Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Curr Med Chem ; 2023 May 19.
Article in English | MEDLINE | ID: covidwho-2322696

ABSTRACT

The application describes compounds, such as compounds of general Formula, with warheads and their use in treating medical diseases or disorders, such as viral infections. Pharmaceutical compositions and synthetic methods of various compounds with warheads are included. The compounds are inhibitors of proteases, such as the 3C, CL- or 3CL-like protease.

2.
PeerJ ; 11: e15086, 2023.
Article in English | MEDLINE | ID: covidwho-2322074

ABSTRACT

Yellow head virus (YHV) is one of the most important pathogens in prawn cultivation. The outbreak of YHV could potentially result in collapses in aquaculture industries. Although a flurry of development has been made in searching for preventive and therapeutic approaches against YHV, there is still no effective therapy available in the market. Previously, computational screening has suggested a few cancer drugs to be used as YHV protease (3CLpro) inhibitors. However, their toxic nature is still of concern. Here, we exploited various computational approaches, such as deep learning-based structural modeling, molecular docking, pharmacological prediction, and molecular dynamics simulation, to search for potential YHV 3CLpro inhibitors. A total of 272 chalcones and flavonoids were in silico screened using molecular docking. The bioavailability, toxicity, and specifically drug-likeness of hits were predicted. Among the hits, molecular dynamics simulation and trajectory analysis were performed to scrutinize the compounds with high binding affinity. Herein, the four selected compounds including chalcones cpd26, cpd31 and cpd50, and a flavonoid DN071_f could be novel potent compounds to prevent YHV and GAV propagation in shrimp. The molecular mechanism at the atomistic level is also enclosed that can be used to further antiviral development.


Subject(s)
Chalcones , Roniviridae , Peptide Hydrolases , Molecular Docking Simulation , Chalcones/pharmacology , Flavonoids/pharmacology , Endopeptidases
3.
Clin Infect Dis ; 76(8): 1403-1411, 2023 04 17.
Article in English | MEDLINE | ID: covidwho-2300490

ABSTRACT

BACKGROUND: This phase 2b part of a randomized phase 2/3 study assessed the efficacy and safety of ensitrelvir for mild-to-moderate coronavirus disease 2019 (COVID-19) during the Omicron epidemic. METHODS: Patients were randomized (1:1:1) to orally receive ensitrelvir fumaric acid 125 mg (375 mg on day 1) or 250 mg (750 mg on day 1) or placebo once daily for 5 days. The co-primary endpoints were the change from baseline in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) titer on day 4 and time-weighted average change from baseline up to 120 hours in the total score of predefined 12 COVID-19 symptoms. Safety was assessed through adverse events. RESULTS: A total of 341 patients (ensitrelvir 125-mg group: 114; ensitrelvir 250-mg group: 116; and placebo group: 111; male: 53.5-64.9%; mean age: 35.3-37.3 years) were included in the efficacy analyses. The change from baseline in SARS-CoV-2 titer on day 4 was significantly greater with both ensitrelvir doses than with placebo (differences from placebo: -0.41 log10 50% tissue-culture infectious dose/mL; P < .0001 for both). The total score of the 12 COVID-19 symptoms did not show a significant difference between the ensitrelvir groups and placebo group. The time-weighted average change from baseline up to 120 hours was significantly greater with ensitrelvir versus placebo in several subtotal scores, including acute symptoms and respiratory symptoms. Most adverse events were mild in severity. CONCLUSIONS: Ensitrelvir treatment demonstrated a favorable antiviral efficacy and potential clinical benefit with an acceptable safety profile. CLINICAL TRIALS REGISTRATION: Japan Registry of Clinical Trials: jRCT2031210350 (https://jrct.niph.go.jp/en-latest-detail/jRCT2031210350).


Subject(s)
COVID-19 , Epidemics , Humans , Male , Adult , SARS-CoV-2 , Antiviral Agents/adverse effects
4.
Vet Microbiol ; 281: 109743, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2304272

ABSTRACT

Infection with porcine epidemic diarrhea virus (PEDV) causes severe watery diarrhea in newborn piglets, leading to substantial financial losses for the swine industry. In this study, we screened small molecule drugs targeting 3 C-like protease (3CLpro) by molecular docking, and further evaluated the antiviral activity of the screened drugs against PEDV. Results showed that octyl gallate (OG), a widely used food additive, exhibited strong binding affinity with the 3CLpro active sites of PEDV. Bio-layer interferometry and fluorescence resonance energy transfer revealed that OG directly interacts with PEDV 3CLpro (KD = 549 nM) and inhibits 3CLpro activity (IC50 = 22.15 µM). OG showed a strong inhibition of PEDV replication in vitro. Virus titers were decreased by 0.58 and 0.71 log10 TCID50/mL for the CV777 and HM2017 strains, respectively. In vivo, all piglets in the PEDV-infected group died at 48 h post-infection (hpi), while 75% of piglets in the OG treatment group showed significant relief from the clinical symptoms, pathological damage, and viral loads in the jejunum and ileum. Moreover, the western blotting results showed that OG also has strong antiviral activity against other swine enteric coronaviruses, including transmissible gastroenteritis virus (TGEV), porcine deltacoronavirus (PDCoV), and swine acute diarrhea syndrome coronavirus (SADS-CoV). Our findings revealed that OG could be developed as a novel antiviral drug against PEDV. The OG exhibited a potential broad-spectrum antiviral drug for control of other swine enteric coronaviruses.


Subject(s)
Coronavirus Infections , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Swine , Porcine epidemic diarrhea virus/physiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Peptide Hydrolases , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Molecular Docking Simulation , Swine Diseases/drug therapy
5.
Eur J Med Chem ; 253: 115311, 2023 May 05.
Article in English | MEDLINE | ID: covidwho-2304178

ABSTRACT

Despite the approval of vaccines, monoclonal antibodies and restrictions during the pandemic, the demand for new efficacious and safe antivirals is compelling to boost the therapeutic arsenal against the COVID-19. The viral 3-chymotrypsin-like protease (3CLpro) is an essential enzyme for replication with high homology in the active site across CoVs and variants showing an almost unique specificity for Leu-Gln as P2-P1 residues, allowing the development of broad-spectrum inhibitors. The design, synthesis, biological activity, and cocrystal structural information of newly conceived peptidomimetic covalent reversible inhibitors are herein described. The inhibitors display an aldehyde warhead, a Gln mimetic at P1 and modified P2-P3 residues. Particularly, functionalized proline residues were inserted at P2 to stabilize the ß-turn like bioactive conformation, modulating the affinity. The most potent compounds displayed low/sub-nM potency against the 3CLpro of SARS-CoV-2 and MERS-CoV and inhibited viral replication of three human CoVs, i.e. SARS-CoV-2, MERS-CoV, and HCoV 229 in different cell lines. Particularly, derivative 12 exhibited nM-low µM antiviral activity depending on the virus, and the highest selectivity index. Some compounds were co-crystallized with SARS-CoV-2 3CLpro validating our design. Altogether, these results foster future work toward broad-spectrum 3CLpro inhibitors to challenge CoVs related pandemics.


Subject(s)
COVID-19 , Middle East Respiratory Syndrome Coronavirus , Peptidomimetics , Humans , SARS-CoV-2 , Protease Inhibitors/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , X-Rays , Peptide Hydrolases , Antiviral Agents/chemistry
6.
Biomed Pharmacother ; 162: 114367, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2262376

ABSTRACT

Despite the need for novel, effective therapeutics for the COVID-19 pandemic, no curative regimen is yet available, therefore patients are forced to rely on supportive and nonspecific therapies. Some SARS-CoV-2 proteins, like the 3 C-like protease (3CLpro) or the major protease (Mpro), have been identified as promising targets for antiviral drugs. The Mpro has major a role in protein processing as well as pathogenesis of the virus, and could be a useful therapeutic target. The antiviral drug nirmatrelvir can keep SARS-CoV-2 from replicating through inhibiting Mpro. Nirmatrelvir was combined with another HIV protease inhibitor, ritonavir, to create Paxlovid (Nirmatrelvir/Ritonavir). The metabolizing enzyme cytochrome P450 3 A is inhibited by ritonavir to lengthen the half-life of nirmatrelvir, so rintonavir acts as a pharmacological enhancer. Nirmatrelvir exhibits potent antiviral activity against current coronavirus variants, despite significant alterations in the SARS-CoV-2 viral genome. Nevertheless, there are still several unanswered questions. This review summarizes the current literature on nirmatrelvir and ritonavir efficacy in treating SARS-CoV-2 infection, and also their safety and possible side effects.


Subject(s)
COVID-19 , HIV Protease Inhibitors , Humans , Ritonavir , SARS-CoV-2 , Pandemics , COVID-19 Drug Treatment , Antiviral Agents , Peptide Hydrolases
7.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: covidwho-2250560

ABSTRACT

Laurus nobilis (bay laurel) is a natural source of biological compounds, and some of its extracts and phytocompounds are also endowed with antiviral activity toward the family of the severe acute respiratory syndrome (SARS)-associated ß-coronaviruses. Some glycosidic laurel compounds such as laurusides were proposed as inhibitors of important protein targets of SARS-CoV-2, which clearly recalls their potential as anti-COVID-19 drugs. Due to the frequent genomic variations of the ß-coronaviruses and the consequent importance of evaluating a new drug candidate with respect to the variants of the target ß-coronavirus, we decided to investigate at an atomistic level the molecular interactions of the potential laurel-derived drugs laurusides 1 and 2 (L01 and L02, respectively) toward a well-conserved and crucial target, the 3C-like protease (Mpro), using the enzymes of both the wild-type of SARS-CoV-2 and of the more recent Omicron variant. Thus, we performed molecular dynamic (MD) simulations of laurusides-SARS-CoV-2 protease complexes to deepen the knowledge on the stability of the interaction and compare the effects of the targeting among the two genomic variants. We found that the Omicron mutation does not significantly impact the lauruside binding and that L02 connects more stably with respect to L01 in the complexes from both variants, even though both compounds prevalently interact within the same binding pocket. Although purely in silico, the current study highlights the potential role of bay laurel phytocompounds in the antiviral and specifically anti-coronavirus research and shows their potential binding toward Mpro, corroborating the important commitment of bay laurel as functional food and disclosing novel scenarios of lauruside-based antiviral therapies.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , Molecular Dynamics Simulation , Peptide Hydrolases/metabolism , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/metabolism , Cysteine Endopeptidases/metabolism , Antiviral Agents/chemistry , Molecular Docking Simulation
8.
Eur J Med Chem ; 244: 114803, 2022 Dec 15.
Article in English | MEDLINE | ID: covidwho-2286080

ABSTRACT

SARS-CoV-2 3CL protease is one of the key targets for drug development against COVID-19. Most known SARS-CoV-2 3CL protease inhibitors act by covalently binding to the active site cysteine. Yet, computational screens against this enzyme were mainly focused on non-covalent inhibitor discovery. Here, we developed a deep learning-based stepwise strategy for selective covalent inhibitor screen. We used a deep learning framework that integrated a directed message passing neural network with a feed-forward neural network to construct two different classifiers for either covalent or non-covalent inhibition activity prediction. These two classifiers were trained on the covalent and non-covalent 3CL protease inhibitors dataset, respectively, which achieved high prediction accuracy. We then successively applied the covalent inhibitor model and the non-covalent inhibitor model to screen a chemical library containing compounds with covalent warheads of cysteine. We experimentally tested the inhibition activity of 32 top-ranking compounds and 12 of them were active, among which 6 showed IC50 values less than 12 µM and the strongest one inhibited SARS-CoV-2 3CL protease with an IC50 of 1.4 µM. Further investigation demonstrated that 5 of the 6 active compounds showed typical covalent inhibition behavior with time-dependent activity. These new covalent inhibitors provide novel scaffolds for developing highly active SARS-CoV-2 3CL covalent inhibitors.


Subject(s)
COVID-19 Drug Treatment , Deep Learning , Humans , SARS-CoV-2 , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Coronavirus 3C Proteases , Cysteine , Antiviral Agents/pharmacology
9.
Journal of Chemical Research ; 47(1), 2023.
Article in English | Scopus | ID: covidwho-2246570

ABSTRACT

The 3C-like protease (also known as Mpro) plays a key role in SARS-CoV-2 replication and has similar substrates across mutant coronaviruses, making it an ideal drug target. We synthesized 19 thiazolidinedione derivatives via the Knoevenagel condensations and Mitsunobu reactions as potential 3C-like protease inhibitors. The activity of these inhibitors is screened in vitro by employing the enzymatic screening model of 3C-like protease using fluorescence resonance energy transfer. Dithiothreitol is included in the enzymatic reaction system to avoid non-specific enzymatic inhibition. Active inhibitors with diverse activity are found in this series of compounds, and two representative inhibitors with potent inhibitory activity are highlighted. © The Author(s) 2023.

10.
Biophys Rev ; : 1-13, 2022 Dec 02.
Article in English | MEDLINE | ID: covidwho-2209554

ABSTRACT

SARS-CoV-2 3C-like protease (3CLpro), a potential therapeutic target for COVID-19, consists of a chymotrypsin fold and a C-terminal α-helical domain (domain III), the latter of which mediates dimerization required for catalytic activation. To gain further understanding of the functional dynamics of SARS-CoV-2 3CLpro, this review extends the scope to the comparative study of many crystal structures of proteases having the chymotrypsin fold (clan PA of the MEROPS database). First, the close correspondence between the zymogen-enzyme transformation in chymotrypsin and the allosteric dimerization activation in SARS-CoV-2 3CLpro is illustrated. Then, it is shown that the 3C-like proteases of family Coronaviridae (the protease family C30), which are closely related to SARS-CoV-2 3CLpro, have the same homodimeric structure and common activation mechanism via domain III mediated dimerization. The survey extended to order Nidovirales reveals that all 3C-like proteases belonging to Nidovirales have domain III, but with various chain lengths, and 3CLpro of family Mesoniviridae (family C107) has the same homodimeric structure as that of C30, even though they have no sequence similarity. As a reference, monomeric 3C proteases belonging to the more distant family Picornaviridae (family C3) lacking domain III are compared with C30, and it is shown that the 3C proteases are rigid enough to maintain their structures in the active state. Supplementary Information: The online version contains supplementary material available at 10.1007/s12551-022-01020-x.

11.
Biochem Biophys Res Commun ; 645: 132-136, 2023 02 19.
Article in English | MEDLINE | ID: covidwho-2176742

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been a public health concern worldwide. Ensitrelvir (S-217622) has been evaluated as an antiviral treatment for COVID-19, targeting SARS-CoV-2 3C-like protease (3CLpro). Ensitrelvir has been reported to have comparable antiviral activity against some of the SARS-CoV-2 variants: alpha, beta, gamma, delta, and omicron (BA.1.18). In this paper, we describe that ensitrelvir is effective against newly emerging SARS-CoV-2 variants and globally prevalent 3CLpro mutations. Ensitrelvir exhibited comparable antiviral activity against SARS-CoV-2 variants, including recently emerging ones: omicron (BA1.1, BA.2, BA.2.75, BA.4, BA.5, BQ.1.1, XBB.1, and XE), mu, lambda, and theta. Genetic surveillance of SARS-CoV-2 3CLpro, the target of ensitrelvir, was conducted using a public database and identified 11 major 3CLpro mutations circulating globally (G15S, T21I, T24I, K88R, L89F, K90R, P108S, P132H, A193V, H246Y, and A255V). The 3CLpro mutation from proline to histidine at amino acid position 132 was especially identified in the omicron variant, with prevalence of 99.69%. Enzyme kinetic assay revealed that these 3CLpro mutants have enzymatic activity comparable to that of the wild type (WT). Next, we assessed the inhibitory effect of ensitrelvir against mutated 3CLpro, with it showing inhibitory effects similar to that against the WT. These in vitro data suggest that ensitrelvir will be effective against currently circulating SARS-CoV-2 variants, including omicron variants and those carrying 3CLpro mutations, which emerging novel SARS-CoV-2 variants could carry.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Peptide Hydrolases , Cysteine Endopeptidases/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Protease Inhibitors/pharmacology
12.
Suranaree Journal of Science and Technology ; 29(6), 2022.
Article in English | Web of Science | ID: covidwho-2101789

ABSTRACT

Objectives : The present study is focused to introduce a better preventive and treatment prospective to fight against the COVID-19 crisis by improving the immune system along with inhibiting the viral activity by a comprehensive review and supportive in-silico study.Methodology : The study was conducted with a systemic review on the phytochemicals possessing immunostimulatory potentiality, which was further supported by an in-silico investigation of thosephytocomponents in improving the immunity of the patient as well as in preventive measures which cancontribute a major part to manage the dangerous consequences of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2).Results : In in-silico study showed that scutellarein, hesperidin andbaicalin exhibited strong binding affinity with ACE2, PAK1, Protease by strong molecular interaction forming several hydrogen bonds.Conclusion : The present finding interprets scutellarein, hesperidin and baicalin to be potential herbal immune boosters with antiviral potentiality which are to be further explored by in-vitro and in-vivo methods.

13.
Pharmaceuticals (Basel) ; 15(11)2022 Oct 28.
Article in English | MEDLINE | ID: covidwho-2090304

ABSTRACT

SARS-CoV-2 is a positive single-strand RNA-based macromolecule that has caused the death of more than 6.3 million people since June 2022. Moreover, by disturbing global supply chains through lockdowns, the virus has indirectly caused devastating damage to the global economy. It is vital to design and develop drugs for this virus and its various variants. In this paper, we developed an in silico study-based hybrid framework to repurpose existing therapeutic agents in finding drug-like bioactive molecules that would cure COVID-19. In the first step, a total of 133 drug-likeness bioactive molecules are retrieved from the ChEMBL database against SARS coronavirus 3CL Protease. Based on the standard IC50, the dataset is divided into three classes: active, inactive, and intermediate. Our comparative analysis demonstrated that the proposed Extra Tree Regressor (ETR)-based QSAR model has improved prediction results related to the bioactivity of chemical compounds as compared to Gradient Boosting-, XGBoost-, Support Vector-, Decision Tree-, and Random Forest-based regressor models. ADMET analysis is carried out to identify thirteen bioactive molecules with the ChEMBL IDs 187460, 190743, 222234, 222628, 222735, 222769, 222840, 222893, 225515, 358279, 363535, 365134, and 426898. These molecules are highly suitable drug candidates for SARS-CoV-2 3CL Protease. In the next step, the efficacy of the bioactive molecules is computed in terms of binding affinity using molecular docking, and then six bioactive molecules are shortlisted, with the ChEMBL IDs 187460, 222769, 225515, 358279, 363535, and 365134. These molecules can be suitable drug candidates for SARS-CoV-2. It is anticipated that the pharmacologist and/or drug manufacturer would further investigate these six molecules to find suitable drug candidates for SARS-CoV-2. They can adopt these promising compounds for their downstream drug development stages.

14.
Pak J Biol Sci ; 25(9): 867-874, 2022 Jan.
Article in English | MEDLINE | ID: covidwho-2030108

ABSTRACT

<b>Background and Objective:</b> Lemongrass (<i>Cymbopogon citratus</i>) and turmeric (<i>Curcuma longa</i>) are widely used by the community for traditional medicinal spices and cooking spices. In the era of the COVID-19 pandemic, people use lemongrass and turmeric to increase immunity and protect the body from infection with the SARS-CoV-2 virus. However, the antiviral mechanisms have not been studied much. This study aims to predict the bioactivity of the phytosterol compounds of lemongrass and turmeric for COVID-19 therapy through inhibition of 3C-like protease (3CLPro) <i>in silico</i>. <b>Materials and Methods:</b> The 3CLPro protein 3D structure was downloaded from the PDB database with the access code 2ZU2 and the phytosterol compounds of lemongrass and turmeric were taken from PubChem. A total of 59 total phytosterol compounds from turmeric and lemongrass were screened for their bioactivity as an antiviral by using online PASS. Compounds with a high activating potential (Pa) were interacted with 3CLPro protein with the PyRx program and analyzed by Discovery Studio version 19.0 and LigPlus. <b>Results:</b> A total of 22 total phytosterol compounds were identified as potential antiviral agents. Based on the Pa value, 15 phytosterol compounds have the potential to act as inhibitor agents for 3CLPro SARS-CoV-2. The phytosterol compounds of lemongrass and turmeric bind to the 3CLPro protein in the N-finger domain region and the A and B domain inhibitors connect residues of the 3CLPro protein. The phytosterols of lemongrass and turmeric show a low binding affinity with 3CLPro SARS-CoV-2, indicating a strong interaction between ligand and protein. The inhibition of phytosterols against 3CLPro protein can be used as a basis for determining candidates for COVID-19 therapeutic agents. <b>Conclusion:</b> The phytosterol compounds contained in lemongrass and turmeric have the potential to act as 3CLPro inhibitors. Further studies both <i>in vitro</i> and <i>in vivo</i> need to be done to prove the inhibitory potential of phytosterol compounds.


Subject(s)
COVID-19 Drug Treatment , Cymbopogon , Phytosterols , Antiviral Agents/pharmacology , Curcuma , Humans , Pandemics , Peptide Hydrolases , Phytosterols/pharmacology , SARS-CoV-2 , Viral Proteins/chemistry , Viral Proteins/metabolism
15.
Antimicrob Agents Chemother ; 66(10): e0069722, 2022 10 18.
Article in English | MEDLINE | ID: covidwho-2029466

ABSTRACT

This multicenter, double-blind, phase 2a part of a phase 2/3 study assessed the efficacy and safety of ensitrelvir, a novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 3C-like protease inhibitor, in Japanese patients with mild-to-moderate coronavirus disease 2019 (COVID-19) or asymptomatic SARS-CoV-2 infection. Sixty-nine patients were randomized (1:1:1) to orally receive 5-day ensitrelvir fumaric acid (375 mg on day 1 followed by 125 mg daily, or 750 mg on day 1 followed by 250 mg daily) or placebo and followed up until day 28. The primary outcome was the change from baseline in the SARS-CoV-2 viral titer. A total of 16, 14, and 17 patients in the ensitrelvir 125 mg, ensitrelvir 250 mg, and placebo groups, respectively, were included in the intention-to-treat population (mean age: 38.0 to 40.4 years). On day 4, the change from baseline in SARS-CoV-2 viral titer (log10 50% tissue culture infectious dose/mL) in patients with positive viral titer and viral RNA at baseline was greater with ensitrelvir 125 mg (mean [standard deviation], -2.42 [1.42]; P = 0.0712) and 250 mg (-2.81 [1.21]; P = 0.0083) versus placebo (-1.54 [0.74]); ensitrelvir treatment reduced SARS-CoV-2 RNA by -1.4 to -1.5 log10 copies/mL versus placebo. The viral titer and viral RNA were similar across groups on and after day 6. The median time to infectious viral clearance decreased by approximately 50 h with ensitrelvir treatment. All adverse events were mild to moderate. Ensitrelvir treatment demonstrated rapid SARS-CoV-2 clearance and was well tolerated (Japan Registry of Clinical Trials identifier: jRCT2031210350).


Subject(s)
Anti-Infective Agents , COVID-19 Drug Treatment , Humans , Adult , SARS-CoV-2 , RNA, Viral , Japan , Protease Inhibitors , Antiviral Agents , Enzyme Inhibitors , Double-Blind Method
16.
Eur J Med Chem ; 238: 114458, 2022 Aug 05.
Article in English | MEDLINE | ID: covidwho-1982956

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), as the pathogen of coronavirus disease 2019 (COVID-19), has infected millions of people and took hundreds of thousands of lives. Unfortunately, there is deficiency of effective medicines to prevent or treat COVID-19. 3C like protease (3CLPro) of SARS-CoV-2 is essential to the viral replication and transcription, and is an attractive target to develop anti-SARS-CoV-2 agents. Targeting on the 3CLPro, we screened our protease inhibitor library and obtained compound 10a as hit to weakly inhibit the SARS-CoV-2 3CLPro, and determined the co-crystal structure of 10a and the protease. Based on the deep understanding on the protein-ligand complexes between the hit and SARS-CoV-2 3CLPro, we designed a series of peptidomimetic inhibitors, with outstanding inhibitory activity against SARS-CoV-2 3CLPro and excellent anti-viral potency against SARS-CoV-2. The protein-ligand complexes of the other key inhibitors with SARS-CoV-2 3CLPro were explicitly described by the X-ray co-crystal study. All such results suggest these peptidomimetic inhibitors could be further applied as encouraging drug candidates.


Subject(s)
COVID-19 Drug Treatment , Peptidomimetics , Antiviral Agents/chemistry , Cysteine Endopeptidases/chemistry , Humans , Ligands , Peptide Hydrolases , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Protease Inhibitors/chemistry , SARS-CoV-2
17.
J Virol ; 96(16): e0102722, 2022 08 24.
Article in English | MEDLINE | ID: covidwho-1973796

ABSTRACT

Protein acetylation plays an important role during virus infection. Thus, it is not surprising that viruses always evolve elaborate mechanisms to regulate the functions of histone deacetylases (HDACs), the essential transcriptional and epigenetic regulators for deacetylation. Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes severe diarrhea in suckling piglets and has the potential to infect humans. In this study, we found that PDCoV infection inhibited cellular HDAC activity. By screening the expressions of different HDAC subfamilies after PDCoV infection, we unexpectedly found that HDAC2 was cleaved. Ectopic expression of HDAC2 significantly inhibited PDCoV replication, while the reverse effects could be observed after treatment with an HDAC2 inhibitor (CAY10683) or the knockdown of HDAC2 expression by specific siRNA. Furthermore, we demonstrated that PDCoV-encoded nonstructural protein 5 (nsp5), a 3C-like protease, was responsible for HDAC2 cleavage through its protease activity. Detailed analyses showed that PDCoV nsp5 cleaved HDAC2 at glutamine 261 (Q261), and the cleaved fragments (amino acids 1 to 261 and 262 to 488) lost the ability to inhibit PDCoV replication. Interestingly, the Q261 cleavage site is highly conserved in HDAC2 homologs from other mammalian species, and the nsp5s encoded by seven tested mammalian coronaviruses also cleaved HDAC2, suggesting that cleaving HDAC2 may be a common strategy used by different mammalian coronaviruses to antagonize the antiviral role of HDAC2. IMPORTANCE As an emerging porcine enteropathogenic coronavirus that possesses the potential to infect humans, porcine deltacoronavirus (PDCoV) is receiving increasing attention. In this work, we found that PDCoV infection downregulated cellular histone deacetylase (HDAC) activity. Of particular interest, the viral 3C-like protease, encoded by the PDCoV nonstructural protein 5 (nsp5), cleaved HDAC2, and this cleavage could be observed in the context of PDCoV infection. Furthermore, the cleavage of HDAC2 appears to be a common strategy among mammalian coronaviruses, including the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), to antagonize the antiviral role of HDAC2. To our knowledge, PDCoV nsp5 is the first identified viral protein that can cleave cellular HDAC2. Results from our study provide new targets to develop drugs combating coronavirus infection.


Subject(s)
COVID-19 , Deltacoronavirus/metabolism , Histone Deacetylase 2/metabolism , Swine Diseases , Animals , Humans , Mammals , Peptide Hydrolases , SARS-CoV-2 , Swine , Swine Diseases/metabolism , Swine Diseases/virology
18.
Comput Struct Biotechnol J ; 20: 3409-3421, 2022.
Article in English | MEDLINE | ID: covidwho-1926353

ABSTRACT

Equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) represent two members of the family Arteriviridae and pose a major threat to the equine- and swine-breeding industries throughout the world. Previously, we and others demonstrated that PRRSV 3C-like protease (3CLpro) had very high glutamic acid (Glu)-specificity at the P1 position (P1-Glu). Comparably, EAV 3CLpro exhibited recognition of both Glu and glutamine (Gln) at the P1 position. However, the underlying mechanisms of the P1 substrate specificity shift of arterivirus 3CLpro remain unclear. We systematically screened the specific amino acids in the S1 subsite of arterivirus 3CLpro using a cyclized luciferase-based biosensor and identified Gly116, His133 and Ser136 (using PRRSV 3CLpro numbering) are important for recognition of P1-Glu, whereas Ser136 is nonessential for recognition of P1-Gln. Molecular dynamics simulations and biochemical experiments highlighted that the PRRSV 3CLpro and EAV 3CLpro formed distinct S1 subsites for the P1 substrate specificity switch. Mechanistically, a specific intermolecular salt bridge between PRRSV 3CLpro and substrate P1-Glu (Lys138/P1-Glu) are invaluable for high Glu-specificity at the P1 position, and the exchange of K138T (salt bridge interruption, from PRRSV to EAV) shifted the specificity of PRRSV 3CLpro toward P1-Gln. In turn, the T139K exchange of EAV 3CLpro showed a noticeable shift in substrate specificity, such that substrates containing P1-Glu are likely to be recognized more efficiently. These findings identify an evolutionarily accessible mechanism for disrupting or reorganizing salt bridge with only a single mutation of arterivirus 3CLpro to trigger a substrate specificity switch.

19.
Molecules ; 27(13)2022 Jul 04.
Article in English | MEDLINE | ID: covidwho-1917637

ABSTRACT

The main protease (Mpro) of the betacoronavirus SARS-CoV-2 is an attractive target for the development of treatments for COVID-19. Structure-based design is a successful approach to discovering new inhibitors of the Mpro. Starting from crystal structures of the Mpro in complexes with the Hepatitis C virus NS3/4A protease inhibitors boceprevir and telaprevir, we optimized the potency of the alpha-ketoamide boceprevir against the Mpro by replacing its P1 cyclobutyl moiety by a γ-lactam as a glutamine surrogate. The resulting compound, MG-78, exhibited an IC50 of 13 nM versus the recombinant Mpro, and similar potency was observed for its P1' N-methyl derivative MG-131. Crystal structures confirmed the validity of our design concept. In addition to SARS-CoV-2 Mpro inhibition, we also explored the activity of MG-78 against the Mpro of the alphacoronavirus HCoV NL63 and against enterovirus 3C proteases. The activities were good (0.33 µM, HCoV-NL63 Mpro), moderate (1.45 µM, Coxsackievirus 3Cpro), and relatively poor (6.7 µM, enterovirus A71 3Cpro), respectively. The structural basis for the differences in activities was revealed by X-ray crystallo-graphy. We conclude that the modified boceprevir scaffold is suitable for obtaining high-potency inhibitors of the coronavirus Mpros but further optimization would be needed to target enterovirus 3Cpros efficiently.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Coronavirus 3C Proteases , Cysteine Endopeptidases/chemistry , Humans , Proline/analogs & derivatives , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins
20.
Molecular Systems Design & Engineering ; : 16, 2022.
Article in English | Web of Science | ID: covidwho-1915302

ABSTRACT

3CLpro is a highly conserved main protease found in coronaviruses, which makes it an attractive target for the development of broad-spectrum coronavirus inhibitors. Its homodimer plays an essential role in coronavirus replication. The SGFRKMAF peptide spanning the N-finger domain of 3CLpro is known to disrupt dimerization, however, its mode of action is not fully understood. We used the conformation of this peptide segment in the observed protein crystal structure as the basis for developing leads for coronavirus dimerization inhibitors. We investigated the interaction of the SGFRKMAF peptide with the 3CLpro monomer in blind docking simulations using a representative ensemble of its conformational states. We identified 8 basins of attraction, distinct regions where the peptide accumulates at the 3CLpro monomer surface. Two binding regions dominate: one, located at the groove between domains II and III of the monomer, and the other at the interface region where dimerization takes place. Peptide binding at these two regions resulted in stable peptide-protein complexes in 90 ns fully solvated molecular dynamics simulations. Using protein-protein docking simulations, we found that peptide binding to at least one of the monomers at the dimer interface region is likely to disrupt 3CLpro dimerization through blocking the "hot spot" residues that predominantly account for dimer stabilization. Peptide binding to the interface region in one of the monomers, and to the groove between domains II and III in the other monomer, leads also to disruption of the native dimer structure. The peptide binding constant at the interface region relative to other regions (K-in/out) was estimated to be similar to 0.12 at 310 K, suggesting that at thermodynamic equilibrium the peptide does not solely bind to one of the basins, corroborating the view of a cooperative mechanism between the two basins. In order to optimize the potential of the SGFRKMAF peptide to disrupt 3CLpro dimerization via preferential binding to the interface region, we conducted systematic mutation of the M6 and F8 residues, sites identified as key to blocking hot spot regions in the observed dimer structure. We found that the [M6F, F8S], [M6I, F8Q], [M6Q, F8T] and [M6T, F8I] mutations result in an increase of K-in/out by at least one order of magnitude, with the [M6F, F8S] mutation (the SGFRKFAS peptide) showing the highest value of K-in/out (similar to 2.53). These mutant peptides are, therefore, candidates for peptide-based lead structures in the development of broad-spectrum coronavirus inhibitors.

SELECTION OF CITATIONS
SEARCH DETAIL