Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (117)

Search Parameters:
Keywords = protease 3CLpro

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
30 pages, 16196 KB  
Article
In Silico Optimization of Inhibitors of the 3-Chymotrypsin-like Protease of SARS-CoV-2
by Issouf Fofana, Brice Dali, Mawa Koné, Katarina Sujova, Eugene Megnassan, Stanislav Miertus and Vladimir Frecer
Life 2026, 16(1), 6; https://doi.org/10.3390/life16010006 - 19 Dec 2025
Viewed by 351
Abstract
In this study, new improved inhibitors of the viral enzyme 3-chymotrypsin-like protease (3CLpro) were designed using structure-based drug design techniques in an effort to discover more effective treatment of coronavirus disease 2019 (COVID-19). Three-dimensional models of 3CLpro–inhibitor complexes were [...] Read more.
In this study, new improved inhibitors of the viral enzyme 3-chymotrypsin-like protease (3CLpro) were designed using structure-based drug design techniques in an effort to discover more effective treatment of coronavirus disease 2019 (COVID-19). Three-dimensional models of 3CLpro–inhibitor complexes were prepared by in situ modification of the crystal structure of the submicromolar covalent inhibitor IPCL6 for a set of 25 known inhibitors with published inhibitory potencies (IC50exp). The QSAR model was prepared with a reasonable correlation between the calculated free energies of formation of the 3CLpro-IPCL complex (∆∆Gcom) and the experimentally determined activities IC50exp, which explained approximately 92% of the variation in the 3CLpro inhibition data. A similar agreement was achieved for the QSAR pharmacophore model (PH4) built on the basis of the active conformations of the IPCL inhibitors bound at the active site of the 3CLpro. The virtual combinatorial library of more than 567,000 IPCL analogues was screened in silico using the PH4 model and resulted in the identification of 39 promising analogues. The best inhibitors designed in this study show high predicted affinity for the 3CLpro protease, as well as favourable predicted ADME properties. For the best new virtual inhibitor candidate IPCL 80-27-74-4, the inhibitory concentration IC50pre was predicted equal to 0.8 nM, which represents a significant improvement in the inhibitory potency of known IPCLs. Ultimately, molecular dynamics simulations of the 12 newly designed top-scoring IPCL inhibitors demonstrated that the 3CLpro–inhibitor complexes exhibited good structural stability, confirming the potential for further development of the designed IPCL analogues. Full article
(This article belongs to the Section Biochemistry, Biophysics and Computational Biology)
Show Figures

Figure 1

22 pages, 5064 KB  
Article
Discovery of Galloyl–Flavonoid Conjugates as SARS-CoV-2 3CLpro Inhibitors: Understanding Binding Interactions Through Computational Approaches
by Nopawit Khamto, Panida Boontawee, Vachira Choommongkol, Kritsada Pruksaphon, Suwicha Patnin, Nuttee Suree, Panchika Prangkio and Puttinan Meepowpan
Int. J. Mol. Sci. 2025, 26(19), 9742; https://doi.org/10.3390/ijms26199742 - 7 Oct 2025
Viewed by 1363
Abstract
The emergence of SARS-CoV-2 in 2019 posed significant global public health challenges. One of the most promising targets for novel antiviral drug development is the SARS-CoV-2 main protease (3CLpro). In this study, fragment molecular orbital (FMO) calculations were conducted to provide [...] Read more.
The emergence of SARS-CoV-2 in 2019 posed significant global public health challenges. One of the most promising targets for novel antiviral drug development is the SARS-CoV-2 main protease (3CLpro). In this study, fragment molecular orbital (FMO) calculations were conducted to provide guidance for the structural modification of natural flavonoids, identifying the pyrogallol moiety as a key candidate. Natural flavonoids were chemically modified to generate 33 semi-synthetic derivatives through the introduction of various functional groups. Our findings revealed that the incorporation of a galloyl moiety significantly enhances anti-proteolytic activity against SARS-CoV-2 3CLpro, achieving up to a 23-fold increase compared to the activity of the parent compounds. Notably, 7-O-galloyl-DMC (40) exhibited the highest anti-proteolytic activity in an enzymatic assay. Additionally, molecular dynamics simulations provided atomic-level insights into the interactions between the galloyl moiety and 3CLpro. All galloylated flavonoid derivatives positioned their galloyl groups within the S1′ sub-pocket, facilitating hydrogen bonding and π-interactions, particularly with Thr26 and Leu27. These findings underscore the potential of the galloyl moiety as a crucial structural element for enhancing the binding affinity of flavonoids with inhibitory activity against SARS-CoV-2 3CLpro. Full article
Show Figures

Graphical abstract

39 pages, 11135 KB  
Article
1,2,4-Thiadiazolidin-3,5-Diones as Inhibitors of Cysteine Proteases
by Maria Aparecida Juliano, Marco Persico, Beatrice Severino, Giuseppe Tumbarello, Debora Okamoto, Karolina Rosa Fernandes, Gabriel Trigo, Aparecida Sadae Tanaka, José Thalles Lacerda, Oleh Tkachuck, Angela Corvino, Ferdinando Fiorino, Antonia Scognamiglio, Francesco Frecentese, Vincenzo Santagada, Stefania Vertuccio, Giuseppe Caliendo, Luiz Juliano and Caterina Fattorusso
Molecules 2025, 30(19), 3896; https://doi.org/10.3390/molecules30193896 - 26 Sep 2025
Viewed by 808
Abstract
A focused library of 1,2,4-thiadiazolidin-3,5-diones (THIA-110), previously characterized as hydrogen sulfide (H2S) donors, was evaluated for inhibitory activity against cysteine proteases. We included two key cysteine proteases aiming at antiviral drug development—SARS-CoV-2 3CLpro (Mpro) and PLpro—alongside reference [...] Read more.
A focused library of 1,2,4-thiadiazolidin-3,5-diones (THIA-110), previously characterized as hydrogen sulfide (H2S) donors, was evaluated for inhibitory activity against cysteine proteases. We included two key cysteine proteases aiming at antiviral drug development—SARS-CoV-2 3CLpro (Mpro) and PLpro—alongside reference enzymes Papain and Cathepsin L. The compounds exhibited distinct selectivity profiles and inhibition mechanisms. The ability to act as covalent inhibitors of 3CLpro in the nanomolar range is of particular interest, with compounds THIA-6, -7, and -10 proving to be the most potent inhibitors of the series, and compounds THIA-1, -2, and -8 proving to be the most selective with respect to the other proteases. We explored the molecular bases of the observed activity profile of THIA-110 through computational studies, which supported and complemented the experimental findings, paving the way for future structure optimization. The results highlight that inhibitory potency depends not only on electrophilicity but also on the ability to access the catalytic cysteine within the active site. The dual functionality of THIA-110 as H2S donors and selective cysteine protease inhibitors underscores its potential as a promising lead for therapeutic development. Full article
Show Figures

Figure 1

16 pages, 3898 KB  
Article
Selective Degradation and Inhibition of SARS-CoV-2 3CLpro by MMP14 Reveals a Novel Strategy for COVID-19 Therapeutics
by Hyun Lee, Yunjeong Hwang, Elizabeth J. Mulder, Yuri Song, Calista Choi, Lijun Rong, Dimitri T. Azar and Kyu-Yeon Han
Int. J. Mol. Sci. 2025, 26(19), 9401; https://doi.org/10.3390/ijms26199401 - 26 Sep 2025
Viewed by 1144
Abstract
Novel therapies to treat infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of respiratory coronavirus disease 2019 (COVID-19), would be of great clinical value to combat the current and future pandemics. Two viral proteases, papain-like protease (PLpro) and [...] Read more.
Novel therapies to treat infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of respiratory coronavirus disease 2019 (COVID-19), would be of great clinical value to combat the current and future pandemics. Two viral proteases, papain-like protease (PLpro) and the main protease 3-chymotrypsin-like protease (3CLpro), are vital in processing the SARS-CoV-2 polyproteins (pp1a and pp1ab) and in releasing 16 nonstructural proteins, making them attractive antiviral drug targets. In this study, we investigated the degradation of the SARS-CoV-2 main protease 3CLpro by matrix metalloproteinase-14 (MMP14). MMP14 is known to recognize over 10 distinct substrate cleavage sequences. Through sequence analysis, we identified 17 and 10 putative MMP14 cleavage motifs within the SARS-CoV-2 3CLpro and PLpro proteases, respectively. Despite the presence of potential sites in both proteins, our in vitro proteolysis assays demonstrated that MMP14 selectively binds to and degrades 3CLpro, but not PLpro. This selective proteolysis by MMP14 results in the complete loss of 3CLpro enzymatic activity. In addition, SARS-CoV-2 pseudovirus replication was inhibited in 293 T cells when either full-length MMP14 or its catalytic domain (cat-MMP14) were overexpressed, presumably due to 3CLpro degradation by MMP14. Finally, to prevent MMP14 from degrading off-target proteins, we propose a new recombinant pro-PL-MMP14 construct that can be activated only by another SARS-CoV-2 protease, PLpro. These findings could open the potential of an alternative therapeutic strategy against SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue Advances in Metalloproteinase)
Show Figures

Graphical abstract

29 pages, 2729 KB  
Article
Computational Evaluation and Multi-Criteria Optimization of Natural Compound Analogs Targeting SARS-CoV-2 Proteases
by Paul Andrei Negru, Andrei-Flavius Radu, Ada Radu, Delia Mirela Tit and Gabriela Bungau
Curr. Issues Mol. Biol. 2025, 47(7), 577; https://doi.org/10.3390/cimb47070577 - 21 Jul 2025
Cited by 1 | Viewed by 1358
Abstract
The global impact of the COVID-19 crisis has underscored the need for novel therapeutic candidates capable of efficiently targeting essential viral proteins. Existing therapeutic strategies continue to encounter limitations such as reduced efficacy against emerging variants, safety concerns, and suboptimal pharmacodynamics, which emphasize [...] Read more.
The global impact of the COVID-19 crisis has underscored the need for novel therapeutic candidates capable of efficiently targeting essential viral proteins. Existing therapeutic strategies continue to encounter limitations such as reduced efficacy against emerging variants, safety concerns, and suboptimal pharmacodynamics, which emphasize the potential of natural-origin compounds as supportive agents with immunomodulatory, anti-inflammatory, and antioxidant benefits. The present study significantly advances prior molecular docking research through comprehensive virtual screening of structurally related analogs derived from antiviral phytochemicals. These compounds were evaluated specifically against the SARS-CoV-2 main protease (3CLpro) and papain-like protease (PLpro). Utilizing chemical similarity algorithms via the ChEMBL database, over 600 candidate molecules were retrieved and subjected to automated docking, interaction pattern analysis, and comprehensive ADMET profiling. Several analogs showed enhanced binding scores relative to their parent scaffolds, with CHEMBL1720210 (a shogaol-derived analog) demonstrating strong interaction with PLpro (−9.34 kcal/mol), and CHEMBL1495225 (a 6-gingerol derivative) showing high affinity for 3CLpro (−8.04 kcal/mol). Molecular interaction analysis revealed that CHEMBL1720210 forms hydrogen bonds with key PLpro residues including GLY163, LEU162, GLN269, TYR265, and TYR273, complemented by hydrophobic interactions with TYR268 and PRO248. CHEMBL1495225 establishes multiple hydrogen bonds with the 3CLpro residues ASP197, ARG131, TYR239, LEU272, and GLY195, along with hydrophobic contacts with LEU287. Gene expression predictions via DIGEP-Pred indicated that the top-ranked compounds could influence biological pathways linked to inflammation and oxidative stress, processes implicated in COVID-19’s pathology. Notably, CHEMBL4069090 emerged as a lead compound with favorable drug-likeness and predicted binding to PLpro. Overall, the applied in silico framework facilitated the rational prioritization of bioactive analogs with promising pharmacological profiles, supporting their advancement toward experimental validation and therapeutic exploration against SARS-CoV-2. Full article
(This article belongs to the Special Issue Novel Drugs and Natural Products Discovery)
Show Figures

Figure 1

15 pages, 2823 KB  
Article
Discovery of Small Molecules Targeting Norovirus 3CL Protease by Multi-Stage Virtual Screening
by Zhongling Shi, Na Liu, Fabao Zhao, Dongwei Kang, Steven De Jonghe, Johan Neyts, Ni Gao and Xinyong Liu
Int. J. Mol. Sci. 2025, 26(12), 5625; https://doi.org/10.3390/ijms26125625 - 12 Jun 2025
Cited by 1 | Viewed by 1455
Abstract
Human noroviruses (HuNoVs) are the primary cause of acute viral gastroenteritis. There are no antivirals or vaccines available to treat and/or prevent HuNoV. Norovirus 3C-like protease (3CLpro) is essential for viral replication; consequently, the inhibition of this enzyme is a fruitful avenue for [...] Read more.
Human noroviruses (HuNoVs) are the primary cause of acute viral gastroenteritis. There are no antivirals or vaccines available to treat and/or prevent HuNoV. Norovirus 3C-like protease (3CLpro) is essential for viral replication; consequently, the inhibition of this enzyme is a fruitful avenue for antinorovirus therapeutics. To discover novel 3CLpro inhibitors with diverse scaffolds, a multi-stage virtual screening approach was performed by docking >10 million compounds into the 3CLpro catalytic site. An initial subset of 18 compounds was selected, and compounds YY-1029 and YY-4204 were identified as the best two molecules. Molecular dynamics (MD) simulations and binding free energy calculations (MM/GBSA) of YY-1029 and YY-4204 were performed to elucidate the binding mechanisms. The ADMET properties were also estimated to predict the potential druggability of representative molecules. All 18 compounds were evaluated for their antinorovirus activity and cytotoxicity in a cell-based replicon system. This work could provide information for the development of 3CL pro inhibitors. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

16 pages, 4257 KB  
Article
Discovery of Small-Molecule Inhibitors Against Norovirus 3CLpro Using Structure-Based Virtual Screening and FlipGFP Assay
by Hao Shen, Shiqi Liu, Limin Shang, Yuchen Liu, Yijin Sha, Dingwei Lei, Yuehui Zhang, Chaozhi Jin, Shanshan Wu, Mingyang Zhang, Han Wen, Chenxi Jia and Jian Wang
Viruses 2025, 17(6), 814; https://doi.org/10.3390/v17060814 - 4 Jun 2025
Cited by 1 | Viewed by 1437
Abstract
Norovirus, a major cause of acute gastroenteritis, possesses a single-stranded positive-sense RNA genome. The viral 3C-like cysteine protease (3CLpro) plays a critical role in processing the viral polyprotein into mature non-structural proteins, a step essential for viral replication. Targeting 3CLpro [...] Read more.
Norovirus, a major cause of acute gastroenteritis, possesses a single-stranded positive-sense RNA genome. The viral 3C-like cysteine protease (3CLpro) plays a critical role in processing the viral polyprotein into mature non-structural proteins, a step essential for viral replication. Targeting 3CLpro has emerged as a promising strategy for developing small-molecule inhibitors against Norovirus. In this study, we employed a combination of virtual screening and the FlipGFP assay to identify potential inhibitors targeting the 3CLpro of Norovirus genotype GII.4. A library of approximately 58,800 compounds was screened using AutoDock Vina tool, yielding 20 candidate compounds based on their Max Affinity scores. These compounds were subsequently evaluated using a cell-based FlipGFP assay. Among them, eight compounds demonstrated significant inhibitory activity against 3CLpro, with Gedatolisib showing the most potent effect (IC50 = 0.06 ± 0.01 μM). Molecular docking and molecular dynamics simulations were conducted to explore the binding mechanisms and structural stability of the inhibitor–3CLpro complexes. Our findings provide valuable insights into the development of antiviral drugs targeting Norovirus 3CLpro, offering potential therapeutic strategies to combat Norovirus infections. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

13 pages, 2709 KB  
Article
Development of Peptidomimetic PROTACs as Potential Degraders of 3-Chymotrypsin-like Protease of SARS-CoV-2
by Chao Wei, Yuhua Li, Lina Guo, Zhiyu Shao and Hua Diao
Int. J. Mol. Sci. 2025, 26(8), 3903; https://doi.org/10.3390/ijms26083903 - 21 Apr 2025
Cited by 3 | Viewed by 1370
Abstract
3CL protease (3CLpro), a key enzyme of SARS-CoV-2 replication, is one of the most selective targets of antivirals, as no homologous protease has been recognized in the human body. As proteolysis-targeting chimeras (PROTACs) are superior to traditional inhibitors, based on the [...] Read more.
3CL protease (3CLpro), a key enzyme of SARS-CoV-2 replication, is one of the most selective targets of antivirals, as no homologous protease has been recognized in the human body. As proteolysis-targeting chimeras (PROTACs) are superior to traditional inhibitors, based on the reported cereblon (CRBN) ligands thalidomide and lenalidomide, 3CLpro ligands of peptidomimetic inhibitors, and suitable linkers, we aimed to develop novel PROTACs that may trigger efficient intracellular 3CLpro degradation through a balance of hydrophilicity and lipophilicity. In brief, we designed and synthesized 5 PROTAC molecules. The 3CLpro degradation efficiency of the PROTACs was assayed in stable SARS-CoV-2 3CLpro expression HEK293 cell models and evaluated by Western blot. All compounds showed prominent 3CLpro degradation activity with tolerable HEK293 cytotoxicity. The most prominent PROTAC compounds, 15 and 16, have DC50 values of approximately 1 µM, and Dmax of 89.3% and 75% respectively, indicating good potential for further application. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

26 pages, 8825 KB  
Article
Biochemical Screening of Phytochemicals and Identification of Scopoletin as a Potential Inhibitor of SARS-CoV-2 Mpro, Revealing Its Biophysical Impact on Structural Stability
by Sarika Bano, Jyotishna Singh, Zainy Zehra, Md Nayab Sulaimani, Taj Mohammad, Seemasundari Yumlembam, Md Imtaiyaz Hassan, Asimul Islam and Sanjay Kumar Dey
Viruses 2025, 17(3), 402; https://doi.org/10.3390/v17030402 - 12 Mar 2025
Cited by 2 | Viewed by 1772
Abstract
The main protease (Mpro or 3CLpro or nsp5) of SARS-CoV-2 is crucial to the life cycle and pathogenesis of SARS-CoV-2, making it an attractive drug target to develop antivirals. This study employed the virtual screening of a few phytochemicals, and the [...] Read more.
The main protease (Mpro or 3CLpro or nsp5) of SARS-CoV-2 is crucial to the life cycle and pathogenesis of SARS-CoV-2, making it an attractive drug target to develop antivirals. This study employed the virtual screening of a few phytochemicals, and the resultant best compound, Scopoletin, was further investigated by a FRET-based enzymatic assay, revealing an experimental IC50 of 15.75 µM. The impact of Scopoletin on Mpro was further investigated by biophysical and MD simulation studies. Fluorescence spectroscopy identified a strong binding constant of 3.17 × 104 M⁻1 for Scopoletin binding to Mpro, as demonstrated by its effective fluorescence quenching of Mpro. Additionally, CD spectroscopy showed a significant reduction in the helical content of Mpro upon interaction with Scopoletin. The findings of thermodynamic measurements using isothermal titration calorimetry (ITC) supported the spectroscopic data, indicating a tight binding of Scopoletin to Mpro with a KA of 2.36 × 103 M−1. Similarly, interaction studies have also revealed that Scopoletin forms hydrogen bonds with the amino acids nearest to the active site, and this has been further supported by molecular dynamics simulation studies. These findings indicate that Scopoletin may be developed as a potential antiviral treatment for SARS-CoV-2 by targeting Mpro. Full article
(This article belongs to the Special Issue Coronaviruses Pathogenesis, Immunity, and Antivirals (2nd Edition))
Show Figures

Figure 1

28 pages, 11579 KB  
Article
Identifying Exifone as a Dual-Target Agent Targeting Both SARS-CoV-2 3CL Protease and the ACE2/S-RBD Interaction Among Clinical Polyphenolic Compounds
by Jiani Lu, Yan Tang, Hongtao Li, Xixiang Chen, Pengcheng Qin, Jianrong Xu, Weihua Li and Lili Chen
Int. J. Mol. Sci. 2025, 26(5), 2243; https://doi.org/10.3390/ijms26052243 - 2 Mar 2025
Cited by 2 | Viewed by 2359
Abstract
The ongoing emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to resistance against multiple coronavirus disease 2019 (COVID-19) vaccines and therapeutic medications, making the development of effective therapeutics against SARS-CoV-2 a high priority. Studies have shown that bioactive polyphenols, [...] Read more.
The ongoing emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has led to resistance against multiple coronavirus disease 2019 (COVID-19) vaccines and therapeutic medications, making the development of effective therapeutics against SARS-CoV-2 a high priority. Studies have shown that bioactive polyphenols, particularly those with triphenol groups, can effectively inhibit the activity of SARS-CoV-2 3-chymotrypsin-like protease (3CLpro). However, the structural instability of polyphenols necessitates further research. To address this, we conducted a literature review to identify triphenol compounds that are either approved or currently undergoing clinical trials, assessing their potential to inhibit SARS-CoV-2 3CLpro. Exifone and benserazide hydrochloride were identified as the inhibitors of SARS-CoV-2 3CLpro among these compounds, using a fluorescence resonance energy transfer (FRET)-based assay. Benserazide hydrochloride was confirmed as a covalent binder to SARS-CoV-2 3CLpro through time-dependent inhibition and kinetic analysis, with its binding mode elucidated by molecular docking. Notably, exifone not only inhibited the protease activity but also blocked the interaction between the host cell receptor angiotensin-converting enzyme 2 (ACE2) and the SARS-CoV-2 spike protein receptor binding domain (S-RBD), as identified by surface plasmon resonance (SPR) and flow cytometry. Additionally, exifone demonstrated antiviral activity against various SARS-CoV-2-S pseudovirus variants. In conclusion, the discovery of exifone and benserazide hydrochloride underscores the potential of polyphenols in developing conserved 3CLpro inhibitors for coronaviruses, offering new strategies for the rapid development of effective drugs against both current and future coronavirus pandemics. Full article
(This article belongs to the Special Issue Molecular Modeling: Latest Advances and Applications)
Show Figures

Figure 1

26 pages, 6506 KB  
Article
Identifying Inhibitor-SARS-CoV2-3CLpro Binding Mechanism Through Molecular Docking, GaMD Simulations, Correlation Network Analysis and MM-GBSA Calculations
by Jianzhong Chen, Jian Wang, Wanchun Yang, Lu Zhao and Xiaoyan Xu
Molecules 2025, 30(4), 805; https://doi.org/10.3390/molecules30040805 - 10 Feb 2025
Cited by 9 | Viewed by 2167
Abstract
The main protease of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), known as 3CLpro, is crucial in the virus’s life cycle and plays a pivotal role in COVID-19. Understanding how small molecules inhibit 3CLpro’s activity is vital for [...] Read more.
The main protease of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), known as 3CLpro, is crucial in the virus’s life cycle and plays a pivotal role in COVID-19. Understanding how small molecules inhibit 3CLpro’s activity is vital for developing anti-COVID-19 therapeutics. To this end, we employed Gaussian accelerated molecular dynamics (GaMD) simulations to enhance the sampling of 3CLpro conformations and conducted correlation network analysis (CNA) to explore the interactions between different structural domains. Our findings indicate that a CNA-identified node in domain II of 3CLpro acts as a conduit, transferring conformational changes from the catalytic regions in domains I and II, triggered by the binding of inhibitors (7YY, 7XB, and Y6G), to domain III, thereby modulating 3CLpro’s activity. Normal mode analysis (NMA) and principal component analysis (PCA) revealed that inhibitor binding affects the structural flexibility and collective movements of the catalytic sites and domain III, influencing 3CLpro’s function. The binding free energies, predicted by both MM-GBSA and QM/MM-GBSA methods, showed a high correlation with experimental data, validating the reliability of our analyses. Furthermore, residues L27, H41, C44, S46, M49, N142, G143, S144, C145, H163, H164, M165, and E166, identified through residue-based free energy decomposition, present promising targets for the design of anti-COVID-19 drugs and could facilitate the development of clinically effective 3CLpro inhibitors. Full article
Show Figures

Figure 1

22 pages, 5934 KB  
Article
Molecular Insights into Structural Dynamics and Binding Interactions of Selected Inhibitors Targeting SARS-CoV-2 Main Protease
by Yuanyuan Wang, Yulin Zhou and Faez Iqbal Khan
Int. J. Mol. Sci. 2024, 25(24), 13482; https://doi.org/10.3390/ijms252413482 - 16 Dec 2024
Cited by 9 | Viewed by 3741
Abstract
The SARS-CoV-2 main protease (Mpro, also known as 3CLpro) is a key target for antiviral therapy due to its critical role in viral replication and maturation. This study investigated the inhibitory effects of Bofutrelvir, Nirmatrelvir, and Selinexor on 3CLpro through molecular docking, molecular [...] Read more.
The SARS-CoV-2 main protease (Mpro, also known as 3CLpro) is a key target for antiviral therapy due to its critical role in viral replication and maturation. This study investigated the inhibitory effects of Bofutrelvir, Nirmatrelvir, and Selinexor on 3CLpro through molecular docking, molecular dynamics (MD) simulations, and free energy calculations. Nirmatrelvir exhibited the strongest binding affinity across docking tools (AutoDock Vina: −8.3 kcal/mol; DiffDock: −7.75 kcal/mol; DynamicBound: 7.59 to 7.89 kcal/mol), outperforming Selinexor and Bofutrelvir. Triplicate 300 ns MD simulations revealed that the Nirmatrelvir-3CLpro complex displayed high conformational stability, reduced root mean square deviation (RMSD), and a modest decrease in solvent-accessible surface area (SASA), indicating enhanced structural rigidity. Gibbs free energy analysis highlighted greater flexibility in unbound 3CLpro, stabilized by Nirmatrelvir binding, supported by stable hydrogen bonds. MolProphet prediction tools, targeting the Cys145 residue, confirmed that Nirmatrelvir exhibited the strongest binding, forming multiple hydrophobic, hydrogen, and π-stacking interactions with key residues, and had the lowest predicted IC50/EC50 (9.18 × 10−8 mol/L), indicating its superior potency. Bofutrelvir and Selinexor showed weaker interactions and higher IC50/EC50 values. MM/PBSA analysis calculated a binding free energy of −100.664 ± 0.691 kJ/mol for the Nirmatrelvir-3CLpro complex, further supporting its stability and binding potency. These results underscore Nirmatrelvir’s potential as a promising therapeutic agent for SARS-CoV-2 and provide novel insights into dynamic stabilizing interactions through AI-based docking and long-term MD simulations. Full article
Show Figures

Figure 1

28 pages, 5658 KB  
Review
Mechanistic Insights into the Mutational Landscape of the Main Protease/3CLPro and Its Impact on Long-Term COVID-19/SARS-CoV-2 Management
by Aganze Gloire-Aimé Mushebenge, Samuel Chima Ugbaja, Nonjabulo Ntombikhona Magwaza, Nonkululeko Avril Mbatha, Tambwe Willy Muzumbukilwa, Mukanda Gedeon Kadima, Fave Yohanna Tata, Mthokosizi Bongani Nxumalo, Riziki Ghislain Manimani, Ntabaza Ndage, Bakari Salvius Amuri, Kahumba Byanga, Manimbulu Nlooto, Rene B. Khan and Hezekiel M. Kumalo
Future Pharmacol. 2024, 4(4), 825-852; https://doi.org/10.3390/futurepharmacol4040044 - 28 Nov 2024
Cited by 2 | Viewed by 4631
Abstract
The main proteinase (Mpro), or 3CLpro, is a critical enzyme in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lifecycle and is responsible for breaking down and releasing vital functional viral proteins crucial for virus development and transmission. As a catalytically active dimer, [...] Read more.
The main proteinase (Mpro), or 3CLpro, is a critical enzyme in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lifecycle and is responsible for breaking down and releasing vital functional viral proteins crucial for virus development and transmission. As a catalytically active dimer, its dimerization interface has become an attractive target for antiviral drug development. Recent research has extensively investigated the enzymatic activity of Mpro, focusing on its role in regulating the coronavirus replication complex and its significance in virus maturation and infectivity. Computational investigations have identified four druggable pockets, suggesting potential allosteric sites beyond the substrate-binding region. Empirical validation through site-directed alanine mutagenesis has targeted residues in both the active and allosteric regions and corroborated these predictions. Structural studies of drug target proteins can inform therapeutic approaches, with metadynamics simulations shedding light on the role of H163 in regulating Mpro function and providing insights into its dynamic equilibrium to the wild-type enzyme. Despite the efficacy of vaccines and drugs in mitigating SARS-CoV-2 spread, its ongoing viral evolution, selective pressures, and continued transmission pose challenges, potentially leading to resistant mutations. Phylogenetic analyses have indicated the existence of several resistant variations predating drug introduction to the human population, emphasizing the likelihood of drug spread. Hydrogen/deuterium-exchange mass spectrometry reveals the structural influence of the mutation. At the same time, clinical trials on 3CLPro inhibitors underscore the clinical significance of reduced enzymatic activity and offer avenues for future therapeutic exploration. Understanding the implications of 3CLPro mutations holds promise for shaping forthcoming therapeutic strategies against COVID-19. This review delves into factors influencing mutation rates and identifies areas warranting further investigation, providing a comprehensive overview of Mpro mutations, categorization, and terminology. Moreover, we examine their associations with clinical outcomes, illness severity, unresolved issues, and future research prospects, including their impact on vaccine efficacy and potential therapeutic targeting. Full article
Show Figures

Figure 1

18 pages, 6075 KB  
Article
A Structural Investigation of the Interaction between a GC-376-Based Peptidomimetic PROTAC and Its Precursor with the Viral Main Protease of Coxsackievirus B3
by Alessia De Santis, Deborah Grifagni, Andrea Orsetti, Elena Lenci, Antonio Rosato, Mariapina D’Onofrio, Andrea Trabocchi, Simone Ciofi-Baffoni, Francesca Cantini and Vito Calderone
Biomolecules 2024, 14(10), 1260; https://doi.org/10.3390/biom14101260 - 6 Oct 2024
Cited by 2 | Viewed by 2417
Abstract
The conservation of the main protease in viral genomes, combined with the absence of a homologous protease in humans, makes this enzyme family an ideal target for developing broad-spectrum antiviral drugs with minimized host toxicity. GC-376, a peptidomimetic 3CL protease inhibitor, has shown [...] Read more.
The conservation of the main protease in viral genomes, combined with the absence of a homologous protease in humans, makes this enzyme family an ideal target for developing broad-spectrum antiviral drugs with minimized host toxicity. GC-376, a peptidomimetic 3CL protease inhibitor, has shown significant efficacy against coronaviruses. Recently, a GC-376-based PROTAC was developed to target and induce the proteasome-mediated degradation of the dimeric SARS-CoV-2 3CLPro protein. Extending this approach, the current study investigates the application of the GC-376 PROTAC to the 3CPro protease of enteroviruses, specifically characterizing its interaction with CVB3 3CPro through X-ray crystallography, NMR (Nuclear Magnetic Resonance) and biochemical techniques. The crystal structure of CVB3 3CPro bound to the GC-376 PROTAC precursor was obtained at 1.9 Å resolution. The crystallographic data show that there are some changes between the binding of CVB3 3CPro and SARS-CoV-2 3CLPro, but the overall similarity is strong (RMSD on C-alpha 0.3 Å). The most notable variation is the orientation of the benzyloxycarbonyl group of GC-376 with the S4 subsite of the proteases. NMR backbone assignment of CVB3 3CPro bound and unbound to the GC-376 PROTAC precursor (80% and 97%, respectively) was obtained. This information complemented the investigation, by NMR, of the interaction of CVB3 3CPro with the GC-376 PROTAC, and its precursor allows us to define that the GC-376 PROTAC binds to CVB3 3CPro in a mode very similar to that of the precursor. The NMR relaxation data indicate that a quench of dynamics of a large part of the protein backbone involving the substrate-binding site and surrounding regions occurs upon GC-376 PROTAC precursor binding. This suggests that the substrate cavity, by sampling different backbone conformations in the absence of the substrate, is able to select the suitable one necessary to covalently bind the substrate, this being the latter reaction, which is the fundamental step required to functionally activate the enzymatic reaction. The inhibition activity assay showed inhibition potency in the micromolar range for GC-376 PROTAC and its precursor. Overall, we can conclude that the GC-376 PROTAC fits well within the binding sites of both proteases, demonstrating its potential as a broad-spectrum antiviral agent. Full article
(This article belongs to the Section Molecular Structure and Dynamics)
Show Figures

Figure 1

14 pages, 4660 KB  
Article
β-Tocotrienol and δ-Tocotrienol as Additional Inhibitors of the Main Protease of Feline Infectious Peritonitis Virus: An In Silico Analysis
by Manos C. Vlasiou, Georgios Nikolaou, Kyriakos Spanoudes and Daphne E. Mavrides
Vet. Sci. 2024, 11(9), 424; https://doi.org/10.3390/vetsci11090424 - 11 Sep 2024
Cited by 1 | Viewed by 3248
Abstract
Feline infectious peritonitis (FIP) is a severe and invariably fatal disease affecting both domestic and wild felines with limited effective therapeutic options available. By considering the significant immunomodulatory effects of vitamin E observed in both animal and human models under physiological and pathological [...] Read more.
Feline infectious peritonitis (FIP) is a severe and invariably fatal disease affecting both domestic and wild felines with limited effective therapeutic options available. By considering the significant immunomodulatory effects of vitamin E observed in both animal and human models under physiological and pathological conditions, we have provided a full in silico investigation of vitamin E and related compounds and their effect on the crystal structure of feline infectious peritonitis virus 3C-like protease (FIPV-3CLpro). This work revealed the β-tocotrienol and δ-tocotrienol analogs as inhibitor candidates for this protein, suggesting their potential as possible drug compounds against FIP or their supplementary use with current medicines against this disease. Full article
(This article belongs to the Section Veterinary Physiology, Pharmacology, and Toxicology)
Show Figures

Figure 1

Back to TopTop