Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
J Virol ; 96(19): e0133222, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36106874

ABSTRACT

Mammalian TRIM7 is an antiviral protein that inhibits multiple human enteroviruses by degrading the viral 2BC protein. Whether TRIM7 is reciprocally targeted by enteroviruses is not known. Here, we report that the 3C protease (3Cpro) from two enteroviruses, coxsackievirus B3 (CVB3) and poliovirus, targets TRIM7 for cleavage. CVB3 3Cpro cleaves TRIM7 at glutamine 24 (Q24), resulting in a truncated TRIM7 that fails to inhibit CVB3 due to dampened E3 ubiquitin ligase activity. TRIM7 Q24 is highly conserved across mammals, except in marsupials, which instead have a naturally occurring histidine (H24) that is not subject to 3Cpro cleavage. Marsupials also express two isoforms of TRIM7, and the two proteins from koalas have distinct antiviral activities. The longer isoform contains an additional exon due to alternate splice site usage. This additional exon contains a unique 3Cpro cleavage site, suggesting that certain enteroviruses may have evolved to target marsupial TRIM7 even if the canonical Q24 is missing. Combined with computational analyses indicating that TRIM7 is rapidly evolving, our data raise the possibility that TRIM7 may be targeted by enterovirus evasion strategies and that evolution of TRIM7 across mammals may have conferred unique antiviral properties. IMPORTANCE Enteroviruses are significant human pathogens that cause viral myocarditis, pancreatitis, and meningitis. Knowing how the host controls these viruses and how the viruses may evade host restriction is important for understanding fundamental concepts in antiviral immunity and for informing potential therapeutic interventions. In this study, we demonstrate that coxsackievirus B3 uses its virally encoded protease to target the host antiviral protein TRIM7 for cleavage, suggesting a potential mechanism of viral immune evasion. We additionally show that TRIM7 has evolved in certain mammalian lineages to express protein variants with distinct antiviral activities and susceptibilities to viral protease-mediated cleavage.


Subject(s)
3C Viral Proteases , Enterovirus Infections , Enterovirus , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , 3C Viral Proteases/metabolism , Animals , Enterovirus/enzymology , Glutamine , Histidine , Host-Pathogen Interactions , Phascolarctidae/virology , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Expert Opin Ther Targets ; 25(6): 479-489, 2021 06.
Article in English | MEDLINE | ID: mdl-34253126

ABSTRACT

Introduction: Enteroviruses are common viruses causing a huge number of acute and chronic infections and producing towering economic costs. Similarly, coronaviruses cause seasonal mild infections, epidemics, and even pandemics and can lead to severe respiratory symptoms. It is important to develop broadly acting antiviral molecules to efficiently tackle the infections caused by thes.Areas covered: This review illuminates the differences and similarities between enteroviruses and coronaviruses and examines the most appealing therapeutic targets to combat both virus groups. Publications of both virus groups and deposited structures discovered through PubMed to March 2021 for viral proteases have been evaluated.Expert opinion: The main protease of coronaviruses and enteroviruses share similarities in their structure and function. These proteases process their viral polyproteins and thus drugs that bind to the active site have potential to target both virus groups. It is important to develop drugs that target more evolutionarily conserved processes and proteins. Moreover, it is a wise strategy to concentrate on processes that are similar between several virus families.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus/physiology , Enterovirus/physiology , Animals , Coronavirus/drug effects , Coronavirus/enzymology , Cysteine Endopeptidases/metabolism , Enterovirus/drug effects , Enterovirus/enzymology , Humans , Substrate Specificity
3.
Commun Biol ; 4(1): 841, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230602

ABSTRACT

Characterizing protein-protein interactions (PPIs) is an effective method to help explore protein function. Here, through integrating a newly identified split human Rhinovirus 3 C (HRV 3 C) protease, super-folder GFP (sfGFP), and ClpXP-SsrA protein degradation machinery, we developed a fluorescence-assisted single-cell methodology (split protease-E. coli ClpXP (SPEC)) to explore protein-protein interactions for both eukaryotic and prokaryotic species in E. coli cells. We firstly identified a highly efficient split HRV 3 C protease with high re-assembly ability and then incorporated it into the SPEC method. The SPEC method could convert the cellular protein-protein interaction to quantitative fluorescence signals through a split HRV 3 C protease-mediated proteolytic reaction with high efficiency and broad temperature adaptability. Using SPEC method, we explored the interactions among effectors of representative type I-E and I-F CRISPR/Cas complexes, which combining with subsequent studies of Cas3 mutations conferred further understanding of the functions and structures of CRISPR/Cas complexes.


Subject(s)
Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Protein Interaction Maps , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Cas Systems , Endopeptidase Clp/genetics , Enterovirus/enzymology , Enterovirus/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histidine Kinase/genetics , Histidine Kinase/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation , Proteolysis , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Virology ; 562: 128-141, 2021 10.
Article in English | MEDLINE | ID: mdl-34315103

ABSTRACT

Picornavirus family members cause disease in humans. Human rhinoviruses (RV), the main causative agents of the common cold, increase the severity of asthma and COPD; hence, effective agents against RVs are required. The 2A proteinase (2Apro), found in all enteroviruses, represents an attractive target; inactivating mutations in poliovirus 2Apro result in an extension of the VP1 protein preventing infectious virion assembly. Variations in sequence and substrate specificity on eIF4G isoforms between RV 2Apro of genetic groups A and B hinder 2Apro as drug targets. Here, we demonstrate that although RV-A2 and RV-B4 2Apro cleave the substrate GAB1 at different sites, the 2Apro from both groups cleave equally efficiently an artificial site containing P1 methionine. We determined the RV-A2 2Apro structure complexed with zVAM.fmk, containing P1 methionine. Analysis of this first 2Apro-inhibitor complex reveals a conserved hydrophobic P4 pocket among enteroviral 2Apro as a potential target for broad-spectrum anti-enteroviral inhibitors.


Subject(s)
Antiviral Agents/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Rhinovirus/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cysteine Endopeptidases/genetics , Enterovirus/chemistry , Enterovirus/enzymology , Eukaryotic Initiation Factor-4G/metabolism , Genetic Variation , HeLa Cells , Humans , Protein Conformation , Rhinovirus/chemistry , Rhinovirus/genetics , Substrate Specificity , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics
5.
mBio ; 12(2)2021 04 13.
Article in English | MEDLINE | ID: mdl-33849973

ABSTRACT

Enteroviruses (EV) deploy two proteases that mediate viral polyprotein cleavage and host cell manipulation. Here, we report that EV 2A proteases cleave all three members of the YTHDF protein family, cytosolic N6-methyladenosine (m6A) "readers" that regulate target mRNA fate. YTHDF protein cleavage occurs very early during infection, before viral translation is detected or cytopathogenic effects are observed. Preemptive YTHDF protein depletion enhanced viral translation and replication but only in cells with restrained viral translation, signs of inefficient 2A protease activity, and protective innate host immune responses. This effect corresponded with repression of interferon (IFN)-stimulated gene (ISG) induction, while type I/III IFN production was not significantly altered. Moreover, YTHDF3 depletion impaired JAK/STAT signaling in cells treated with type I, but not type II, IFN. YTHDF3 depletion's stimulatory effect on viral dynamics was dampened by JAK/STAT blockade and enhanced by type I IFN pretreatment of cells. We propose that EV 2A proteases cleave YTHDF proteins to antagonize ISG induction in infected cells.IMPORTANCE It is believed that ∼7,000 messenger RNAs (mRNAs) are subject to N6-methyladenosine modification. The biological significance of this remains mysterious. The YTHDF m6A readers are three related proteins with high affinity for m6A-modified mRNA, yet their biological functions remain obscure. We discovered that polio/enteroviruses elicit very early proteolysis of YTHDF1 to 3 in infected cells. Our research demonstrates that YTHDF3 acts as a positive regulator of antiviral JAK/STAT signaling in response to positive single-strand RNA virus infection, enabling type I interferon (IFN)-mediated gene regulatory programs to unfurl in infected cells. Our observation of viral degradation of the YTHDF proteins demonstrates that they are key response modifiers in the innate antiviral immune response.


Subject(s)
Enterovirus/genetics , Interferon Type I/metabolism , Janus Kinases/metabolism , RNA-Binding Proteins/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Viral Proteins/metabolism , Cell Line , Enterovirus/enzymology , HeLa Cells , Humans , Immunity, Innate , Interferon Type I/immunology , Janus Kinases/genetics , Proteolysis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , STAT Transcription Factors/genetics , Viral Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 117(24): 13499-13508, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32467165

ABSTRACT

The existence of multiple serotypes renders vaccine development challenging for most viruses in the Enterovirus genus. An alternative and potentially more viable strategy for control of these viruses is to develop broad-spectrum antivirals by targeting highly conserved proteins that are indispensable for the virus life cycle, such as the 3C protease. Previously, two single-chain antibody fragments, YDF and GGVV, were reported to effectively inhibit human rhinovirus 14 proliferation. Here, we found that both single-chain antibody fragments target sites on the 3C protease that are distinct from its known drug site (peptidase active site) and possess different mechanisms of inhibition. YDF does not block the active site but instead noncompetitively inhibits 3C peptidase activity through an allosteric effect that is rarely seen for antibody protease inhibitors. Meanwhile, GGVV antagonizes the less-explored regulatory function of 3C in genome replication. The interaction between 3C and the viral genome 5' noncoding region has been reported to be important for enterovirus genome replication. Here, the interface between human rhinovirus 14 3C and its 5' noncoding region was probed by hydrogen-deuterium exchange coupled mass spectrometry and found to partially overlap with the interface between GGVV and 3C. Consistently, prebinding of GGVV completely abolishes interaction between human rhinovirus 14 3C and its 5' noncoding region. The epitopes of YDF and GGVV, therefore, represent two additional sites of therapeutic vulnerability in rhinovirus. Importantly, the GGVV epitope appears to be conserved across many enteroviruses, suggesting that it is a promising target for pan-enterovirus inhibitor screening and design.


Subject(s)
Antiviral Agents/pharmacology , Cysteine Endopeptidases/chemistry , Enterovirus/drug effects , Single-Chain Antibodies/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , 3C Viral Proteases , 5' Untranslated Regions , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Cysteine Endopeptidases/metabolism , Enterovirus/enzymology , Epitopes , Genome, Viral , RNA, Viral/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Viral Proteins/metabolism
7.
Nucleic Acids Res ; 48(10): 5591-5602, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32286652

ABSTRACT

RNA virus survival depends on efficient viral genome replication, which is performed by the viral RNA dependent RNA polymerase (RdRp). The recent development of high throughput magnetic tweezers has enabled the simultaneous observation of dozens of viral RdRp elongation traces on kilobases long templates, and this has shown that RdRp nucleotide addition kinetics is stochastically interrupted by rare pauses of 1-1000 s duration, of which the short-lived ones (1-10 s) are the temporal signature of a low fidelity catalytic pathway. We present a simple and precise temperature controlled system for magnetic tweezers to characterize the replication kinetics temperature dependence between 25°C and 45°C of RdRps from three RNA viruses, i.e. the double-stranded RNA bacteriophage Φ6, and the positive-sense single-stranded RNA poliovirus (PV) and human rhinovirus C (HRV-C). We found that Φ6 RdRp is largely temperature insensitive, while PV and HRV-C RdRps replication kinetics are activated by temperature. Furthermore, the activation energies we measured for PV RdRp catalytic state corroborate previous estimations from ensemble pre-steady state kinetic studies, further confirming the catalytic origin of the short pauses and their link to temperature independent RdRp fidelity. This work will enable future temperature controlled study of biomolecular complex at the single molecule level.


Subject(s)
RNA Viruses/enzymology , RNA-Dependent RNA Polymerase/metabolism , Temperature , Virus Replication , Bacteriophage phi 6/enzymology , Enterovirus/enzymology , Enzyme Activation , Kinetics , Microscopy , Poliovirus/enzymology
8.
J Med Chem ; 63(9): 4562-4578, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32045235

ABSTRACT

The main protease of coronaviruses and the 3C protease of enteroviruses share a similar active-site architecture and a unique requirement for glutamine in the P1 position of the substrate. Because of their unique specificity and essential role in viral polyprotein processing, these proteases are suitable targets for the development of antiviral drugs. In order to obtain near-equipotent, broad-spectrum antivirals against alphacoronaviruses, betacoronaviruses, and enteroviruses, we pursued a structure-based design of peptidomimetic α-ketoamides as inhibitors of main and 3C proteases. Six crystal structures of protease-inhibitor complexes were determined as part of this study. Compounds synthesized were tested against the recombinant proteases as well as in viral replicons and virus-infected cell cultures; most of them were not cell-toxic. Optimization of the P2 substituent of the α-ketoamides proved crucial for achieving near-equipotency against the three virus genera. The best near-equipotent inhibitors, 11u (P2 = cyclopentylmethyl) and 11r (P2 = cyclohexylmethyl), display low-micromolar EC50 values against enteroviruses, alphacoronaviruses, and betacoronaviruses in cell cultures. In Huh7 cells, 11r exhibits three-digit picomolar activity against the Middle East Respiratory Syndrome coronavirus.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus/drug effects , Enterovirus/drug effects , Lactams/pharmacology , Peptidomimetics/pharmacology , Virus Replication/drug effects , 3C Viral Proteases , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Binding Sites , Cell Line, Tumor , Chlorocebus aethiops , Coronavirus/enzymology , Coronavirus 3C Proteases , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Drug Design , Enterovirus/enzymology , Humans , Lactams/chemical synthesis , Lactams/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/metabolism , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Protein Binding , Vero Cells , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/metabolism
9.
Viruses ; 12(1)2020 01 09.
Article in English | MEDLINE | ID: mdl-31936473

ABSTRACT

BACKGROUND: Enteroviruses are a group of common non-enveloped RNA viruses that cause symptoms ranging from mild respiratory infections to paralysis. Due to the abundance of enterovirus infections it is hard to distinguish between on-going and previous infections using immunological assays unless the IgM fraction is studied. METHODS: In this study we show using Indirect ELISA and capture IgM ELISA that an IgG antibody response against the nonstructural enteroviral proteins 2A and 3C can be used to distinguish between IgM positive (n = 22) and IgM negative (n = 20) human patients with 83% accuracy and a diagnostic odds ratio of 30. Using a mouse model, we establish that the antibody response to the proteases is short-lived compared to the antibody response to the structural proteins in. As such, the protease antibody response serves as a potential marker for an acute infection. CONCLUSIONS: Antibody responses against enterovirus proteases are shorter-lived than against structural proteins and can differentiate between IgM positive and negative patients, and therefore they are a potential marker for acute infections.


Subject(s)
Antibodies, Viral/blood , Enterovirus/enzymology , Enterovirus/immunology , Immunoglobulin G/blood , Immunoglobulin M/blood , Peptide Hydrolases/immunology , 3C Viral Proteases , Acute Disease , Adult , Animals , Antibodies, Viral/immunology , Antibody Formation , Biomarkers/blood , Cysteine Endopeptidases/immunology , Enterovirus Infections/diagnosis , Enterovirus Infections/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Infant , Mice , Mice, Inbred C57BL , Peptide Hydrolases/classification , Viral Proteins/immunology
10.
Biol Chem ; 400(3): 405-415, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30521472

ABSTRACT

Proteases are crucial for regulating biological processes in organisms through hydrolysis of peptide bonds. Recombinant proteases have moreover become important tools in biotechnological, and biomedical research and as therapeutics. We have developed a label-free high-throughput method for quantitative assessment of proteolytic activity in Escherichia coli. The screening method is based on co-expression of a protease of interest and a reporter complex. This reporter consists of an aggregation-prone peptide fused to a fluorescent protein via a linker that contains the corresponding substrate sequence. Cleavage of the substrate rescues the fluorescent protein from aggregation, resulting in increased fluorescence that correlates to proteolytic activity, which can be monitored using flow cytometry. In one round of flow-cytometric cell sorting, we isolated an efficiently cleaved tobacco etch virus (TEV) substrate from a 1:100 000 background of non-cleavable sequences, with around 6000-fold enrichment. We then engineered the 3C protease from coxsackievirus B3 (CVB3 3Cpro) towards improved proteolytic activity on the substrate LEVLFQ↓GP. We isolated highly proteolytic active variants from a randomly mutated CVB3 3Cpro library with up to 4-fold increase in activity. The method enables simultaneous measurement of proteolytic activity and protease expression levels and can therefore be applied for protease substrate profiling, as well as directed evolution of proteases.


Subject(s)
Cysteine Endopeptidases/metabolism , Enterovirus/enzymology , Flow Cytometry , Fluorescence , Viral Proteins/metabolism , 3C Viral Proteases , Escherichia coli/cytology , Escherichia coli/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Protein Engineering
11.
Angew Chem Int Ed Engl ; 57(39): 12702-12706, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30118570

ABSTRACT

Analogous to reversible post-translational protein modifications, the ability to attach and subsequently remove modifications on proteins would be valuable for protein and biological research. Although bioorthogonal functionalities have been developed to conjugate or cleave protein modifications, they are introduced into proteins on separate residues and often with bulky side chains, limiting their use to one type of control and primarily protein surface. Here we achieved dual control on one residue by genetically encoding S-propargyl-cysteine (SprC), which has bioorthogonal alkyne and propargyl groups in a compact structure, permitting usage in protein interior in addition to surface. We demonstrated its incorporation at the dimer interface of glutathione transferase for in vivo crosslinking via thiol-yne click chemistry, and at the active site of human rhinovirus 3C protease for masking and then turning on enzyme activity via Pd-cleavage of SprC into Cys. In addition, we installed biotin onto EGFP via Sonogashira coupling of SprC and then tracelessly removed it via Pd cleavage. SprC is small in size, commercially available, nontoxic, and allows for bond building and breaking on a single residue. Genetically encoded SprC will be valuable for chemically controlling proteins with an essential Cys and for reversible protein modifications.


Subject(s)
Cysteine Endopeptidases/metabolism , Cysteine/chemistry , Green Fluorescent Proteins/chemistry , Viral Proteins/metabolism , 3C Viral Proteases , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Biotin/chemistry , Catalysis , Catalytic Domain , Click Chemistry , Cysteine/metabolism , Cysteine Endopeptidases/chemistry , Enterovirus/enzymology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Methanosarcina/metabolism , Mutagenesis, Site-Directed , Palladium/chemistry , Pargyline/chemistry , Thioredoxins/chemistry , Thioredoxins/genetics , Thioredoxins/metabolism , Viral Proteins/chemistry
12.
J Med Chem ; 61(3): 1218-1230, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29328649

ABSTRACT

Lead structure discovery mainly focuses on the identification of noncovalently binding ligands. Covalent linkage, however, is an essential binding mechanism for a multitude of successfully marketed drugs, although discovered by serendipity in most cases. We present a concept for the design of fragments covalently binding to proteases. Covalent linkage enables fragment binding unrelated to affinity to shallow protein binding sites and at the same time allows differentiated targeted hit verification and binding location verification through mass spectrometry. We describe a systematic and rational computational approach for the identification of covalently binding fragments from compound collections inhibiting enteroviral 3C protease, a target with high therapeutic potential. By implementing reactive groups potentially forming covalent bonds as a chemical feature in our 3D pharmacophore methodology, covalent binders were discovered by high-throughput virtual screening. We present careful experimental validation of the virtual hits using enzymatic assays and mass spectrometry unraveling a novel, previously unknown irreversible inhibition of the 3C protease by phenylthiomethyl ketone-based fragments. Subsequent synthetic optimization through fragment growing and reactivity analysis against catalytic and noncatalytic cysteines revealed specific irreversible 3C protease inhibition.


Subject(s)
Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Enterovirus/enzymology , Ketones/chemistry , Ketones/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , 3C Viral Proteases , Catalytic Domain , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/metabolism , Drug Design , High-Throughput Screening Assays , Ketones/metabolism , Models, Molecular , Structure-Activity Relationship , Substrate Specificity , Viral Proteins/chemistry
13.
Virol Sin ; 32(6): 548-552, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29164396

ABSTRACT

The RNA-dependent RNA polymerases (RdRPs) encoded by RNA viruses represent a unique class of nucleic acid polymerases. Unlike other classes of single-subunit polymerases, viral RdRPs have evolved a unique conformational change in their palm domain to close the active site during catalysis. The hallmark of this conformational change is the backbone shift of the polymerase motif A from an "open" state to a "closed" state, allowing two universally conserved aspartic acid residues to orient toward each other for divalent metal binding and catalysis. The "closed" motif A conformation was only observed upon the binding of correct NTP in RdRP catalytic complexes or under rare conditions such as induced by a bound lutetium ion or a bound glutamate molecule. By solving the crystal structure of the catalytic elongation complex of the coxsackievirus RdRP, we in this work observed for the first time the "closed" motif A conformation in the absence of an NTP substrate or other conformational-change-inducing factors. This observation emphasizes the intrinsic dynamic features of viral RdRP motif A, and solidifies the structural basis for how this important structural element participates in catalytic events of the RdRPs.


Subject(s)
Enterovirus/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Crystallography, X-Ray , Models, Molecular
14.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28490584

ABSTRACT

Enteroviruses (EVs) are implicated in a wide range of diseases in humans and animals. In this study, a novel enterovirus (enterovirus species G [EVG]) (EVG 08/NC_USA/2015) was isolated from a diagnostic sample from a neonatal pig diarrhea case and identified by using metagenomics and complete genome sequencing. The viral genome shares 75.4% nucleotide identity with a prototypic EVG strain (PEV9 UKG/410/73). Remarkably, a 582-nucleotide insertion, flanked by 3Cpro cleavage sites at the 5' and 3' ends, was found in the 2C/3A junction region of the viral genome. This insertion encodes a predicted protease with 54 to 68% amino acid identity to torovirus (ToV) papain-like protease (PLP) (ToV-PLP). Structural homology modeling predicts that this protease adopts a fold and a catalytic site characteristic of minimal PLP catalytic domains. This structure is similar to those of core catalytic domains of the foot-and-mouth disease virus leader protease and coronavirus PLPs, which act as deubiquitinating and deISGylating (interferon [IFN]-stimulated gene 15 [ISG15]-removing) enzymes on host cell substrates. Importantly, the recombinant ToV-PLP protein derived from this novel enterovirus also showed strong deubiquitination and deISGylation activities and demonstrated the ability to suppress IFN-ß expression. Using reverse genetics, we generated a ToV-PLP knockout recombinant virus. Compared to the wild-type virus, the ToV-PLP knockout mutant virus showed impaired growth and induced higher expression levels of innate immune genes in infected cells. These results suggest that ToV-PLP functions as an innate immune antagonist; enterovirus G may therefore gain fitness through the acquisition of ToV-PLP from a recombination event.IMPORTANCE Enteroviruses comprise a highly diversified group of viruses. Genetic recombination has been considered a driving force for viral evolution; however, recombination between viruses from two different orders is a rare event. In this study, we identified a special case of cross-order recombination between enterovirus G (order Picornavirales) and torovirus (order Nidovirales). This naturally occurring recombination event may have broad implications for other picornaviral and/or nidoviral species. Importantly, we demonstrated that the exogenous ToV-PLP gene that was inserted into the EVG genome encodes a deubiquitinase/deISGylase and potentially suppresses host cellular innate immune responses. Our results provide insights into how a gain of function through genetic recombination, in particular cross-order recombination, may improve the ability of a virus to evade host immunity.


Subject(s)
Deubiquitinating Enzymes/genetics , Enterovirus/enzymology , Enterovirus/genetics , Feces/virology , Mutagenesis, Insertional , Torovirus/enzymology , Torovirus/genetics , Animals , Animals, Newborn , Diarrhea/veterinary , Enterovirus/isolation & purification , Metagenomics , RNA, Viral/genetics , Recombination, Genetic , Sequence Analysis, DNA , Swine , Swine Diseases/virology , United States
15.
Article in English | MEDLINE | ID: mdl-28461310

ABSTRACT

Hand-foot-and-mouth disease (HFMD), caused by enterovirus, is a threat to public health worldwide. To date, enterovirus 71 (EV71) has been one of the major causative agents of HFMD in the Pacific-Asia region, and outbreaks with EV71 cause millions of infections. However, no drug is currently available for clinical therapeutics. In our previous works, we developed a set of protease inhibitors (PIs) targeting the EV71 3C protease (3Cpro). Among these are NK-1.8k and NK-1.9k, which have various active groups and high potencies and selectivities. In the study described here, we determined the structures of the PI NK-1.8k in complex with wild-type (WT) and drug-resistant EV71 3Cpro Comparison of these structures with the structure of unliganded EV71 3Cpro and its complex with AG7088 indicated that the mutation of N69 to a serine residue destabilized the S2 pocket. Thus, the mutation influenced the cleavage activity of EV71 3Cpro and the inhibitory activity of NK-1.8k in an in vitro protease assay and highlighted that site 69 is an additional key site for PI design. More information for the optimization of the P1' to P4 groups of PIs was also obtained from these structures. Together with the results of our previous works, these in-depth results elucidate the inhibitory mechanism of PIs and shed light to develop PIs for the clinical treatment of infections caused by EV71 and other enteroviruses.


Subject(s)
Antiviral Agents/metabolism , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Enterovirus/enzymology , Protease Inhibitors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , 3C Viral Proteases , Antiviral Agents/chemistry , Hand, Foot and Mouth Disease/enzymology , Hand, Foot and Mouth Disease/metabolism , Isoxazoles/chemistry , Isoxazoles/metabolism , Mutation , Phenylalanine/analogs & derivatives , Protease Inhibitors/chemistry , Protein Structure, Tertiary , Pyrrolidinones/chemistry , Pyrrolidinones/metabolism , Valine/analogs & derivatives
16.
Biophys Chem ; 219: 9-16, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27668727

ABSTRACT

Hand foot and mouth disease (HFMD) epidemic has occurred in many countries. Coxsackievirus A16 (CV-A16) and Enterovirus A71 (EV-A71) are the main causes of HFMD. Up to now, there are no anti-HFMD drugs available. Rupintrivir, a broad-spectrum inhibitor, is a drug candidate for HFMD treatment, while other HFMD inhibitors designed from several studies have a relatively low efficiency. Therefore, in this work we aim to study the binding mechanisms of rupintrivir and a peptidic α,ß-unsaturated ethyl ester (SG85) against both CV-A16 and EV-A71 3C proteases (3Cpro) using all-atoms molecular dynamics simulation. The obtained results indicate that SG85 shows a stronger binding affinity than rupintrivir against CV-A16. Both inhibitors exhibit a comparable affinity against EV-A71 3Cpro. The molecular information of the binding of the two inhibitors to the proteases will be elucidated. Thus, it is implied that these two compounds may be used as leads for further anti-HFMD drug design and development.


Subject(s)
Enterovirus A, Human/enzymology , Enterovirus/enzymology , Molecular Dynamics Simulation , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Cysteine Endopeptidases , Drug Design , Enzyme Inhibitors/pharmacology , Hand, Foot and Mouth Disease/virology , Humans , In Vitro Techniques , Isoxazoles/pharmacology , Oligopeptides/pharmacology , Peptides/pharmacology , Phenylalanine/analogs & derivatives , Protein Binding , Pyrrolidinones/pharmacology , Valine/analogs & derivatives
17.
Nucleic Acids Res ; 44(14): 6883-95, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27317698

ABSTRACT

Genetic recombination in single-strand, positive-sense RNA viruses is a poorly understand mechanism responsible for generating extensive genetic change and novel phenotypes. By moving a critical cis-acting replication element (CRE) from the polyprotein coding region to the 3' non-coding region we have further developed a cell-based assay (the 3'CRE-REP assay) to yield recombinants throughout the non-structural coding region of poliovirus from dually transfected cells. We have additionally developed a defined biochemical assay in which the only protein present is the poliovirus RNA dependent RNA polymerase (RdRp), which recapitulates the strand transfer events of the recombination process. We have used both assays to investigate the role of the polymerase fidelity and nucleotide turnover rates in recombination. Our results, of both poliovirus intertypic and intratypic recombination in the CRE-REP assay and using a range of polymerase variants in the biochemical assay, demonstrate that RdRp fidelity is a fundamental determinant of recombination frequency. High fidelity polymerases exhibit reduced recombination and low fidelity polymerases exhibit increased recombination in both assays. These studies provide the basis for the analysis of poliovirus recombination throughout the non-structural region of the virus genome and provide a defined biochemical assay to further dissect this important evolutionary process.


Subject(s)
Enterovirus/enzymology , Enterovirus/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombination, Genetic , Animals , Base Sequence , Biological Assay , DNA Replication , DNA, Intergenic/genetics , Genome, Viral , HeLa Cells , Humans , Mice , Mutation/genetics , Nucleotides/metabolism , Poliovirus/genetics , RNA-Dependent RNA Polymerase/genetics , Templates, Genetic , Virus Replication
18.
Rev Med Virol ; 26(4): 251-67, 2016 07.
Article in English | MEDLINE | ID: mdl-27145174

ABSTRACT

Enteroviruses are common human pathogens, and infections are particularly frequent in children. Severe infections can lead to a variety of diseases, including poliomyelitis, aseptic meningitis, myocarditis and neonatal sepsis. Enterovirus infections have also been implicated in asthmatic exacerbations and type 1 diabetes. The large disease spectrum of the closely related enteroviruses may be partially, but not fully, explained by differences in tissue tropism. The molecular mechanisms by which enteroviruses cause disease are poorly understood, but there is increasing evidence that the two enteroviral proteases, 2A(pro) and 3C(pro) , are important mediators of pathology. These proteases perform the post-translational proteolytic processing of the viral polyprotein, but they also cleave several host-cell proteins in order to promote the production of new virus particles, as well as to evade the cellular antiviral immune responses. Enterovirus-associated processing of cellular proteins may also contribute to pathology, as elegantly demonstrated by the 2A(pro) -mediated cleavage of dystrophin in cardiomyocytes contributing to Coxsackievirus-induced cardiomyopathy. It is likely that improved tools to identify targets for these proteases will reveal additional host protein substrates that can be linked to specific enterovirus-associated diseases. Here, we discuss the function of the enteroviral proteases in the virus replication cycle and review the current knowledge regarding how these proteases modulate the infected cell in order to favour virus replication, including ways to avoid detection by the immune system. We also highlight new possibilities for the identification of protease-specific cellular targets and thereby a way to discover novel mechanisms contributing to disease. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cysteine Endopeptidases/metabolism , Enterovirus/enzymology , Enterovirus/physiology , Host-Pathogen Interactions , Virus Replication , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Enterovirus/pathogenicity , Humans , Immune Evasion , Protein Conformation , Protein Processing, Post-Translational , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
19.
J Gen Virol ; 97(6): 1368-1380, 2016 06.
Article in English | MEDLINE | ID: mdl-26935471

ABSTRACT

Type I IFNs play an important role in the immune response to enterovirus infections. Their importance is underscored by observations showing that many enteroviruses including coxsackie B viruses (CVBs) have developed strategies to block type I IFN production. Recent studies have highlighted a role for the type III IFNs (also called IFNλs) in reducing permissiveness to infections with enteric viruses including coxsackievirus. However, whether or not CVBs have measures to evade the effects of type III IFNs remains unknown. By combining virus infection studies and different modes of administrating the dsRNA mimic poly I : C, we discovered that CVBs target both TLR3- and MDA5/RIG-I-mediated type III IFN expression. Consistent with this, the cellular protein expression levels of the signal transduction proteins TRIF and IPS1 were reduced and no hyperphosphorylation of IRF-3 was observed following infection with the virus. Notably, decreased expression of full-length TRIF and IPS1 and the appearance of cleavage products was observed upon both CVB3 infection and in cellular protein extracts incubated with recombinant 2Apro, indicating an important role for the viral protease in subverting the cellular immune system. Collectively, our study reveals that CVBs block the expression of type III IFNs, and that this is achieved by a similar mechanism as the virus uses to block type I IFN production. We also demonstrate that the virus blocks several intracellular viral recognition pathways of importance for both type I and III IFN production. The simultaneous targeting of numerous arms of the host immune response may be required for successful viral replication and dissemination.


Subject(s)
Cysteine Endopeptidases/metabolism , Enterovirus/immunology , Enterovirus/pathogenicity , Immune Evasion , Immunity, Innate , Interleukins/antagonists & inhibitors , Viral Proteins/metabolism , Enterovirus/enzymology , Interferon-Induced Helicase, IFIH1/metabolism , Interferons , Signal Transduction , Toll-Like Receptor 3/metabolism
20.
PLoS One ; 10(10): e0141383, 2015.
Article in English | MEDLINE | ID: mdl-26509685

ABSTRACT

Unc93b is an endoplasmic reticulum (ER)-resident transmembrane protein that serves to bind and traffic toll-like receptors (TLRs) from the ER to their appropriate subcellular locations for ligand sensing. Because of its role in TLR trafficking, Unc93b is necessary for an effective innate immune response to coxsackievirus B3 (CVB), a positive-sense single stranded RNA virus belonging to the enterovirus family. Here, we show that Unc93b is cleaved by a CVB-encoded cysteine protease (3Cpro) during viral replication. Further, we define a role for Unc93b in the induction of apoptotic cell death and show that expression of wild-type Unc93b, but not a mutant incapable of binding TLRs or exiting the ER (H412R), induces apoptosis. Furthermore, we show that cellular caspases activated during apoptosis directly cleave Unc93b. Interestingly, we show that the 3Cpro- and caspase-mediated cleavage of Unc93b both occur within ten amino acids in the distal N-terminus of Unc93b. Mechanistically, neither caspase-mediated nor 3Cpro-mediated cleavage of Unc93b altered its trafficking function, inhibited its role in facilitating TLR3 or TLR8 signaling, or altered its apoptosis-inducing effects. Taken together, our studies show that Unc93b is targeted by both viral- and host cell-specific proteases and identify a function of Unc93b in the induction of apoptotic cell death.


Subject(s)
Apoptosis/genetics , Enterovirus/enzymology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Peptide Hydrolases/metabolism , 3C Viral Proteases , Caspases/metabolism , Cell Line , Cells, Cultured , Cysteine Endopeptidases/metabolism , Enterovirus Infections/genetics , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Gene Expression , Humans , Mutation , Protein Transport , Proteolysis , Signal Transduction , Toll-Like Receptors/metabolism , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...