Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Vaccine ; 26(5): 664-76, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18096276

ABSTRACT

Vaccinia virus (VACV) has been used as the vaccine to protect against smallpox, and recombinant VACVs have been used to develop vaccine candidates against numerous cancers and infectious diseases. Although relatively safe for use in humans, the strains of VACV that were used as smallpox vaccines led to several complications including, progressive infection in immune compromised individuals, eczema vaccination in individuals with a history of atopic dermatitis, and encephalitis and perimyocarditis in apparently healthy individuals. The work described in this paper focuses on attenuated strains of VACV that may have the potential for use as vaccine vectors with reduced pathogenicity. We have generated several VACV mutants in a WR background with specific mutations in the E3L gene that were at least a 1000-fold less pathogenic compared to wtVACV upon intra-nasal infection of mice. Many of these mutant viruses replicated to high titers in the nasal mucosa of mice following intra-nasal administration. Despite replication to high titers in the nose, there was little spread to other organs in infected animals. Intra-nasal vaccination with doses as low as 100-1000 pfu (plaque forming units) of these replicating VACV constructs were sufficient to protect the host from challenge with large doses of wtVACV. Similar constructs in a Copenhagen and a NYCBH background were highly attenuated, yet effective as vaccines in the mouse model. These recombinant VACV constructs may be promising vector candidates for use in vaccination strategies against smallpox and other pathogens.


Subject(s)
RNA-Binding Proteins/genetics , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/genetics , Smallpox/prevention & control , Vaccination , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics , Administration, Intranasal , Animals , Cell Line , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mutation , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccinia virus/pathogenicity , Virulence
2.
Virology ; 333(2): 263-70, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15721360

ABSTRACT

Encephalitis is a rare, but serious complication from vaccination against smallpox using replication competent strains of vaccinia virus. In this report we describe mutants of vaccinia virus, containing N-terminal deletions of the vaccinia virus interferon resistance gene, E3L, that are attenuated for neuropathogenesis in a mouse model system. These recombinant viruses replicated to high titers in the nasal mucosa after intra-nasal infection of C57BL/6 mice but failed to spread to the lungs or brain. These viruses demonstrated reduced pathogenicity after intra-cranial infection as well, indicating a decrease in neurovirulence. Intra-nasal inoculation or inoculation by scarification with a low dose of recombinant virus containing a deletion of the entire N-terminal domain of E3L protected against challenge with a high dose of wild-type vaccinia virus, suggesting that this replication competent, but attenuated strain of vaccinia virus may have promise as an improved vaccine for protecting against smallpox, and as a vector for inducing mucosal immunity to heterologous pathogenic organisms.


Subject(s)
RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Vaccinia virus/pathogenicity , Viral Proteins/chemistry , Viral Proteins/physiology , Animals , Brain/virology , Female , Genes, Viral , Immunity, Mucosal , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Sequence Deletion , Vaccinia/etiology , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/physiology , Viral Proteins/genetics , Viral Proteins/immunology , Virulence , Virus Replication
3.
Virology ; 314(1): 305-14, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14517083

ABSTRACT

Orf virus (OV), the prototypic parapoxvirus, is resistant to the effects of interferon (IFN) and this function of OV has been mapped to the OV20.0L gene. The protein product of this gene shares 31% amino acid identity to the E3L-encoded protein of vaccinia virus (VV) that is required for the broad host range and IFN-resistant phenotype of VV in cells in culture and for virulence of the virus in vivo. In this study we investigated whether the distantly related OV E3L homologue could complement the deletion of E3L in VV. The recombinant VV (VV/ORF-E3L) expressing the OV E3L homologue in place of VV E3L was indistinguishable from wt VV in its cell-culture phenotype. But VV/ORF-E3L was over a 1000-fold less pathogenic than wt VV (LD(50) > 5 x 10(6) PFU, compared to LD(50) of wtVV = 4 x 10(3) PFU) following intranasal infection of mice. While wt VV spread to the lungs and brain and replicated to high titers in the brain of infected mice, VV/ORF-E3L could not be detected in the lungs or brain following intranasal infection. VV/ORF-E3L was at least 100,000-fold less pathogenic than wt VV on intracranial injection. Domain swap experiments demonstrate that the difference in pathogenesis maps to the C-terminal domain of these proteins. This domain has been shown to be required for the dsRNA binding function of the VV E3L.


Subject(s)
Gene Deletion , Genetic Complementation Test , Orf virus/pathogenicity , RNA-Binding Proteins/genetics , Sequence Homology, Amino Acid , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Humans , Interferon-beta/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Orf virus/genetics , RNA-Binding Proteins/metabolism , Recombination, Genetic , Sequence Alignment , Vaccinia/physiopathology , Vaccinia/virology , Vaccinia virus/genetics , Viral Plaque Assay , Viral Proteins/metabolism , Virulence
4.
J Virol ; 76(18): 9207-17, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12186904

ABSTRACT

Hepatitis C virus (HCV) sets up a persistent infection in patients that likely involves a complex virus-host interaction. We previously found that the HCV nonstructural 5A (NS5A) protein interacts with growth factor receptor-binding protein 2 (Grb2) adaptor protein and inhibits the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) by epidermal growth factor (EGF). In the present study, we extended this analysis and investigated the specificity of the Grb2-NS5A interaction and whether the subversion of mitogenic signaling involves additional pathways. NS5A containing mutations within the C-terminal proline-rich motif neither bound Grb2 nor inhibited ERK1/2 activation by EGF, demonstrating that NS5A-Grb2 binding and downstream effects were due to direct interactions. Interestingly, NS5A could also form a complex with the Grb2-associated binder 1 (Gab1) protein in an EGF treatment-dependent manner. However, the NS5A-Gab1 association, which appeared indirect, was not mediated by direct NS5A-Grb2 interaction but was likely dependent on direct NS5A interaction with the p85 subunit of phosphatidylinositol 3-kinase (PI3K). The in vivo association of NS5A with p85 PI3K required the N-terminal, but not the C-terminal, region of NS5A. The downstream effects of the NS5A-p85 PI3K interaction included increased tyrosine phosphorylation of p85 PI3K in response to EGF. Consistent with this observation and the antiapoptotic properties of NS5A, we also detected enhanced tyrosine phosphorylation of the downstream AKT protein kinase and increased serine phosphorylation of BAD, a proapoptotic factor and an AKT substrate, in the presence of NS5A. These results collectively suggest a model in which NS5A interacts with Grb2 to inhibit mitogenic signaling while simultaneously promoting the PI3K-AKT cell survival pathway by interaction with p85 PI3K, which may represent a crucial step in HCV persistence and pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Hepacivirus/pathogenicity , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Apoptosis , Enzyme Activation , GRB2 Adaptor Protein , Gene Expression Regulation , HeLa Cells , Hepacivirus/physiology , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Viral Nonstructural Proteins/genetics
5.
J Virol ; 76(10): 5251-9, 2002 May.
Article in English | MEDLINE | ID: mdl-11967338

ABSTRACT

The vaccinia virus E3L gene encodes two double-stranded RNA binding proteins that promote viral growth and pathogenesis through suppression of innate immunity. To explore how E3L enables vaccinia virus to evade the interferon system, cells and mice deficient in the principal interferon-regulated antiviral enzymes, PKR and RNase L, were infected with wild-type vaccinia virus and strains of vaccinia virus from which E3L had been deleted (E3L-deleted strains). While wild-type virus was unaffected by RNase L and PKR, virus lacking E3L replicated only in the deficient cells. Nevertheless, E3L-deleted virus failed to replicate to high titers or to cause significant morbidity or mortality in triply deficient mice lacking RNase L, PKR, and Mx1. To investigate the underlying cause, we determined the effect of E3L on interferon regulatory factor 3 (IRF3), a transcription factor required for viral induction of subtypes of type I interferons. Results showed that IRF3 activation and interferon-beta induction occurred after infections with E3L-deleted virus but not with wild-type virus. These findings demonstrate that E3L plays an essential role in the pathogenesis of vaccinia virus by blocking the interferon system at multiple levels. Furthermore, our results indicate the existence of an interferon-mediated antipoxvirus pathway that operates independently of PKR, Mx1, or the 2-5A/RNase L system.


Subject(s)
GTP-Binding Proteins , Interferon-beta/biosynthesis , RNA-Binding Proteins/pharmacology , Vaccinia virus/pathogenicity , Vaccinia/virology , Viral Proteins/pharmacology , Animals , Cricetinae , DNA-Binding Proteins/immunology , Disease Models, Animal , Endoribonucleases/deficiency , Endoribonucleases/genetics , Female , Gene Deletion , Humans , Interferon Regulatory Factor-3 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myxovirus Resistance Proteins , Proteins/genetics , RNA-Binding Proteins/immunology , Transcription Factors/immunology , Vaccinia/immunology , Vaccinia virus/metabolism , Viral Proteins/immunology , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL