Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 11(11)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-38006014

ABSTRACT

TNX-1800 is a synthetically derived live recombinant chimeric horsepox virus (rcHPXV) vaccine candidate expressing Wuhan SARS-CoV-2 spike (S) protein. The primary objective of this study was to evaluate the immunogenicity and efficacy of TNX-1800 in two nonhuman primate species challenged with USA-WA1/2020 SARS-CoV-2. TNX-1800 vaccination was well tolerated with no serious adverse events or significant changes in clinical parameters. A single dose of TNX-1800 generated humoral responses in African Green Monkeys and Cynomolgus Macaques, as measured by the total binding of anti-SARS-CoV-2 S IgG and neutralizing antibody titers against the USA-WA1/2020 strain. In addition, a single dose of TNX-1800 induced an interferon-gamma (IFN-γ)-mediated T-cell response in Cynomolgus Macaques. Following challenge with SARS-CoV-2, African Green and Cynomolgus Macaques exhibited rapid clearance of virus in the upper and lower respiratory tract. Future studies will assess the efficacy of TNX-1800 against newly emerging variants and demonstrate its safety in humans.

2.
Viruses ; 15(10)2023 10 21.
Article in English | MEDLINE | ID: mdl-37896908

ABSTRACT

TNX-1800 is a preclinical stage synthetic-derived live attenuated chimeric horsepox virus vaccine engineered to express the SARS-CoV-2 spike (S) gene. The objectives of this study were to assess the safety, tolerability, and immunogenicity of TNX-1800 administration in Syrian golden hamsters and New Zealand white rabbits. Animals were vaccinated at three doses via percutaneous inoculation. The data showed that the single percutaneous administration of three TNX-1800 vaccine dose levels was well tolerated in both hamsters and rabbits. At all dose levels, rabbits were more decerning regarding vaccine site reaction than hamsters. Lastly, no TNX-1800 genomes could be detected at the site of vaccination. Post-vaccination, all animals had anti-SARS-CoV-2 spike protein IgG specific antibody responses. These data demonstrate that TNX-1800 infection was limited, asymptomatic, and cleared by the end of this study, and a single dose was able to generate immune responses.


Subject(s)
COVID-19 , Poxviridae , Cricetinae , Rabbits , Animals , Mesocricetus , SARS-CoV-2/genetics , Vaccines, Attenuated/adverse effects , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Antibodies, Viral , Immunoglobulin G , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing
3.
Antiviral Res ; 210: 105513, 2023 02.
Article in English | MEDLINE | ID: mdl-36592670

ABSTRACT

Antibody-based therapy is emerging as a critical therapeutic countermeasure to treat acute viral infections by offering rapid protection against clinical disease. The advancements in structural biology made it feasible to rationalize monoclonal antibodies (mAbs) by identifying key and, possibly, neutralizing epitopes of viral proteins for therapeutic purposes. A critical component in assessing mAbs during pandemics requires the development of rapid but detailed methods to detect and quantitate the neutralization activity. In this study, we developed and optimized two high-content image (HCI)-based assays: one to detect viral proteins by staining and the second to quantify cytopathic viral effects by a label-free phenotypic assay. These assays were employed to screen for therapeutic antibodies against the monkeypox virus (MPXV) using surrogate poxviruses such as vaccinia virus (VACV). Plaque-based neutralization results confirmed the HCI data. The phenotypic assay found pox virus-induced syncytia formation in various cells, and we were able to quantitate and use this phenotype to screen mAbs. The HCI identified several potent VACV-neutralizing antibodies that showed in vitro efficacy against both clades of MPXV. In addition, a combination study of ST-246/tecovirimat/TPOXX a single neutralizing antibody Ab-40, showed synergistic activity against VACV in an in-vitro neutralization assay. This rapid high-content method utilizing state-of-the-art technologies enabled the evaluation of hundreds of mAbs quickly to identify several potent anti-MPXV neutralizing mAbs for further development.


Subject(s)
Antibodies, Viral , Monkeypox virus , Antibodies, Neutralizing , Vaccinia virus/genetics , Viral Proteins , Antibodies, Monoclonal/pharmacology , Neutralization Tests
4.
J Virol ; 84(19): 10276-88, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660183

ABSTRACT

The coronavirus nucleocapsid (N) protein plays an essential role in virion assembly via interactions with the large, positive-strand RNA viral genome and the carboxy-terminal endodomain of the membrane protein (M). To learn about the functions of N protein domains in the coronavirus mouse hepatitis virus (MHV), we replaced the MHV N gene with its counterpart from the closely related bovine coronavirus (BCoV). The resulting viral mutant was severely defective, even though individual domains of the N protein responsible for N-RNA, N-M, or N-N interactions were completely interchangeable between BCoV and MHV. The lesion in the BCoV N substitution mutant could be compensated for by reverting mutations in the central, serine- and arginine-rich (SR) domain of the N protein. Surprisingly, a second class of reverting mutations were mapped to the amino terminus of a replicase subunit, nonstructural protein 3 (nsp3). A similarly defective MHV N mutant bearing an insertion of the SR region from the severe acute respiratory syndrome coronavirus N protein was rescued by the same two classes of reverting mutations. Our genetic results were corroborated by the demonstration that the expressed amino-terminal segment of nsp3 bound selectively to N protein from infected cells, and this interaction was RNA independent. Moreover, we found a direct correlation between the N-nsp3 interaction and the ability of N protein to stimulate the infectivity of transfected MHV genomic RNA (gRNA). Our results suggest a role for this previously unknown N-nsp3 interaction in the localization of genomic RNA to the replicase complex at an early stage of infection.


Subject(s)
Coronavirus, Bovine/physiology , DNA-Directed RNA Polymerases/physiology , Murine hepatitis virus/physiology , Nucleocapsid Proteins/physiology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/physiology , Amino Acid Sequence , Animals , Cattle , Coronavirus Nucleocapsid Proteins , Coronavirus, Bovine/genetics , Coronavirus, Bovine/pathogenicity , DNA-Directed RNA Polymerases/genetics , Genome, Viral , Humans , Mice , Molecular Sequence Data , Murine hepatitis virus/genetics , Murine hepatitis virus/pathogenicity , Mutation , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Protein Structure, Tertiary , RNA-Dependent RNA Polymerase/genetics , Recombination, Genetic , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/physiology , Sequence Homology, Amino Acid , Transfection , Virulence/genetics , Virulence/physiology
5.
J Virol ; 84(16): 8262-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20519394

ABSTRACT

The type I interferon (IFN) response plays an essential role in the control of in vivo infection by the coronavirus mouse hepatitis virus (MHV). However, in vitro, most strains of MHV are largely resistant to the action of this cytokine, suggesting that MHV encodes one or more functions that antagonize or evade the IFN system. A particular strain of MHV, MHV-S, exhibited orders-of-magnitude higher sensitivity to IFN than prototype strain MHV-A59. Through construction of interstrain chimeric recombinants, the basis for the enhanced IFN sensitivity of MHV-S was found to map entirely to the region downstream of the spike gene, at the 3' end of the genome. Sequence analysis revealed that the major difference between the two strains in this region is the absence of gene 5a from MHV-S. Creation of a gene 5a knockout mutant of MHV-A59 demonstrated that a major component of IFN resistance maps to gene 5a. Conversely, insertion of gene 5a, or its homologs from related group 2 coronaviruses, at an upstream genomic position in an MHV-A59/S chimera restored IFN resistance. This is the first demonstration of a coronavirus gene product that can protect that same virus from the antiviral state induced by IFN. Neither protein kinase R, which phosphorylates eukaryotic initiation factor 2, nor oligoadenylate synthetase, which activates RNase L, was differentially activated in IFN-treated cells infected with MHV-A59 or MHV-S. Thus, the major IFN-induced antiviral activities that are specifically inhibited by MHV, and possibly by other coronaviruses, remain to be identified.


Subject(s)
Interferons/antagonists & inhibitors , Murine hepatitis virus/immunology , Murine hepatitis virus/pathogenicity , Viral Proteins/physiology , Virulence Factors/physiology , Animals , Base Sequence , Cell Line , Chromosome Mapping , DNA Mutational Analysis , Gene Knockout Techniques , Genetic Complementation Test , Immune Evasion , Immune Tolerance , Interferons/immunology , Mice , Molecular Sequence Data , RNA, Viral/genetics , Sequence Analysis, DNA , Viral Plaque Assay , Viral Proteins/immunology , Virulence Factors/immunology
6.
J Virol ; 82(3): 1214-28, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032506

ABSTRACT

The upstream end of the 3' untranslated region (UTR) of the mouse hepatitis virus genome contains two essential and overlapping RNA secondary structures, a bulged stem-loop and a pseudoknot, which have been proposed to be elements of a molecular switch that is critical for viral RNA synthesis. It has previously been shown that a particular six-base insertion in loop 1 of the pseudoknot is extremely deleterious to the virus. We have now isolated multiple independent second-site revertants of the loop 1 insertion mutant, and we used reverse-genetics methods to confirm the identities of suppressor mutations that could compensate for the original insertion. The suppressors were localized to two separate regions of the genome. Members of one class of suppressor were mapped to the portions of gene 1 that encode nsp8 and nsp9, thereby providing the first evidence for specific interactions between coronavirus replicase gene products and a cis-acting genomic RNA element. The second class of suppressor was mapped to the extreme 3' end of the genome, a result which pointed to the existence of a direct base-pairing interaction between loop 1 of the pseudoknot and the genomic terminus. The latter finding was strongly supported by phylogenetic evidence and by the construction of a deletion mutant that reduced the 3' UTR to its minimal essential elements. Taken together, the interactions revealed by the two classes of suppressors suggest a model for the initiation of coronavirus negative-strand RNA synthesis.


Subject(s)
3' Untranslated Regions/metabolism , Murine hepatitis virus/genetics , Murine hepatitis virus/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , 3' Untranslated Regions/chemistry , 3' Untranslated Regions/genetics , Animals , Mice , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Suppression, Genetic , Transcription, Genetic , Virus Replication/physiology
7.
J Virol ; 81(22): 12709-14, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17855523

ABSTRACT

An unknown virus was isolated from a lung biopsy sample and multiple other samples from a patient who developed a lethal case of pneumonia following a peripheral blood stem cell transplant. A random PCR-based molecular screening method was used to identify the infectious agent as avian paramyxovirus 1 (APMV-1; a group encompassing Newcastle disease virus), which is a highly contagious poultry pathogen that has only rarely been found in human infections. Immunohistochemical analysis confirmed the presence of APMV-1 antigen in sloughed alveolar cells in lung tissue from autopsy. Sequence from the human isolate showed that it was most closely related to virulent pigeon strains of APMV-1. This is the most completely documented case of a systemic human infection caused by APMV-1 and is the first report of an association between this virus and a fatal disease in a human.


Subject(s)
Newcastle Disease/diagnosis , Newcastle Disease/virology , Newcastle disease virus/isolation & purification , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Adult , Animals , Antigens, Viral/analysis , Birds , Fatal Outcome , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Newcastle Disease/pathology , Newcastle disease virus/genetics , Pneumonia, Viral/pathology , Stem Cell Transplantation/adverse effects
8.
J Virol ; 81(3): 1274-87, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17093194

ABSTRACT

The 3' cis-acting element for mouse hepatitis virus (MHV) RNA synthesis resides entirely within the 301-nucleotide 3' untranslated region (3' UTR) of the viral genome and consists of three regions. Encompassing the upstream end of the 3' UTR are a bulged stem-loop and an overlapping RNA pseudoknot, both of which are essential to MHV and common to all group 2 coronaviruses. At the downstream end of the genome is the minimal signal for initiation of negative-strand RNA synthesis. Between these two ends is a hypervariable region (HVR) that is only poorly conserved between MHV and other group 2 coronaviruses. Paradoxically, buried within the HVR is an octanucleotide motif (oct), 5'-GGAAGAGC-3', which is almost universally conserved in coronaviruses and is therefore assumed to have a critical biological function. We conducted an extensive mutational analysis of the HVR. Surprisingly, this region tolerated numerous deletions, rearrangements, and point mutations. Most striking, a mutant deleted of the entire HVR was only minimally impaired in tissue culture relative to the wild type. By contrast, the HVR deletion mutant was highly attenuated in mice, causing no signs of clinical disease and minimal weight loss compared to wild-type virus. Correspondingly, replication of the HVR deletion mutant in the brains of mice was greatly reduced compared to that of the wild type. Our results show that neither the HVR nor oct is essential for the basic mechanism of MHV RNA synthesis in tissue culture. However, the HVR appears to play a significant role in viral pathogenesis.


Subject(s)
3' Untranslated Regions/physiology , Murine hepatitis virus/pathogenicity , RNA, Viral/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Mice , Murine hepatitis virus/genetics , Murine hepatitis virus/physiology , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , Virus Replication
9.
Antimicrob Agents Chemother ; 50(4): 1320-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16569847

ABSTRACT

Triaryl pyrazoline {[5-(4-chloro-phenyl)-3-thiophen-2-yl-4,5-dihydro-pyrazol-1-yl]-phenyl-methanone} inhibits flavivirus infection in cell culture. The inhibitor was identified through high-throughput screening of a compound library using a luciferase-expressing West Nile (WN) virus infection assay. The compound inhibited an epidemic strain of WN virus without detectable cytotoxicity (a 50% effective concentration of 28 microM and a compound concentration of >or=300 microM required to reduce 50% cell viability). Besides WN virus, the compound also inhibited other flaviviruses (dengue, yellow fever, and St. Louis encephalitis viruses), an alphavirus (Western equine encephalitis virus), a coronavirus (mouse hepatitis virus), and a rhabdovirus (vesicular stomatitis virus). However, the compound did not suppress an orthomyxovirus (influenza virus) or a retrovirus (human immunodeficiency virus type 1). Mode-of-action analyses in WN virus showed that the compound did not inhibit viral entry or virion assembly but specifically suppressed viral RNA synthesis. To examine the mechanism of inhibition of dengue virus, we developed two replicon systems for dengue type 1 virus: (i) a stable cell line that harbored replicons containing a luciferase reporter and a neomycin phosphotransferase selection marker and (ii) a luciferase-expressing replicon that could differentiate between viral translation and RNA replication. Analyses of the compound in the dengue type 1 virus replicon systems showed that it weakly suppressed viral translation but significantly inhibited viral RNA synthesis. Overall, the results demonstrate that triaryl pyrazoline exerts a broad spectrum of antiflavivirus activity through potent inhibition of viral RNA replication. This novel inhibitor could be developed for potential treatment of flavivirus infection.


Subject(s)
Antiviral Agents/pharmacology , Flavivirus/drug effects , RNA, Viral/biosynthesis , Animals , Chlorocebus aethiops , Dengue Virus/drug effects , Dengue Virus/genetics , Flavivirus/genetics , Pyrazoles/pharmacology , Vero Cells , Virus Replication/drug effects , West Nile virus/drug effects , West Nile virus/genetics
10.
J Virol ; 78(14): 7846-51, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15220462

ABSTRACT

The 3' untranslated region (3' UTR) of the genome of the severe acute respiratory syndrome coronavirus can functionally replace its counterpart in the prototype group 2 coronavirus mouse hepatitis virus (MHV). By contrast, the 3' UTRs of representative group 1 or group 3 coronaviruses cannot operate as substitutes for the MHV 3' UTR.


Subject(s)
3' Untranslated Regions/genetics , Enhancer Elements, Genetic , Murine hepatitis virus/genetics , Recombination, Genetic , Severe acute respiratory syndrome-related coronavirus/genetics , Virus Replication , Animals , Base Sequence , Genome, Viral , Humans , Mice , Molecular Sequence Data
11.
J Virol ; 78(2): 669-82, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14694098

ABSTRACT

RNA virus genomes contain cis-acting sequence and structural elements that participate in viral replication. We previously identified a bulged stem-loop secondary structure at the upstream end of the 3' untranslated region (3' UTR) of the genome of the coronavirus mouse hepatitis virus (MHV). This element, beginning immediately downstream of the nucleocapsid gene stop codon, was shown to be essential for virus replication. Other investigators discovered an adjacent downstream pseudoknot in the 3' UTR of the closely related bovine coronavirus (BCoV). This pseudoknot was also shown to be essential for replication, and it has a conserved counterpart in every group 1 and group 2 coronavirus. In MHV and BCoV, the bulged stem-loop and pseudoknot are, in part, mutually exclusive, because of the overlap of the last segment of the stem-loop and stem 1 of the pseudoknot. This led us to hypothesize that they form a molecular switch, possibly regulating a transition occurring during viral RNA synthesis. We have now performed an extensive genetic analysis of the two components of this proposed switch. Our results define essential and nonessential components of these structures and establish the limits to which essential parts of each element can be destabilized prior to loss of function. Most notably, we have confirmed the interrelationship of the two putative switch elements. Additionally, we have identified a pseudoknot loop insertion mutation that appears to point to a genetic interaction between the pseudoknot and a distant region of the genome.


Subject(s)
3' Untranslated Regions/genetics , Enhancer Elements, Genetic/genetics , Murine hepatitis virus/genetics , Mutation , RNA, Viral/genetics , Recombination, Genetic , 3' Untranslated Regions/chemistry , 3' Untranslated Regions/metabolism , Animals , Base Sequence , Mice , Molecular Sequence Data , Murine hepatitis virus/physiology , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...