-13580. [PMID: 34699337 DOI: 10.1080/07391102.2021.1993342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Molecular Dynamics simulations are often used in drug design. However, such simulations do not account for the physiological environment of the receptor; hence overlook its impact on biomolecular interactions. To address this lacuna, we identified three objectives to pursue - develop models of physiological environment, study a drug-receptor complex in such environments, and identify methods to analyze these complicated simulations. Two novel physiological models were developed and studied. The first, called 'm10', comprises of 10 of the most abundant cytoplasmic metabolites at physiological concentrations. The second, called 'phy', supplements m10 with an additional crowder protein to elicit macromolecular crowding effect. The main protease (Mpro) of SARS-CoV-2, being essential for viral replication, is an attractive drug target for COVID-19. Hence, we chose Mpro docked with multiple drugs as our model drug-receptor system. With a plethora of compounds, physiological systems can be exceedingly large and complex. A novel Spark-based software (SparkTraj) was developed to rapidly analyze non-specific contacts and water interactions. Our study shows that crowding enhances the difference in the dynamics of apo- vs drug-bound complexes. Metabolites, at times as a cluster, were seen interacting with the protease, drugs, and binding sites in drug-free receptor. Except one that crawled to an adjacent pocket in phy, the drugs remained in their respective pockets in all simulations. Given these observations, we hope that the models and approach presented here would help the optimization, evaluation, and selection of potential drugs. Generic biomolecular dynamics could also benefit from such models and tools.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- B S Sanjeev
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, India
| | - Dheeraj Chitara
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, India
| | - Arumugam Madhumalar
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
9
|
Effects of Site-Directed Mutations on the Communicability between Local Segments and Binding Pocket Distortion of Engineered GH11 Xylanases Visualized through Network Topology Analysis. Catalysts 2022. [DOI: 10.3390/catal12101165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mutations occurred within the binding pocket of enzymes directly modified the interaction network between an enzyme and its substrate. However, some mutations affecting the catalytic efficiency occurred far from the binding pocket and the explanation regarding mechanisms underlying the transmission of the mechanical signal from the mutated site to the binding pocket was lacking. In this study, network topology analysis was used to characterize and visualize the changes of interaction networks caused by site-directed mutations on a GH11 xylanase from our previous study. For each structure, coordinates from molecular dynamics (MD) trajectory were obtained to create networks of representative atoms from all protein and xylooligosaccharide substrate residues, in which edges were defined between pairs of residues within a cutoff distance. Then, communicability matrices were extracted from the network to provide information on the mechanical signal transmission from the number of possible paths between any residue pairs or local protein segments. The analysis of subgraph centrality and communicability clearly showed that site-direct mutagenesis at non-reducing or reducing ends caused binding pocket distortion close to the opposite ends and created denser interaction networks. However, site-direct mutagenesis at both ends cancelled the binding pocket distortion, while enhancing the thermostability. Therefore, the network topology analysis tool on the atomistic simulations of engineered proteins could play some roles in protein design for the minimization to the correction of binding pocket tilting, which could affect the functionality and efficacy of enzymes.
Collapse
|
10
|
Sajid Jamal QM, Alharbi AH, Ahmad V. Identification of doxorubicin as a potential therapeutic against SARS-CoV-2 (COVID-19) protease: a molecular docking and dynamics simulation studies. J Biomol Struct Dyn 2022; 40:7960-7974. [PMID: 33826483 PMCID: PMC8043163 DOI: 10.1080/07391102.2021.1905551] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 03/15/2021] [Indexed: 02/08/2023]
Abstract
After one year, the COVID-19 pandemic caused by SARS-CoV-2 is still the largest concern for the scientific community. Of the many recognized drug targets of SARS-CoV-2, the main protease is one of the most important target due to its function in viral replication. We conducted an in silico study with repurposing drugs of antibiotics class against virus protease and peptidase using AutoDock tool. The following significant binding energy interaction was observed with protease (PDB: 6LU7) like piperacillin -7.25; tobramycin -9.20 and doxorubicin (Doxo) -10.04 kcal/mol and with peptidase (PDB: 2GTB) piperacillin -7.08; tobramycin -8.54 and Doxo -9.89 kcal/mol. Furthermore, the interaction and stability behavior of the Doxo-protease and Doxo-peptidase complexes were analyzed for a 100-nanosecond (ns) time. Calculated RMSD values observed using molecular dynamics simulation (MDS) were found to be 0.15-0.25 nm, RMSF calculation per residues showed a value near 0.2 nm and Rg values remained approximately 2.25 nm. MM-PBSA analysis of total binding energy calculation of Doxo-protease and Doxo-peptidase complexes are found to be -148.692 and -105.367 kJ/mol, respectively. Moreover, amino acid residue ASP-197 showed the lowest contribution binding energy i.e. -18.1185 kJ/mol, and amino acid residue ASP-187 showed -17.0267 kJ/mol contribution energy. Thus, significant docking interaction and stable dynamicity of Doxo-protease complex with time was suggested that Doxo could be a choice to inhibit potentially the viral proteases that could prevent the entry inside the host cell to control the COVID-19 disease. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia
| | - Ali H. Alharbi
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia
| | - Varish Ahmad
- Health Information Technology Department, Faculty of Applied Studies, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
11
|
Santos ES, Silva PC, Sousa PSA, Aquino CC, Pacheco G, Teixeira LFLS, Araujo AR, Sousa FBM, Barros RO, Ramos RM, Rocha JA, Nicolau LAD, Medeiros JVR. Antiviral potential of diminazene aceturate against SARS-CoV-2 proteases using computational and in vitro approaches. Chem Biol Interact 2022; 367:110161. [PMID: 36116513 PMCID: PMC9476334 DOI: 10.1016/j.cbi.2022.110161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Diminazene aceturate (DIZE), an antiparasitic, is an ACE2 activator, and studies show that activators of this enzyme may be beneficial for COVID-19, disease caused by SARS-CoV-2. Thus, the objective was to evaluate the in silico and in vitro affinity of diminazene aceturate against molecular targets of SARS-CoV-2. 3D structures from DIZE and the proteases from SARS-CoV-2, obtained through the Protein Data Bank and Drug Database (Drubank), and processed in computer programs like AutodockTools, LigPlot, Pymol for molecular docking and visualization and GROMACS was used to perform molecular dynamics. The results demonstrate that DIZE could interact with all tested targets, and the best binding energies were obtained from the interaction of Protein S (closed conformation −7.87 kcal/mol) and Mpro (−6.23 kcal/mol), indicating that it can act both by preventing entry and viral replication. The results of molecular dynamics demonstrate that DIZE was able to promote a change in stability at the cleavage sites between S1 and S2, which could prevent binding to ACE2 and fusion with the membrane. In addition, in vitro tests confirm the in silico results showing that DIZE could inhibit the binding between the spike receptor-binding domain protein and ACE2, which could promote a reduction in the virus infection. However, tests in other experimental models with in vivo approaches are needed.
Collapse
Affiliation(s)
- Esley S Santos
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Priscila C Silva
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Paulo S A Sousa
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Cristhyane C Aquino
- Postgraduate Program in Medical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Gabriella Pacheco
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Luiz F L S Teixeira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Alyne R Araujo
- Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Francisca B M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Romulo O Barros
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Ricardo M Ramos
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Jefferson A Rocha
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Lucas A D Nicolau
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Jand V R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil.
| |
Collapse
|
12
|
Clemente CM, Freiberger MI, Ravetti S, Beltramo DM, Garro AG. An in silico analysis of Ibuprofen enantiomers in high concentrations of sodium chloride with SARS-CoV-2 main protease. J Biomol Struct Dyn 2022; 40:5653-5664. [PMID: 33459192 PMCID: PMC7832455 DOI: 10.1080/07391102.2021.1872420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/02/2021] [Indexed: 12/01/2022]
Abstract
2020 will be remembered worldwide for the outbreak of Coronavirus disease (COVID-19), which quickly spread until it was declared as a global pandemic. The main protease (Mpro) of SARS-CoV-2, a key enzyme in coronavirus, represents an attractive pharmacological target for inhibition of SARS-CoV-2 replication. Here, we evaluated whether the anti-inflammatory drug Ibuprofen, may act as a potential SARS-CoV-2 Mpro inhibitor, using an in silico study. From molecular dynamics (MD) simulations, we also evaluated the influence of ionic strength on the affinity and stability of the Ibuprofen-Mpro complexes. The docking analysis shows that R(-)Ibuprofen and S(+)Ibuprofen isomers can interact with multiple key residues of the main protease, through hydrophobic interactions and hydrogen bonds, with favourable binding energies (-6.2 and -5.7 kcal/mol, respectively). MM-GBSA and MM-PBSA calculations confirm the affinity of these complexes, in terms of binding energies. It also demonstrates that the ionic strength modifies significantly their binding affinities. Different structural parameters calculated from the MD simulations (120 ns) reveal that these complexes are conformational stable in the different conditions analysed. In this context, the results suggest that the condition 2 (0.25 NaCl) bind more tightly the Ibuprofen to Mpro than the others conditions. From the frustration analysis, we could characterize two important regions (Cys44-Pro52 and Linker loop) of this protein involved in the interaction with Ibuprofen. In conclusion, our findings allow us to propose that racemic mixtures of the Ibuprofen enantiomers might be a potential treatment option against SARS-CoV-2 Mpro. However, further research is necessary to determinate their possible medicinal use.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- C. M. Clemente
- Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María, Villa María, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - M. I. Freiberger
- Protein Physiology Lab, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET-IQUIBICEN, Buenos Aires, Argentina
| | - S. Ravetti
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Centro de Investigaciones y Transferencia de Villa María (CIT VM), Instituto Académico Pedagógico de Ciencias Humanas, Universidad Nacional de Villa María, Villa María, Argentina
| | - D. M. Beltramo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Centro de Excelencia en Productos y Procesos de Córdoba (CEPROCOR), Córdoba, Argentina
- Cátedra de Biotecnología, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - A. G. Garro
- Centro de Investigaciones y Transferencia de Villa María (CIT VM), Instituto Académico Pedagógico de Ciencias Humanas, Universidad Nacional de Villa María, Villa María, Argentina
- Centro de Excelencia en Productos y Procesos de Córdoba (CEPROCOR), Córdoba, Argentina
| |
Collapse
|
13
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
14
|
Flynn JM, Samant N, Schneider-Nachum G, Barkan DT, Yilmaz NK, Schiffer CA, Moquin SA, Dovala D, Bolon DNA. Comprehensive fitness landscape of SARS-CoV-2 M pro reveals insights into viral resistance mechanisms. eLife 2022; 11:e77433. [PMID: 35723575 PMCID: PMC9323007 DOI: 10.7554/elife.77433] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
With the continual evolution of new strains of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) that are more virulent, transmissible, and able to evade current vaccines, there is an urgent need for effective anti-viral drugs. The SARS-CoV-2 main protease (Mpro) is a leading target for drug design due to its conserved and indispensable role in the viral life cycle. Drugs targeting Mpro appear promising but will elicit selection pressure for resistance. To understand resistance potential in Mpro, we performed a comprehensive mutational scan of the protease that analyzed the function of all possible single amino acid changes. We developed three separate high throughput assays of Mpro function in yeast, based on either the ability of Mpro variants to cleave at a defined cut-site or on the toxicity of their expression to yeast. We used deep sequencing to quantify the functional effects of each variant in each screen. The protein fitness landscapes from all three screens were strongly correlated, indicating that they captured the biophysical properties critical to Mpro function. The fitness landscapes revealed a non-active site location on the surface that is extremely sensitive to mutation, making it a favorable location to target with inhibitors. In addition, we found a network of critical amino acids that physically bridge the two active sites of the Mpro dimer. The clinical variants of Mpro were predominantly functional in our screens, indicating that Mpro is under strong selection pressure in the human population. Our results provide predictions of mutations that will be readily accessible to Mpro evolution and that are likely to contribute to drug resistance. This complete mutational guide of Mpro can be used in the design of inhibitors with reduced potential of evolving viral resistance.
Collapse
Affiliation(s)
- Julia M Flynn
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Neha Samant
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Gily Schneider-Nachum
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - David T Barkan
- Novartis Institutes for Biomedical ResearchEmeryvilleUnited States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | | | - Dustin Dovala
- Novartis Institutes for Biomedical ResearchEmeryvilleUnited States
| | - Daniel NA Bolon
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| |
Collapse
|
15
|
Chen X, Leyendecker S, van den Bedem H. Kinematic Vibrational Entropy Assessment and Analysis of SARS CoV-2 Main Protease. J Chem Inf Model 2022; 62:2869-2879. [PMID: 35594568 DOI: 10.1021/acs.jcim.2c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The three-dimensional conformations of a protein influence its function and select for the ligands it can interact with. The total free energy change during protein-ligand complex formation includes enthalphic and entropic components, which together report on the binding affinity and conformational states of the complex. However, determining the entropic contribution is computationally burdensome. Here, we apply kinematic flexibility analysis (KFA) to efficiently estimate vibrational frequencies from static protein and protein-ligand structures. The vibrational frequencies, in turn, determine the vibrational entropies of the structures and their complexes. Our estimates of the vibrational entropy change caused by ligand binding compare favorably to values obtained from a dynamic Normal Mode Analysis (NMA). Higher correlation factors can be achieved by increasing the distance cutoff in the potential energy model. Furthermore, we apply our new method to analyze the entropy changes of the SARS CoV-2 main protease when binding with different ligand inhibitors, which is relevant for the design of potential drugs.
Collapse
Affiliation(s)
- Xiyu Chen
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Sigrid Leyendecker
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Henry van den Bedem
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 94720 San Francisco, California, United States
| |
Collapse
|
16
|
Antiviral Effects of Artemisinin and Its Derivatives against SARS-CoV-2 Main Protease: Computational Evidences and Interactions with ACE2 Allelic Variants. Pharmaceuticals (Basel) 2022; 15:ph15020129. [PMID: 35215242 PMCID: PMC8877620 DOI: 10.3390/ph15020129] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Fighting against the emergent coronavirus disease (COVID-19) remains a big challenge at the front of the world communities. Recent research has outlined the potential of various medicinal herbs to counteract the infection. This study aimed to evaluate the interaction of artemisinin, a sesquiterpene lactone extracted from the Artemisia genus, and its derivatives with the SARS-CoV-2 main protease. To assess their potential use against COVID-19, the interactions of the main active principle of Artemisia with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro) was investigated through in silico probing. Our results showed that artemesinin and its derivatives manifested good oral absorption and bioavailability scores (0.55). They potently bound to the Mpro site of action—specifically, to its Cys145 residue. The selected compounds established two to three conventional hydrogen bonds with binding affinities ranging between −5.2 and −8.1 kcal/mol. Furthermore, artemisinin interactions with angiotensin converting enzyme 2 (ACE2) were dependent on the ACE2 allelic variants. The best score was recorded with rs961360700. A molecular dynamic simulation showed sufficient stability of the artemisinin–Mpro complex on the trajectory of 100 ns simulation frame. These binding interactions, together with drug-likeness and pharmacokinetic findings, confirmed that artemisinin might inhibit Mpro activity and explain the ethnopharmacological use of the herb and its possible antiviral activity against SARS-CoV-2 infection inducing COVID-19. Nevertheless, it interacted differently with the various ACE2 allelic variants reported to bind with the SARS-CoV-2 spike protein.
Collapse
|
17
|
Hasib RA, Ali MC, Rahman MS, Rahman MM, Ahmed FF, Mashud MAA, Islam MA, Jamal MAHM. A computational biology approach for the identification of potential SARS-CoV-2 main protease inhibitors from natural essential oil compounds. F1000Res 2021. [DOI: 10.12688/f1000research.73999.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has fomented a climate of fear worldwide due to its rapidly spreading nature, and high mortality rate. The World Health Organization (WHO) declared it as a global pandemic on 11th March, 2020. Many endeavors have been made to find appropriate medications to restrain the SARS CoV-2 infection from spreading but there is no specific antiviral therapy to date. However, a computer-aided drug design approach can be an alternative to identify probable drug candidates within a short time. SARS-CoV-2 main protease is a proven drug target, and it plays a pivotal role in viral replication and transcription. Methods: In this study, we identified a total of 114 essential oil compounds as a feasible anti-SARS-CoV-2 agent from several online reservoirs. These compounds were screened by incorporating ADMET profiling, molecular docking, and 50 ns of molecular dynamics simulation to identify potential drug candidates against the SARS-CoV-2 main protease. The crystallized SARS-CoV-2 main protease structure was collected from the RCSB PDB database (PDB ID 6LU7). Results: According to the results of the ADMET study, none of the compounds have any side effects that could reduce their druglikeness or pharmacokinetic properties. Out of 114 compounds, we selected bisabololoxide B, eremanthin, and leptospermone as our top drug candidates based on their higher binding affinity scores, and strong interaction with the Cys 145-His 41 catalytic dyad. Finally, the molecular dynamics simulation was implemented to evaluate the structural stability of the ligand-receptor complex. MD simulations disclosed that all the hits showed conformational stability compared to the positive control α-ketoamide. Conclusions: Our study showed that the top three hits might work as potential anti-SARS-CoV-2 agents, which can pave the way for discovering new drugs, but for experimental validation, they will require more in vivo trials.
Collapse
|
18
|
Al-Wahaibi LH, Mostafa A, Mostafa YA, Abou-Ghadir OF, Abdelazeem AH, Gouda AM, Kutkat O, Abo Shama NM, Shehata M, Gomaa HAM, Abdelrahman MH, Mohamed FAM, Gu X, Ali MA, Trembleau L, Youssif BGM. Discovery of novel oxazole-based macrocycles as anti-coronaviral agents targeting SARS-CoV-2 main protease. Bioorg Chem 2021; 116:105363. [PMID: 34555629 PMCID: PMC8445767 DOI: 10.1016/j.bioorg.2021.105363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 12/25/2022]
Abstract
We have discovered a family of synthetic oxazole-based macrocycles to be active against SARS-CoV-2. The synthesis, pharmacological properties, and docking studies of the compounds are reported in this study. The structure of the new macrocycles was confirmed by NMR spectroscopy and mass spectrometry. Compounds 13, 14, and 15a-c were evaluated for their anti-SARS-CoV-2 activity on SARS-COV-2 (NRC-03-nhCoV) virus in Vero-E6 cells. Isopropyl triester 13 and triacid 14 demonstrated superior inhibitory activities against SARS-CoV-2 compared to carboxamides 15a-c. MTT cytotoxicity assays showed that the CC50 (50% cytotoxicity concentration) of 13, 14, and 15a-c ranged from 159.1 to 741.8 μM and their safety indices ranged from 2.50 to 39.1. Study of the viral inhibition via different mechanisms of action (viral adsorption, replication, or virucidal property) showed that 14 had mild virucidal (60%) and inhibitory effects on virus adsorption (66%) at 20 μM concentrations. Compound 13 displayed several inhibitory effects at three levels, but the potency of its action is primarily virucidal. The inhibitory activity of compounds 13, 14, and 15a-c against the enzyme SARS-CoV-2 Mpro was evaluated. Isopropyl triester 13 had a significant inhibition activity against SARS-CoV-2 Mpro with an IC50 of 2.58 µM. Large substituents on the macrocyclic template significantly reduced the inhibitory effects of the compounds. Study of the docking of the compounds in the SARS CoV-2-Mpro active site showed that the most potent macrocycles 13 and 14 exhibited the best fit and highest affinity for the active site binding pocket. Taken together, the present study shows that the new macrocyclic compounds constitute a new family of SARS CoV-2-Mpro inhibitors that are worth being further optimized and developed.
Collapse
Affiliation(s)
- Lamya H Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah Bint Abdulrahman University, Saudi Arabia.
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Yaser A Mostafa
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Ola F Abou-Ghadir
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Ahmed H Abdelazeem
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Riyadh Elm University, Riyadh 11681, Saudi Arabia
| | - Ahmed M Gouda
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Noura M Abo Shama
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Mahmoud Shehata
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Hesham A M Gomaa
- Pharmacology Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Mostafa H Abdelrahman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt; Chemistry Department, School of Natural and Computing Sciences, University of Aberdeen, Meston Building, Aberdeen AB24 3UE, United Kingdom
| | - Fatma A M Mohamed
- Clinical Laboratory Science Department, College of Applied Medical Sciences, Jouf University, Aljouf 72341, Saudi Arabia; Chemistry Department, Faculty of Science, Alexandria University, Alexandria 21321, Egypt
| | - Xuyuan Gu
- Chemistry Department, School of Natural and Computing Sciences, University of Aberdeen, Meston Building, Aberdeen AB24 3UE, United Kingdom
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Laurent Trembleau
- Chemistry Department, School of Natural and Computing Sciences, University of Aberdeen, Meston Building, Aberdeen AB24 3UE, United Kingdom.
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| |
Collapse
|
19
|
Selvaraj C, Dinesh DC, Krafcikova P, Boura E, Aarthy M, Pravin MA, Singh SK. Structural Understanding of SARS-CoV-2 Drug Targets, Active Site Contour Map Analysis and COVID-19 Therapeutics. Curr Mol Pharmacol 2021; 15:418-433. [PMID: 34488601 DOI: 10.2174/1874467214666210906125959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
The most iconic word of the year 2020 is 'COVID-19', the shortened name for coronavirus disease 2019. The pandemic, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is responsible for multiple worldwide lockdowns, an economic crisis, and a substantial increase in hospitalizations for viral pneumonia along with respiratory failure and multiorgan dysfunctions. Recently, the first few vaccines were approved by World Health Organization (WHO) and can eventually save millions of lives. Even though, few emergency use drugs like Remdesivir and several other repurposed drugs, still there is no approved drug for COVID-19. The coronaviral encoded proteins involved in host-cell entry, replication, and host-cell invading mechanism are potentially therapeutic targets. This perspective review provides the molecular overview of SARS-CoV-2 life cycle for summarizing potential drug targets, structural insights, active site contour map analyses of those selected SARS-CoV-2 protein targets for drug discovery, immunology, and pathogenesis.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi-630004, Tamil Nadu. India
| | | | - Petra Krafcikova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2, 166 10 Prague 6. Czech Republic
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2, 166 10 Prague 6. Czech Republic
| | - Murali Aarthy
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi-630004, Tamil Nadu. India
| | - Muthuraja Arun Pravin
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi-630004, Tamil Nadu. India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi-630004, Tamil Nadu. India
| |
Collapse
|
20
|
Mahmud S, Biswas S, Kumar Paul G, Mita MA, Afrose S, Robiul Hasan M, Sharmin Sultana Shimu M, Uddin MAR, Salah Uddin M, Zaman S, Kaderi Kibria KM, Arif Khan M, Bin Emran T, Abu Saleh M. Antiviral peptides against the main protease of SARS-CoV-2: A molecular docking and dynamics study. ARAB J CHEM 2021; 14:103315. [PMID: 34909064 PMCID: PMC8277949 DOI: 10.1016/j.arabjc.2021.103315] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
The recent coronavirus outbreak has changed the world's economy and health sectors due to the high mortality and transmission rates. Because the development of new effective vaccines or treatments against the virus can take time, an urgent need exists for the rapid development and design of new drug candidates to combat this pathogen. Here, we obtained antiviral peptides obtained from the data repository of antimicrobial peptides (DRAMP) and screened their predicted tertiary structures for the ability to inhibit the main protease of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using multiple combinatorial docking programs, including PatchDock, FireDock, and ClusPro. The four best peptides, DRAMP00877, DRAMP02333, DRAMP02669, and DRAMP03804, had binding energies of -1125.3, -1084.5, -1005.2, and -924.2 Kcal/mol, respectively, as determined using ClusPro, and binding energies of -55.37, -50.96, -49.25, -54.81 Kcal/mol, respectively, as determined using FireDock, which were better binding energy values than observed for other peptide molecules. These peptides were found to bind with the active cavity of the SARS-CoV-2 main protease; at Glu166, Cys145, Asn142, Phe140, and Met165, in addition to the substrate-binding sites, Domain 2 and Domain 3, whereas fewer interactions were observed with Domain 1. The docking studies were further confirmed by a molecular dynamics simulation study, in which several descriptors, including the root-mean-square difference (RMSD), root-mean-square fluctuation (RMSF), solvent-accessible surface area (SASA), radius of gyration (Rg), and hydrogen bond formation, confirmed the stable nature of the peptide-main protease complexes. Toxicity and allergenicity studies confirmed the non-allergenic nature of the peptides. This present study suggests that these identified antiviral peptide molecules might inhibit the main protease of SARS-CoV-2, although further wet-lab experiments remain necessary to verify these findings.
Collapse
Affiliation(s)
- Shafi Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Suvro Biswas
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Gobindo Kumar Paul
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mohasana Akter Mita
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Shamima Afrose
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Robiul Hasan
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mst Sharmin Sultana Shimu
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mohammad Abu Raihan Uddin
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong, Bangladesh
| | - Md Salah Uddin
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Shahriar Zaman
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - K M Kaderi Kibria
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Arif Khan
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University, Chittagong 4381, Bangladesh
| | - Md Abu Saleh
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| |
Collapse
|
21
|
Padhi AK, Rath SL, Tripathi T. Accelerating COVID-19 Research Using Molecular Dynamics Simulation. J Phys Chem B 2021; 125:9078-9091. [PMID: 34319118 PMCID: PMC8340580 DOI: 10.1021/acs.jpcb.1c04556] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/12/2021] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic has emerged as a global medico-socio-economic disaster. Given the lack of effective therapeutics against SARS-CoV-2, scientists are racing to disseminate suggestions for rapidly deployable therapeutic options, including drug repurposing and repositioning strategies. Molecular dynamics (MD) simulations have provided the opportunity to make rational scientific breakthroughs in a time of crisis. Advancements in these technologies in recent years have become an indispensable tool for scientists studying protein structure, function, dynamics, interactions, and drug discovery. Integrating the structural data obtained from high-resolution methods with MD simulations has helped in comprehending the process of infection and pathogenesis, as well as the SARS-CoV-2 maturation in host cells, in a short duration of time. It has also guided us to identify and prioritize drug targets and new chemical entities, and to repurpose drugs. Here, we discuss how MD simulation has been explored by the scientific community to accelerate and guide translational research on SARS-CoV-2 in the past year. We have also considered future research directions for researchers, where MD simulations can help fill the existing gaps in COVID-19 research.
Collapse
Affiliation(s)
- Aditya K. Padhi
- Laboratory for Structural Bioinformatics, Center for
Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi,
Yokohama, Kanagawa 230-0045, Japan
| | - Soumya Lipsa Rath
- Department of Biotechnology, National
Institute of Technology, Warangal, Telangana 506004,
India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory,
Department of Biochemistry, North-Eastern Hill University,
Shillong 793022, India
| |
Collapse
|
22
|
Pluskota-Karwatka D, Hoffmann M, Barciszewski J. Reducing SARS-CoV-2 pathological protein activity with small molecules. J Pharm Anal 2021; 11:383-397. [PMID: 33842018 PMCID: PMC8020608 DOI: 10.1016/j.jpha.2021.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/21/2021] [Accepted: 03/29/2021] [Indexed: 01/18/2023] Open
Abstract
Coronaviruses are dangerous human and animal pathogens. The newly identified coronavirus SARS-CoV-2 is the causative agent of COVID-19 outbreak, which is a real threat to human health and life. The world has been struggling with this epidemic for about a year, yet there are still no targeted drugs and effective treatments are very limited. Due to the long process of developing new drugs, reposition of existing ones is one of the best ways to deal with an epidemic of emergency infectious diseases. Among the existing drugs, there are candidates potentially able to inhibit the SARS-CoV-2 replication, and thus inhibit the infection of the virus. Some therapeutics target several proteins, and many diseases share molecular paths. In such cases, the use of existing pharmaceuticals for more than one purpose can reduce the time needed to design new drugs. The aim of this review was to analyze the key targets of viral infection and potential drugs acting on them, as well as to discuss various strategies and therapeutic approaches, including the possible use of natural products. We highlighted the approach based on increasing the involvement of human deaminases, particularly APOBEC deaminases in editing of SARS-CoV-2 RNA. This can reduce the cytosine content in the viral genome, leading to the loss of its integrity. We also indicated the nucleic acid technologies as potential approaches for COVID-19 treatment. Among numerous promising natural products, we pointed out curcumin and cannabidiol as good candidates for being anti-SARS-CoV-2 agents.
Collapse
Affiliation(s)
| | - Marcin Hoffmann
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, 61-614, Poznań, Poland
| | - Jan Barciszewski
- NanoBiomedical Center of the Adam Mickiewicz University, 61-614, Poznań, Poland
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences 61-704, Poznań, Poland
| |
Collapse
|
23
|
Sladek V, Yamamoto Y, Harada R, Shoji M, Shigeta Y, Sladek V. pyProGA-A PyMOL plugin for protein residue network analysis. PLoS One 2021; 16:e0255167. [PMID: 34329304 PMCID: PMC8323899 DOI: 10.1371/journal.pone.0255167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/11/2021] [Indexed: 11/18/2022] Open
Abstract
The field of protein residue network (PRN) research has brought several useful methods and techniques for structural analysis of proteins and protein complexes. Many of these are ripe and ready to be used by the proteomics community outside of the PRN specialists. In this paper we present software which collects an ensemble of (network) methods tailored towards the analysis of protein-protein interactions (PPI) and/or interactions of proteins with ligands of other type, e.g. nucleic acids, oligosaccharides etc. In parallel, we propose the use of the network differential analysis as a method to identify residues mediating key interactions between proteins. We use a model system, to show that in combination with other, already published methods, also included in pyProGA, it can be used to make such predictions. Such extended repertoire of methods allows to cross-check predictions with other methods as well, as we show here. In addition, the possibility to construct PRN models from various kinds of input is so far a unique asset of our code. One can use structural data as defined in PDB files and/or from data on residue pair interaction energies, either from force-field parameters or fragment molecular orbital (FMO) calculations. pyProGA is a free open-source software available from https://gitlab.com/Vlado_S/pyproga.
Collapse
Affiliation(s)
- Vladimir Sladek
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro, Tokyo, Japan
| | - Ryuhei Harada
- Center for Computational Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mitsuo Shoji
- Center for Computational Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Vladimir Sladek
- Institute of Construction and Architecture, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
24
|
Lata S, Akif M. Comparative protein structure network analysis on 3CL pro from SARS-CoV-1 and SARS-CoV-2. Proteins 2021; 89:1216-1225. [PMID: 33983654 PMCID: PMC8242809 DOI: 10.1002/prot.26143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/10/2021] [Accepted: 05/05/2021] [Indexed: 12/29/2022]
Abstract
The main protease Mpro, 3CLpro is an important target from coronaviruses. In spite of having 96% sequence identity among Mpros from SARS‐CoV‐1 and SARS‐CoV‐2; the inhibitors used to block the activity of SARS‐CoV‐1 Mpro so far, were found to have differential inhibitory effect on Mpro of SARS‐CoV‐2. The possible reason could be due to the difference of few amino acids among the peptidases. Since, overall 3‐D crystallographic structure of Mpro from SARS‐CoV‐1 and SARS‐CoV‐2 is quite similar and mapping a subtle structural variation is seemingly impossible. Hence, we have attempted to study a structural comparison of SARS‐CoV‐1 and SARS‐CoV‐2 Mpro in apo and inhibitor bound states using protein structure network (PSN) based approach at contacts level. The comparative PSNs analysis of apo Mpros from SARS‐CoV‐1 and SARS‐CoV‐2 uncovers small but significant local changes occurring near the active site region and distributed throughout the structure. Additionally, we have shown how inhibitor binding perturbs the PSG and the communication pathways in Mpros. Moreover, we have also investigated the network connectivity on the quaternary structure of Mpro and identified critical residue pairs for complex formation using three centrality measurement parameters along with the modularity analysis. Taken together, these results on the comparative PSN provide an insight into conformational changes that may be used as an additional guidance towards specific drug development.
Collapse
Affiliation(s)
- Surabhi Lata
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Mohd Akif
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
25
|
Abstract
In the global health emergency caused by coronavirus disease 2019 (COVID-19), efficient and specific therapies are urgently needed. Compared with traditional small-molecular drugs, antibody therapies are relatively easy to develop; they are as specific as vaccines in targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); and they have thus attracted much attention in the past few months. This article reviews seven existing antibodies for neutralizing SARS-CoV-2 with 3D structures deposited in the Protein Data Bank (PDB). Five 3D antibody structures associated with the SARS-CoV spike (S) protein are also evaluated for their potential in neutralizing SARS-CoV-2. The interactions of these antibodies with the S protein receptor-binding domain (RBD) are compared with those between angiotensin-converting enzyme 2 and RBD complexes. Due to the orders of magnitude in the discrepancies of experimental binding affinities, we introduce topological data analysis, a variety of network models, and deep learning to analyze the binding strength and therapeutic potential of the 14 antibody-antigen complexes. The current COVID-19 antibody clinical trials, which are not limited to the S protein target, are also reviewed.
Collapse
Affiliation(s)
- Jiahui Chen
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, USA;
| | - Kaifu Gao
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, USA;
| | - Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, USA;
| | - Duc Duy Nguyen
- Department of Mathematics, University of Kentucky, Lexington, Kentucky 40506, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, USA;
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
26
|
Amaral JL, Oliveira JTA, Lopes FES, Freitas CDT, Freire VN, Abreu LV, Souza PFN. Quantum biochemistry, molecular docking, and dynamics simulation revealed synthetic peptides induced conformational changes affecting the topology of the catalytic site of SARS-CoV-2 main protease. J Biomol Struct Dyn 2021; 40:8925-8937. [PMID: 33949286 PMCID: PMC8108194 DOI: 10.1080/07391102.2021.1920464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
The recent outbreak caused by SARS-CoV-2 continues to threat and take many lives all over the world. The lack of an efficient pharmacological treatments are serious problems to be faced by scientists and medical staffs worldwide. In this work, an in silico approach based on the combination of molecular docking, dynamics simulations, and quantum biochemistry revealed that the synthetic peptides RcAlb-PepI, PepGAT, and PepKAA, strongly interact with the main protease (Mpro) a pivotal protein for SARS-CoV-2 replication. Although not binding to the proteolytic site of SARS-CoV-2 Mpro, RcAlb-PepI, PepGAT, and PepKAA interact with other protein domain and allosterically altered the protease topology. Indeed, such peptide-SARS-CoV-2 Mpro complexes provoked dramatic alterations in the three-dimensional structure of Mpro leading to area and volume shrinkage of the proteolytic site, which could affect the protease activity and thus the virus replication. Based on these findings, it is suggested that RcAlb-PepI, PepGAT, and PepKAA could interfere with SARS-CoV-2 Mpro role in vivo. Also, unlike other antiviral drugs, these peptides have no toxicity to human cells. This pioneering in silico investigation opens up opportunity for further in vivo research on these peptides, towards discovering new drugs and entirely new perspectives to treat COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jackson L. Amaral
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Jose T. A. Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Francisco E. S. Lopes
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
- Center for Permanent Education in Health Care, CEATS/School of Public Health of Ceará-ESP-CE, Fortaleza, Brazil
| | - Cleverson D. T. Freitas
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Valder N. Freire
- Department of Physics, Federal University of Ceará, Fortaleza, Brazil
| | - Leonardo V. Abreu
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Pedro F. N. Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
27
|
Bhat ZA, Chitara D, Iqbal J, Sanjeev BS, Madhumalar A. Targeting allosteric pockets of SARS-CoV-2 main protease M pro. J Biomol Struct Dyn 2021; 40:6603-6618. [PMID: 33645457 DOI: 10.1080/07391102.2021.1891141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Repurposing of antivirals is an attractive therapeutic option for the treatment of COVID-19. Main protease (Mpro), also called 3 C-like protease (3CLpro) is a key protease of SARS-CoV-2 involved in viral replication, and is a promising drug target for antivirals. A major challenge to test the efficacy of antivirals is the conformational plasticity of Mpro and its future mutation prone flexibility. Suitable choice of drugs in catalytic and allosteric pockets appear to be essential for combination therapy. Current study, based on docking and extensive set of MD simulations, finds the combination of Elbasvir, Glecaprevir and Ritonavir to be a viable candidate for further experimental drug testing/pharmacophore design for Mpro.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zahoor Ahmad Bhat
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Dheeraj Chitara
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Allahabad, Uttar Pradesh, India
| | - Jawed Iqbal
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - B S Sanjeev
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Allahabad, Uttar Pradesh, India
| | - Arumugam Madhumalar
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
28
|
Wang R, Chen J, Gao K, Hozumi Y, Yin C, Wei GW. Analysis of SARS-CoV-2 mutations in the United States suggests presence of four substrains and novel variants. Commun Biol 2021; 4:228. [PMID: 33589648 PMCID: PMC7884689 DOI: 10.1038/s42003-021-01754-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 has been mutating since it was first sequenced in early January 2020. Here, we analyze 45,494 complete SARS-CoV-2 geneome sequences in the world to understand their mutations. Among them, 12,754 sequences are from the United States. Our analysis suggests the presence of four substrains and eleven top mutations in the United States. These eleven top mutations belong to 3 disconnected groups. The first and second groups consisting of 5 and 8 concurrent mutations are prevailing, while the other group with three concurrent mutations gradually fades out. Moreover, we reveal that female immune systems are more active than those of males in responding to SARS-CoV-2 infections. One of the top mutations, 27964C > T-(S24L) on ORF8, has an unusually strong gender dependence. Based on the analysis of all mutations on the spike protein, we uncover that two of four SASR-CoV-2 substrains in the United States become potentially more infectious.
Collapse
Affiliation(s)
- Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, MI, 48824, USA
| | - Jiahui Chen
- Department of Mathematics, Michigan State University, East Lansing, MI, 48824, USA
| | - Kaifu Gao
- Department of Mathematics, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuta Hozumi
- Department of Mathematics, Michigan State University, East Lansing, MI, 48824, USA
| | - Changchuan Yin
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
29
|
Verma S, Pandey AK. Factual insights of the allosteric inhibition mechanism of SARS-CoV-2 main protease by quercetin: an in silico analysis. 3 Biotech 2021; 11:67. [PMID: 33457176 PMCID: PMC7802979 DOI: 10.1007/s13205-020-02630-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/28/2020] [Indexed: 01/19/2023] Open
Abstract
SARS-CoV-2 main protease (Mpro) cleaves the viral polypeptide 1a and 1ab in a site-specific ((L-Q|(S, A, G)) manner and produce functional enzymes for mediating viral replication. Numerous studies have reported synthetic competitive inhibitors against this target enzyme but increase in substrate concentration often reduces the effectiveness of such inhibitors. Allosteric inhibition by natural compound can provide safe and effective treatment by alleviating this limitation. Present study deals with in silico allosteric inhibition analysis of quercetin, against SARS-CoV-2-Mpro. Molecular docking of quercetin with Mpro revealed consistent binding of quercetin at a site other than active site in multiple runs, with the highest binding energy of - 8.31 kcal/mol, forming 6 H-bonds with residues Gln127, Cys128, Lys137, Asp289 and Glu290. Molecular dynamic simulation of 50 ns revealed high stability of Mpro-quercetin complex with RMSD values ranging from 0.1 to 0.25 nm. Moreover, native-Mpro and Mpro-quercetin complex conformations extracted at different time points from simulation trajectories were subjected to active site-specific docking with modelled substrate peptide (AVLQSGFR) by ZDOCK server. Results displayed site-specific cleavage of peptide when docked with native-Mpro. While substrate peptide remained intact when docked with Mpro-quercetin complex, also the binding energy of peptide reduced from 785 to 86 from 1 to 50 ns as quercetin induced alterations in the active site cavity reducing its affinity for the substrate. Further, no interactions were noticed between peptide and active site residues of Mpro-quercetin complex conformations at 40 and 50 ns. Hence, quercetin displayed effective allosteric inhibition potential against SARS-CoV-2 Mpro, and can be developed into an efficient treatment for COVID-19.
Collapse
Affiliation(s)
- Shalja Verma
- Department of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi, Uttar Pradesh 284128 India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi, 110016 India
| | - Anand Kumar Pandey
- Department of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi, Uttar Pradesh 284128 India
| |
Collapse
|
30
|
Dubanevics I, McLeish TCB. Computational analysis of dynamic allostery and control in the SARS-CoV-2 main protease. J R Soc Interface 2021; 18:20200591. [PMID: 33402024 PMCID: PMC7879766 DOI: 10.1098/rsif.2020.0591] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/07/2020] [Indexed: 01/06/2023] Open
Abstract
The COVID-19 pandemic caused by the novel coronavirus SARS-CoV-2 has no publicly available vaccine or antiviral drugs at the time of writing. An attractive coronavirus drug target is the main protease (Mpro, also known as 3CLpro) because of its vital role in the viral cycle. A significant body of work has been focused on finding inhibitors which bind and block the active site of the main protease, but little has been done to address potential non-competitive inhibition, targeting regions other than the active site, partly because the fundamental biophysics of such allosteric control is still poorly understood. In this work, we construct an elastic network model (ENM) of the SARS-CoV-2 Mpro homodimer protein and analyse its dynamics and thermodynamics. We found a rich and heterogeneous dynamical structure, including allosterically correlated motions between the homodimeric protease's active sites. Exhaustive 1-point and 2-point mutation scans of the ENM and their effect on fluctuation free energies confirm previously experimentally identified bioactive residues, but also suggest several new candidate regions that are distant from the active site, yet control the protease function. Our results suggest new dynamically driven control regions as possible candidates for non-competitive inhibiting binding sites in the protease, which may assist the development of current fragment-based binding screens. The results also provide new insights into the active biophysical research field of protein fluctuation allostery and its underpinning dynamical structure.
Collapse
Affiliation(s)
- Igors Dubanevics
- School of Natural Sciences, University of York, York, UK
- Department of Physics, University of York, York, UK
| | | |
Collapse
|
31
|
Wang R, Chen J, Hozumi Y, Yin C, Wei GW. Decoding Asymptomatic COVID-19 Infection and Transmission. J Phys Chem Lett 2020; 11:10007-10015. [PMID: 33179934 PMCID: PMC8150094 DOI: 10.1021/acs.jpclett.0c02765] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
One of the major challenges in controlling the coronavirus disease 2019 (COVID-19) outbreak is its asymptomatic transmission. The pathogenicity and virulence of asymptomatic COVID-19 remain mysterious. On the basis of the genotyping of 75775 SARS-CoV-2 genome isolates, we reveal that asymptomatic infection is linked to SARS-CoV-2 11083G>T mutation (i.e., L37F at nonstructure protein 6 (NSP6)). By analyzing the distribution of 11083G>T in various countries, we unveil that 11083G>T may correlate with the hypotoxicity of SARS-CoV-2. Moreover, we show a global decaying tendency of the 11083G>T mutation ratio indicating that 11083G>T hinders the SARS-CoV-2 transmission capacity. Artificial intelligence, sequence alignment, and network analysis are applied to show that NSP6 mutation L37F may have compromised the virus's ability to undermine the innate cellular defense against viral infection via autophagy regulation. This assessment is in good agreement with our genotyping of the SARS-CoV-2 evolution and transmission across various countries and regions over the past few months.
Collapse
Affiliation(s)
| | | | | | - Changchuan Yin
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | | |
Collapse
|
32
|
Hâncean MG, Slavinec M, Perc M. The impact of human mobility networks on the global spread of COVID-19. JOURNAL OF COMPLEX NETWORKS 2020; 8:cnaa041. [PMID: 34191993 PMCID: PMC7989546 DOI: 10.1093/comnet/cnaa041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/14/2020] [Indexed: 05/23/2023]
Abstract
Human mobility networks are crucial for a better understanding and controlling the spread of epidemics. Here, we study the impact of human mobility networks on the COVID-19 onset in 203 different countries. We use exponential random graph models to perform an analysis of the country-to-country global spread of COVID-19. We find that most countries had similar levels of virus spreading, with only a few acting as the main global transmitters. Our evidence suggests that migration and tourism inflows increase the probability of COVID-19 case importations while controlling for contiguity, continent co-location and sharing a language. Moreover, we find that air flights were the dominant mode of transportation while male and returning travellers were the main carriers. In conclusion, a mix of mobility and geography factors predicts the COVID-19 global transmission from one country to another. These findings have implications for non-pharmaceutical public health interventions and the management of transborder human circulation.
Collapse
Affiliation(s)
| | - Mitja Slavinec
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Matjaž Perc
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan & Complexity Science Hub Vienna, Vienna, Austria
| |
Collapse
|
33
|
Singh N, Rai SN, Singh V, Singh MP. Molecular characterization, pathogen-host interaction pathway and in silico approaches for vaccine design against COVID-19. J Chem Neuroanat 2020; 110:101874. [PMID: 33091590 PMCID: PMC7571424 DOI: 10.1016/j.jchemneu.2020.101874] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022]
Abstract
COVID-19 has forsaken the world because of extremely high infection rates and high mortality rates. At present we have neither medicine nor vaccine to prevent this pandemic. Lockdowns, curfews, isolations, quarantines, and social distancing are the only ways to mitigate their infection. This is badly affecting the mental health of people. Hence, there is an urgent need to address this issue. Coronavirus disease 2019 (COVID-19) is caused by a novel Betacorona virus named SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) which has emerged in the city of Wuhan in China and declared a pandemic by WHO since it affected almost all the countries the world, infected 24,182,030 people and caused 825,798 death as per data are compiled from John Hopkins University (JHU). The genome of SARS-CoV-2 has a single-stranded positive (+) sense RNA of ∼30 kb nucleotides. Phylogenetic analysis reveals that SARS-CoV-2 shares the highest nucleotide sequence similarity (∼79 %) with SARS-CoV. Envelope and nucleocapsids are two evolutionary conserved regions of SARS-CoV-2 having a sequence identity of about 96 % and 89.6 %, respectively as compared to SARS-CoV. The characterization of SARS-CoV-2 is based on polymerase chain reaction (PCR) and metagenomic next-generation sequencing. Transmission of this virus in the human occurs through the respiratory tract and decreases the respiration efficiency of lungs. Humans are generally susceptible to SARS-CoV-2 with an incubation period of 2-14 days. The virus first infects the lower airway and bind with angiotensin-converting enzyme 2 (ACE2) of alveolar epithelial cells. Due to the unavailability of drugs or vaccines, it is very urgent to design potential vaccines or drugs for COVID-19. Reverse vaccinology and immunoinformatic play an important role in designing potential vaccines against SARS-CoV-2. The suitable vaccine selects for SARS-CoV-2 based on binding energy between the target protein and the designed vaccine. The stability and activity of the designed vaccine can be estimated by using molecular docking and dynamic simulation approaches. This review mainly focused on the brief up to date information about COVID-19, molecular characterization, pathogen-host interaction pathways involved during COVID-19 infection. It also covers potential vaccine design against COVID-19 by using various computational approaches. SARS-CoV-2 enters brain tissue through the different pathway and harm human's brain and causes severe neurological disruption.
Collapse
Affiliation(s)
- Nidhi Singh
- Centre of Bioinformatics, University of Allahabad, Prayagraj, 211002, India
| | - Sachchida Nand Rai
- Centre of Biotechnology, University of Allahabad, Prayagraj, 211002, India
| | - Veer Singh
- School of Biochemical Engineering, IIT (BHU) Varanasi, 221005, India
| | - Mohan P Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj, 211002, India.
| |
Collapse
|
34
|
Vlachakis D, Papakonstantinou E, Mitsis T, Pierouli K, Diakou I, Chrousos G, Bacopoulou F. Molecular mechanisms of the novel coronavirus SARS-CoV-2 and potential anti-COVID19 pharmacological targets since the outbreak of the pandemic. Food Chem Toxicol 2020; 146:111805. [PMID: 33038452 PMCID: PMC7543766 DOI: 10.1016/j.fct.2020.111805] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
The novel coronavirus SARS-CoV-2 has emerged as a severe threat against public health and global economies. COVID-19, the disease caused by this virus, is highly contagious and has led to an ongoing pandemic. SARS-CoV-2 affects, mainly, the respiratory system, with most severe cases primarily showcasing acute respiratory distress syndrome. Currently, no targeted therapy exists, and since the number of infections and death toll keeps rising, it has become a necessity to study possible therapeutic targets. Antiviral drugs can target various stages of the viral infection, and in the case of SARS-CoV-2, both structural and non-structural proteins have been proposed as potential drug targets. This review focuses on the most researched SARS-CoV-2 proteins, their structure, function, and possible therapeutic approaches.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, Athens, 11855, Greece; University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 8 Levadias Street, Athens, 11527, Greece; Lab of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece; Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Strand, London WC2R 2LS, UK
| | - Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, Athens, 11855, Greece
| | - Thanasis Mitsis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, Athens, 11855, Greece
| | - Katerina Pierouli
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, Athens, 11855, Greece
| | - Io Diakou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, Athens, 11855, Greece
| | - George Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 8 Levadias Street, Athens, 11527, Greece; Lab of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Street, Athens, 11527, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 8 Levadias Street, Athens, 11527, Greece.
| |
Collapse
|
35
|
Wang R, Chen J, Gao K, Hozumi Y, Yin C, Wei GW. Characterizing SARS-CoV-2 mutations in the United States. RESEARCH SQUARE 2020:rs.3.rs-49671. [PMID: 32818213 PMCID: PMC7430589 DOI: 10.21203/rs.3.rs-49671/v1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been mutating since it was first sequenced in early January 2020. The genetic variants have developed into a few distinct clusters with different properties. Since the United States (US) has the highest number of viral infected patients globally, it is essential to understand the US SARS-CoV-2. Using genotyping, sequence-alignment, time-evolution, k-means clustering, protein-folding stability, algebraic topology, and network theory, we reveal that the US SARS-CoV-2 has four substrains and five top US SARS-CoV-2 mutations were first detected in China (2 cases), Singapore (2 cases), and the United Kingdom (1 case). The next three top US SARS-CoV-2 mutations were first detected in the US. These eight top mutations belong to two disconnected groups. The first group consisting of 5 concurrent mutations is prevailing, while the other group with three concurrent mutations gradually fades out. We identify that one of the top mutations, 27964C>T-(S24L) on ORF8, has an unusually strong gender dependence. Based on the analysis of all mutations on the spike protein, we further uncover that three of four US SASR-CoV-2 substrains become more infectious. Our study calls for effective viral control and containing strategies in the US.
Collapse
Affiliation(s)
- Rui Wang
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Jiahui Chen
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Kaifu Gao
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Yuta Hozumi
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Changchuan Yin
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, MI 48824, USA
- Department of Electrical and Computer Engineering, Michigan State University, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI 48824, USA
| |
Collapse
|
36
|
Abadias L, Estrada-Rodriguez G, Estrada E. Fractional-Order Susceptible-Infected Model: Definition and Applications to the Study of COVID-19 Main Protease. FRACTIONAL CALCULUS & APPLIED ANALYSIS 2020; 23:635-655. [PMID: 34849076 PMCID: PMC8617368 DOI: 10.1515/fca-2020-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Indexed: 05/02/2023]
Abstract
We propose a model for the transmission of perturbations across the amino acids of a protein represented as an interaction network. The dynamics consists of a Susceptible-Infected (SI) model based on the Caputo fractional-order derivative. We find an upper bound to the analytical solution of this model which represents the worse-case scenario on the propagation of perturbations across a protein residue network. This upper bound is expressed in terms of Mittag-Leffler functions of the adjacency matrix of the network of inter-amino acids interactions. We then apply this model to the analysis of the propagation of perturbations produced by inhibitors of the main protease of SARS CoV-2. We find that the perturbations produced by strong inhibitors of the protease are propagated far away from the binding site, confirming the long-range nature of intra-protein communication. On the contrary, the weakest inhibitors only transmit their perturbations across a close environment around the binding site. These findings may help to the design of drug candidates against this new coronavirus.
Collapse
Affiliation(s)
- Luciano Abadias
- Departamento de Matemáticas, Facultad de Ciencias Universidad de Zaragoza, 50009 Zaragoza, Spain
- Instituto Universitario de Matemáticas y Aplicaciones, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | | | - Ernesto Estrada
- Instituto Universitario de Matemáticas y Aplicaciones, Universidad de Zaragoza, 50009 Zaragoza, Spain
- 50018, Zaragoza, Spain
| |
Collapse
|
37
|
Estrada E. COVID-19 and SARS-CoV-2. Modeling the present, looking at the future. PHYSICS REPORTS 2020; 869:1-51. [PMID: 32834430 PMCID: PMC7386394 DOI: 10.1016/j.physrep.2020.07.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 05/21/2023]
Abstract
Since December 2019 the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has produced an outbreak of pulmonary disease which has soon become a global pandemic, known as COronaVIrus Disease-19 (COVID-19). The new coronavirus shares about 82% of its genome with the one which produced the 2003 outbreak (SARS CoV-1). Both coronaviruses also share the same cellular receptor, which is the angiotensin-converting enzyme 2 (ACE2) one. In spite of these similarities, the new coronavirus has expanded more widely, more faster and more lethally than the previous one. Many researchers across the disciplines have used diverse modeling tools to analyze the impact of this pandemic at global and local scales. This includes a wide range of approaches - deterministic, data-driven, stochastic, agent-based, and their combinations - to forecast the progression of the epidemic as well as the effects of non-pharmaceutical interventions to stop or mitigate its impact on the world population. The physical complexities of modern society need to be captured by these models. This includes the many ways of social contacts - (multiplex) social contact networks, (multilayers) transport systems, metapopulations, etc. - that may act as a framework for the virus propagation. But modeling not only plays a fundamental role in analyzing and forecasting epidemiological variables, but it also plays an important role in helping to find cures for the disease and in preventing contagion by means of new vaccines. The necessity for answering swiftly and effectively the questions: could existing drugs work against SARS CoV-2? and can new vaccines be developed in time? demands the use of physical modeling of proteins, protein-inhibitors interactions, virtual screening of drugs against virus targets, predicting immunogenicity of small peptides, modeling vaccinomics and vaccine design, to mention just a few. Here, we review these three main areas of modeling research against SARS CoV-2 and COVID-19: (1) epidemiology; (2) drug repurposing; and (3) vaccine design. Therefore, we compile the most relevant existing literature about modeling strategies against the virus to help modelers to navigate this fast-growing literature. We also keep an eye on future outbreaks, where the modelers can find the most relevant strategies used in an emergency situation as the current one to help in fighting future pandemics.
Collapse
Affiliation(s)
- Ernesto Estrada
- Instituto Universitario de Matemáticas y Aplicaciones, Universidad de Zaragoza, 50009 Zaragoza, Spain
- ARAID Foundation, Government of Aragón, 50018 Zaragoza, Spain
| |
Collapse
|