Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Vaccine ; 29(52): 9691-6, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-21983358

ABSTRACT

The New York City Board of Health (NYCBH) vaccinia virus (VACV) vaccine strain was deleted for the immune evasion gene, E3L, and tested for its pathogenicity and ability to protect mice from heterologous challenge with ectromelia virus (ECTV). NYCBHΔE3L was found to be highly attenuated for pathogenicity in a newborn mouse model and showed a similar attenuated phenotype as the NYVAC strain of vaccinia virus. Scarification with one or two doses of the attenuated NYCBHΔE3L was able to protect mice equally as well as NYCBH from death, weight loss, and viral spread to visceral organs. A single dose of NYCBHΔE3L resulted in low poxvirus-specific antibodies, and a second dose increased levels of poxvirus-specific antibodies to a level similar to that seen in animals vaccinated with a single dose of NYCBH. However, similar neutralizing antibody titers were observed following one or two doses of NYCBHΔE3L or NYCBH. Thus, NYCBHΔE3L shows potential as a candidate for a safer human smallpox vaccine since it protects mice from challenge with a heterologous poxvirus.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Gene Deletion , RNA-Binding Proteins/genetics , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Viral Proteins/genetics , Virulence Factors/genetics , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Body Weight , Cricetinae , Ectromelia, Infectious/immunology , Ectromelia, Infectious/mortality , Ectromelia, Infectious/pathology , Female , Mice , Pregnancy , Smallpox Vaccine/administration & dosage , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viremia/prevention & control
2.
Antimicrob Agents Chemother ; 51(2): 689-95, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17116683

ABSTRACT

ST-246 was evaluated for activity against cowpox virus (CV), vaccinia virus (VV), and ectromelia virus (ECTV) and had an in vitro 50% effective concentration (EC50) of 0.48 microM against CV, 0.05 microM against VV, and 0.07 microM against ECTV. The selectivity indices were >208 and >2,000 for CV and VV, respectively. The in vitro antiviral activity of ST-246 was significantly greater than that of cidofovir, which had an EC50 of 41.1 microM against CV and 29.2 microM against VV, with selectivity indices of >7 and >10, respectively. ST-246 administered once daily by oral gavage to mice infected intranasally with CV beginning 4 h or delayed until 72 h postinoculation was highly effective when given for a 14-day duration using 100, 30, or 10 mg/kg of body weight. When 100 mg/kg of ST-246 was administered to VV-infected mice, a duration of 5 days was sufficient to significantly reduce mortality even when treatment was delayed 24 h postinoculation. Viral replication in liver, spleen, and kidney, but not lung, of CV- or VV-infected mice was reduced by ST-246 compared to levels for vehicle-treated mice. When 100 mg/kg of ST-246 was given once daily to mice infected by the intranasal route with ECTV, treatment for 10 days prevented mortality even when treatment was delayed up to 72 h after viral inoculation. Viral replication in target organs of ECTV-infected mice was also reduced.


Subject(s)
Benzamides/administration & dosage , Indoles/administration & dosage , Orthopoxvirus/drug effects , Poxviridae Infections/drug therapy , Administration, Oral , Animals , Disease Models, Animal , Female , Isoindoles , Mice , Organ Specificity , Orthopoxvirus/physiology , Time Factors , Treatment Outcome , Virus Replication/drug effects
3.
J Gen Virol ; 86(Pt 11): 2969-2977, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227218

ABSTRACT

Rabbitpox virus (RPXV) is highly virulent for rabbits and it has long been suspected to be a close relative of vaccinia virus. To explore these questions, the complete coding region of the rabbitpox virus genome was sequenced to permit comparison with sequenced strains of vaccinia virus and other orthopoxviruses. The genome of RPXV strain Utrecht (RPXV-UTR) is 197 731 nucleotides long, excluding the terminal hairpin structures at each end of the genome. The RPXV-UTR genome has 66.5 % A + T content, 184 putative functional genes and 12 fragmented ORF regions that are intact in other orthopoxviruses. The sequence of the RPXV-UTR genome reveals that two RPXV-UTR genes have orthologues in variola virus (VARV; the causative agent of smallpox), but not in vaccinia virus (VACV) strains. These genes are a zinc RING finger protein gene (RPXV-UTR-008) and an ankyrin repeat family protein gene (RPXV-UTR-180). A third gene, encoding a chemokine-binding protein (RPXV-UTR-001/184), is complete in VARV but functional only in some VACV strains. Examination of the evolutionary relationship between RPXV and other orthopoxviruses was carried out using the central 143 kb DNA sequence conserved among all completely sequenced orthopoxviruses and also the protein sequences of 49 gene products present in all completely sequenced chordopoxviruses. The results of these analyses both confirm that RPXV-UTR is most closely related to VACV and suggest that RPXV has not evolved directly from any of the sequenced VACV strains, since RPXV contains a 719 bp region not previously identified in any VACV.


Subject(s)
Genome, Viral , Vaccinia virus/genetics , Base Sequence , DNA, Viral/chemistry , DNA, Viral/genetics , Molecular Sequence Data , Open Reading Frames , Vaccinia virus/classification
4.
J Immunol ; 167(8): 4553-9, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591783

ABSTRACT

The Janus kinase/STAT pathway has emerged as the paradigm of IFN-induced protection from viral infections. However, the possible participation of other signaling proteins in this protection is not clearly understood. In this report, we demonstrate that activation of phosphatidylinositol 3-kinase (PI3K) by either serum factors or IFNs blocks cell death induced by encephalomyocarditis virus (EMCV) and HSV. This increased resistance to virus-induced cell death does not involve the activation of the STAT pathway and occurs in the presence of normal viral replication. Interestingly, the cell uses two different PI3K regulated pathways to block EMCV- and HSV-induced cell death. The increased sensitivity of p85alpha(-/-) embryonic fibroblasts to EMCV-induced cell death is specifically corrected by overexpression of an activated allele of Akt/protein kinase B, but not activated mitogen-activated protein kinase extracellular kinase. Conversely, the augmented sensitivity of p85alpha(-/-) cells to HSV-induced cell death was compensated for by expression of an activated form of mitogen-activated protein kinase extracellular kinase, but not by activated Akt/protein kinase B. We conclude from these data that PI3K-activated pathways function in parallel with the Janus kinase/STAT pathway to protect cells from the lethal effects of viruses.


Subject(s)
Cell Death/physiology , Encephalomyocarditis virus/pathogenicity , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Simplexvirus/pathogenicity , Animals , Clone Cells , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/cytology , Interferons/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Trans-Activators/metabolism , Virus Replication
5.
Am J Pathol ; 159(1): 273-83, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438474

ABSTRACT

Viral infection is one environmental factor that has been implicated as a precipitating event that may initiate beta-cell damage during the development of diabetes. This study examines the mechanisms by which the viral replicative intermediate, double-stranded (ds) RNA impairs beta-cell function and induces beta-cell death. The synthetic dsRNA molecule polyinosinic-polycytidylic acid (poly IC) stimulates beta-cell DNA damage and apoptosis without impairing islet secretory function. In contrast, the combination of poly IC and interferon (IFN)-gamma stimulates DNA damage, apoptosis, and necrosis of islet cells, and this damage is associated with the inhibition of glucose-stimulated insulin secretion. Nitric oxide mediates the inhibitory and destructive actions of poly IC + IFN-gamma on insulin secretion and islet cell necrosis. Inhibitors of nitric oxide synthase, aminoguanidine, and N(G)-monomethyl-L-arginine, attenuate poly IC + IFN-gamma-induced DNA damage to levels observed in response to poly IC alone, prevent islet cell necrosis, and prevent the inhibitory actions on glucose-stimulated insulin secretion. N(G)-monomethyl-L-arginine fails to prevent poly IC- and poly IC + IFN-gamma-induced islet cell apoptosis. PKR, the dsRNA-dependent protein kinase that mediates the antiviral response in infected cells, is required for poly IC- and poly IC + IFN-gamma-induced islet cell apoptosis, but not nitric oxide-mediated islet cell necrosis. Alone, poly IC fails to stimulate DNA damage in islets isolated from PKR-deficient mice; however, nitric oxide-dependent DNA damage induced by the combination of poly IC + IFN-gamma is not attenuated by the genetic absence of PKR. These findings indicate that dsRNA stimulates PKR-dependent islet cell apoptosis, an event that is associated with normal islet secretory function. In contrast, poly IC + IFN-gamma-induced inhibition of glucose-stimulated insulin secretion and islet cell necrosis are events that are mediated by islet production of nitric oxide. These findings suggest that at least one IFN-gamma-induced antiviral response (islet cell necrosis) is mediated through a PKR-independent pathway.


Subject(s)
Interferon-gamma/pharmacology , Islets of Langerhans/physiology , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , Animals , Apoptosis/physiology , Cell Death/physiology , Cells, Cultured , DNA Damage/physiology , Drug Synergism , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Microscopy, Electron , Necrosis , Nitric Oxide/physiology , Protein Kinases/physiology , Rats , Rats, Sprague-Dawley , eIF-2 Kinase/physiology
6.
Virology ; 274(1): 17-25, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10936084

ABSTRACT

The poxvirus molluscum contagiosum (MC) has a worldwide distribution and its prevalence is on the rise. Here we report that the MCV MC013L protein inhibits glucocorticoid and vitamin D, but not retinoid or estrogen, nuclear receptor transactivation. A direct interaction of MC013L with glucocorticoid and vitamin D receptor is supported by yeast two-hybrid, GST pull-down, and far Western blot analyses. Glucocorticoids act as potent inhibitors of keratinocyte proliferation, while vitamin D and retinoids promote and block terminal differentiation, respectively. Therefore, MC013L may promote efficient virus replication by blocking the differentiation of infected keratinocytes. MC013L may be the first member of a new class of poxvirus proteins that directly modulate nuclear receptor-mediated transcription.


Subject(s)
Heat-Shock Proteins/physiology , Molluscum contagiosum virus/physiology , Receptors, Steroid/antagonists & inhibitors , Viral Proteins/physiology , Amino Acid Sequence , Animals , COS Cells , Calcitriol/metabolism , Cell Nucleus , Gene Expression , Genes, Reporter , Growth Substances/genetics , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Molecular Sequence Data , Molluscum contagiosum virus/genetics , Molluscum contagiosum virus/metabolism , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Steroid/genetics , Sequence Homology, Amino Acid , Transcription, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/physiology
7.
EMBO J ; 19(14): 3630-8, 2000 Jul 17.
Article in English | MEDLINE | ID: mdl-10899117

ABSTRACT

In this study, the role of the double-stranded (ds) RNA-dependent protein kinase (PKR) in macrophage activation was examined. dsRNA [polyinosinic:polycytidylic acid (poly IC)]-stimulated inducible nitric oxide synthase, interleukin (IL)-1alpha and IL-1beta mRNA expression, nitrite formation and IL-1 release are attenuated in RAW264.7 cells stably expressing dominant negative (dn) mutants of PKR. The transcriptional regulator nuclear factor (NF)-kappaB is activated by dsRNA, and appears to be required for dsRNA-induced macrophage activation. While dnPKR mutants prevent macrophage activation, they fail to attenuate dsRNA-induced IkappaB degradation or NF-kappaB nuclear localization. The inhibitory actions of dnPKR on dsRNA-induced macrophage activation can be overcome by treatment with interferon (IFN)-gamma, an event associated with PKR degradation. Furthermore, dsRNA + IFN-gamma stimulate inducible nitric oxide synthase expression, IkappaB degradation and NF-kappaB nuclear localization to similar levels in macrophages isolated from PKR(-/-) and PKR(+/+) mice. These findings indicate that both NF-kappaB and PKR are required for dsRNA-induced macrophage activation; however, dsRNA-induced NF-kappaB activation occurs by PKR-independent mechanisms in macrophages. In addition, the PKR dependence of dsRNA-induced macrophage activation can be overcome by IFN-gamma.


Subject(s)
Macrophage Activation/drug effects , Macrophages, Peritoneal/enzymology , RNA, Double-Stranded/pharmacology , eIF-2 Kinase/metabolism , Animals , Cell Line , Cells, Cultured , Enzyme Induction/drug effects , I-kappa B Proteins/metabolism , Interferon-gamma/pharmacology , Interleukin-1/genetics , Interleukin-1/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Mutation , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Poly I-C/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Messenger/metabolism , Signal Transduction/drug effects , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
8.
J Immunol ; 164(6): 3246-54, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706717

ABSTRACT

IL-18 induces IFN-gamma and NK cell cytotoxicity, making it a logical target for viral antagonism of host defense. We demonstrate that the ectromelia poxvirus p13 protein, bearing homology to the mammalian IL-18 binding protein, binds IL-18, and inhibits its activity in vitro. Binding of IL-18 to the viral p13 protein was compared with binding to the cellular IL-18R. The dissociation constant of p13 for murine IL-18 is 5 nM, compared with 0.2 nM for the cellular receptor heterodimer. Mice infected with a p13 deletion mutant of ectromelia virus had elevated cytotoxicity for YAC-1 tumor cell targets compared with control animals. Additionally, the p13 deletion mutant virus exhibited decreased levels of infectivity. Our data suggest that inactivation of IL-18, and subsequent impairment of NK cell cytotoxicity, may be one mechanism by which ectromelia evades the host immune response.


Subject(s)
Cytotoxicity, Immunologic , Ectromelia virus/immunology , Immunosuppressive Agents/metabolism , Interleukin-18/metabolism , Killer Cells, Natural/immunology , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Binding, Competitive/immunology , COS Cells , Ectromelia virus/growth & development , Ectromelia virus/metabolism , Female , Immunosuppressive Agents/immunology , Injections, Intraperitoneal , Interleukin-18/antagonists & inhibitors , Interleukin-18 Receptor alpha Subunit , Killer Cells, Natural/virology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Binding/immunology , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/metabolism , Receptors, Interleukin-18 , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Proteins/physiology , Virus Replication
9.
Virus Res ; 66(2): 155-73, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10725549

ABSTRACT

From the right-hand end of the ectromelia virus (strain Moscow) genome, 32318 bps have been sequenced, and characterized to include a total of 18 open reading frames (ORFs) and six regions which apparently no longer code for functional proteins. At least six of the ORFs appear to be involved in blocking the inflammatory/immune host response to infection, and therefore probably contribute significantly to the virulence of this virus in its natural host, the mouse. One of these genes encoded an isolog of the poxvirus chemokine binding protein, and was shown to be the most abundant protein secreted from ectromelia virus infected cells. Two regions were found to have significant similarity to poxvirus genes encoding tumor necrosis factor (TNF) binding proteins. Both are distinct from cytokine response modifier (crm)B and crmC but only one is predicted to encode a functional TNF binding protein. A novel similarity between the C-terminal domain of poxvirus TNF binding proteins and several other poxvirus proteins is also presented. The results are discussed in the context of ectromelia virus pathogenesis of mice.


Subject(s)
Ectromelia virus/genetics , Ectromelia, Infectious/virology , Genome, Viral , Immunologic Factors/genetics , Amino Acid Sequence , Animals , Antigens, Viral/analysis , Cell Line , DNA Transposable Elements , Ectromelia virus/chemistry , Ectromelia virus/pathogenicity , Female , Glycoproteins/genetics , Humans , Immunohistochemistry , Membrane Proteins/genetics , Mice , Mice, Inbred A , Molecular Sequence Data , Open Reading Frames , Receptors, Chemokine/genetics , Receptors, Interferon/genetics , Receptors, Interleukin-1/genetics , Receptors, Tumor Necrosis Factor/genetics , Sequence Alignment , Sequence Deletion , Serpins/genetics , Skin Diseases, Viral/veterinary , Viral Proteins/genetics , Virulence
10.
Proc Natl Acad Sci U S A ; 95(7): 3786-91, 1998 Mar 31.
Article in English | MEDLINE | ID: mdl-9520445

ABSTRACT

Cowpox virus Brighton red strain (CPV) contains a gene, crmD, which encodes a 320-aa tumor necrosis factor receptor (TNFR) of 44% and 22% identity, respectively, to the CPV TNFR-like proteins, cytokine response modifiers (crm) CrmB and CrmC. The crmD gene was interrupted in three other cowpox strains examined and absent in various other orthopoxviruses; however, four strains of ectromelia virus (ECT) examined contained an intact crmD (97% identity to CPV crmD) and lacked cognates of crmB and crmC. The protein, CrmD, contains a transport signal; a 151-aa cysteine-rich region with 21 cysteines that align with human TNFRII ligand-binding region cysteines; and C-terminal region sequences that are highly diverged from cellular TNFR C-terminal region sequences involved in signal transduction. Bacterial maltose-binding proteins containing the CPV or ECT CrmD cysteine-rich region bound TNF and lymphotoxin-alpha (LTalpha) and blocked their in vitro cytolytic activity. Secreted viral CrmD bound TNF and LTalpha and was detectable after the early stage of replication, using nonreducing conditions, as 60- to 70-kDa predominant and 90- to 250-kDa minor disulfide-linked complexes that were able to be reduced to a 46-kDa form and deglycosylated to a 38-kDa protein. Cells infected with CPV produced extremely low amounts of CrmD compared with ECT. Possessing up to three TNFRs, including CrmD, which is secreted as disulfide-linked complexes in varied amounts by CPV and ECT, likely enhances the dynamics of the immune modulating mechanisms of orthopoxviruses.


Subject(s)
Glycoproteins/analysis , Orthopoxvirus/metabolism , Receptors, Tumor Necrosis Factor/analysis , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Glycoproteins/genetics , Humans , Molecular Sequence Data , Orthopoxvirus/genetics , Receptors, Tumor Necrosis Factor/genetics , Sequence Alignment , Viral Proteins/analysis , Viral Proteins/genetics
11.
Immunity ; 8(4): 473-82, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9586637

ABSTRACT

The vaccinia virus A39R protein is a member of the semaphorin family. A39R.Fc protein was used to affinity purify an A39R receptor from a human B cell line. Tandem mass spectrometry of receptor peptides yielded partial amino acid sequences that allowed the identification of corresponding cDNA clones. Sequence analysis of this receptor indicated that it is a novel member of the plexin family and identified a semaphorin-like domain within this family, thus suggesting an evolutionary relationship between receptor and ligand. A39R up-regulated ICAM-1 on, and induced cytokine production from, human monocytes. These data, then, describe a receptor for an immunologically active semaphorin and suggest that it may serve as a prototype for other plexin-semaphorin binding pairs.


Subject(s)
Cytokines/biosynthesis , Membrane Proteins/physiology , Monocytes/immunology , Receptors, Cell Surface/metabolism , Receptors, Virus , Vaccinia virus/immunology , Viral Proteins/immunology , Viral Proteins/metabolism , Amino Acid Sequence , B-Lymphocytes/metabolism , Cell Adhesion Molecules/genetics , Cell Line , Cloning, Molecular , Evolution, Molecular , Humans , Intercellular Adhesion Molecule-1/metabolism , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Monocytes/metabolism , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Vaccinia virus/genetics , Viral Proteins/genetics
12.
Virus Res ; 52(2): 157-67, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9495531

ABSTRACT

A novel poxvirus gene has been characterized within the genome of ectromelia virus. It has significant similarity to a family of lysophospholipases suggesting that it may function in the degradation of lysophospholipids. Since these molecules are active in the stimulation of inflammation, we hypothesize that this gene may play a role in virus virulence. This gene is expressed early in the ectromelia virus replication cycle, before DNA replication. We have also characterized a human cDNA that encodes a protein which is 49.5% identical to the ectromelia virus protein. By its presence in multiple cDNA libraries, this human gene is known to be expressed in a variety of body tissues and is likely to function in the normal regulation of lysophospholipid levels. This family of proteins have conserved blocks of amino acids that are indicative of a serine-aspartic acid-histidine catalytic triad, similar to those used by true lipases and a number of esterases.


Subject(s)
Ectromelia virus/enzymology , Escherichia coli/enzymology , Genes, Viral , Lysophospholipase/genetics , Amino Acid Sequence , Base Sequence , Cell Line , DNA, Viral , Ectromelia virus/genetics , Gene Expression , Humans , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid
13.
J Virol ; 70(12): 8301-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8970949

ABSTRACT

The importance of T-lymphocyte subsets in the control of poxvirus infections is controversial. To determine the relative contribution of lymphocyte subsets important for recovery from infection with ectromelia virus (EV), a natural murine poxvirus pathogen, C57BL/6 (B6) mice lacking functional CD8+ T cells because of disruption of the beta2-microglobulin gene or lacking functional CD4+ T cells because of disruption of the I-(A)beta gene, acutely depleted of CD8+ or CD4+ T cells with monoclonal antibody, or depleted of macrophage subsets by the macrophage suicide technique were used. Recovery from infection was strictly dependent on the effector functions of CD8+ T cells, in the absence of which 100% mortality resulted. This lymphocyte population had demonstrable antiviral activity early in the infection process even before class I major histocompatibility complex (MHC)-restricted CD8+ cytotoxic T-lymphocyte (CTL) activity was detectable. CD4+ T cells were found to be necessary for the generation of an optimal virus-specific, class I MHC-restricted CD8+ CTL response and contributed to virus clearance not involving cytolytic mechanisms. In both models of CD4+ T-cell deficiency, virus clearance was incomplete and persisted at low levels in most organs and at very high levels in the skin, but the animals did not die. The elimination of macrophage subpopulations impeded virus clearance, impaired the generation of class I MHC-restricted antiviral CTL response, and resulted in 100% mortality. These findings establish an absolute requirement for CD8+ and CD4+ T lymphocytes and macrophage subsets in the elimination of a natural murine poxvirus infection and support the idea that macrophages may be essential accessory cells for the generation of class I MHC-restricted antiviral CTL responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Macrophages/immunology , Animals , Cell Line , Chlorocebus aethiops , Cytotoxicity, Immunologic/immunology , Ectromelia virus/physiology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/immunology , Virus Replication/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
14.
Lab Anim Sci ; 46(6): 602-11, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9001171

ABSTRACT

Mousepox was diagnosed in and eradicated from a laboratory mouse colony at the Naval Medical Research Institute. The outbreak began with increased mortality in a single room; subsequently, small numbers of animals in separate cages in other rooms were involved. Signs of disease were often mild, and overall mortality was low; BALB/cByJ mice were more severely affected, and many of them died spontaneously. Conjunctivitis was the most common clinical sign of disease in addition to occasional small, crusty scabs on sparsely haired or hairless areas of skin. Necropsy findings included conjunctivitis, enlarged spleen, and pale liver. Hemorrhage into the pyloric region of the stomach and proximal portion of the small intestine was observed in experimentally infected animals. In immune competent and immune deficient mice, the most common histologic finding was multifocal to coalescing splenic necrosis; necrosis was seen less frequently in liver, lymph nodes, and Peyer's patches. Necrosis was rarely observed in ovary, vagina, uterus, colon, or lung. Splenic necrosis often involved over 50% of the examined tissue, including white and red pulp. Hepatic necrosis was evident as either large, well-demarcated areas of coagulative necrosis or as multiple, random, interlacing bands of necrosis. Intracytoplasmic eosinophilic inclusion bodies were seen in conjunctival mucosae and haired palpebra. Ectromelia virus was confirmed as the causative agent of the epizootic by electron microscopy, immunohistochemistry, animal inoculations, serologic testing, virus isolation, and polymerase chain reaction. Serologic testing was of little value in the initial stages of the outbreak, although 6 weeks later, orthopoxvirus-specific antibody was detected in colony mice by indirect fluorescent antibody and enzyme-linked immunosorbent assay procedures. The outbreak originated from injection of mice with a contaminated, commercially produced, pooled mouse serum. The most relevant concern may be the unknown location of the source of the virus and the presence of a reservoir for this virus within the United States.


Subject(s)
Animals, Laboratory , Ectromelia, Infectious/epidemiology , Mice, Inbred BALB C , Animals , Antibodies, Viral/blood , Conjunctivitis/pathology , Conjunctivitis/virology , DNA, Viral/analysis , Ectromelia virus/genetics , Ectromelia virus/immunology , Ectromelia virus/isolation & purification , Ectromelia, Infectious/diagnosis , Ectromelia, Infectious/pathology , Liver/pathology , Mice , Microscopy, Electron , Necrosis , Polymerase Chain Reaction , Skin Diseases, Viral/pathology , Spleen/pathology
15.
Proc Natl Acad Sci U S A ; 93(1): 383-7, 1996 Jan 09.
Article in English | MEDLINE | ID: mdl-8552643

ABSTRACT

ICSBP is a member of the interferon (IFN) regulatory factor (IRF) family that regulates expression of type I interferon (IFN) and IFN-regulated genes. To study the role of the IRF family in viral infection, a cDNA for the DNA-binding domain (DBD) of ICSBP was stably transfected into U937 human monocytic cells. Clones that expressed DBD exhibited a dominant negative phenotype and did not elicit antiviral activity against vesicular stomatitis virus (VSV) infection upon IFN treatment. Most notably, cells expressing DBD were refractory to infection by vaccinia virus (VV) and human immunodeficiency virus type 1 (HIV-1). The inhibition of VV infection was attributed to defective virion assembly, and that of HIV-1 to low CD4 expression and inhibition of viral transcription in DBD clones. HIV-1 and VV were found to have sequences in their regulatory regions similar to the IFN-stimulated response element (ISRE) to which IRF family proteins bind. Accordingly, these viral sequences and a cellular ISRE bound a shared factor(s) expressed in U937 cells. These observations suggest a novel host-virus relationship in which the productive infection of some viruses is regulated by the IRF-dependent transcription pathway through the ISRE.


Subject(s)
Carrier Proteins/physiology , Gene Expression Regulation, Viral , HIV Infections/prevention & control , HIV-1/genetics , Repressor Proteins/genetics , Vaccinia virus/genetics , Vaccinia/prevention & control , Base Sequence , Cells, Cultured , DNA-Binding Proteins/metabolism , Genes, Dominant , Humans , Interferon Regulatory Factors , Interferons/physiology , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism
16.
J Virol ; 70(1): 165-71, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8523521

ABSTRACT

Induction of the cytopathic effect (CPE) in cells infected with poxvirus seems ubiquitous in that it has been associated with all different strains and preparations of poxviruses, regardless of the replicating status of these viruses. The study of the mechanisms by which CPE is induced by nonreplicating poxviruses is hampered by the lack of any noncytopathic mutant strains and preparations. In this paper, we report on the patterns of gene expression and induction of CPE by vaccinia viruses treated by limited cross-linking with psoralen and long-wave UV light (PLWUV). We show that treatment of cell-free virus with PLWUV could inactivate viral replication without abolishing the ability of the virus to infect cells. Viral transcription as indicated by reporter genes was generally enhanced and prolonged under early viral promoters and abolished under late promoters. Furthermore, increasing the levels of cross-linking with PLWUV resulted in a decrease and abolishment of viral expression of a large reporter gene and a concomitant loss of the induction of CPE. Cells infected with such a virus were able to express the reporter genes and proliferate. The generation of nonreplicating and noncytopathic recombinant vaccinia viruses may help in studies of the mechanisms of CPE induction by poxvirus and may facilitate the use of poxviral vectors in broader areas of research and clinical applications.


Subject(s)
Cross-Linking Reagents/pharmacology , Cytopathogenic Effect, Viral/drug effects , Ficusin/pharmacology , Gene Expression Regulation, Viral/drug effects , Photosensitizing Agents/pharmacology , Ultraviolet Rays , Vaccinia virus/drug effects , Animals , Cell Line , Cytopathogenic Effect, Viral/radiation effects , DNA, Viral/metabolism , Genes, Reporter , Promoter Regions, Genetic , Transcription, Genetic , Tumor Cells, Cultured , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Vaccinia virus/radiation effects , Virus Replication/drug effects , Virus Replication/radiation effects
17.
Cancer J Sci Am ; 2(1): 21-7, 1996.
Article in English | MEDLINE | ID: mdl-9166494

ABSTRACT

PURPOSE: This study investigates the feasibility of systemic gene delivery in a tumor-bearing host using a vaccinia virus-based in vivo gene delivery and expression system. METHODS: A recombinant vaccinia virus encoding human interleukin-1beta (hIL-1beta) was constructed with a strong synthetic vaccinia virus late promoter driving hIL-1beta gene expression. C57BL/6 mice bearing established subcutaneous pancreatic tumors were injected intravenously in a blinded, randomized fashion with different doses of either the recombinant vaccinia virus(vMJ601hIL-Ibeta), control vaccinia (wild-type or TK-deficient), or saline. Toxicity was assessed, serial tumor sizes were measured, and viral titers and the amount of hIL-1beta in tumor, liver, and spleen were determined. RESULTS: High viral titers (10(6) PFU/g) were detected in tumors for all three viruses on postinjection day 3, and tumor viral titers persisted at high levels until day 9. In contrast, viral titers were initially 104-fold lower in nontumor tissues and decreased to undetectable levels by day 9. vMJ60hIL-1beta was rapidly cleared from liver and spleen by day 3 (titer levels < 100 PFU/g), while tumor titer levels persisted at 8.5 x 10(6) PFU/g. hIL-1beta was measurable in three of three tumors from vMJ601hIL-1beta treated mice on postinjection day 3, one of three on day 6, and one of three on day 9; no hIL-1beta was detected in any other tumors or normal tissues. Wild-type vaccinia had no antitumor effects. Treatment with two different doses of vMJ601hIL-1beta resulted in a consistent and significant decrease in tumor size in repeatable experiments as compared to controls. Histologic analysis revealed tumor cell necrosis with a surrounding neutrophil infiltrate in the vMJ601hIL-1beta treated tumor. CONCLUSION: These data show that recombinant vaccinia virus encoding hIL-1beta given intravenously preferentially localizes and amplifies in tumor tissue, is rapidly cleared from liver and spleen, produces measurable hIL-1beta in tumor but not normal tissues, and inhibits growth of established solid tumors in mice. Recombinant vaccinia virus encoding therapeutic genes may be a practical, efficient vehicle for direct in vivo gene transfer and expression in the treatment of cancer.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Interleukin-1/genetics , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/therapy , Vaccinia virus , Animals , Gene Expression , Humans , Immunohistochemistry , Interleukin-1/analysis , Liver/virology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Neoplasms, Experimental/virology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Recombinant Proteins , Spleen/virology
18.
Virology ; 213(2): 655-9, 1995 Nov 10.
Article in English | MEDLINE | ID: mdl-7491789

ABSTRACT

Molluscum contagiosum virus (MCV) infects preadolescent children and sexually active adults, frequently causing a disfiguring cutaneous disease in immunosuppressed HIV-infected individuals. The development of an efficacious treatment regime has been hampered by the failure to replicate the virus in the laboratory. Here we report the first demonstration of MCV replication in an experimental system. In human foreskin grafts to athymic mice, MCV induced morphological changes which were indistinguishable from patient biopsies and included the development and migration of molluscum bodies containing mature virions to the epidermal surface.


Subject(s)
DNA, Viral/analysis , Molluscum Contagiosum/virology , Molluscum contagiosum virus/physiology , Skin/virology , Animals , Humans , Infant, Newborn , Male , Mice , Mice, Nude , Molluscum Contagiosum/pathology , Molluscum contagiosum virus/isolation & purification , Skin/pathology , Skin Transplantation , Transplantation, Heterologous , Virion/metabolism , Virus Replication
19.
J Virol ; 69(7): 4103-11, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7769668

ABSTRACT

We have previously described a gene of ectromelia virus (EV) that codes for a 28-kDa RING zinc finger-containing protein (p28) that is nonessential for virus growth in cell culture but is critical for EV pathogenicity in mice (T. G. Senkevich, E. V. Koonin, and R. M. L. Buller, Virology 198:118-128; 1994). Here, we show that, unlike all tested cell cultures, the expression of p28 is required for in vitro replication of EV in murine resident peritoneal macrophages. In macrophages infected with the p28- mutant, viral DNA replication was not detected, whereas the synthesis of at least two early proteins was observed. Immunofluorescence and biochemical analyses showed that in EV-infected macrophages or BSC-1 cells, p28 is associated with virus factories. By use of a vaccinia virus expression system to examine different truncated versions of p28, it was shown that the disruption of the specific structure of the RING domain had no influence on the intracellular localization of this protein. When viral DNA replication was inhibited with cytosine arabinoside, p28 was found in distinct, focal structures that may be precursors to the factories. We hypothesize that in macrophages, which are highly specialized, nondividing cells, p28 substitutes for an unknown cellular factor(s) that may be required for viral DNA replication or a stage of virus reproduction between the expression of early genes and the onset of DNA synthesis. In the absence of p28, the attenuation of EV pathogenicity can be explained by a failure of the virus to replicate in macrophage lineage cells at all successive steps in the spread of virus from the skin to its target organ, the liver.


Subject(s)
Ectromelia virus/physiology , Macrophages/virology , Viral Proteins/physiology , Virus Replication , Zinc Fingers , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA Replication , Female , Mice , Molecular Sequence Data , Rabbits , Viral Proteins/analysis
20.
J Biol Chem ; 270(27): 15974-8, 1995 Jul 07.
Article in English | MEDLINE | ID: mdl-7608155

ABSTRACT

Poxviruses encode a large number of proteins that attenuate the inflammatory and immune responses to infection. In this report we demonstrate that a number of orthopoxviruses express a type I interferon (IFN)-binding protein, which is encoded by the B18R open reading frame in the WR strain of vaccinia virus. The B18R protein has significant regions of homology with the alpha subunits of the mouse, human, and bovine type I IFN receptors, bound human IFN alpha 2 with high affinity, and inhibited transmembrane signaling as demonstrated by inhibition of Fc receptor factor gamma 1/gamma 2 and interferon-stimulated gene factor-3 formation as well as inhibition of the IFN alpha antiviral response. Among viral host response modifiers, the B18R protein is unique inasmuch as it exists as a soluble extracellular as well as a cell surface protein and thus should effectively block both autocrine and paracrine functions of IFN.


Subject(s)
Interferon-alpha/antagonists & inhibitors , Orthopoxvirus/genetics , Receptors, Interferon/genetics , Signal Transduction , Viral Proteins/genetics , Amino Acid Sequence , DNA-Binding Proteins/metabolism , Genes, Viral/genetics , Molecular Sequence Data , Receptors, Interferon/metabolism , STAT1 Transcription Factor , STAT2 Transcription Factor , Sequence Homology, Amino Acid , Trans-Activators/metabolism , Vaccinia virus/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...