Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Viruses ; 14(2)2022 01 29.
Article in English | MEDLINE | ID: covidwho-1715766

ABSTRACT

Stephen Oroszlan received his early education in Hungary, graduating in 1950 from the Technical University in Budapest with a degree in chemical engineering [...].


Subject(s)
Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , History, 20th Century , History, 21st Century , Humans , Male , Retroviridae/drug effects , Retroviridae/metabolism , Viral Protease Inhibitors/pharmacology , Viral Proteases/chemistry , Viral Proteases/metabolism
2.
Viruses ; 13(10)2021 10 02.
Article in English | MEDLINE | ID: covidwho-1441885

ABSTRACT

Viral proteases are indispensable for successful virion maturation, thus making them a prominent drug target. Their enzyme activity is tightly spatiotemporally regulated by expression in the precursor form with little or no activity, followed by activation via autoprocessing. These cleavage events are frequently triggered upon transportation to a specific compartment inside the host cell. Typically, precursor oligomerization or the presence of a co-factor is needed for activation. A detailed understanding of these mechanisms will allow ligands with non-canonical mechanisms of action to be designed, which would specifically modulate the initial irreversible steps of viral protease autoactivation. Binding sites exclusive to the precursor, including binding sites beyond the protease domain, can be exploited. Both inhibition and up-regulation of the proteolytic activity of viral proteases can be detrimental for the virus. All these possibilities are discussed using examples of medically relevant viruses including herpesviruses, adenoviruses, retroviruses, picornaviruses, caliciviruses, togaviruses, flaviviruses, and coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Viral Protease Inhibitors/pharmacology , Viral Proteases/metabolism , Virus Diseases/drug therapy , Adenoviruses, Human/drug effects , Adenoviruses, Human/metabolism , Flavivirus/drug effects , Flavivirus/metabolism , HIV-1/drug effects , Herpesviridae/drug effects , Herpesviridae/metabolism , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Viral Proteases/biosynthesis
3.
Nat Commun ; 12(1): 5553, 2021 09 21.
Article in English | MEDLINE | ID: covidwho-1434104

ABSTRACT

SARS-CoV-2 is the causative agent behind the COVID-19 pandemic, responsible for over 170 million infections, and over 3.7 million deaths worldwide. Efforts to test, treat and vaccinate against this pathogen all benefit from an improved understanding of the basic biology of SARS-CoV-2. Both viral and cellular proteases play a crucial role in SARS-CoV-2 replication. Here, we study proteolytic cleavage of viral and cellular proteins in two cell line models of SARS-CoV-2 replication using mass spectrometry to identify protein neo-N-termini generated through protease activity. We identify previously unknown cleavage sites in multiple viral proteins, including major antigens S and N: the main targets for vaccine and antibody testing efforts. We discover significant increases in cellular cleavage events consistent with cleavage by SARS-CoV-2 main protease, and identify 14 potential high-confidence substrates of the main and papain-like proteases. We show that siRNA depletion of these cellular proteins inhibits SARS-CoV-2 replication, and that drugs targeting two of these proteins: the tyrosine kinase SRC and Ser/Thr kinase MYLK, show a dose-dependent reduction in SARS-CoV-2 titres. Overall, our study provides a powerful resource to understand proteolysis in the context of viral infection, and to inform the development of targeted strategies to inhibit SARS-CoV-2 and treat COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/metabolism , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Animals , Cell Line , Dipeptides/pharmacology , Humans , Mutation , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Proteolysis , Proteomics , RNA, Small Interfering/pharmacology , SARS-CoV-2/genetics , Viral Proteases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism , COVID-19 Drug Treatment
4.
J Pharmacol Exp Ther ; 378(2): 166-172, 2021 08.
Article in English | MEDLINE | ID: covidwho-1403003

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to be a global threat since its emergence. Although several COVID-19 vaccines have become available, the prospective timeframe for achieving effective levels of vaccination across global populations remains uncertain. Moreover, the emergence of SARS-CoV-2 variants presents continuing potential challenges for future vaccination planning. Therefore, development of effective antiviral therapies continues to be an urgent unmet need for COVID-19. Successful antiviral regimens for the treatment of human immunodeficiency virus and hepatitis C virus infections have established viral proteases as validated targets for antiviral drug development. In this context, we review protease targets in drug development, currently available antiviral protease inhibitors, and therapeutic development efforts on SARS-CoV-2 main protease and papain-like protease. SIGNIFICANCE STATEMENT: Coronavirus disease 2019 (COVID-19) continues to be a global threat since its emergence. The development of effective antiviral therapeutics for COVID-19 remains an urgent and long-term need. Because viral proteases are validated drug targets, specific severe acute respiratory syndrome coronavirus 2 protease inhibitors are critical therapeutics to be developed for treatment of COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Drug Development , Protease Inhibitors/pharmacology , SARS-CoV-2/enzymology , Viral Proteases/metabolism , Animals , Antiviral Agents/therapeutic use , Humans , Protease Inhibitors/therapeutic use , SARS-CoV-2/drug effects
5.
Nat Commun ; 12(1): 668, 2021 01 28.
Article in English | MEDLINE | ID: covidwho-1387328

ABSTRACT

Except remdesivir, no specific antivirals for SARS-CoV-2 infection are currently available. Here, we characterize two small-molecule-compounds, named GRL-1720 and 5h, containing an indoline and indole moiety, respectively, which target the SARS-CoV-2 main protease (Mpro). We use VeroE6 cell-based assays with RNA-qPCR, cytopathic assays, and immunocytochemistry and show both compounds to block the infectivity of SARS-CoV-2 with EC50 values of 15 ± 4 and 4.2 ± 0.7 µM for GRL-1720 and 5h, respectively. Remdesivir permitted viral breakthrough at high concentrations; however, compound 5h completely blocks SARS-CoV-2 infection in vitro without viral breakthrough or detectable cytotoxicity. Combination of 5h and remdesivir exhibits synergism against SARS-CoV-2. Additional X-ray structural analysis show that 5h forms a covalent bond with Mpro and makes polar interactions with multiple active site amino acid residues. The present data suggest that 5h might serve as a lead Mpro inhibitor for the development of therapeutics for SARS-CoV-2 infection.


Subject(s)
COVID-19 Drug Treatment , Coronavirus Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Viral Proteases/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Humans , Indoles/pharmacology , Pyridines/pharmacology , Vero Cells , Viral Proteases/metabolism
6.
ACS Infect Dis ; 7(6): 1483-1502, 2021 06 11.
Article in English | MEDLINE | ID: covidwho-1387146

ABSTRACT

Viral proteases are highly specific and recognize conserved cleavage site sequences of ∼6-8 amino acids. Short stretches of homologous host-pathogen sequences (SSHHPS) can be found spanning the viral protease cleavage sites. We hypothesized that these sequences corresponded to specific host protein targets since >40 host proteins have been shown to be cleaved by Group IV viral proteases and one Group VI viral protease. Using PHI-BLAST and the viral protease cleavage site sequences, we searched the human proteome for host targets and analyzed the hit results. Although the polyprotein and host proteins related to the suppression of the innate immune responses may be the primary targets of these viral proteases, we identified other cleavable host proteins. These proteins appear to be related to the virus-induced phenotype associated with Group IV viruses, suggesting that information about viral pathogenesis may be extractable directly from the viral genome sequence. Here we identify sequences cleaved by the SARS-CoV-2 papain-like protease (PLpro) in vitro within human MYH7 and MYH6 (two cardiac myosins linked to several cardiomyopathies), FOXP3 (an X-linked Treg cell transcription factor), ErbB4 (HER4), and vitamin-K-dependent plasma protein S (PROS1), an anticoagulation protein that prevents blood clots. Zinc inhibited the cleavage of these host sequences in vitro. Other patterns emerged from multispecies sequence alignments of the cleavage sites, which may have implications for the selection of animal models and zoonosis. SSHHPS/nsP is an example of a sequence-specific post-translational silencing mechanism.


Subject(s)
Papain , Peptide Hydrolases , SARS-CoV-2/enzymology , Viral Proteases/metabolism , Amino Acid Sequence , Cardiac Myosins/chemistry , Forkhead Transcription Factors/chemistry , Humans , Myosin Heavy Chains/chemistry , Papain/metabolism , Peptide Hydrolases/metabolism , Protein S/chemistry , Receptor, ErbB-4/chemistry
7.
J Phys Chem Lett ; 12(1): 65-72, 2021 Jan 14.
Article in English | MEDLINE | ID: covidwho-1387117

ABSTRACT

We analyzed a 100 µs MD trajectory of the SARS-CoV-2 main protease by a non-parametric data analysis approach which allows characterizing a free energy landscape as a simultaneous function of hundreds of variables. We identified several conformations that, when visited by the dynamics, are stable for several hundred nanoseconds. We explicitly characterize and describe these metastable states. In some of these configurations, the catalytic dyad is less accessible. Stabilizing them by a suitable binder could lead to an inhibition of the enzymatic activity. In our analysis we keep track of relevant contacts between residues which are selectively broken or formed in the states. Some of these contacts are formed by residues which are far from the catalytic dyad and are accessible to the solvent. Based on this analysis we propose some relevant contact patterns and three possible binding sites which could be targeted to achieve allosteric inhibition.


Subject(s)
COVID-19 , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , SARS-CoV-2/metabolism , Viral Proteases/chemistry , Viral Proteases/metabolism , Binding Sites , Humans , Models, Molecular , Protease Inhibitors/chemistry , Protein Binding , Protein Conformation
8.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: covidwho-1323269

ABSTRACT

In the last year, the COVID-19 pandemic has highly affected the lifestyle of the world population, encouraging the scientific community towards a great effort on studying the infection molecular mechanisms. Several vaccine formulations are nowadays available and helping to reach immunity. Nevertheless, there is a growing interest towards the development of novel anti-covid drugs. In this scenario, the main protease (Mpro) represents an appealing target, being the enzyme responsible for the cleavage of polypeptides during the viral genome transcription. With the aim of sharing new insights for the design of novel Mpro inhibitors, our research group developed a machine learning approach using the support vector machine (SVM) classification. Starting from a dataset of two million commercially available compounds, the model was able to classify two hundred novel chemo-types as potentially active against the viral protease. The compounds labelled as actives by SVM were next evaluated through consensus docking studies on two PDB structures and their binding mode was compared to well-known protease inhibitors. The best five compounds selected by consensus docking were then submitted to molecular dynamics to deepen binding interactions stability. Of note, the compounds selected via SVM retrieved all the most important interactions known in the literature.


Subject(s)
COVID-19 Drug Treatment , Coronavirus Protease Inhibitors/pharmacology , Drug Evaluation, Preclinical/methods , SARS-CoV-2/drug effects , Support Vector Machine , Antiviral Agents/pharmacology , COVID-19/virology , Coronavirus Protease Inhibitors/metabolism , Databases, Pharmaceutical , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Pandemics , SARS-CoV-2/enzymology , Small Molecule Libraries , Supervised Machine Learning , Viral Nonstructural Proteins/metabolism , Viral Proteases/metabolism
9.
Expert Rev Clin Pharmacol ; 14(10): 1305-1315, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1322577

ABSTRACT

BACKGROUND: The high transmission and pathogenicity of SARS-CoV-2 has led to a pandemic that has halted the world's economy and health. The newly evolved strains and scarcity of vaccines has worsened the situation. The main protease (Mpro) of SARS-CoV-2 can act as a potential target due to its role in viral replication and conservation level. METHODS: In this study, we have enlisted more than 1100 phytochemicals from Asian plants based on deep literature mining. The compounds library was screened against the Mpro of SARS-CoV-2. RESULTS: The selected three ligands, Flemichin, Delta-Oleanolic acid, and Emodin 1-O-beta-D-glucoside had a binding energy of -8.9, -8.9, -8.7 KJ/mol respectively. The compounds bind to the active groove of the main protease at; Cys145, Glu166, His41, Met49, Pro168, Met165, Gln189. The multiple descriptors from the simulation study; root mean square deviation, root mean square fluctuation, radius of gyration, hydrogen bond, solvent accessible surface area confirms the stable nature of the protein-ligand complexes. Furthermore, post-md analysis confirms the rigidness in the docked poses over the simulation trajectories. CONCLUSIONS: Our combinatorial drug design approaches may help researchers to identify suitable drug candidates against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Phytochemicals/pharmacology , SARS-CoV-2/enzymology , Viral Proteases/metabolism , Antiviral Agents/chemistry , Databases, Chemical , Gene Expression Regulation, Viral/drug effects , Molecular Docking Simulation , Molecular Structure , Phytochemicals/chemistry , Viral Proteases/genetics
10.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: covidwho-1298153

ABSTRACT

The non-structural protein 2 (nsP2) of alphavirus Venezuelan equine encephalitis virus (VEEV) is a cysteine protease that is responsible for processing of the viral non-structural polyprotein and is an important drug target owing to the clinical relevance of VEEV. In this study we designed two recombinant VEEV nsP2 constructs to study the effects of an N-terminal extension on the protease activity and to investigate the specificity of the elongated enzyme in vitro. The N-terminal extension was found to have no substantial effect on the protease activity. The amino acid preferences of the VEEV nsP2 protease were investigated on substrates representing wild-type and P5, P4, P2, P1, P1', and P2' variants of Semliki forest virus nsP1/nsP2 cleavage site, using a His6-MBP-mEYFP recombinant substrate-based protease assay which has been adapted for a 96-well plate-based format. The structural basis of enzyme specificity was also investigated in silico by analyzing a modeled structure of VEEV nsP2 complexed with oligopeptide substrate. To our knowledge, in vitro screening of P1' amino acid preferences of VEEV nsP2 protease remains undetermined to date, thus, our results may provide valuable information for studies and inhibitor design of different alphaviruses or other Group IV viruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/enzymology , Viral Proteases/chemistry , Catalytic Domain , Molecular Dynamics Simulation , Oligopeptides/chemistry , Oligopeptides/metabolism , Substrate Specificity , Viral Proteases/genetics , Viral Proteases/metabolism
11.
Future Med Chem ; 13(16): 1353-1366, 2021 08.
Article in English | MEDLINE | ID: covidwho-1282697

ABSTRACT

Background: The new coronavirus pandemic has had a significant impact worldwide, and therapeutic treatment for this viral infection is being strongly pursued. Efforts have been undertaken by medicinal chemists to discover molecules or known drugs that may be effective in COVID-19 treatment - in particular, targeting the main protease (Mpro) of the virus. Materials & methods: We have employed an innovative strategy - application of ligand- and structure-based virtual screening - using a special compilation of an approved and diverse set of SARS-CoV-2 crystallographic complexes that was recently published. Results and conclusion: We identified seven drugs with different original indications that might act as potential Mpro inhibitors and may be preferable to other drugs that have been repurposed. These drugs will be experimentally tested to confirm their potential Mpro inhibition and thus their effectiveness against COVID-19.


Subject(s)
Antiviral Agents/chemistry , COVID-19 Drug Treatment , Protease Inhibitors/chemistry , SARS-CoV-2/drug effects , Small Molecule Libraries/chemistry , Viral Proteases/metabolism , Antiviral Agents/pharmacology , Databases, Chemical , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Protease Inhibitors/pharmacology , Protein Binding , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
12.
Chem Biol Drug Des ; 98(4): 604-619, 2021 10.
Article in English | MEDLINE | ID: covidwho-1273079

ABSTRACT

3CLpro is essential for SARS-CoV-2 replication and infection; its inhibition using small molecules is a potential therapeutic strategy. In this study, a comprehensive crystallography-guided fragment-based drug discovery approach was employed to design new inhibitors for SARS-CoV-2 3CLpro. All small molecules co-crystallized with SARS-CoV-2 3CLpro with structures deposited in the Protein Data Bank were used as inputs. Fragments sitting in the binding pocket (87) were grouped into eight geographical types. They were interactively coupled using various synthetically reasonable linkers to generate larger molecules with divalent binding modes taking advantage of two different fragments' interactions. In total, 1,251 compounds were proposed, and 7,158 stereoisomers were screened using Glide (standard precision and extra precision), AutoDock Vina, and Prime MMGBSA. The top 22 hits having conformations approaching the linear combination of their constituent fragments were selected for MD simulation on Desmond. MD simulation suggested 15 of these did adopt conformations very close to their constituent pieces with far higher binding affinity than either constituent domain alone. These structures could provide a starting point for the further design of SARS-CoV-2 3CLpro inhibitors with improved binding, and structures are provided.


Subject(s)
Antiviral Agents/chemistry , COVID-19 Drug Treatment , SARS-CoV-2/drug effects , Viral Protease Inhibitors/chemistry , Viral Proteases/metabolism , Antiviral Agents/pharmacology , Crystallization , Drug Design , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Multivariate Analysis , Protein Binding , Protein Conformation , Stereoisomerism , Structure-Activity Relationship , Viral Protease Inhibitors/pharmacology
13.
Biomolecules ; 11(6)2021 05 28.
Article in English | MEDLINE | ID: covidwho-1256422

ABSTRACT

The urgent need for novel and effective drugs against the SARS-CoV-2 coronavirus pandemic has stimulated research worldwide. The Papain-like protease (PLpro), which is essential for viral replication, shares a similar active site structural architecture to other cysteine proteases. Here, we have used representatives of the Ovarian Tumor Domain deubiquitinase family OTUB1 and OTUB2 along with the PLpro of SARS-CoV-2 to validate and rationalize the binding of inhibitors from previous SARS-CoV candidate compounds. By forming a new chemical bond with the cysteine residue of the catalytic triad, covalent inhibitors irreversibly suppress the protein's activity. Modeling covalent inhibitor binding requires detailed knowledge about the compounds' reactivities and binding. Molecular Dynamics refinement simulations of top poses reveal detailed ligand-protein interactions and show their stability over time. The recently discovered selective OTUB2 covalent inhibitors were used to establish and validate the computational protocol. Structural parameters and ligand dynamics are in excellent agreement with the ligand-bound OTUB2 crystal structures. For SARS-CoV-2 PLpro, recent covalent peptidomimetic inhibitors were simulated and reveal that the ligand-protein interaction is very dynamic. The covalent and non-covalent docking plus subsequent MD refinement of known SARS-CoV inhibitors into DUBs and the SARS-CoV-2 PLpro point out a possible approach to target the PLpro cysteine protease from SARS-CoV-2. The results show that such an approach gives insight into ligand-protein interactions, their dynamic character, and indicates a path for selective ligand design.


Subject(s)
Deubiquitinating Enzymes/antagonists & inhibitors , Protease Inhibitors/chemistry , SARS-CoV-2/metabolism , Viral Proteases/chemistry , Binding Sites , COVID-19/pathology , Catalytic Domain , Deubiquitinating Enzymes/metabolism , Drug Design , Female , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Dynamics Simulation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protease Inhibitors/metabolism , SARS-CoV-2/isolation & purification , Viral Proteases/metabolism
14.
Pharmacol Rep ; 73(3): 926-938, 2021 Jun.
Article in English | MEDLINE | ID: covidwho-1220621

ABSTRACT

INTRODUCTION: Drug repurposing is the need of the hour considering the medical emergency caused by the COVID-19 pandemic. Recently, cytokine storm by the host immune system has been linked with high viral load, loss of lung function, acute respiratory distress syndrome (ARDS), multiple organ failure, and subsequent fatal outcome. OBJECTIVE: This study aimed to identify potential FDA approved drugs that can be repurposed for COVID-19 treatment using an in-silico analysis. METHODS: In this study, virtual screening of selected FDA approved drugs was performed by targeting the main protease (Mpro) of SARS-CoV-2 and the key molecules involved in the 'Cytokine storm' in COVID-19 patients. Based on our preliminary screening supported by extensive literature search, we selected FDA approved drugs to target the SARS-CoV-2 main protease (Mpro) and the key players of cytokine storm, TNF-α, IL-6, and IL-1ß. These compounds were examined based on systematic docking studies and further validated using a combination of molecular dynamics simulations and molecular mechanic/generalized/Born/Poisson-Boltzmann surface area (MM/G/P/BSA) free energy calculations. RESULTS: Based on the findings, Rifampicin and Letermovir appeared as the most promising drug showing a very good binding affinity with the main protease of SARS-CoV-2 and TNF-α, IL-6, and IL-1ß. However, it is pertinent to mention here that our findings need further validation by in vitro analysis and clinical trials. CONCLUSION: This study provides an insight into the drug repurposing approach in which several FDA approved drugs were examined to inhibit COVID-19 infection by targeting the main protease of SARS-COV-2 and the cytokine storm.


Subject(s)
Acetates/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Quinazolines/therapeutic use , Rifampin/therapeutic use , COVID-19/metabolism , Cytokine Release Syndrome/drug therapy , Cytokines/metabolism , Drug Repositioning/methods , Humans , Molecular Docking Simulation , SARS-CoV-2/drug effects , Viral Proteases/metabolism
15.
Eur J Pharm Sci ; 160: 105744, 2021 May 01.
Article in English | MEDLINE | ID: covidwho-1056567

ABSTRACT

The current global pandemic outbreak of COVID-19, caused by the SARS-CoV-2, strikes an invincible damage to both daily life and the global economy. WHO guidelines for COVID-19 clinical management includes infection control and prevention, social distancing and supportive care using supplemental oxygen and mechanical ventilator support. Currently, evolving researches and clinical reports regarding infected patients with SARS-CoV-2 suggest a potential list of repurposed drugs that may produce appropriate pharmacological therapeutic efficacies in treating COVID-19 infected patients. In this study, we performed virtual screening and evaluated the obtained results of US-FDA approved small molecular database library (302 drug molecule) against two important different protein targets in COVID-19. Best compounds in molecular docking were used as a training set for generation of two different pharmacophores. The obtained pharmacophores were employed for virtual screening of ChEMBL database. The filtered compounds were clustered using Finger print model to obtain two compounds that will be subjected to molecular docking simulations against the two targets. Compounds complexes with SARS-CoV-2 main protease and S-protein were studied using molecular dynamics (MD) simulation. MD simulation studies suggest the potential inhibitory activity of ChEMBL398869 against SARS-CoV-2 main protease and restress the importance of Gln189 flexibility in inhibitors recognition through increasing S2 subsite plasticity.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/virology , Databases, Protein , Molecular Dynamics Simulation , SARS-CoV-2/enzymology , Viral Proteases/metabolism , Amino Acid Substitution , Antiviral Agents/chemistry , Humans , Models, Chemical , Molecular Structure , Protein Conformation , SARS-CoV-2/genetics , Structure-Activity Relationship , Viral Protease Inhibitors , Viral Proteases/chemistry , Viral Proteases/genetics
16.
Biochimie ; 182: 177-184, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1039291

ABSTRACT

The main protease (Mpro) of SARS-CoV and SARS-CoV-2 is a key enzyme in viral replication and a promising target for the development of antiviral therapeutics. The understanding of this protein is based on a number of observations derived from earlier x-ray structures, which mostly consider substrates or ligands as the main reason behind modulation of the active site. This lead to the concept of substrate-induced subsite cooperativity as an initial attempt to explain the dual binding specificity of this enzyme in recognizing the cleavage sequences at its N- and C-termini, which are important processing steps in obtaining the mature protease. The presented hypothesis proposes that structural heterogeneity is a property of the enzyme, independent of the presence of a substrate or ligand. Indeed, the analysis of Mpro structures of SARS-CoV and SARS-CoV-2 reveals a conformational diversity for the catalytically competent state in ligand-free structures. Variation in the binding site appears to result from flexibility at residues lining the S1 subpocket and segments incorporating methionine 49 and glutamine 189. The structural evidence introduces "structure-based recognition" as a new paradigm in substrate proteolysis by Mpro. In this concept, the binding space in subpockets of the enzyme varies in a non-cooperative manner, causing distinct conformations, which recognize and process different cleavage sites, as the N- and C-termini. Insights into the recognition basis of the protease provide explanation to the ordered processing of cleavage sites. The hypothesis expands the conformational space of the enzyme and consequently opportunities for drug development and repurposing efforts.


Subject(s)
COVID-19/virology , Protein Conformation , SARS-CoV-2/enzymology , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteases/chemistry , Viral Proteases/metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Catalytic Domain , Drug Design , Humans , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Substrate Specificity
17.
J R Soc Interface ; 18(174): 20200591, 2021 01.
Article in English | MEDLINE | ID: covidwho-1010695

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.


Subject(s)
COVID-19/virology , SARS-CoV-2/metabolism , Viral Proteases/chemistry , Computer Simulation , Crystallization , Humans , Models, Molecular , Protein Conformation , SARS-CoV-2/enzymology , Thermodynamics , Viral Proteases/drug effects , Viral Proteases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL