Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Chem Biol Interact ; 396: 111040, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38735453

ABSTRACT

Global warming and climate change have made dengue disease a global health issue. More than 50 % of the world's population is at danger of dengue virus (DENV) infection, according to the World Health Organization (WHO). Therefore, a clinically approved dengue fever vaccination and effective treatment are needed. Peptide medication development is new pharmaceutical research. Here we intend to recognize the structural features inhibiting the DENV NS2B/NS3 serine protease for a series of peptide-hybrid inhibitors (R1-R2-Lys-R3-NH2) by the 3D-QSAR technique. Comparative molecular field analysis (q2 = 0.613, r2 = 0.938, r2pred = 0.820) and comparative molecular similarity indices analysis (q2 = 0.640, r2 = 0.928, r2pred = 0.693) were established, revealing minor, electropositive, H-bond acceptor groups at the R1 position, minor, electropositive, H-bond donor groups at the R2 position, and bulky, hydrophobic groups at the R3 position for higher inhibitory activity. Docking studies revealed extensive H-bond and hydrophobic interactions in the binding of tripeptide analogues to the NS2B/NS3 protease. This study provides an insight into the key structural features for the design of peptide-based inhibitors of DENV NS2B/NS3 protease.


Subject(s)
Dengue Virus , Molecular Docking Simulation , Peptides , Quantitative Structure-Activity Relationship , Serine Endopeptidases , Viral Nonstructural Proteins , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Dengue Virus/drug effects , Dengue Virus/enzymology , Serine Endopeptidases/metabolism , Serine Endopeptidases/chemistry , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/metabolism , Binding Sites , Hydrogen Bonding , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hydrophobic and Hydrophilic Interactions , Viral Proteases
2.
Int J Biol Macromol ; 267(Pt 2): 131629, 2024 May.
Article in English | MEDLINE | ID: mdl-38631585

ABSTRACT

Current management of HCV infection is based on Direct-Acting Antiviral Drugs (DAAs). However, resistance-associated mutations, especially in the NS3 and NS5B regions are gradually decreasing the efficacy of DAAs. Among the most effective HCV NS3/4A protease drugs, Sofosbuvir also develops resistance due to mutations in the NS3 and NS5B regions. Four mutations at positions A156Y, L36P, Q41H, and Q80K are classified as high-level resistance mutations. The resistance mechanism of HCV NS3/4A protease toward Sofosbuvir caused by these mutations is still unclear, as there is less information available regarding the structural and functional effects of the mutations against Sofosbuvir. In this work, we combined molecular dynamics simulation, molecular mechanics/Generalized-Born surface area calculation, principal component analysis, and free energy landscape analysis to explore the resistance mechanism of HCV NS3/4A protease due to these mutations, as well as compare interaction changes in wild-type. Subsequently, we identified that the mutant form of HCV NS3/4A protease affects the activity of Sofosbuvir. In this study, the resistance mechanism of Sofosbuvir at the atomic level is proposed. The proposed drug-resistance mechanism will provide valuable guidance for the design of HCV drugs.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Hepacivirus , Molecular Dynamics Simulation , Mutation , Sofosbuvir , Viral Nonstructural Proteins , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , DEAD-box RNA Helicases , Drug Resistance, Viral/genetics , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/enzymology , Nucleoside-Triphosphatase , Serine Endopeptidases , Serine Proteases , Sofosbuvir/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Proteases
3.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673962

ABSTRACT

In the global pandemic scenario, dengue and zika viruses (DENV and ZIKV, respectively), both mosquito-borne members of the flaviviridae family, represent a serious health problem, and considering the absence of specific antiviral drugs and available vaccines, there is a dire need to identify new targets to treat these types of viral infections. Within this drug discovery process, the protease NS2B/NS3 is considered the primary target for the development of novel anti-flavivirus drugs. The NS2B/NS3 is a serine protease that has a dual function both in the viral replication process and in the elusion of the innate immunity. To date, two main classes of NS2B/NS3 of DENV and ZIKV protease inhibitors have been discovered: those that bind to the orthosteric site and those that act at the allosteric site. Therefore, this perspective article aims to discuss the main features of the use of the most potent NS2B/NS3 inhibitors and their impact at the social level.


Subject(s)
Antiviral Agents , Dengue , Protease Inhibitors , Zika Virus Infection , Animals , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , DEAD-box RNA Helicases , Dengue/drug therapy , Dengue/virology , Dengue Virus/drug effects , Nucleoside-Triphosphatase , Protease Inhibitors/therapeutic use , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Serine Endopeptidases/metabolism , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Proteases , Zika Virus/drug effects , Zika Virus/enzymology , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
4.
Antiviral Res ; 226: 105878, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582134

ABSTRACT

Flaviviruses can cause severe illness in humans. Effective and safe vaccines are available for some species; however, for many flaviviruses disease prevention or specific treatments remain unavailable. The viral replication cycle depends on the proteolytic activity of the NS2B-NS3 protease, which releases functional viral proteins from a non-functional polyprotein precursor, rendering the protease a promising drug target. In this study, we characterised recombinant NS2B-NS3 proteases from ten flaviviruses including three unreported proteases from the Usutu, Kyasanur forest disease and Powassan viruses. All protease constructs comprise a covalent Gly4-Ser-Gly4 linker connecting the NS3 serine protease domain with its cofactor NS2B. We conducted a comprehensive cleavage site analysis revealing areas of high conversion. While all proteases were active in enzymatic assays, we noted a 1000-fold difference in catalytic efficiency across proteases from different flaviviruses. Two bicyclic peptide inhibitors displayed anti-pan-flaviviral protease activity with inhibition constants ranging from 10 to 1000 nM.


Subject(s)
Antiviral Agents , Flavivirus , Serine Endopeptidases , Viral Nonstructural Proteins , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Flavivirus/drug effects , Flavivirus/enzymology , Serine Endopeptidases/metabolism , Serine Endopeptidases/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , RNA Helicases/metabolism , RNA Helicases/chemistry , RNA Helicases/genetics , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Viral Proteases , Nucleoside-Triphosphatase , DEAD-box RNA Helicases
5.
Virus Res ; 344: 199368, 2024 06.
Article in English | MEDLINE | ID: mdl-38588924

ABSTRACT

Several viruses are now known to code for deubiquitinating proteases in their genomes. Ubiquitination is an essential post-translational modification of cellular substrates involved in many processes in the cell, including in innate immune signalling. This post-translational modification is regulated by the ubiquitin conjugation machinery, as well as various host deubiquitinating enzymes. The conjugation of ubiquitin chains to several innate immune related factors is often needed to induce downstream signalling, shaping the antiviral response. Viral deubiquitinating proteins, besides often having a primary function in the viral replication cycle by cleaving the viral polyprotein, are also able to cleave ubiquitin chains from such host substrates, in that way exerting a function in innate immune evasion. The presence of viral deubiquitinating enzymes has been firmly established for numerous animal-infecting viruses, such as some well-researched and clinically important nidoviruses, and their presence has now been confirmed in several plant viruses as well. Viral proteases in general have long been highlighted as promising drug targets, with a current focus on small molecule inhibitors. In this review, we will discuss the range of viral deubiquitinating proteases known to date, summarise the various avenues explored to inhibit such proteases and discuss novel strategies and models intended to inhibit and study these specific viral enzymes.


Subject(s)
Deubiquitinating Enzymes , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/genetics , Humans , Viral Proteases/metabolism , Protein Processing, Post-Translational , Ubiquitination , Animals , Virus Replication , Antiviral Agents/pharmacology , Protease Inhibitors/pharmacology , Viruses/drug effects , Viruses/enzymology , Viral Proteins/metabolism , Viral Proteins/genetics , Ubiquitin/metabolism , Immunity, Innate
6.
Comput Biol Chem ; 110: 108061, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574417

ABSTRACT

Being widely accepted tools in computational drug search, the (Q)SAR methods have limitations related to data incompleteness. The proteochemometrics (PCM) approach expands the applicability area by using description for both protein and ligand structures. The PCM algorithms are urgently required for the development of new antiviral agents. We suggest the PCM method using the TLMNA descriptors, combining the MNA descriptors of ligands and protein sequence N-grams. Our method was validated on the viral chymotrypsin-like proteases and their ligands. We have developed an original protocol allowing us to collect a comprehensive set of 15 protein sequences and more than 9000 ligands from the ChEMBL database. The N-grams were derived from the 3D-based alignment, accurately superposing ligand-binding regions. In testing the ligand set in SAR mode with MNA descriptors, an accuracy above 0.95 was determined that shows the perspective of the antiviral drug search in virtual chemical libraries. The effective PCM models were built with the TLMNA descriptor. The strong validation procedure with pair exclusion simulated the prediction of interactions between the new ligands and new targets, resulting in accuracy estimation up to 0.89. The PCM approach shows slightly lower accuracy caused by more uncertainty compared with SAR, but it overcomes the problem of data incompleteness.


Subject(s)
Antiviral Agents , Protease Inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Ligands , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Algorithms , Viral Proteases/chemistry , Viral Proteases/metabolism
7.
J Basic Microbiol ; 64(6): e2300279, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38616711

ABSTRACT

Hepatitis C virus (HCV) is the most common infection worldwide. The correlation between HCV and renal cell carcinoma (RCC) is still mysterious. Therefore, the relationship between HCV and RCC was investigated. The study included 100 patients with RCC; 32 with HCV infection, and 68 without HCV infection. Expressions of viral proteins (NS3 and NS5A) were tested using an immune electron-microscope (IEM) and immunohistochemistry (IHC). IHC and quantitative real time-PCR investigated the presentation of human proteins TP53 and p21 genes. Transmission electron (TEM) detected viral-like particles in infected RCC tissues. The gene and protein expression of P53 was higher in HCV positive versus HCV negative patients and p21 was lower in HCV positive versus HCV negative in both tumor and normal tissue samples. Viral like particles were observed by TEM in the infected tumor and normal portion of the RCC tissues and the plasma samples. The IEM showed the depositions of NS3 and NS5A in infected renal tissues, while in noninfected samples, were not observed. The study hypothesizes that a correlation between HCV and RCC could exist through successfully detecting HCV-like particles, HCV proteins, and (p53 and p21) in RCC-infected patients.


Subject(s)
Carcinoma, Renal Cell , Genotype , Hepacivirus , Kidney Neoplasms , Tumor Suppressor Protein p53 , Viral Nonstructural Proteins , Humans , Carcinoma, Renal Cell/virology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Hepacivirus/genetics , Viral Nonstructural Proteins/genetics , Kidney Neoplasms/virology , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Male , Tumor Suppressor Protein p53/genetics , Female , Middle Aged , Hepatitis C/virology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Aged , Adult , Immunohistochemistry , Viral Proteases , RNA-Dependent RNA Polymerase , DEAD-box RNA Helicases , Nucleoside-Triphosphatase , Serine Endopeptidases
8.
Viruses ; 16(3)2024 02 22.
Article in English | MEDLINE | ID: mdl-38543704

ABSTRACT

The continuous emergence of SARS-CoV-2 variants caused the persistence of the COVID-19 epidemic and challenged the effectiveness of the existing vaccines. The viral proteases are the most attractive targets for developing antiviral drugs. In this scenario, our study explores the use of HIV-1 protease inhibitors against SARS-CoV-2. An in silico screening of a library of HIV-1 proteases identified four anti-HIV compounds able to interact with the 3CLpro of SARS-CoV-2. Thus, in vitro studies were designed to evaluate their potential antiviral effectiveness against SARS-CoV-2. We employed pseudovirus technology to simulate, in a highly safe manner, the adsorption of the alpha (α-SARS-CoV-2) and omicron (ο-SARS-CoV-2) variants of SARS-CoV-2 and study the inhibitory mechanism of the selected compounds for cell-virus interaction. The results reported a mild activity against the viral proteases 3CLpro and PLpro, but efficient inhibitory effects on the internalization of both variants mediated by cathepsin B/L. Our findings provide insights into the feasibility of using drugs exhibiting antiviral effects for other viruses against the viral and host SARS-CoV-2 proteases required for entry.


Subject(s)
COVID-19 , Cysteine Proteases , Humans , SARS-CoV-2/genetics , Protease Inhibitors/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cysteine Endopeptidases/genetics , Viral Proteases , Molecular Docking Simulation
9.
Viruses ; 16(3)2024 02 27.
Article in English | MEDLINE | ID: mdl-38543732

ABSTRACT

Viral proteases are an important target for drug development, since they can modulate vital pathways in viral replication, maturation, assembly and cell entry. With the (re)appearance of several new viruses responsible for causing diseases in humans, like the West Nile virus (WNV) and the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), understanding the mechanisms behind blocking viral protease's function is pivotal for the development of new antiviral drugs and therapeutical strategies. Apart from directly inhibiting the target protease, usually by targeting its active site, several new pathways have been explored to impair its activity, such as inducing protein aggregation, targeting allosteric sites or by inducing protein degradation by cellular proteasomes, which can be extremely valuable when considering the emerging drug-resistant strains. In this review, we aim to discuss the recent advances on a broad range of viral proteases inhibitors, therapies and molecular approaches for protein inactivation or degradation, giving an insight on different possible strategies against this important class of antiviral target.


Subject(s)
Antiviral Agents , Peptide Hydrolases , Humans , Peptide Hydrolases/metabolism , Antiviral Agents/therapeutic use , Endopeptidases , SARS-CoV-2/metabolism , Viral Proteases
10.
Angew Chem Int Ed Engl ; 63(16): e202400599, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38407550

ABSTRACT

Proteins capable of switching between distinct active states in response to biochemical cues are ideal for sensing and controlling biological processes. Activatable CRISPR-Cas systems are significant in precise genetic manipulation and sensitive molecular diagnostics, yet directly controlling Cas protein function remains challenging. Herein, we explore anti-CRISPR (Acr) proteins as modules to create synthetic Cas protein switches (CasPSs) based on computational chemistry-directed rational protein interface engineering. Guided by molecular fingerprint analysis, electrostatic potential mapping, and binding free energy calculations, we rationally engineer the molecular interaction interface between Cas12a and its cognate Acr proteins (AcrVA4 and AcrVA5) to generate a series of orthogonal protease-responsive CasPSs. These CasPSs enable the conversion of specific proteolytic events into activation of Cas12a function with high switching ratios (up to 34.3-fold). These advancements enable specific proteolysis-inducible genome editing in mammalian cells and sensitive detection of viral protease activities during virus infection. This work provides a promising strategy for developing CRISPR-Cas tools for controllable gene manipulation and regulation and clinical diagnostics.


Subject(s)
CRISPR-Associated Proteins , Gene Editing , Animals , CRISPR-Cas Systems/genetics , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Endopeptidases/metabolism , Viral Proteases/genetics , Viral Proteases/metabolism , Mammals/metabolism
11.
J Virol ; 97(12): e0092823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38047713

ABSTRACT

IMPORTANCE: Most protease-targeted antiviral development evaluates the ability of small molecules to inhibit the cleavage of artificial substrates. However, before they can cleave any other substrates, viral proteases need to cleave themselves out of the viral polyprotein in which they have been translated. This can occur either intra- or inter-molecularly. Whether this process occurs intra- or inter-molecularly has implications for the potential for precursors to accumulate and for the effectiveness of antiviral drugs. We argue that evaluating candidate antivirals for their ability to block these cleavages is vital to drug development because the buildup of uncleaved precursors can be inhibitory to the virus and potentially suppress the selection of drug-resistant variants.


Subject(s)
Antiviral Agents , Enterovirus , Viral Protease Inhibitors , Viral Proteases , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Proteolysis , Viral Proteases/metabolism , Viral Protease Inhibitors/pharmacology , Enterovirus/drug effects , Enterovirus/physiology , Polyproteins/metabolism
12.
Antiviral Res ; 220: 105758, 2023 12.
Article in English | MEDLINE | ID: mdl-38008194

ABSTRACT

Coronavirus (CoV) replication requires efficient cleavage of viral polyproteins into an array of non-structural proteins involved in viral replication, organelle formation, viral RNA synthesis, and host shutoff. Human CoVs (HCoVs) encode two viral cysteine proteases, main protease (Mpro) and papain-like protease (PLpro), that mediate polyprotein cleavage. Using a structure-guided approach, a phenothiazine urea derivative that inhibits both SARS-CoV-2 Mpro and PLpro protease activity was identified. In silico docking studies also predicted the binding of the phenothiazine urea to the active sites of structurally similar Mpro and PLpro proteases from distantly related alphacoronavirus, HCoV-229 E (229 E), and the betacoronavirus, HCoV-OC43 (OC43). The lead phenothiazine urea derivative displayed broad antiviral activity against all three HCoVs tested in cellulo. It was further demonstrated that the compound inhibited 229 E and OC43 at an early stage of viral replication, with diminished formation of viral replication organelles, and the RNAs that are made within them, as expected following viral protease inhibition. These observations suggest that the phenothiazine urea derivative readily inhibits viral replication and may broadly inhibit proteases of diverse coronaviruses.


Subject(s)
Peptide Hydrolases , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , Papain/chemistry , Viral Proteases , Phenothiazines/pharmacology , Protease Inhibitors/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
13.
J Biol Chem ; 299(12): 105388, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37890782

ABSTRACT

The main protease of severe acute respiratory syndrome coronavirus 2, Mpro, is a key viral protein essential for viral infection and replication. Mpro has been the target of many pharmacological efforts; however, the host-specific regulation of Mpro protein remains unclear. Here, we report the ubiquitin-proteasome-dependent degradation of Mpro protein in human cells, facilitated by the human E3 ubiquitin ligase ZBTB25. We demonstrate that Mpro has a short half-life that is prolonged via proteasomal inhibition, with its Lys-100 residue serving as a potential ubiquitin acceptor. Using in vitro binding assays, we observed ZBTB25 and Mpro bind to each other in vitro, and using progressive deletional mapping, we further uncovered the required domains for this interaction. Finally, we used an orthologous beta-coronavirus infection model and observed that genetic ablation of ZBTB25 resulted in a more highly infective virus, an effect lost upon reconstitution of ZBTB25 to deleted cells. In conclusion, these data suggest a new mechanism of Mpro protein regulation as well as identify ZBTB25 as an anticoronaviral E3 ubiquitin ligase.


Subject(s)
Coronavirus 3C Proteases , DNA-Binding Proteins , SARS-CoV-2 , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Viral Proteases/genetics , Viral Proteases/metabolism , Viral Proteins/metabolism , SARS-CoV-2/physiology , Coronavirus 3C Proteases/metabolism , COVID-19/virology
14.
J Virol ; 97(8): e0059723, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37578235

ABSTRACT

Multiple coronaviruses (CoVs) can cause respiratory diseases in humans. While prophylactic vaccines designed to prevent infection are available for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), incomplete vaccine efficacy, vaccine hesitancy, and the threat of other pathogenic CoVs for which vaccines do not exist have highlighted the need for effective antiviral therapies. While antiviral compounds targeting the viral polymerase and protease are already in clinical use, their sensitivity to potential resistance mutations as well as their breadth against the full range of human and preemergent CoVs remain incompletely defined. To begin to fill that gap in knowledge, we report here the development of an improved, noninfectious, cell-based fluorescent assay with high sensitivity and low background that reports on the activity of viral proteases, which are key drug targets. We demonstrate that the assay is compatible with not only the SARS-CoV-2 Mpro protein but also orthologues from a range of human and nonhuman CoVs as well as clinically reported SARS-CoV-2 drug-resistant Mpro variants. We then use this assay to define the breadth of activity of two clinically used protease inhibitors, nirmatrelvir and ensitrelvir. Continued use of this assay will help define the strengths and limitations of current therapies and may also facilitate the development of next-generation protease inhibitors that are broadly active against both currently circulating and preemergent CoVs. IMPORTANCE Coronaviruses (CoVs) are important human pathogens with the ability to cause global pandemics. Working in concert with vaccines, antivirals specifically limit viral disease in people who are actively infected. Antiviral compounds that target CoV proteases are already in clinical use; their efficacy against variant proteases and preemergent zoonotic CoVs, however, remains incompletely defined. Here, we report an improved, noninfectious, and highly sensitive fluorescent method of defining the sensitivity of CoV proteases to small molecule inhibitors. We use this approach to assay the activity of current antiviral therapies against clinically reported SARS-CoV-2 protease mutants and a panel of highly diverse CoV proteases. Additionally, we show this system is adaptable to other structurally nonrelated viral proteases. In the future, this assay can be used to not only better define the strengths and limitations of current therapies but also help develop new, broadly acting inhibitors that more broadly target viral families.


Subject(s)
Antiviral Agents , Protease Inhibitors , Viral Proteases , Humans , Antiviral Agents/pharmacology , COVID-19 , Protease Inhibitors/pharmacology , SARS-CoV-2
15.
Anal Chem ; 95(28): 10728-10735, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37410966

ABSTRACT

Viral proteases play a crucial role in viral infection and are regarded as promising targets for antiviral drug development. Consequently, biosensing methods that target viral proteases have contributed to the study of virus-related diseases. This work presents a ratiometric electrochemical sensor that enables highly sensitive detection of viral proteases through the integration of target proteolysis-activated in vitro transcription and the DNA-functionalized electrochemical interface. In particular, each viral protease-mediated proteolysis triggers the transcription of multiple RNA outputs, leading to amplified ratiometric signals on the electrochemical interface. Using the NS3/4A protease of the hepatitis C virus as a model, this method achieves robust and specific NS3/4A protease sensing with sub-femtomolar sensitivity. The feasibility of this sensor was demonstrated by monitoring NS3/4A protease activities in virus-infected cell samples with varying viral loads and post-infection times. This study provides a new approach to analyzing viral proteases and holds the potential for developing direct-acting antivirals and novel therapies for viral infections.


Subject(s)
Electrochemical Techniques , Proteolysis , Viral Proteases/metabolism , Hepatitis C/enzymology , Electrochemical Techniques/methods , Humans , Cell Line
16.
Curr Opin Immunol ; 83: 102354, 2023 08.
Article in English | MEDLINE | ID: mdl-37311351

ABSTRACT

Host innate immune sensors are vital for the initial detection of pathogen infection. Such sensors thus need to constantly adapt in escalating evolutionary arms races with pathogens. Recently, two inflammasome-forming proteins, CARD8 and NLRP1, have emerged as innate immune sensors for the enzymatic activity of virus-encoded proteases. When cleaved within a rapidly evolving 'tripwire' region, CARD8 and NLRP1 assemble into inflammasomes that initiate pyroptotic cell death and pro-inflammatory cytokine release as a form of effector-triggered immunity. Short motifs in the CARD8 and NLRP1 tripwires mimic the protease-specific cleavage sites of picornaviruses, coronaviruses, and HIV-1, providing virus-specific sensing that can rapidly change between closely related hosts and within the human population. Recent work highlights the evolutionary arms races between viral proteases and NLRP1 and CARD8, including insights into the mechanisms of inflammasome activation, host diversity of viral sensing, and means that viruses have evolved to avoid tripping the wire.


Subject(s)
Inflammasomes , Peptide Hydrolases , Humans , Inflammasomes/metabolism , Peptide Hydrolases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , NLR Proteins/metabolism , Apoptosis Regulatory Proteins , Viral Proteases/metabolism , CARD Signaling Adaptor Proteins , Neoplasm Proteins/metabolism
17.
J Biol Chem ; 299(6): 104831, 2023 06.
Article in English | MEDLINE | ID: mdl-37201587

ABSTRACT

Viral proteases play key roles in viral replication, and they also facilitate immune escape by proteolyzing diverse target proteins. Deep profiling of viral protease substrates in host cells is beneficial for understanding viral pathogenesis and for antiviral drug discovery. Here, we utilized substrate phage display coupled with protein network analysis to identify human proteome substrates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteases, including papain-like protease (PLpro) and 3C-like protease (3CLpro). We first performed peptide substrates selection of PLpro and 3CLpro, and we then used the top 24 preferred substrate sequences to identify a total of 290 putative protein substrates. Protein network analysis revealed that the top clusters of PLpro and 3CLpro substrate proteins contain ubiquitin-related proteins and cadherin-related proteins, respectively. We verified that cadherin-6 and cadherin-12 are novel substrates of 3CLpro, and CD177 is a novel substrate of PLpro using in vitro cleavage assays. We thus demonstrated that substrate phage display coupled with protein network analysis is a simple and high throughput method to identify human proteome substrates of SARS-CoV-2 viral proteases for further understanding of virus-host interactions.


Subject(s)
COVID-19 , SARS-CoV-2 , Viral Proteases , Humans , Peptide Hydrolases/metabolism , Proteome , SARS-CoV-2/enzymology , SARS-CoV-2/metabolism
18.
ACS Chem Biol ; 18(5): 1228-1236, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37140437

ABSTRACT

Chemical control of protein activity is a powerful tool for scientific study, synthetic biology, and cell therapy; however, for broad use, effective chemical inducer systems must minimally crosstalk with endogenous processes and exhibit desirable drug delivery properties. Accordingly, the drug-controllable proteolytic activity of hepatitis C cis-protease NS3 and its associated antiviral drugs have been used to regulate protein activity and gene modulation. These tools advantageously exploit non-eukaryotic and non-prokaryotic proteins and clinically approved inhibitors. Here, we expand the toolkit by utilizing catalytically inactive NS3 protease as a high affinity binder to genetically encoded, antiviral peptides. Through our approach, we create NS3-peptide complexes that can be displaced by FDA-approved drugs to modulate transcription, cell signaling, and split-protein complementation. With our developed system, we invented a new mechanism to allosterically regulate Cre recombinase. Allosteric Cre regulation with NS3 ligands enables orthogonal recombination tools in eukaryotic cells and functions in divergent organisms to control prokaryotic recombinase activity.


Subject(s)
Antiviral Agents , Viral Proteases , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Hepacivirus , Peptide Hydrolases , Peptides/pharmacology , Peptides/chemistry , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/metabolism
19.
PLoS Comput Biol ; 19(4): e1011054, 2023 04.
Article in English | MEDLINE | ID: mdl-37098073

ABSTRACT

Biochemical processes in cells, including enzyme-catalyzed reactions, occur in crowded conditions with various background macromolecules occupying up to 40% of cytoplasm's volume. Viral enzymes in the host cell also encounter such crowded conditions as they often function at the endoplasmic reticulum membranes. We focus on an enzyme encoded by the hepatitis C virus, the NS3/4A protease, which is crucial for viral replication. We have previously found experimentally that synthetic crowders, polyethylene glycol (PEG) and branched polysucrose (Ficoll), differently affect the kinetic parameters of peptide hydrolysis catalyzed by NS3/4A. To gain understanding of the reasons for such behavior, we perform atomistic molecular dynamics simulations of NS3/4A in the presence of either PEG or Ficoll crowders and with and without the peptide substrates. We find that both crowder types make nanosecond long contacts with the protease and slow down its diffusion. However, they also affect the enzyme structural dynamics; crowders induce functionally relevant helical structures in the disordered parts of the protease cofactor, NS4A, with the PEG effect being more pronounced. Overall, PEG interactions with NS3/4A are slightly stronger but Ficoll forms more hydrogen bonds with NS3. The crowders also interact with substrates; we find that the substrate diffusion is reduced much more in the presence of PEG than Ficoll. However, contrary to NS3, the substrate interacts more strongly with Ficoll than with PEG crowders, with the substrate diffusion being similar to crowder diffusion. Importantly, crowders also affect the substrate-enzyme interactions. We observe that both PEG and Ficoll enhance the presence of substrates near the active site, especially near catalytic H57 but Ficoll crowders increase substrate binding more than PEG molecules.


Subject(s)
Peptide Hydrolases , Viral Nonstructural Proteins , Ficoll , Viral Nonstructural Proteins/chemistry , Peptides , Hepacivirus/chemistry , Viral Proteases
20.
Signal Transduct Target Ther ; 8(1): 79, 2023 02 24.
Article in English | MEDLINE | ID: mdl-36823147

ABSTRACT

Innate immunity represents one of the main host responses to viral infection.1-3 STING (Stimulator of interferon genes), a crucial immune adapter functioning in host cells, mediates cGAS (Cyclic GMP-AMP Synthase) sensing of exogenous and endogenous DNA fragments and generates innate immune responses.4 Whether STING activation was involved in infection and replication of enterovirus remains largely unknown. In the present study, we discovered that human enterovirus A71 (EV-A71) infection triggered STING activation in a cGAS dependent manner. EV-A71 infection caused mitochondrial damage and the discharge of mitochondrial DNA into the cytosol of infected cells. However, during EV-A71 infection, cGAS-STING activation was attenuated. EV-A71 proteins were screened and the viral protease 2Apro had the greatest capacity to inhibit cGAS-STING activation. We identified TRAF3 as an important factor during STING activation and as a target of 2Apro. Supplement of TRAF3 rescued cGAS-STING activation suppression by 2Apro. TRAF3 supported STING activation mediated TBK1 phosphorylation. Moreover, we found that 2Apro protease activity was essential for inhibiting STING activation. Furthermore, EV-D68 and CV-A16 infection also triggered STING activation. The viral protease 2Apro from EV-D68 and CV-A16 also had the ability to inhibit STING activation. As STING activation prior to EV-A71 infection generated cellular resistance to EV-A71 replication, blocking EV-A71-mediated STING suppression represents a new anti-viral target.


Subject(s)
Enterovirus A, Human , Membrane Proteins , TNF Receptor-Associated Factor 3 , Humans , Antigens, Viral , Enterovirus A, Human/physiology , Nucleotidyltransferases/genetics , TNF Receptor-Associated Factor 3/genetics , Viral Proteases , Immunity, Innate
SELECTION OF CITATIONS
SEARCH DETAIL
...