Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Journal of Computational Biophysics & Chemistry ; : 1-16, 2023.
Article in English | Academic Search Complete | ID: covidwho-2319742

ABSTRACT

The COVID-19 pandemic raised an unprecedented race in biotechnology in search for effective therapies and a preventive vaccine. Scientists worldwide have been attempting to stop the viral infection by interfering with the biological function of the SARS-CoV-2 main protease (Mpro), a critical protein required for viral transcription and replication during infection. In this study, we employed an effective approach integrating deep learning model calculations and steered molecular dynamic simulations to generate highly promising inhibitors of SARS-CoV-2 Mpro. First, using deep learning calculations, a natural molecule that was identified as a potential inhibitor of SARS-CoV-2 Mpro was chemically altered to boost its ligand-binding affinity to the Mpro protease. The proposed compounds were then verified using steered molecular dynamic simulations to estimate their binding free energies to SARS-CoV-2 Mpro. The procedure was repeated until the binding free energies of the proposed compounds did not improve further. Overall, one proposed compound was shown to have a high nanomolar affinity, and two others were estimated to possess nanomolar affinities for SARS-CoV-2 Mpro, indicating that they are highly promising inhibitors of the protease. Absorption, distribution, metabolism, and excretion and toxicity analysis show that all three chemicals are drug-like compounds following the MACCS-II Drug Data Report database, orally absorbed, tightly attached to the plasma membrane, and noncarcinogenic in rats. The results obtained potentially support COVID-19 treatment. [ FROM AUTHOR] Copyright of Journal of Computational Biophysics & Chemistry is the property of World Scientific Publishing Company and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full . (Copyright applies to all s.)

2.
Chem Phys ; 569: 111859, 2023 May 01.
Article in English | MEDLINE | ID: covidwho-2271420

ABSTRACT

SARS-CoV-2-encoded accessory protein ORF3a was found to be a conserved coronavirus protein that shows crucial roles in apoptosis in cells as well as in virus release and replications. To complete the knowledge and identify the unknown of this protein, further comprehensive research is needed to clarify the leading role of ORF3a in the functioning of the coronavirus. One of the efficient approaches to determining the functionality of this protein is to investigate the mechanical properties and study its structural dynamics in the presence of physical stimuli. Herein, performing all-atom steered molecular dynamics (SMD) simulations, the mechanical properties of the force-bearing components of the ORF3a channel are calculated in different physiological conditions. As variations occurring in ORF3a may lead to alteration in protein structure and function, the G49V mutation was also simulated to clarify the relationship between the mechanical properties and chemical stability of the protein by comparing the behavior of the wild-type and mutant Orf3a. From a physiological conditions point of view, it was observed that in the solvated system, the presence of water molecules reduces Young's modulus of TM1 by ∼30 %. Our results also show that by substitution of Gly49 with valine, Young's modulus of the whole helix increases from 1.61 ± 0.20 to 2.08 ± 0.15 GPa, which is consistent with the calculated difference in free energy of wild-type and mutant helices. In addition to finding a way to fight against Covid-19 disease, understanding the mechanical behavior of these biological nanochannels can lead to the development of the potential applications of the ORF3a protein channel, such as tunable nanovalves in smart drug delivery systems, nanofilters in the new generation of desalination systems, and promising applications in DNA sequencing.

3.
Eur J Med Chem ; 251: 115226, 2023 May 05.
Article in English | MEDLINE | ID: covidwho-2280022

ABSTRACT

By combining docking and molecular dynamics simulations, we explored a library of 65 mostly axially chiral naphthylisoquinoline alkaloids and their analogues, with most different molecular architectures and structural analogues, for their activity against SARS-CoV-2. Although natural biaryls are often regarded without consideration of their axial chirality, they can bind to protein targets in an atroposelective manner. By combining docking results with steered molecular dynamics simulations, we identified one alkaloid, korupensamine A, that atropisomer-specifically inhibited the main protease (Mpro) activity of SARS-CoV-2 significantly in comparison to the reference covalent inhibitor GC376 (IC50 = 2.52 ± 0.14 and 0.88 ± 0.15 µM, respectively) and reduced viral growth by five orders of magnitude in vitro (EC50 = 4.23 ± 1.31 µM). To investigate the binding pathway and mode of interaction of korupensamine A within the active site of the protease, we utilized Gaussian accelerated molecular dynamics simulations, which reproduced the docking pose of korupensamine A inside the active site of the enzyme. The study presents naphthylisoquinoline alkaloids as a new class of potential anti-COVID-19 agents.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , Protease Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Hydrolases/metabolism
4.
J Mol Graph Model ; 118: 108345, 2023 01.
Article in English | MEDLINE | ID: covidwho-2239079

ABSTRACT

Human norovirus (HuNoV) causes acute viral gastroenteritis in all age groups, and dehydration and severe diarrhea in the elderly. The World Health Organization reports ∼1.45 million deaths from acute gastroenteritis annually in the world. Rupintrivir, an inhibitory medicine against the human rhinovirus C3 protease, has been reported to inhibit HuNoV 3C protease. However, several HuNoV 3C protease mutations have been revealed to reduce the susceptibility of HuNoV to rupintrivir. The structural details behind rupintrivir-resistance of these single-point mutations (A105V and I109V) are not still clear. Hence, in this study, a combination of computational techniques were used to determine the rupintrivir-resistance mechanism and to propose an inhibitor against wild-type and mutant HuNoV 3C protease through structure-based virtual screening. Dynamic structural results indicated the unstable binding of rupintrivir at the cleft binding site of the wild-type and mutant 3C proteases, leading to its detachment. Our findings presented that the domain II of the HuNoV 3C protease had a critical role in binding of inhibitory molecules. Binding energy computations, steered molecular dynamics and umbrella sampling simulations confirmed that amentoflavone, the novel suggested inhibitor, strongly binds to the cleft site of all protease models and has a good structural stability in the complex system along the molecular dynamic simulations. Our in silico study proposed the selected compound as a potential inhibitor against the HuNoV 3C protease. However, additional experimental and clinical studies are required to corroborate the therapeutic efficacy of the compound.


Subject(s)
Antiviral Agents , Norovirus , Protease Inhibitors , Humans , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Gastroenteritis/drug therapy , Gastroenteritis/virology , Norovirus/drug effects , Norovirus/metabolism , Peptide Hydrolases , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry
5.
Comput Struct Biotechnol J ; 20: 6078-6086, 2022.
Article in English | MEDLINE | ID: covidwho-2095240

ABSTRACT

SARS-CoV-2 variants often include surface mutations in the Spike protein that are important for viruses to recognize host receptors and evade antibody neutralization. The Spike protein also has mutations in the interior of the protein likely to affect the Spike protein S1 - S2 subunit's separation propensity, the most important of which is the D614G mutation. Remarkably, the Omicron variant contains a large number of internal mutations at the S2: S1 interface, which have not been investigated yet. In this study, we examined the effects of such interfacial mutations on the S2: S1 and subunit domain interactions and on the subunit's dissociation process. We found that the interaction with S2 is mainly contributed by the three encapsulation domains, named INT, ED1 and ED2 of S1, which are sandwiched between the S1 RBD and N-terminal NTD domain. We found that D614 is the strongest contributor for the S2: S1 interaction which is greatly weakened by the D614G mutation. Surprisingly, we found that, mutations T547K, H655Y, N764K, N856K, N969K, L981F in the Omicron variant largely enhance the S2: ED1 interaction, partially compensating the loss of S2: ED2 interaction due to the D614G mutation. Lastly, these results, together with biological considerations, allow us to suggest that in addition to the binding strength of between the RBD and ACE2, the stability of the Spike protein and the propensity of Spike protein S2: S1 separation are critical factors which likely exist in a balance for a particular infectivity and pathogenicity of the virus.

6.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: covidwho-2032988

ABSTRACT

The coronavirus E proteins are small membrane proteins found in the virus envelope of alpha and beta coronaviruses that have a high degree of overlap in their biochemical and functional properties despite minor sequence variations. The SARS-CoV-2 E is a 75-amino acid transmembrane protein capable of acting as an ion channel when assembled in a pentameric fashion. Various studies have found that hexamethylene amiloride (HMA) can inhibit the ion channel activity of the E protein in bilayers and also inhibit viral replication in cultured cells. Here, we use the available structural data in conjunction with homology modelling to build a comprehensive model of the E protein to assess potential binding sites and molecular interactions of HMA derivatives. Furthermore, we employed an iterative cycle of molecular modelling, extensive docking simulations, molecular dynamics and leveraging steered molecular dynamics to better understand the pore characteristics and quantify the affinity of the bound ligands. Results from this work highlight the potential of acylguanidines as blockers of the E protein and guide the development of subsequent small molecule inhibitors.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Amiloride/analogs & derivatives , Amiloride/pharmacology , Amino Acids , Humans , Ion Channels/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation
7.
Scientia Sinica Chimica ; 52(5):721-730, 2022.
Article in Chinese | Scopus | ID: covidwho-1892268

ABSTRACT

After the first large-scale outbreak of pneumonia (COVID-19) caused by novel coronavirus (SARS-CoV-2) infection in December 2019, it was declared a global pandemic by the World Health Organization in March 2020. Significant infectivity and lethality rate pose a huge threat to the life and safety of all mankind, and we urgently need to grasp the infection/transmission behavior of the virus at the molecular level. Novel coronavirus and the 2003 outbreak of severe acute respiratory syndrome coronavirus (SARS-CoV) belong to bat SARS-like coronavirus (SARSR-COV) species, and their surface spike glycoproteins are highly homologous. The key step of their invasion into human cells is the binding of the receptor binding domain (RBD) part of the surface spike protein and human angiotensin converting enzyme 2(ACE2). Many experimental studies have reported that SARS-CoV-2 possesses a stronger binding ability than SARS-CoV. In order to explain the experimental results, molecular dynamics simulations, steered molecular dynamics simulations and umbrella sampling were performed on the binding of wild-type RBD (RBDWT) of the two viruses and ACE2. The results showed that different behaviors of the β-loop region lead to differences in key residues and interactions, and further result in differences in binding/unbinding mechanisms. The binding affinity of SARS-COV-2RBD and ACE2 is nearly 30 times that of SARS-COV-RBD. © 2022. All rights reserved.

8.
J Mol Model ; 28(5): 128, 2022 Apr 24.
Article in English | MEDLINE | ID: covidwho-1802772

ABSTRACT

In COVID-19 infection, the SARS-CoV-2 spike protein S1 interacts to the ACE2 receptor of human host, instigating the viral infection. To examine the competitive inhibitor efficacy of broad spectrum alpha helical AMPs extracted from frog skin, a comparative study of intermolecular interactions between viral S1 and AMPs was performed relative to S1-ACE2p interactions. The ACE2 binding region with S1 was extracted as ACE2p from the complex for ease of computation. Surprisingly, the Spike-Dermaseptin-S9 complex had more intermolecular interactions than the other peptide complexes and importantly, the S1-ACE2p complex. We observed how atomic displacements in docked complexes impacted structural integrity of a receptor-binding domain in S1 through conformational sampling analysis. Notably, this geometry-based sampling approach confers the robust interactions that endure in S1-Dermaseptin-S9 complex, demonstrating its conformational transition. Additionally, QM calculations revealed that the global hardness to resist chemical perturbations was found more in Dermaseptin-S9 compared to ACE2p. Moreover, the conventional MD through PCA and the torsional angle analyses indicated that Dermaseptin-S9 altered the conformations of S1 considerably. Our analysis further revealed the high structural stability of S1-Dermaseptin-S9 complex and particularly, the trajectory analysis of the secondary structural elements established the alpha helical conformations to be retained in S1-Dermaseptin-S9 complex, as substantiated by SMD results. In conclusion, the functional dynamics proved to be significant for viral Spike S1 and Dermaseptin-S9 peptide when compared to ACE2p complex. Hence, Dermaseptin-S9 peptide inhibitor could be a strong candidate for therapeutic scaffold to prevent infection of SARS-CoV-2.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antimicrobial Cationic Peptides , COVID-19 Drug Treatment , COVID-19 , Spike Glycoprotein, Coronavirus , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/therapeutic use , Anura/metabolism , COVID-19/prevention & control , Humans , Peptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
9.
Mar Drugs ; 20(3)2022 Feb 24.
Article in English | MEDLINE | ID: covidwho-1725847

ABSTRACT

The COVID-19 pandemic and its continuing emerging variants emphasize the need to discover appropriate treatment, where vaccines alone have failed to show complete protection against the new variants of the virus. Therefore, treatment of the infected cases is critical. This paper discusses the bio-guided isolation of three indole diketopiperazine alkaloids, neoechinulin A (1), echinulin (2), and eurocristatine (3), from the Red Sea-derived Aspergillus fumigatus MR2012. Neoechinulin A (1) exhibited a potent inhibitory effect against SARS-CoV-2 Mpro with IC50 value of 0.47 µM, which is comparable to the reference standard GC376. Despite the structural similarity between the three compounds, only 1 showed a promising effect. The mechanism of inhibition is discussed in light of a series of extensive molecular docking, classical and steered molecular dynamics simulation experiments. This paper sheds light on indole diketopiperazine alkaloids as a potential structural motif against SARS-CoV-2 Mpro. Additionally, it highlights the potential of different molecular docking and molecular dynamics simulation approaches in the discrimination between active and inactive structurally related Mpro inhibitors.


Subject(s)
Antiviral Agents/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Indole Alkaloids/chemistry , Piperazines/chemistry , SARS-CoV-2/enzymology , Alkaloids/chemistry , Alkaloids/isolation & purification , Antiviral Agents/isolation & purification , Aspergillus fumigatus/chemistry , Cysteine Proteinase Inhibitors/isolation & purification , Indole Alkaloids/isolation & purification , Molecular Docking Simulation , Molecular Dynamics Simulation , Piperazines/isolation & purification
10.
Molecular Simulation ; : 8, 2021.
Article in English | Web of Science | ID: covidwho-1585510

ABSTRACT

Ivermectin (D1), an FDA-approved drug, is mainly used as an inhibitor for the treatment of parasitic infections. This drug was drawn attention due to its effectiveness in COVID-19 treatment. In this work, we investigated how ivermectin and its analogues are interacted with Imp alpha/beta 1 heterodimer by molecular docking, steered molecular dynamics (SMD), classical molecular dynamics (MD), and MM/PBSA binding free energy analysis. Docking results showed that ivermectin dimer showed the highest binding affinity of -12.2 kcal/mol compared to its monomer. In SMD, the highest acceleration of 600 pm/ps(2) is noticed for D1-Imp alpha/beta 1 complex in the distance from 18.27 to 18.36 angstrom. The pulling force of 1745.86 pN is also detected for D1-Imp alpha/beta 1. To validate the docking and SMD results, 100 ns molecular dynamics (MD) simulation is performed on the D1-Imp alpha/beta 1 complex. The average RMSD value indicates a good structural stability of the complex despite some significant changes at the beginning. It is noted that most of the residues are stable over the simulation time with an average RMSF value of less than 2.65 angstrom. The MM/PBSA free energy of ivermectin also shows strong and spontaneous binding with the Imp alpha/beta 1.

11.
J Biomol Struct Dyn ; 40(5): 2099-2112, 2022 03.
Article in English | MEDLINE | ID: covidwho-889353

ABSTRACT

COVID-19, which is caused by a novel coronavirus known as SARS-CoV-2, has spread rapidly around the world, and it has infected more than 29 million individuals as recorded on 16 September 2020. Much effort has been made to stop the virus from spreading, and there are currently no approved pharmaceutical products to treat COVID-19. Here, we apply an in silico approach to investigate more than 3800 FDA approved drugs on the viral RBD S1-ACE2 interface as a target. The compounds were investigated through flexible ligand docking, ADME property calculations and protein-ligand interaction maps. Molecular dynamics (MD) simulations were also performed on eleven compounds to study the stability and the interactions of the protein-ligand complexes. The MD simulations show that bagrosin, chidamide, ebastine, indacaterol, regorafenib, salazosulfadimidine, silodosin and tasosartan are relatively stable near the C terminal domain (CTD1) of the S1 subunit of the viral S protein. The relative MMGBSA binding energies show that silodosin has the best binding to the target. The constant velocity steered molecular dynamics (SMD) simulations show that silodosin preferentially interacts with the RBD S1 and has potential to act as an interfering compound between viral spike-host ACE2 interactions. Communicated by Ramaswamy H. Sarma.


Subject(s)
COVID-19 Drug Treatment , Pharmaceutical Preparations , Glycoproteins , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL