Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932170

ABSTRACT

The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has triggered a global COVID-19 pandemic, challenging healthcare systems worldwide. Effective therapeutic strategies against this novel coronavirus remain limited, underscoring the urgent need for innovative approaches. The present research investigates the potential of cannabis compounds as therapeutic agents against SARS-CoV-2 through their interaction with the virus's papain-like protease (PLpro) protein, a crucial element in viral replication and immune evasion. Computational methods, including molecular docking and molecular dynamics (MD) simulations, were employed to screen cannabis compounds against PLpro and analyze their binding mechanisms and interaction patterns. The results showed cannabinoids with binding affinities ranging from -6.1 kcal/mol to -4.6 kcal/mol, forming interactions with PLpro. Notably, Cannabigerolic and Cannabidiolic acids exhibited strong binding contacts with critical residues in PLpro's active region, indicating their potential as viral replication inhibitors. MD simulations revealed the dynamic behavior of cannabinoid-PLpro complexes, highlighting stable binding conformations and conformational changes over time. These findings shed light on the mechanisms underlying cannabis interaction with SARS-CoV-2 PLpro, aiding in the rational design of antiviral therapies. Future research will focus on experimental validation, optimizing binding affinity and selectivity, and preclinical assessments to develop effective treatments against COVID-19.


Subject(s)
Antiviral Agents , Cannabinoids , Molecular Docking Simulation , Molecular Dynamics Simulation , SARS-CoV-2 , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Cannabinoids/pharmacology , Cannabinoids/chemistry , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism , Protein Binding , COVID-19 Drug Treatment , Virus Replication/drug effects , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/metabolism
2.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928340

ABSTRACT

Papain-like protease PLpro, a domain within a large polyfunctional protein, nsp3, plays key roles in the life cycle of SARS-CoV-2, being responsible for the first events of cleavage of a polyprotein into individual proteins (nsp1-4) as well as for the suppression of cellular immunity. Here, we developed a new genetically encoded fluorescent sensor, named PLpro-ERNuc, for detection of PLpro activity in living cells using a translocation-based readout. The sensor was designed as follows. A fragment of nsp3 protein was used to direct the sensor on the cytoplasmic surface of the endoplasmic reticulum (ER) membrane, thus closely mimicking the natural target of PLpro. The fluorescent part included two bright fluorescent proteins-red mScarlet I and green mNeonGreen-separated by a linker with the PLpro cleavage site. A nuclear localization signal (NLS) was attached to ensure accumulation of mNeonGreen into the nucleus upon cleavage. We tested PLpro-ERNuc in a model of recombinant PLpro expressed in HeLa cells. The sensor demonstrated the expected cytoplasmic reticular network in the red and green channels in the absence of protease, and efficient translocation of the green signal into nuclei in the PLpro-expressing cells (14-fold increase in the nucleus/cytoplasm ratio). Then, we used PLpro-ERNuc in a model of Huh7.5 cells infected with the SARS-CoV-2 virus, where it showed robust ER-to-nucleus translocation of the green signal in the infected cells 24 h post infection. We believe that PLpro-ERNuc represents a useful tool for screening PLpro inhibitors as well as for monitoring virus spread in a culture.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , HeLa Cells , COVID-19/virology , COVID-19/diagnosis , COVID-19/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Coronavirus Papain-Like Proteases/metabolism , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Coronavirus 3C Proteases/metabolism , Protein Transport , Biosensing Techniques/methods
3.
J Med Chem ; 67(12): 10211-10232, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38871484

ABSTRACT

Papain-like protease (PLpro) is a promising therapeutic target for its pivotal role in the life cycle of SARS-CoV-2. A series of 1,2,4-oxadiazole derivatives was designed and synthesized via a ring formation strategy based on SARS-CoV-2 PLpro-GRL0617 complex structure. Systematic structure-activity relationship studies revealed that introducing oxadiazole and aryl carboxylic acid moieties to GRL0617 enhanced the enzymatic inhibition activity, affinity, and deubiquitination capacity toward PLpro. 1,2,4-Oxadiazole compounds 13f and 26r, which had PLpro inhibition activity (IC50 = 1.8 and 1.0 µM) and antiviral activity against SARS-CoV-2 (EC50 = 5.4 and 4.3 µM), exhibited good metabolic stability (t1/2 > 93.2 min) and higher plasma exposure (AUC0-t = 17,380.08 and 24,289.76 ng·h/mL) in mice. Especially, compound 26r with moderate oral bioavailability of 39.1% and potent antiviral activity is worthy of further studies in vivo. Our findings provide a new insight for the discovery of antiviral agents targeting PLpro.


Subject(s)
Antiviral Agents , Drug Design , Oxadiazoles , SARS-CoV-2 , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Animals , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Structure-Activity Relationship , SARS-CoV-2/drug effects , Mice , Humans , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Carboxylic Acids/chemical synthesis , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , COVID-19 Drug Treatment , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism
4.
Gigascience ; 132024 Jan 02.
Article in English | MEDLINE | ID: mdl-38869150

ABSTRACT

Viral helicases are promising targets for the development of antiviral therapies. Given their vital function of unwinding double-stranded nucleic acids, inhibiting them blocks the viral replication cycle. Previous studies have elucidated key structural details of these helicases, including the location of substrate binding sites, flexible domains, and the discovery of potential inhibitors. Here we present a series of new Galaxy tools and workflows for performing and analyzing molecular dynamics simulations of viral helicases. We first validate them by demonstrating recapitulation of data from previous simulations of Zika (NS3) and SARS-CoV-2 (NSP13) helicases in apo and complex with inhibitors. We further demonstrate the utility and generalizability of these Galaxy workflows by applying them to new cases, proving their usefulness as a widely accessible method for exploring antiviral activity.


Subject(s)
Molecular Dynamics Simulation , SARS-CoV-2 , SARS-CoV-2/enzymology , Zika Virus/enzymology , Workflow , RNA Helicases/chemistry , RNA Helicases/metabolism , Humans , DNA Helicases/chemistry , DNA Helicases/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , Binding Sites , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
5.
Biol Pharm Bull ; 47(5): 965-966, 2024.
Article in English | MEDLINE | ID: mdl-38763750

ABSTRACT

The emergence of coronavirus disease 2019 (COVID-19), a novel identified pneumonia resulting from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, has significantly impacted and posed significant challenges to human society. The papain-like protease (PLpro) found in the nonstructural protein 3 of SARS-CoV-2 plays a vital role in viral replication. Moreover, PLpro disrupts the host immune response by cleaving ubiquitin and interferon-stimulated gene 15 from host proteins. Consequently, PLpro has emerged as a promising drug target against SARS-CoV-2 infection. Computational studies have reported that ciclesonide can bind to SARS-CoV-2 PLpro. However, the inhibitory effects of ciclenoside on the PLpro have not been experimentally evaluated. Here, we evaluated the inhibitory effects of synthetic glucocorticoids (sGCs), including ciclesonide, on SARS-CoV-2 PLpro in vitro assay. Ciclesonide significantly inhibited the enzymatic activity of PLpro, compared with other sGCs and its IC50 was 18.4 ± 1.89 µM. These findings provide insights into the development of PLpro inhibitors.


Subject(s)
Pregnenediones , SARS-CoV-2 , Pregnenediones/pharmacology , SARS-CoV-2/drug effects , Humans , COVID-19 Drug Treatment , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism , Antiviral Agents/pharmacology , Molecular Docking Simulation , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Glucocorticoids/pharmacology , COVID-19/virology
6.
J Nat Med ; 78(3): 784-791, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38512650

ABSTRACT

Papain-like protease (PLpro) enzyme plays a vital role in viral replication as it breaks down polyproteins and disrupts the host's immune response. There are few reports on Kampo formulas that focus on PLpro activity. In this study, we evaluated the inhibitory effects of senkyuchachosan, a traditional Japanese medicine, on PLpro of SARS-CoV-2, the virus responsible for causing COVID-19. We purified the PLpro enzyme and conducted in vitro enzymatic assays using specific substrates. Among the nine crude drugs present in senkyuchachosan, four (Cyperi Rhizoma, Schizonepetae Spica, Menthae Herba, and Camelliae sinensis Folium [CsF]) strongly inhibited PLpro activity. CsF, derived from Camellia sinensis (green tea), contains polyphenols, including catechins and tannins. To confirm that the PLpro inhibitory effects of senkyuchachosan predominantly stem from tannins, the tannins were removed from the decoction using polyvinylpolypyrrolidone (PVPP). The inhibitory effect of senkyuchachosan on PLpro activity was reduced by the removal of PVPP. In addition, the tannin fraction obtained from the CsF extracts showed significant PLpro inhibitory effects. These findings lay the groundwork for the potential development of therapeutic agents that target SARS-CoV-2 infection by intervening in proteolytic cleavage of the virus.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Plant Extracts , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism , COVID-19/virology , Medicine, Kampo , Plant Extracts/pharmacology , Plant Extracts/chemistry , SARS-CoV-2/drug effects , Tannins/pharmacology
7.
PLoS Pathog ; 20(3): e1012100, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38527094

ABSTRACT

The coronavirus papain-like protease (PLpro) is crucial for viral replicase polyprotein processing. Additionally, PLpro can subvert host defense mechanisms by its deubiquitinating (DUB) and deISGylating activities. To elucidate the role of these activities during SARS-CoV-2 infection, we introduced mutations that disrupt binding of PLpro to ubiquitin or ISG15. We identified several mutations that strongly reduced DUB activity of PLpro, without affecting viral polyprotein processing. In contrast, mutations that abrogated deISGylating activity also hampered viral polyprotein processing and when introduced into the virus these mutants were not viable. SARS-CoV-2 mutants exhibiting reduced DUB activity elicited a stronger interferon response in human lung cells. In a mouse model of severe disease, disruption of PLpro DUB activity did not affect lethality, virus replication, or innate immune responses in the lungs. This suggests that the DUB activity of SARS-CoV-2 PLpro is dispensable for virus replication and does not affect innate immune responses in vivo. Interestingly, the DUB mutant of SARS-CoV replicated to slightly lower titers in mice and elicited a diminished immune response early in infection, although lethality was unaffected. We previously showed that a MERS-CoV mutant deficient in DUB and deISGylating activity was strongly attenuated in mice. Here, we demonstrate that the role of PLpro DUB activity during infection can vary considerably between highly pathogenic coronaviruses. Therefore, careful considerations should be taken when developing pan-coronavirus antiviral strategies targeting PLpro.


Subject(s)
COVID-19 , Coronavirus Papain-Like Proteases , Humans , Animals , Mice , Coronavirus Papain-Like Proteases/genetics , SARS-CoV-2/metabolism , Immunity, Innate , Papain/genetics , Papain/metabolism , Peptide Hydrolases/metabolism , Virus Replication , Polyproteins
8.
J Virol ; 98(4): e0157523, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38483167

ABSTRACT

As for all single-stranded, positive-sense RNA (+RNA) viruses, intracellular RNA synthesis relies on extensive remodeling of host cell membranes that leads to the formation of specialized structures. In the case of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronavirus causing COVID-19, endoplasmic reticulum membranes are modified, resulting in the formation of double-membrane vesicles (DMVs), which contain the viral dsRNA intermediate and constitute membrane-bound replication organelles. The non-structural and transmembrane protein nsp3 is a key player in the biogenesis of DMVs and, therefore, represents an interesting antiviral target. However, as an integral transmembrane protein, it is challenging to express for structural biology. The C-terminus of nsp3 encompasses all the membrane-spanning, -interacting, and -remodeling elements. By using a cell-free expression system, we successfully produced the C-terminal region of nsp3 (nsp3C) and reconstituted purified nsp3C into phospholipid nanodiscs, opening the way for structural studies. Negative-stain transmission electron microscopy revealed the presence of nsp3C oligomers very similar to the region abutting and spanning the membrane on the cytosolic side of DMVs in a recent subtomogram average of the SARS-CoV-2 nsp3-4 pore (1). AlphaFold-predicted structural models fit particularly well with our experimental data and support a pore-forming hexameric assembly. Altogether, our data give unprecedented clues to understand the structural organization of nsp3, the principal component that shapes the molecular pore that spans the DMVs and is required for the export of RNA in vivo. IMPORTANCE: Membrane remodeling is at the heart of intracellular replication for single-stranded, positive-sense RNA viruses. In the case of coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), this leads to the formation of a network of double-membrane vesicles (DMVs). Targeting DMV biogenesis offers promising prospects for antiviral therapies. This requires a better understanding of the molecular mechanisms and proteins involved. Three non-structural proteins (nsp3, nsp4, and nsp6) direct the intracellular membrane rearrangements upon SARS-CoV-2 infection. All of them contain transmembrane helices. The nsp3 component, the largest and multi-functional protein of the virus, plays an essential role in this process. Aiming to understand its structural organization, we used a cell-free protein synthesis assay to produce and reconstitute the C-terminal part of nsp3 (nsp3C) including transmembrane domains into phospholipid nanodiscs. Our work reveals the oligomeric organization of one key player in the biogenesis of SARS-CoV-2 DMVs, providing basis for the design of future antiviral strategies.


Subject(s)
COVID-19 , RNA, Viral , SARS-CoV-2 , Viral Nonstructural Proteins , Humans , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , COVID-19/virology , Endoplasmic Reticulum/metabolism , Phospholipids , RNA, Viral/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
9.
Science ; 383(6690): 1434-1440, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38547259

ABSTRACT

The emergence of SARS-CoV-2 variants and drug-resistant mutants calls for additional oral antivirals. The SARS-CoV-2 papain-like protease (PLpro) is a promising but challenging drug target. We designed and synthesized 85 noncovalent PLpro inhibitors that bind to a recently discovered ubiquitin binding site and the known BL2 groove pocket near the S4 subsite. Leads inhibited PLpro with the inhibitory constant Ki values from 13.2 to 88.2 nanomolar. The co-crystal structures of PLpro with eight leads revealed their interaction modes. The in vivo lead Jun12682 inhibited SARS-CoV-2 and its variants, including nirmatrelvir-resistant strains with EC50 from 0.44 to 2.02 micromolar. Oral treatment with Jun12682 improved survival and reduced lung viral loads and lesions in a SARS-CoV-2 infection mouse model, suggesting that PLpro inhibitors are promising oral SARS-CoV-2 antiviral candidates.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Coronavirus Papain-Like Proteases , Coronavirus Protease Inhibitors , Drug Design , SARS-CoV-2 , Animals , Mice , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/chemistry , Disease Models, Animal , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Coronavirus Protease Inhibitors/administration & dosage , Coronavirus Protease Inhibitors/chemistry , Coronavirus Protease Inhibitors/pharmacology , Administration, Oral , Crystallography, X-Ray , Structure-Activity Relationship , Viral Load/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred BALB C
10.
Science ; 383(6681): 354, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38271509

ABSTRACT

Viral protein prods nerve cells, may be treatment target.


Subject(s)
COVID-19 , Coronavirus Papain-Like Proteases , Nociceptors , SARS-CoV-2 , Sneezing , Humans , COVID-19/physiopathology , SARS-CoV-2/enzymology , Animals , Mice , Nociceptors/virology
11.
Phytomedicine ; 123: 155176, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37976697

ABSTRACT

BACKGROUND: Selected natural compounds exhibit very good antiviral properties. Especially, the medicinal plant Humulus lupulus (hop) contains several secondary plant metabolites some of which have previously shown antiviral activities. Among them, the prenylated chalcone xanthohumol (XN) demonstrated to be a potent inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro). HYPOTHESIS/PURPOSE: Following the finding that xanthohumol (XN) is a potent inhibitor of SARS-CoV-2 Mpro, the effect of XN and its major derivatives isoxanthohumol (IXN), 6-prenylnaringenin (6-PN), and 8-prenylnaringenin (8-PN) from hops on SARS-CoV-2 papain-like protease (PLpro) were investigated. STUDY DESIGN: The modulatory effect of the hop compounds on PLpro were studied first in silico and then in vitro. In addition, the actual effect of hop compounds on the replication of SARS-CoV-2 in host cells was investigated. METHODS: In silico docking analysis was used to predict the binding affinity of hop compounds to the active site of PLpro. A recombinant PLpro was cloned, purified, characterized, and analyzed by small-angle X-ray scattering (SAXS), deISGylation assays, and kinetic analyses. Antiviral activity of hop compounds was assessed using the fluorescently labeled wildtype SARS-CoV-2 (icSARS-CoV-2-mNG) in Caco-2 host cells. RESULTS: Our in silico docking suggests that the purified hop compounds bind to the active site of SARS-CoV-2 PLpro blocking the access of its natural substrates. The hop-derived compounds inhibit SARS-CoV-2 PLpro with half maximal inhibitory concentration (IC50) values in the range of 59-162 µM. Furthermore, we demonstrate that XN and 6-PN, in particular, impede viral replication with IC50 values of 3.3 µM and 7.3 µM, respectively. CONCLUSION: In addition to the already known inhibition of Mpro by XN, our results show, for the first time, that hop-derived compounds target also SARS-CoV-2 PLpro which is a promising therapeutic target as it contributes to both viral replication and modulation of the immune system. These findings support the possibility to develop new hop-derived antiviral drugs targeting human coronaviruses.


Subject(s)
COVID-19 , Coronavirus Papain-Like Proteases , Flavonoids , Humulus , Propiophenones , Humans , Humulus/chemistry , Caco-2 Cells , Scattering, Small Angle , SARS-CoV-2 , X-Ray Diffraction , Virus Replication , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Molecular Docking Simulation
12.
ACS Chem Biol ; 19(1): 22-36, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38150587

ABSTRACT

The papain-like protease of SARS-COV-2 is essential for viral replication and pathogenesis. Its location within a much larger multifunctional protein, NSP3, makes it an ideal candidate for a targeted degradation approach capable of eliminating multiple functions with a single-molecule treatment. In this work, we have developed a HiBiT-based cellular model to study NSP3 degradation and used this platform for the discovery of monovalent NSP3 degraders. We present previously unreported degradation activity of published papain-like protease inhibitors. Follow-up exploration of structure-activity relationships and mechanism-of-action studies points to the recruitment of the ubiquitin-proteasome machinery that is solely driven by site occupancy, regardless of molecular features of the ligand. Supported by HDX data, we hypothesize that binding-induced structural changes in NSP3 trigger the recruitment of an E3 ligase and lead to proteasomal degradation.


Subject(s)
COVID-19 , Coronavirus Papain-Like Proteases , Papain , Humans , Papain/metabolism , Viral Nonstructural Proteins/metabolism , SARS-CoV-2/chemistry , Protease Inhibitors/metabolism
13.
Chem Pharm Bull (Tokyo) ; 71(12): 897-905, 2023.
Article in English | MEDLINE | ID: mdl-38044142

ABSTRACT

Virtual screening with high-performance computers is a powerful and cost-effective technique in drug discovery. A chemical database is searched to find candidate compounds firmly bound to a target protein, judging from the binding poses and/or binding scores. The severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) infectious disease has spread worldwide for the last three years, causing severe slumps in economic and social activities. SARS-Cov-2 has two viral proteases: 3-chymotrypsin-like (3CL) and papain-like (PL) protease. While approved drugs have already been released for the 3CL protease, no approved agent is available for PL protease. In this work, we carried out in silico screening for the PL protease inhibitors, combining docking simulation and molecular mechanics calculation. Docking simulations were applied to 8,820 molecules in a chemical database of approved and investigational compounds. Based on the binding poses generated by the docking simulations, molecular mechanics calculations were performed to optimize the binding structures and to obtain the binding scores. Based on the binding scores, 57 compounds were selected for in vitro assay of the inhibitory activity. Five inhibitory compounds were identified from the in vitro measurement. The predicted binding structures of the identified five compounds were examined, and the significant interaction between the individual compound and the protease catalytic site was clarified. This work demonstrates that computational virtual screening by combining docking simulation with molecular mechanics calculation is effective for searching candidate compounds in drug discovery.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Molecular Docking Simulation , Viral Nonstructural Proteins , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Coronavirus Papain-Like Proteases/metabolism , Molecular Dynamics Simulation , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
14.
J Mol Biol ; 435(24): 168337, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37918563

ABSTRACT

Identifying residues critical to protein-protein binding and efficient design of stable and specific protein binders are challenging tasks. Extending beyond the direct contacts in a protein-protein binding interface, our study employs computational modeling to reveal the essential network of residue interactions and dihedral angle correlations critical in protein-protein recognition. We hypothesized that mutating residues exhibiting highly correlated dynamic motion within the interaction network could efficiently optimize protein-protein interactions to create tight and selective protein binders. We tested this hypothesis using the ubiquitin (Ub) and MERS coronaviral papain-like protease (PLpro) complex, since Ub is a central player in multiple cellular functions and PLpro is an antiviral drug target. Our designed ubiquitin variant (UbV) hosting three mutated residues displayed a ∼3,500-fold increase in functional inhibition relative to wild-type Ub. Further optimization of two C-terminal residues within the Ub network resulted in a KD of 1.5 nM and IC50 of 9.7 nM for the five-point Ub mutant, eliciting 27,500-fold and 5,500-fold enhancements in affinity and potency, respectively, as well as improved selectivity, without destabilizing the UbV structure. Our study highlights residue correlation and interaction networks in protein-protein interactions, and introduces an effective approach to design high-affinity protein binders for cell biology research and future therapeutics.


Subject(s)
Coronavirus Papain-Like Proteases , Middle East Respiratory Syndrome Coronavirus , Ubiquitin , Middle East Respiratory Syndrome Coronavirus/enzymology , Protein Binding , Ubiquitin/chemistry , Ubiquitin/metabolism , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism
15.
J Mol Biol ; 435(23): 168316, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37858708

ABSTRACT

Deubiquitinases (DUBs) are proteolytic enzymes that catalyze the removal of ubiquitin from protein substrates. The critical role of DUBs in regulating protein ubiquitination makes them attractive drug targets in oncology, neurodegenerative disease, and antiviral development. Biochemical assays for quantifying DUB activity have enabled characterization of substrate preferences and discovery of small molecule inhibitors. However, assessing the efficacy of these inhibitors in cellular contexts to support clinical drug development has been limited by a lack of tractable cell-based assays. To address this gap, we developed a two-color flow cytometry-based assay that allows for sensitive quantification of DUB activity and inhibition in living cells. The utility of this system was demonstrated by quantifying the potency of GRL0617 against the viral DUB SARS-CoV-2 PLpro, identifying potential GRL0617 resistance mutations, and performing structure-function analysis of the vOTU domain from the recently emerged Yezo virus. In addition, the system was optimized for cellular DUBs by modifying a GFP-targeting nanobody to recruit USP7 and USP28 to benchmark a panel of reported inhibitors and assess inhibition kinetics. Together, these results demonstrate the utility of these assays for studying DUB biology in a cellular context with potential to aid in inhibitor discovery and development.


Subject(s)
Deubiquitinating Enzymes , Flow Cytometry , Protease Inhibitors , Humans , Aniline Compounds/pharmacology , Benzamides/pharmacology , Deubiquitinating Enzymes/analysis , Deubiquitinating Enzymes/antagonists & inhibitors , Neurodegenerative Diseases/enzymology , Ubiquitin/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitination/drug effects , Flow Cytometry/methods , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Coronavirus Papain-Like Proteases/analysis , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Single-Domain Antibodies
16.
Bioorg Chem ; 140: 106830, 2023 11.
Article in English | MEDLINE | ID: mdl-37683544

ABSTRACT

Global coronavirus disease 2019 (COVID-19) pandemic still threatens human health and public safety, and the development of effective antiviral agent is urgently needed. The SARS-CoV-2 main protease (Mpro) and papain-like protease (PLpro) are vital proteins in viral replication and promising therapeutic targets. Additionally, PLpro also modulates host immune response by cleaving ubiquitin and interferon-stimulated gene product 15 (ISG15) from ISGylated host proteins. In this report, we identified [1,2]selenazolo[5,4-c]pyridin-3(2H)-one and benzo[d]isothiazol-3(2H)-one as attractive scaffolds of PLpro and Mpro inhibitors. The representative compounds 6c and 7e exhibited excellent PLpro inhibition with percent inhibition of 42.9% and 44.9% at 50 nM, respectively. The preliminary enzyme kinetics experiment and fluorescent labelling experiment results determined that 6c was identified as a covalent PLpro inhibitor, while 7e was a non-covalent inhibitor. Molecular docking and dynamics simulations revealed that 6c and 7e bound to Zn-finger domain of PLpro. Compounds 6c and 7e were also identified to potent Mpro inhibitors, and they exhibited potent antiviral activities in SARS-CoV-2 infected Vero E6 cells, with EC50 value of 3.9 µM and 7.4 µM, respectively. In addition, the rat liver homogenate half-life of 6c and 7e exceeded 24 h. These findings suggest that 6c and 7e are promising led compounds for further development of PLpro/Mpro dual-target antiviral drugs.


Subject(s)
COVID-19 , Coronavirus Papain-Like Proteases , Coronavirus Protease Inhibitors , Animals , Humans , Rats , Antiviral Agents/pharmacology , Coloring Agents , Endopeptidases , Molecular Docking Simulation , Peptide Hydrolases , SARS-CoV-2 , Coronavirus Protease Inhibitors/chemistry , Coronavirus Protease Inhibitors/pharmacology , Coronavirus Papain-Like Proteases/antagonists & inhibitors
17.
J Virol ; 97(10): e0050723, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37768083

ABSTRACT

IMPORTANCE: Generation of virus-host protein-protein interactions (PPIs) maps may provide clues to uncover SARS-CoV-2-hijacked cellular processes. However, these PPIs maps were created by expressing each viral protein singularly, which does not reflect the life situation in which certain viral proteins synergistically interact with host proteins. Our results reveal the host-viral protein-protein interactome of SARS-CoV-2 NSP3, NSP4, and NSP6 expressed individually or in combination. Furthermore, REEP5/TRAM1 complex interacts with NSP3 at ROs and promotes viral replication. The significance of our research is identifying virus-host interactions that may be targeted for therapeutic intervention.


Subject(s)
Coronavirus Papain-Like Proteases , Host Microbial Interactions , Membrane Glycoproteins , Membrane Proteins , Membrane Transport Proteins , SARS-CoV-2 , Virus Replication , Humans , COVID-19/virology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Protein Binding , Protein Interaction Maps , SARS-CoV-2/growth & development , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/metabolism , Coronavirus Papain-Like Proteases/metabolism
18.
PLoS Pathog ; 19(8): e1011614, 2023 08.
Article in English | MEDLINE | ID: mdl-37651466

ABSTRACT

Despite unprecedented efforts, our therapeutic arsenal against SARS-CoV-2 remains limited. The conserved macrodomain 1 (Mac1) in NSP3 is an enzyme exhibiting ADP-ribosylhydrolase activity and a possible drug target. To determine the role of Mac1 catalytic activity in viral replication, we generated recombinant viruses and replicons encoding a catalytically inactive NSP3 Mac1 domain by mutating a critical asparagine in the active site. While substitution to alanine (N40A) reduced catalytic activity by ~10-fold, mutations to aspartic acid (N40D) reduced activity by ~100-fold relative to wild-type. Importantly, the N40A mutation rendered Mac1 unstable in vitro and lowered expression levels in bacterial and mammalian cells. When incorporated into SARS-CoV-2 molecular clones, the N40D mutant only modestly affected viral fitness in immortalized cell lines, but reduced viral replication in human airway organoids by 10-fold. In mice, the N40D mutant replicated at >1000-fold lower levels compared to the wild-type virus while inducing a robust interferon response; all animals infected with the mutant virus survived infection. Our data validate the critical role of SARS-CoV-2 NSP3 Mac1 catalytic activity in viral replication and as a promising therapeutic target to develop antivirals.


Subject(s)
Coronavirus Papain-Like Proteases , SARS-CoV-2 , Virus Replication , Animals , Humans , Mice , Alanine , Antiviral Agents , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/genetics , Coronavirus Papain-Like Proteases/metabolism
19.
J Cell Biol ; 222(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37318453

ABSTRACT

SARS-CoV-2 depends on host proteins for successful replication. In this issue, Williams et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202203060) report that the ER membrane-modulating proteins RTN3 and RTN4 are required for the formation of SARS-CoV-2 replication organelles via direct interaction with viral proteins NSP3 and NSP4.


Subject(s)
Carrier Proteins , Membrane Proteins , Nerve Tissue Proteins , Nogo Proteins , SARS-CoV-2 , Virus Replication , Humans , Carrier Proteins/genetics , COVID-19 , Endoplasmic Reticulum , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nogo Proteins/genetics , SARS-CoV-2/physiology , Viral Nonstructural Proteins , Coronavirus Papain-Like Proteases
20.
Sci Signal ; 16(783): eadd0082, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37130168

ABSTRACT

The SARS-CoV-2 papain-like protease (PLpro), which has deubiquitinating activity, suppresses the type I interferon (IFN-I) antiviral response. We investigated the mechanism by which PLpro antagonizes cellular antiviral responses. In HEK392T cells, PLpro removed K63-linked polyubiquitin chains from Lys289 of the stimulator of interferon genes (STING). PLpro-mediated deubiquitination of STING disrupted the STING-IKKε-IRF3 complex that induces the production of IFN-ß and IFN-stimulated cytokines and chemokines. In human airway cells infected with SARS-CoV-2, the combined treatment with the STING agonist diABZi and the PLpro inhibitor GRL0617 resulted in the synergistic inhibition of SARS-CoV-2 replication and increased IFN-I responses. The PLpros of seven human coronaviruses (SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-229E, HCoV-HKU1, HCoV-OC43, and HCoV-NL63) and four SARS-CoV-2 variants of concern (α, ß, γ, and δ) all bound to STING and suppressed STING-stimulated IFN-I responses in HEK293T cells. These findings reveal how SARS-CoV-2 PLpro inhibits IFN-I signaling through STING deubiquitination and a general mechanism used by seven human coronaviral PLpros to dysregulate STING and to facilitate viral innate immune evasion. We also identified simultaneous pharmacological STING activation and PLpro inhibition as a potentially effective strategy for antiviral therapy against SARS-CoV-2.


Subject(s)
Coronavirus Papain-Like Proteases , Immune Evasion , Interferon Type I , Membrane Proteins , Humans , Interferon Type I/immunology , Coronavirus Papain-Like Proteases/metabolism , HEK293 Cells , Ubiquitination , Membrane Proteins/metabolism , SARS-CoV-2/physiology , Virus Replication , COVID-19/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...