Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Sci Adv ; 10(35): eado4288, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213347

ABSTRACT

Vaccines and first-generation antiviral therapeutics have provided important protection against COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, there remains a need for additional therapeutic options that provide enhanced efficacy and protection against potential viral resistance. The SARS-CoV-2 papain-like protease (PLpro) is one of the two essential cysteine proteases involved in viral replication. While inhibitors of the SARS-CoV-2 main protease have demonstrated clinical efficacy, known PLpro inhibitors have, to date, lacked the inhibitory potency and requisite pharmacokinetics to demonstrate that targeting PLpro translates to in vivo efficacy in a preclinical setting. Here, we report the machine learning-driven discovery of potent, selective, and orally available SARS-CoV-2 PLpro inhibitors, with lead compound PF-07957472 (4) providing robust efficacy in a mouse-adapted model of COVID-19 infection.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Coronavirus Papain-Like Proteases , Disease Models, Animal , SARS-CoV-2 , Animals , Mice , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/metabolism , Humans , COVID-19/virology , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/therapeutic use , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Machine Learning , Female , Virus Replication/drug effects
2.
Sci Adv ; 10(30): eadl4013, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39047088

ABSTRACT

To facilitate the detection and management of potential clinical antiviral resistance, in vitro selection of drug-resistant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) against the virus Mpro inhibitor nirmatrelvir (Paxlovid active component) was conducted. Six Mpro mutation patterns containing T304I alone or in combination with T21I, L50F, T135I, S144A, or A173V emerged, with A173V+T304I and T21I+S144A+T304I mutations showing >20-fold resistance each. Biochemical analyses indicated inhibition constant shifts aligned to antiviral results, with S144A and A173V each markedly reducing nirmatrelvir inhibition and Mpro activity. SARS-CoV-2 surveillance revealed that in vitro resistance-associated mutations from our studies and those reported in the literature were rarely detected in the Global Initiative on Sharing All Influenza Data database. In the Paxlovid Evaluation of Protease Inhibition for COVID-19 in High-Risk Patients trial, E166V was the only emergent resistance mutation, observed in three Paxlovid-treated patients, none of whom experienced COVID-19-related hospitalization or death.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Drug Resistance, Viral , Mutation , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/drug effects , Drug Resistance, Viral/genetics , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/virology , COVID-19/genetics , COVID-19/epidemiology , Coronavirus 3C Proteases/genetics , Coronavirus 3C Proteases/antagonists & inhibitors , Lactams , Leucine , Nitriles , Proline
3.
Antiviral Res ; 230: 105970, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39067667

ABSTRACT

Variants of SARS-CoV-2 pose significant challenges in public health due to their increased transmissibility and ability to evade natural immunity, vaccine protection, and monoclonal antibody therapeutics. The emergence of the highly transmissible Omicron variant and subsequent subvariants, characterized by an extensive array of over 32 mutations within the spike protein, intensifies concerns regarding vaccine evasion. In response, multiple antiviral therapeutics have received FDA emergency use approval, targeting the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and main protease (Mpro) regions, known to have relatively fewer mutations across novel variants. In this study, we evaluated the efficacy of nirmatrelvir (PF-07321332) and other clinically significant SARS-CoV-2 antivirals against a diverse panel of SARS-CoV-2 variants, encompassing the newly identified Omicron subvariants XBB1.5 and JN.1, using live-virus antiviral assays. Our findings demonstrate that while the last Omicron subvariants exhibited heightened pathogenicity in our animal model, nirmatrelvir and other clinically relevant antivirals consistently maintained their efficacy against all tested variants, including the XBB1.5 subvariant.

4.
J Med Chem ; 67(16): 13550-13571, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-38687966

ABSTRACT

Despite the record-breaking discovery, development and approval of vaccines and antiviral therapeutics such as Paxlovid, coronavirus disease 2019 (COVID-19) remained the fourth leading cause of death in the world and third highest in the United States in 2022. Here, we report the discovery and characterization of PF-07817883, a second-generation, orally bioavailable, SARS-CoV-2 main protease inhibitor with improved metabolic stability versus nirmatrelvir, the antiviral component of the ritonavir-boosted therapy Paxlovid. We demonstrate the in vitro pan-human coronavirus antiviral activity and off-target selectivity profile of PF-07817883. PF-07817883 also demonstrated oral efficacy in a mouse-adapted SARS-CoV-2 model at plasma concentrations equivalent to nirmatrelvir. The preclinical in vivo pharmacokinetics and metabolism studies in human matrices are suggestive of improved oral pharmacokinetics for PF-07817883 in humans, relative to nirmatrelvir. In vitro inhibition/induction studies against major human drug metabolizing enzymes/transporters suggest a low potential for perpetrator drug-drug interactions upon single-agent use of PF-07817883.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Protease Inhibitors , SARS-CoV-2 , Humans , Animals , Mice , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Antiviral Agents/chemistry , Administration, Oral , Protease Inhibitors/pharmacology , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/therapeutic use , Protease Inhibitors/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Rats , COVID-19/virology
6.
Chemosphere ; 352: 141307, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38307338

ABSTRACT

The present study compares the effect of using different solvents on the electrochemical properties of the reduced TiO2 nanotubes (TiO2-rNTs) layered Ti/TiO2-rNTs/SnO2-Sb/PbO2 anodes. The electrodes are prepared using three different solvent-based precursors: (i) isopropanol, (ii) ethylene glycol and citric acid (Pechini method), and (iii) 2-hydroxyethylammonium acetate (2HEAA) ionic liquid (IL) via the thermal decomposition route. The decomposition mechanism of precursor solutions was explored using the thermogravimetric (TGA) analysis. Further, the physicochemical properties of the electrodes are examined using Field emission Scanning Electron microscopy (FE-SEM), X-ray diffraction spectroscopy (XRD), and X-ray photoelectron emission spectroscopy (XPS). The results revealed that solvents with higher viscosity and slower decomposition rates support better film uniformity and higher stability of the electrode. The TiO2 -rNTs bottom layer and PbO2 top layer helped obtain higher film stability, increased working potential window (2.2 V vs. SHE) of the electrode, and the repeatability of the results. The performance of different electrodes based on the precursor solution is found as IL â‰« Pechini > Isopropanol. 4-chlorophenol (4-CP) is used as a model pollutant to test the performance of IL-Ti/TiO2-rNTs/SnO2-Sb/PbO2 anode in an anodic oxidation (AO) coupled electro-Fenton (EF) treatment. Further, the reliability of the electrode is evaluated by mineralizing other persistent organic pollutants (POPs) like tetracyclin, phenol, 2-chlorophenol (2-CP), and 2,4-dichlorophenol (2,4-DCP). Under the optimized conditions, the proposed system was able to mineralize the tetracyclin, phenol, 2-CP, 2,4-DCP, and 4-CP up to 78.91, 82.07, 74.96, 78.78, and 69.3 %, respectively. Moreover, the degradation mechanism of chlorophenols is proposed.


Subject(s)
Oxides , Water Pollutants, Chemical , Oxides/chemistry , Persistent Organic Pollutants , 2-Propanol , Reproducibility of Results , Titanium/chemistry , Oxidation-Reduction , Phenols , Phenol/chemistry , Electrodes , Solvents , Water Pollutants, Chemical/chemistry
7.
Chemosphere ; 336: 139225, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37356583

ABSTRACT

The influence of anode materials on the electrochemical treatment of tannery wastewater (TWW) was evaluated using Pt, Ti/RuO2-IrO2 (DSA), Ti/SnO2-Sb, Ti/PbO2, and Ti/SnO2-Sb/PbO2 electrodes. The comparison of the degradation mechanism of these electrodes in the electro-Fenton (EF) treatment was evaluated. The Ti/SnO2-Sb/PbO2 anode was efficient, with high electrocatalytic activity, stability, and reproducibility of the degradation results. Further, the study was extended to define the ability of sequential EF and electrocoagulation (EC) processes to clean TWW. The EC treatment was conducted using Al electrodes, and the performance of the combined treatment was evaluated by the removal of chemical oxygen demand (COD), turbidity, total suspended solids (TSS), sulfide, and Cr removal. The role of chlorides and sulfate salts during both treatments was evaluated by monitoring the concentration changes of these anions during the whole treatment using ion chromatography (IC). A sequential 1.5 h EF and 1 h EC treatment were applied to achieve a satisfactory degradation of (81.2 ± 3.9)% COD, >98% Cr, >99% turbidity, TSS, and sulfide removal. Additionally, the combined treatment was found to be more efficient towards the COD removal, achieving about 22.5% higher COD removal consuming almost the same amount of electrical energy.


Subject(s)
Water Pollutants, Chemical , Water Purification , Oxidation-Reduction , Reproducibility of Results , Electrocoagulation , Wastewater , Electrodes , Water Purification/methods , Water Pollutants, Chemical/chemistry , Titanium/chemistry
8.
Proc Natl Acad Sci U S A ; 120(16): e2214997120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37043537

ABSTRACT

While somatic variants of TRAF7 (Tumor necrosis factor receptor-associated factor 7) underlie anterior skull-base meningiomas, here we report the inherited mutations of TRAF7 that cause congenital heart defects. We show that TRAF7 mutants operate in a dominant manner, inhibiting protein function via heterodimerization with wild-type protein. Further, the shared genetics of the two disparate pathologies can be traced to the common origin of forebrain meninges and cardiac outflow tract from the TRAF7-expressing neural crest. Somatic and inherited mutations disrupt TRAF7-IFT57 interactions leading to cilia degradation. TRAF7-mutant meningioma primary cultures lack cilia, and TRAF7 knockdown causes cardiac, craniofacial, and ciliary defects in Xenopus and zebrafish, suggesting a mechanistic convergence for TRAF7-driven meningiomas and developmental heart defects.


Subject(s)
Heart Defects, Congenital , Meningeal Neoplasms , Meningioma , Animals , Adaptor Proteins, Signal Transducing/metabolism , Heart Defects, Congenital/genetics , Meningeal Neoplasms/genetics , Meningioma/genetics , Meningioma/pathology , Mutation , Skull/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Humans , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins
9.
Front Vet Sci ; 9: 1028077, 2022.
Article in English | MEDLINE | ID: mdl-36387381

ABSTRACT

The foot-and-mouth disease virus (FMDV) leader proteinase (Lpro) is a papain like protease that cleaves the viral polyprotein and several host factors affecting host cell translation and induction of innate immunity. Introduction of Lpro mutations ablating catalytic activity is not tolerated by the virus, however, complete coding sequence deletion or introduction of targeted amino acid substitutions can render viable progeny. In proof-of-concept studies, we have previously identified and characterized FMDV Lpro mutants that are attenuated in cell culture and in animals, while retaining their capacity for inducing a strong adaptive immunity. By using molecular modeling, we have now identified a His residue (H138), that resides outside the substrate binding and catalytic domain, and is highly conserved across serotypes. Mutation of H138 renders possible FMDV variants of reduced virulence in vitro and in vivo. Kinetics studies showed that FMDV A12-LH138L mutant replicates similarly to FMDV A12-wild type (WT) virus in cells that do not offer immune selective pressure, but attenuation is observed upon infection of primary or low passage porcine epithelial cells. Western blot analysis on protein extracts from these cells, revealed that while processing of translation initiation factor eIF-4G was slightly delayed, no degradation of innate sensors or effector molecules such as NF-κB or G3BP2 was observed, and higher levels of interferon (IFN) and IFN-stimulated genes (ISGs) were induced after infection with A12-LH138L as compared to WT FMDV. Consistent with the results in porcine cells, inoculation of swine with this mutant resulted in a mild, or in some cases, no clinical disease but induction of a strong serological adaptive immune response. These results further support previous evidence that Lpro is a reliable target to derive numerous viable FMDV strains that alone or in combination could be exploited for the development of novel FMD vaccine platforms.

10.
Antiviral Res ; 208: 105429, 2022 12.
Article in English | MEDLINE | ID: mdl-36208677

ABSTRACT

Vero cells are widely used for antiviral tests and virology research for SARS-CoV-2 as well as viruses from various other families. However, Vero cells generally express high levels of multi-drug resistance 1 (MDR1) or Pgp protein, the efflux transporter of foreign substances including many antiviral compounds, affecting the antiviral activity as well as interpretation of data. To address this, a Pgp gene knockout VeroE6 cell line (VeroE6-Pgp-KO) was generated using CRISPR-CAS9 technology. These cells no longer expressed the Pgp protein as indicated by flow cytometry analysis following staining with a Pgp-specific monoclonal antibody. They also showed significantly reduced efflux transporter activity in the calcein acetoxymethyl ester (calcein AM) assay. The VeroE6-Pgp-KO cells and the parental VeroE6 cells were each infected with SARS-CoV-2 to test antiviral activities of remdesivir and nirmatrelvir, two known Pgp substrates, in the presence or absence of a Pgp inhibitor. The compounds showed antiviral activities in VeroE6-Pgp-KO cells similar to that observed in the presence of the Pgp inhibitor. Thus, the newly established VeroE6-Pgp-KO cell line adds a new in vitro virus infection system for SARS-CoV-2 and possibly other viruses to test antiviral therapies without a need to control the Pgp activity. Removal of the Pgp inhibitor for antiviral assays will lead to less data variation and prevent failed assays.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Chlorocebus aethiops , Animals , SARS-CoV-2/genetics , Antiviral Agents/pharmacology , Gene Knockout Techniques , Vero Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line
11.
Nat Med ; 27(12): 2165-2175, 2021 12.
Article in English | MEDLINE | ID: mdl-34887573

ABSTRACT

Intracranial aneurysm (IA) rupture leads to subarachnoid hemorrhage, a sudden-onset disease that often causes death or severe disability. Although genome-wide association studies have identified common genetic variants that increase IA risk moderately, the contribution of variants with large effect remains poorly defined. Using whole-exome sequencing, we identified significant enrichment of rare, deleterious mutations in PPIL4, encoding peptidyl-prolyl cis-trans isomerase-like 4, in both familial and index IA cases. Ppil4 depletion in vertebrate models causes intracerebral hemorrhage, defects in cerebrovascular morphology and impaired Wnt signaling. Wild-type, but not IA-mutant, PPIL4 potentiates Wnt signaling by binding JMJD6, a known angiogenesis regulator and Wnt activator. These findings identify a novel PPIL4-dependent Wnt signaling mechanism involved in brain-specific angiogenesis and maintenance of cerebrovascular integrity and implicate PPIL4 gene mutations in the pathogenesis of IA.


Subject(s)
Brain/blood supply , Cyclophilins/genetics , Intracranial Aneurysm/genetics , Neovascularization, Pathologic/genetics , RNA-Binding Proteins/genetics , Cyclophilins/physiology , Humans , Mutation , RNA-Binding Proteins/physiology , Exome Sequencing , Wnt Signaling Pathway/physiology
12.
Science ; 374(6575): 1586-1593, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34726479

ABSTRACT

The worldwide outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. Alongside vaccines, antiviral therapeutics are an important part of the healthcare response to countering the ongoing threat presented by COVID-19. Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity and excellent off-target selectivity and in vivo safety profiles. PF-07321332 has demonstrated oral activity in a mouse-adapted SARS-CoV-2 model and has achieved oral plasma concentrations exceeding the in vitro antiviral cell potency in a phase 1 clinical trial in healthy human participants.


Subject(s)
COVID-19 Drug Treatment , Lactams/pharmacology , Lactams/therapeutic use , Leucine/pharmacology , Leucine/therapeutic use , Nitriles/pharmacology , Nitriles/therapeutic use , Proline/pharmacology , Proline/therapeutic use , SARS-CoV-2/drug effects , Viral Protease Inhibitors/pharmacology , Viral Protease Inhibitors/therapeutic use , Administration, Oral , Animals , COVID-19/virology , Clinical Trials, Phase I as Topic , Coronavirus/drug effects , Disease Models, Animal , Drug Therapy, Combination , Humans , Lactams/administration & dosage , Lactams/pharmacokinetics , Leucine/administration & dosage , Leucine/pharmacokinetics , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Nitriles/administration & dosage , Nitriles/pharmacokinetics , Proline/administration & dosage , Proline/pharmacokinetics , Randomized Controlled Trials as Topic , Ritonavir/administration & dosage , Ritonavir/therapeutic use , SARS-CoV-2/physiology , Viral Protease Inhibitors/administration & dosage , Viral Protease Inhibitors/pharmacokinetics , Virus Replication/drug effects
13.
J Virol ; 94(17)2020 08 17.
Article in English | MEDLINE | ID: mdl-32581111

ABSTRACT

Many RNA viruses encode a proof-reading deficient, low-fidelity RNA-dependent polymerase (RdRp), which generates genetically diverse populations that can adapt to changing environments and thwart antiviral therapies. 3Dpol, the RdRp of the foot-and-mouth disease virus (FMDV), is responsible for replication of viral genomes. The 3Dpol N terminus encodes a nuclear localization signal (NLS) sequence,MRKTKLAPT, important for import of the protein to host nucleus. Previous studies showed that substitutions at residues 18 and 20 of the NLS are defective in proper incorporation of nucleotides and RNA binding. Here, we use a systematic alanine scanning mutagenesis approach to understand the role of individual residues of the NLS in nuclear localization and nucleotide incorporation activities of 3Dpol We identify two residues of 3Dpol NLS, T19 and L21, that are important for the maintenance of enzyme fidelity. The 3Dpol NLS alanine substitutions of T19 and L21 results in aberrant incorporation of nucleoside analogs, conferring a low fidelity phenotype of the enzyme. A molecular dynamics simulation of RNA- and mutagen (RTP)-bound 3Dpol revealed that the T19 residue participates in a hydrogen bond network, including D165 in motif F and R416 at the C terminus of the FMDV 3Dpol and RNA template-primer. Based on these findings and previous studies, we conclude that at least the first six residues of theMRKTKLAPT sequence motif play a vital role in the maintenance of faithful RNA synthesis activity (fidelity) of FMDV 3Dpol, suggesting that the role of the NLS motif in similar viral polymerases needs to be revisited.IMPORTANCE In this study, we employed genetic and molecular dynamics approaches to analyze the role of individual amino acids of the FMDV 3Dpol nuclear localization signal (NLS). The NLS residues were mutated to alanine using a type A full-genome cDNA clone, and the virus progeny was analyzed for defects in growth and in competition with the parental virus. We identified two mutants in 3Dpol, T19A and L21A, that exhibited high rate of mutation, were sensitive to nucleotide analogs, and displayed reduced replicative fitness compared to the parental virus. Using molecular dynamics simulation, we demonstrated that residues T19 and L21 played a role in the structural configuration of the interaction network at the 3Dpol palm subdomain. Cumulatively, our data suggest that the T19 and L21 3Dpol amino acids are important for maintaining the fidelity of the FMDV polymerase and ensuring faithful replication of the FMDV genome.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/physiology , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Amino Acid Substitution , Animals , Cell Line , Genome, Viral , Molecular Dynamics Simulation , Mutagenesis , Mutation , Nuclear Localization Signals/chemistry , Nucleotides , Protein Conformation , RNA, Viral , Virus Replication
14.
PLoS One ; 14(4): e0210847, 2019.
Article in English | MEDLINE | ID: mdl-31022193

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious viral disease that severely impacts global food security and is one of the greatest constraints on international trade of animal products. Extensive viral population diversity and rapid, continuous mutation of circulating FMD viruses (FMDVs) pose significant obstacles to the control and ultimate eradication of this important transboundary pathogen. The current study investigated mechanisms contributing to within-host evolution of FMDV in a natural host species (cattle). Specifically, vaccinated and non-vaccinated cattle were infected with FMDV under controlled, experimental conditions and subsequently sampled for up to 35 days to monitor viral genomic changes as related to phases of disease and experimental cohorts. Consensus-level genomic changes across the entire FMDV coding region were characterized through three previously defined stages of infection: early, transitional, and persistent. The overall conclusion was that viral evolution occurred via a combination of two mechanisms: emergence of full-genomic minority haplotypes from within the inoculum super-swarm, and concurrent continuous point mutations. Phylogenetic analysis indicated that individuals were infected with multiple distinct haplogroups that were pre-existent within the ancestral inoculum used to infect all animals. Multiple shifts of dominant viral haplotype took place during the early and transitional phases of infection, whereas few shifts occurred during persistent infection. Overall, this work suggests that the establishment of the carrier state is not associated with specific viral genomic characteristics. These insights into FMDV population dynamics have important implications for virus sampling methodology and molecular epidemiology.


Subject(s)
Carrier State/veterinary , Evolution, Molecular , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease/virology , Genome, Viral/genetics , Animals , Capsid Proteins/genetics , Carrier State/immunology , Carrier State/virology , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/isolation & purification , Haplotypes , Longitudinal Studies , Mutation , Phylogeny , RNA, Viral/genetics , Viral Vaccines/administration & dosage
15.
J Vet Diagn Invest ; 30(5): 699-707, 2018 09.
Article in English | MEDLINE | ID: mdl-29916768

ABSTRACT

The highly contagious foot-and-mouth disease virus (FMDV) afflicts cloven-hoofed animals, resulting in significant costs because of loss of trade and recovery from disease. We developed a sensitive, specific, and rapid competitive ELISA (cELISA) to detect serum antibodies to FMDV. The cELISA utilized a monoclonal blocking antibody specific for a highly conserved FMDV nonstructural 3B epitope, a recombinant mutant FMDV 3ABC coating protein, and optimized format variables including serum incubation for 90 min at 20-25°C. Samples from 16 animals experimentally infected with one FMDV serotype (A, O, Asia, or SAT-1) demonstrated early detection capacity beginning 7 d post-inoculation. All samples from 55 vesicular stomatitis virus antibody-positive cattle and 44 samples from cloven-hoofed animals affected by non-FMD vesicular diseases were negative in the cELISA, demonstrating 100% analytical specificity. The diagnostic sensitivity was 100% against sera from 128 cattle infected with isolates of all FMDV serotypes, emphasizing serotype-agnostic results. Diagnostic specificities of U.S. cattle ( n = 1135) and swine ( n = 207) sera were 99.4% and 100%, respectively. High repeatability and reproducibility were demonstrated with 3.1% coefficient of variation in percent inhibition data and 100% agreement using 2 kit lots and 400 negative control serum samples, with no difference between bench and biosafety cabinet operation. Negative results from vaccinated, uninfected cattle, pig, and sheep sera confirmed the DIVA (differentiate infected from vaccinated animals) capability. This rapid (<3 h), select agent-free assay with high sensitivity and specificity, DIVA capability, and room temperature processing capability will serve as a useful tool in FMDV surveillance, emergency preparedness, response, and outbreak recovery programs.


Subject(s)
Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Foot-and-Mouth Disease Virus/isolation & purification , Foot-and-Mouth Disease/diagnosis , Viral Nonstructural Proteins/isolation & purification , Animals , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/virology , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/immunology , Foot-and-Mouth Disease/virology , Sheep , Sheep Diseases/diagnosis , Sheep Diseases/virology , Swine , Swine Diseases/diagnosis , Swine Diseases/virology
16.
Front Microbiol ; 9: 485, 2018.
Article in English | MEDLINE | ID: mdl-29616004

ABSTRACT

Viral genomes have evolved to maximize their potential of overcoming host defense mechanisms and to induce a variety of disease syndromes. Structurally, a genome of a virus consists of coding and noncoding regions, and both have been shown to contribute to initiation and progression of disease. Accumulated work in picornaviruses has stressed out the importance of the noncoding RNAs, or untranslated 5'- and 3'-regions (UTRs), in both replication and translation of viral genomes. Unsurprisingly, defects in these processes have been reported to cause viral attenuation and affect viral pathogenicity. However, substantial evidence suggests that these untranslated RNAs may influence the outcome of the host innate immune response. This review discusses the involvement of 5'- and 3'-terminus UTRs in induction and regulation of host immunity and its consequences for viral life cycle and virulence.

17.
Virology ; 512: 132-143, 2017 12.
Article in English | MEDLINE | ID: mdl-28961454

ABSTRACT

The S fragment of the FMDV 5' UTR is predicted to fold into a long stem-loop structure and it has been implicated in virus-host protein interactions. In this study, we report the minimal S fragment sequence required for virus viability and show a direct correlation between the extent of the S fragment deletion mutations and attenuated phenotypes. Furthermore, we provide novel insight into the role of the S fragment in modulating the host innate immune response. Importantly, in an FMDV mouse model system, all animals survive the inoculation with the live A24 FMDV-S4 mutant, containing a 164 nucleotide deletion in the upper S fragment loop, at a dose 1000 higher than the one causing lethality by parental A24 FMDV, indicating that the A24 FMDV-S4 virus is highly attenuated in vivo. Additionally, mice exposed to high doses of live A24 FMDV-S4 virus are fully protected when challenged with parental A24 FMDV virus.


Subject(s)
5' Untranslated Regions/genetics , Foot-and-Mouth Disease Virus/physiology , Immunity, Innate/physiology , Virus Replication/physiology , Animals , Cattle , Cell Line , Cricetinae , Foot-and-Mouth Disease Virus/genetics , RNA, Viral/genetics , Sequence Deletion , Virus Replication/genetics
18.
J Virol ; 91(15)2017 08 01.
Article in English | MEDLINE | ID: mdl-28515297

ABSTRACT

Foot-and-mouth disease virus (FMDV) RNA-dependent RNA polymerase (RdRp) (3Dpol) catalyzes viral RNA synthesis. Its characteristic low fidelity and absence of proofreading activity allow FMDV to rapidly mutate and adapt to dynamic environments. In this study, we used the structure of FMDV 3Dpol in combination with previously reported results from similar picornaviral polymerases to design point mutations that would alter replication fidelity. In particular, we targeted Trp237 within conserved polymerase motif A because of the low reversion potential inherent in the single UGG codon. Using biochemical and genetic tools, we show that the replacement of tryptophan 237 with phenylalanine imparts higher fidelity, but replacements with isoleucine and leucine resulted in lower-fidelity phenotypes. Viruses containing these W237 substitutions show in vitro growth kinetics and plaque morphologies similar to those of the wild-type (WT) A24 Cruzeiro strain in BHK cells, and both high- and low-fidelity variants retained fitness during coinfection with the wild-type virus. The higher-fidelity W237F (W237FHF) mutant virus was more resistant to the mutagenic nucleoside analogs ribavirin and 5-fluorouracil than the WT virus, whereas the lower-fidelity W237I (W237ILF) and W237LLF mutant viruses exhibited lower ribavirin resistance. Interestingly, the variant viruses showed heterogeneous and slightly delayed growth kinetics in primary porcine kidney cells, and they were significantly attenuated in mouse infection experiments. These data demonstrate, for a single virus, that either increased or decreased RdRp fidelity attenuates virus growth in animals, which is a desirable feature for the development of safer and genetically more stable vaccine candidates.IMPORTANCE Foot-and-mouth disease (FMD) is the most devastating disease affecting livestock worldwide. Here, using structural and biochemical analyses, we have identified FMDV 3Dpol mutations that affect polymerase fidelity. Recombinant FMDVs containing substitutions at 3Dpol tryptophan residue 237 were genetically stable and displayed plaque phenotypes and growth kinetics similar to those of the wild-type virus in cell culture. We further demonstrate that viruses harboring either a W237FHF substitution or W237ILF and W237LLF mutations were highly attenuated in animals. Our study shows that obtaining 3Dpol fidelity variants by protein engineering based on polymerase structure and function could be exploited for the development of attenuated FMDV vaccine candidates that are safer and more stable than strains obtained by selective pressure via mutagenic nucleotides or adaptation approaches.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/metabolism , Foot-and-Mouth Disease Virus/enzymology , Foot-and-Mouth Disease Virus/pathogenicity , Protein Engineering , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Substitution , Animals , Antiviral Agents , Cells, Cultured , Cricetinae , DNA Mutational Analysis , Disease Models, Animal , Drug Resistance, Viral , Fluorouracil/pharmacology , Foot-and-Mouth Disease/pathology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/growth & development , Mice , Mutagenesis, Site-Directed , Point Mutation , Ribavirin/pharmacology , Swine , Tryptophan/genetics , Tryptophan/metabolism , Viral Plaque Assay
19.
Virus Res ; 223: 181-9, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27497620

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal disease of domestic pigs that has significant economic consequences for the swine industry. The viral genome encodes for more than 150 genes, and only a select few of these genes have been studied in some detail. Here we report the characterization of open reading frame Ep152R that has a predicted complement control module/SCR domain. This domain is found in Vaccinia virus proteins that are involved in blocking the immune response during viral infection. A recombinant ASFV harboring a HA tagged version of the Ep152R protein was developed (ASFV-G-Ep152R-HA) and used to demonstrate that Ep152R is an early virus protein. Attempts to construct recombinant viruses having a deleted Ep152R gene were consistently unsuccessful indicating that Ep152R is an essential gene. Interestingly, analysis of host-protein interactions for Ep152R using a yeast two-hybrid screen, identified BAG6, a protein previously identified as being required for ASFV replication. Furthermore, fluorescent microscopy analysis confirms that Ep152R-BAG6 interaction actually occurs in cells infected with ASFV.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/metabolism , African Swine Fever/virology , Genes, Essential , Molecular Chaperones/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Conserved Sequence , Host-Pathogen Interactions , Macrophages/metabolism , Macrophages/virology , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Open Reading Frames , Protein Binding , Protein Conformation , Protein Interaction Mapping/methods , Protein Transport , Sequence Deletion , Swine , Two-Hybrid System Techniques , Viral Proteins/chemistry , Virus Replication
20.
Virology ; 495: 136-47, 2016 08.
Article in English | MEDLINE | ID: mdl-27209448

ABSTRACT

Here, we engineered two FMD viruses with histidine residues inserted into or fused to the FMDV capsid. Both 6xHis viruses exhibited growth kinetics, plaque morphologies and antigenic characteristics similar to wild-type virus. The 6xHis tag allowed one-step purification of the mutant virions by Co(2+) affinity columns. Electron microscopy and biochemical assays showed that the 6xHis FMDVs readily assembled into antigen: adjuvant complexes in solution, by conjugating with Ni(2+)-chelated nanolipoprotein and monophosphoryl lipid A adjuvant (MPLA:NiNLP). Animals Immunized with the inactivated 6xHis-FMDV:MPLA:NiNLP vaccine acquired enhanced protective immunity against FMDV challenge compared to virions alone. Induction of anti-6xHis and anti-FMDV neutralizing antibodies in the immunized animals could be exploited in the differentiation of vaccinated from infected animals needed for the improvement of FMD control measures. The novel marker vaccine/nanolipid technology described here has broad applications for the development of distinctive and effective immune responses to other pathogens of importance.


Subject(s)
Adjuvants, Immunologic , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Ions , Metals , Nanoparticles , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Foot-and-Mouth Disease Virus/genetics , Gene Order , Genome, Viral , Lipoproteins/immunology , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL