Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Rev Esp Quimioter ; 35(6): 509-518, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35785957

ABSTRACT

Infection caused by Monkeypox Virus (MPVX) has small rodents as its natural reservoir and both monkeys and humans are occasional hosts. The causative agent is an Orthopoxvirus (MPVX) that was isolated in monkeys in 1958 and proved capable of passing to humans in 1970. It remained contained in Africa, causing isolated episodes of infection, until 2003 when an outbreak occurred in the United States following importation of animals from that continent. Since then, anecdotal cases have continued to be reported outside Africa, usually very clearly linked to travelers to those countries, but in May 2022, a broad outbreak of this disease has begun, now affecting several continents, with the emergence of human cases of MPVX (H-MPVX) infection mainly among Men that have Sex with Men (MSM). The disease has an incubation time ranging from 5 to 15 days and is characterized by the presence of pustules, fever, malaise and headache. The presence of significant regional lymphadenopathy is a differential feature with episodes of classical smallpox. Proctitis and pharyngitis, with minimal skin lesions, may be another form of presentation. Diagnosis can be confirmed by PCR testing of lesions or by demonstration of MPVX in other body fluids or tissues, although in the appropriate epidemiologic setting the clinical picture is highly suggestive of the disease. Effective drug treatment has been developed as part of programs to protect against potential bioterrorist agents and smallpox vaccinees are known to have high protection against monkeypox. New vaccines are available, but neither the drugs nor the vaccines are yet freely available on the market. The prognosis of the disease appears, at least in adults in developed countries, to be good, with very low mortality figures and much less aggressive behavior than that described in classical smallpox. Isolation measures, essential for the control of the outbreak, have been published by the health authorities.


Subject(s)
Mpox (monkeypox) , Sexual and Gender Minorities , Smallpox , Male , Adult , Animals , Humans , United States , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/diagnosis , Smallpox/epidemiology , Homosexuality, Male , Monkeypox virus , Disease Outbreaks
2.
Mol Ecol ; 24(19): 4812-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26198078

ABSTRACT

RNA viruses exist as complex mixtures of genotypes, known as quasispecies, where the evolution potential resides in the whole community of related genotypes. Quasispecies structure and dynamics have been studied in detail for virus infecting animals and plants but remain unexplored for those infecting micro-organisms in environmental samples. We report the first metagenomic study of RNA viruses in an Antarctic lake (Lake Limnopolar, Livingston Island). Similar to low-latitude aquatic environments, this lake harbours an RNA virome dominated by positive single-strand RNA viruses from the order Picornavirales probably infecting micro-organisms. Antarctic picorna-like virus 1 (APLV1), one of the most abundant viruses in the lake, does not incorporate any mutation in the consensus sequence from 2006 to 2010 and shows stable quasispecies with low-complexity indexes. By contrast, APLV2-APLV3 are detected in the lake water exclusively in summer samples and are major constituents of surrounding cyanobacterial mats. Their quasispecies exhibit low complexity in cyanobacterial mat, but their run-off-mediated transfer to the lake results in a remarkable increase of complexity that may reflect the convergence of different viral quasispecies from the catchment area or replication in a more diverse host community. This is the first example of viral quasispecies from natural aquatic ecosystems and points to ecological connectivity as a modulating factor of quasispecies complexity.


Subject(s)
Ecosystem , Genome, Viral , RNA Viruses/classification , RNA Viruses/isolation & purification , Antarctic Regions , Cyanobacteria/virology , Lakes , Metagenomics , Molecular Sequence Data , Phylogeny , Picornaviridae/classification , Picornaviridae/isolation & purification , Sequence Analysis, RNA
3.
Mucosal Immunol ; 3(6): 633-44, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20664576

ABSTRACT

Tumor necrosis factor α (TNFα) is a key pathogenic factor in Crohn's disease and rheumatoid arthritis. TNF(ΔARE) mice express high levels of TNFα and present Crohn's-like ileitis and arthritis. Alterations in the chemokine network could underline the TNF-driven ileitis. The aim of this study was to evaluate the role of TNF and chemokines in ileitis using ectromelia virus cytokine response modifier D (CrmD), a protein that binds TNFα and a limited number of chemokines. We generated transgenic mice expressing CrmD in intestinal epithelial cells (vCrmD mice) and crossed them with the TNF(ΔARE) mice to test whether CrmD could affect TNF-driven inflammatory processes. During homeostasis, only the number of B cells in the lamina propria was reduced by CrmD expression. Interestingly, CrmD expression in the intestine markedly attenuated the inflammatory infiltrates in the ileum of TNF(ΔARE) mice, but did not affect development of arthritis. Our results suggest that CrmD affects development of ileitis by locally affecting both TNF and chemokine function in the ileum.


Subject(s)
B-Lymphocytes/metabolism , Crohn Disease/immunology , Ectromelia virus/immunology , Intestinal Mucosa/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Rheumatic Fever/immunology , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/metabolism , Animals , Arthritis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cells, Cultured , Crohn Disease/genetics , Crohn Disease/pathology , Crohn Disease/physiopathology , Disease Models, Animal , Humans , Ileitis , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutation/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Rheumatic Fever/genetics , Rheumatic Fever/pathology , Rheumatic Fever/physiopathology , Transgenes/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/genetics , Viral Proteins/immunology
4.
J Gen Virol ; 86(Pt 12): 3209-3214, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16298965

ABSTRACT

Recently, glycoprotein G (gG) of several alphaherpesviruses infecting large herbivores was shown to belong to a new family of chemokine-binding proteins (vCKBPs). In the present study, the function of Felid herpesvirus 1 (FeHV-1) gG as a vCKBP was investigated and the following conclusions were reached: (i) FeHV-1 secreted gG is a high-affinity broad-spectrum vCKBP that binds CC, CXC and C chemokines; (ii) gG is the only vCKBP expressed by FeHV-1 that binds CCL3 and CXCL1; (iii) secreted gG blocks chemokine activity by preventing their interaction with high-affinity cellular receptors; (iv) the membrane-anchored form of gG expressed on the surface of infected cells is also able to bind chemokines; and (v) the vCKBP activity is conserved among different field isolates of FeHV-1. Altogether, these data demonstrate that FeHV-1 gG is a new member of the vCKBP-4 family. Moreover, this study is the first to demonstrate that gG expressed at the surface of FeHV-1-infected cells can also bind chemokines.


Subject(s)
Chemokines/metabolism , Varicellovirus/physiology , Viral Envelope Proteins/metabolism , Chemokines/antagonists & inhibitors , Chemokines, C/metabolism , Chemokines, CC/metabolism , Chemokines, CXC/metabolism , DNA, Viral/chemistry , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA
5.
J Virol ; 75(1): 226-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119592

ABSTRACT

Cytokines and chemokines play a critical role in both the innate and acquired immune responses and constitute prime targets for pathogen sabotage. Molecular mimicry of cytokines and cytokine receptors is a mechanism encoded by large DNA viruses to modulate the host immune response. Three tumor necrosis factor receptors (TNFRs) have been identified in the poxvirus cowpox virus. Here we report the identification and characterization of a fourth distinct soluble TNFR, named cytokine response modifier E (CrmE), encoded by cowpox virus. The crmE gene has been sequenced in strains of the orthopoxviruses cowpox virus, ectromelia virus, and camelpox virus, and was found to be active in cowpox virus. crmE is expressed as a secreted 18-kDa protein with TNF binding activity. CrmE was produced in the baculovirus and vaccinia virus expression systems and was shown to bind human, mouse, and rat TNF, but not human lymphotoxin alpha, conjugates of lymphotoxins alpha and beta, or seven other ligands of the TNF superfamily. However, CrmE protects cells only from the cytolytic activity of human TNF. CrmE is a new member of the TNFR superfamily which is expressed as a soluble molecule that blocks the binding of TNF to high-affinity TNFRs on the cell surface. The remarkable finding of a fourth poxvirus-encoded TNFR suggests that modulation of TNF activity is complex and represents a novel viral immune evasion mechanism.


Subject(s)
Poxviridae/genetics , Receptors, Tumor Necrosis Factor/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , DNA, Viral/chemistry , Humans , Mice , Molecular Sequence Data , Rats , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Virol ; 74(23): 11230-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11070021

ABSTRACT

Poxviruses encode a broad range of proteins that interfere with host immune functions, such as soluble versions of receptors for the cytokines tumor necrosis factor, interleukin-1 beta, gamma interferon (IFN-gamma), IFN-alpha/beta, and chemokines. These virus-encoded cytokine receptors have a profound effect on virus pathogenesis and enable the study of the role of cytokines in virus infections. The vaccinia virus (VV) Western Reserve gene B18R encodes a secreted protein with 3 immunoglobulin domains that functions as a soluble receptor for IFN-alpha/beta. We have found that after secretion B18R binds to both uninfected and infected cells. The B18R protein present at the cell surface maintains the properties of the soluble receptor, binding IFN-alpha/beta with high affinity and with broad species specificity, and protects cells from the antiviral state induced by IFN-alpha/beta. VV strain Wyeth expressed a truncated B18R protein lacking the C-terminal immunoglobulin domain. This protein binds IFN with lower affinity and retains its ability to bind to cells, indicating that the C-terminal region of B18R contributes to IFN binding. The replication of a VV B18R deletion mutant in tissue culture was restricted in the presence of IFN-alpha, whereas the wild-type virus replicated normally. Binding of soluble recombinant B18R to cells protected the cultures from IFN and allowed VV replication. This represents a novel strategy of virus immune evasion in which secreted IFN-alpha/beta receptors not only bind the soluble cytokine but also bind to uninfected cells and protect them from the antiviral effects of IFN-alpha/beta, maintaining the cells' susceptibility to virus infections. The adaptation of this soluble receptor to block IFN-alpha/beta activity locally will help VV to replicate in the host and spread in tissues. This emphasizes the importance of local effects of IFN-alpha/beta against virus infections.


Subject(s)
Antiviral Agents/pharmacology , Interferons/pharmacology , Receptors, Interferon/physiology , Vaccinia virus/drug effects , Amino Acid Sequence , Animals , Base Sequence , Humans , Interferons/metabolism , Molecular Sequence Data , Open Reading Frames , Rabbits , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Signal Transduction , Vaccinia virus/physiology , Virus Replication
8.
Trends Microbiol ; 8(9): 410-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10989308

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Antibody Formation , Apoptosis , Immunity, Cellular , Interferons , Major Histocompatibility Complex , Receptors, Cytokine
9.
J Virol ; 74(18): 8460-71, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954546

ABSTRACT

The production of secreted proteins that bind cytokines and block their activity has been well characterized as an immune evasion strategy of the orthopoxviruses vaccinia virus (VV) and cowpox virus (CPV). However, very limited information is available on the expression of similar cytokine inhibitors by ectromelia virus (EV), a virulent natural mouse pathogen that causes mousepox. We have characterized the expression and binding properties of three major secreted immunomodulatory activities in 12 EV strains and isolates. Eleven of the 12 EVs expressed a soluble, secreted 35-kDa viral chemokine binding protein with properties similar to those of homologous proteins from VV and CPV. All of the EVs expressed soluble, secreted receptors that bound to mouse, human, and rat tumor necrosis factor alpha. We also detected the expression of a soluble, secreted interleukin-1beta (IL-1beta) receptor (vIL-1betaR) by all of the EVs. EV differed from VV and CPV in that binding of human (125)I-IL-1beta to the EV vIL-1betaR could not be detected. Nevertheless, the EV vIL-1betaR prevented the interaction of human and mouse IL-1beta with cellular receptors. There are significant differences in amino acid sequence between the EV vIL-1betaR and its VV and CPV homologs which may account for the results of the binding studies. The conservation of these activities in EV suggests evolutionary pressure to maintain them in a natural poxvirus infection. Mousepox represents a useful model for the study of poxvirus pathogenesis and immune evasion. These findings will facilitate future study of the role of EV immunomodulatory factors in the pathogenesis of mousepox.


Subject(s)
Chemokines, CC/metabolism , Cytokines/metabolism , Ectromelia virus/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Cell Line , Chemokines, CC/antagonists & inhibitors , Chlorocebus aethiops , Cytokines/antagonists & inhibitors , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Mice , Molecular Sequence Data , Rats , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Sequence Homology, Amino Acid , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/isolation & purification
10.
Mol Med Today ; 6(9): 365-72, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954870

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Virus Diseases/virology , Animals , Antibody Formation , Apoptosis/immunology , Chemokines/immunology , Cytokines/immunology , Forecasting , Humans , Interferons/immunology , Killer Cells, Natural/immunology , Major Histocompatibility Complex/immunology , T-Lymphocytes, Cytotoxic/immunology
11.
Immunol Today ; 21(9): 447-55, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953097

ABSTRACT

During the millions of years they have coexisted with their hosts, viruses have learned how to manipulate host immune control mechanisms. Viral gene functions provide an overview of many relevant principles in cell biology and immunology. Our knowledge of viral gene functions must be integrated into virus-host interaction networks to understand viral pathogenesis, and could lead to new anti-viral strategies and the ability to exploit viral functions as tools in medicine.


Subject(s)
Virus Diseases/immunology , Animals , Apoptosis , Chemokines/physiology , Cytokines/physiology , Histocompatibility Antigens/physiology , Humans , Immune Tolerance , Interferons/physiology , T-Lymphocytes, Cytotoxic/immunology
12.
J Gen Virol ; 81(Pt 5): 1223-30, 2000 May.
Article in English | MEDLINE | ID: mdl-10769064

ABSTRACT

Interleukin-18 (IL-18) is a proinflammatory cytokine that plays a key role in the activation of natural killer and T helper 1 cell responses principally by inducing interferon-gamma (IFN-gamma). Human and mouse secreted IL-18-binding proteins (IL-18BPs) have recently been described which block IL-18 activity but have no sequence similarity to membrane IL-18 receptors. Several poxvirus genes encode proteins with sequence similarity to IL-18BPs. Here we show that vaccinia, ectromelia and cowpox viruses secrete from infected cells a soluble IL-18BP (vIL-18BP) that may modulate the host antiviral response. The ectromelia virus protein was found to block NF-kappaB activation and induction of IFN-gamma in response to IL-18. The highly attenuated vaccinia virus modified virus Ankara encodes IL-18-binding activity, and thus deletion of the vIL-18BP may improve further the safety and immunogenicity of this promising human vaccine candidate. We confirm that molluscum contagiosum virus, a molluscipoxvirus that produces small skin tumours in immunocompetent individuals and opportunistic infections in immunodeficient AIDS patients, also encodes a related, larger vIL-18BP (gene MC54L). This protein may contribute to the lack of inflammatory response characteristic of molluscum contagiosum virus lesions. The expression of vIL-18BPs by distinct poxvirus genera that cause local or general viral dissemination, or persistent or acute infections in the host, emphasizes the importance of IL-18 in response to viral infections.


Subject(s)
Glycoproteins/genetics , Glycoproteins/metabolism , Orthopoxvirus/genetics , Orthopoxvirus/metabolism , Amino Acid Sequence , Animals , Baculoviridae/genetics , Cell Line , Cowpox virus/genetics , Cowpox virus/metabolism , Culture Media , Ectromelia virus/genetics , Ectromelia virus/metabolism , Glycoproteins/chemistry , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molluscum contagiosum virus/genetics , Molluscum contagiosum virus/metabolism , Recombinant Proteins/metabolism , Spleen/cytology , Spleen/metabolism , Vaccinia virus/genetics , Vaccinia virus/metabolism
13.
J Virol ; 74(7): 3404-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10708458

ABSTRACT

We have generated mice lacking the gene for beta interferon and report that they are highly susceptible to vaccinia virus infection. Furthermore, in cultured embryo fibroblasts, viral induction of alpha interferon and of 2-5A synthetase genes is impaired. We also show that beta interferon does not prime its own expression.


Subject(s)
Antiviral Agents/physiology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Animals , Base Sequence , Cells, Cultured , DNA Primers , Genetic Predisposition to Disease , Mice , Mice, Knockout , Virus Diseases/genetics
14.
J Immunol ; 164(5): 2396-404, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10679075

ABSTRACT

The human herpesvirus 6 (HHV-6) U51 gene defines a new family of betaherpesvirus-specific genes encoding multiple transmembrane glycoproteins with similarity to G protein-coupled receptors, in particular, human chemokine receptors. These are distinct from the HHV-6 U12 and HCMV US28 family. In vitro transcription and translation as well as transient cellular expression of U51 showed properties of a multiple transmembrane protein with a 30-kDa monomer as well as high m.w. aggregates or oligomers. Transient cellularly expressed U51 also appeared to form dimeric intermediates. Despite having only limited sequence similarity to chemokine receptors, U51 stably expressed in cell lines showed specific binding of the CC chemokine RANTES and competitive binding with other beta chemokines, such as eotaxin; monocyte chemoattractant protein 1, 3, and 4; as well as the HHV-8 chemokine vMIPII. In epithelial cells already secreting RANTES, U51 expression resulted in specific transcriptional down-regulation. This correlated with reduced secretion of RANTES protein into the culture supernatants. Regulation of RANTES levels may alter selective recruitment of circulating inflammatory cells that the virus can infect and thus could mediate the systemic spread of the virus from initial sites of infection in epithelia. Alternatively, chemokine regulation could modulate a protective inflammatory response to aid the spread of virus by immune evasion. Such mimicry, by viral proteins, of host receptors leading to down-regulation of chemokine expression is a novel immunomodulatory mechanism.


Subject(s)
Chemokine CCL5/metabolism , Chemokines, CC/metabolism , Down-Regulation , Herpesvirus 6, Human/metabolism , Receptors, Chemokine/physiology , Receptors, Virus/physiology , Amino Acid Sequence , Animals , Cell Line , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/biosynthesis , Down-Regulation/genetics , Epithelial Cells/metabolism , Epithelial Cells/virology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Genes, Viral , Herpesvirus 6, Human/genetics , Humans , K562 Cells , Ligands , Molecular Sequence Data , Protein Binding/genetics , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Receptors, Virus/chemistry , Receptors, Virus/genetics , Tumor Cells, Cultured , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/physiology , Viral Structural Proteins/genetics
15.
J Exp Med ; 191(3): 573-8, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10662803

ABSTRACT

Chemokines are a family of small proteins that interact with seven-transmembrane domain receptors and modulate the migration of immune cells into sites of inflammation and infection. The murine gammaherpesvirus 68 M3 gene encodes a secreted 44-kD protein with no sequence similarity to known chemokine receptors. We show that M3 binds a broad range of chemokines, including CC, CXC, C, and CX(3)C chemokines, but does not bind human B cell-specific nor mouse neutrophil-specific CXC chemokines. This herpesvirus chemokine binding protein (hvCKBP) blocks the interaction of chemokines with high-affinity cellular receptors and inhibits chemokine-induced elevation of intracellular calcium levels. hvCKBP is the first soluble chemokine receptor identified in herpesviruses; it represents a novel protein structure with the ability to bind all subfamilies of chemokines in solution and has potential therapeutic applications.


Subject(s)
Gammaherpesvirinae/genetics , Receptors, Chemokine/genetics , Viral Proteins/genetics , Animals , Binding, Competitive , Cell Line , Chemokine CCL4 , Chemokines/pharmacology , Cricetinae , Heparin , Heparitin Sulfate , Humans , Interleukin-8/metabolism , Iodine Radioisotopes , Macrophage Inflammatory Proteins/metabolism , Mice , Open Reading Frames , Protein Binding/drug effects , Receptors, Chemokine/metabolism , Viral Proteins/metabolism
16.
Arch Virol Suppl ; 15: 111-29, 1999.
Article in English | MEDLINE | ID: mdl-10470273

ABSTRACT

Vaccinia virus comprises the live vaccine that was used for vaccination against smallpox. Following the eradication of smallpox, vaccinia virus was developed as an expression vector that is now used widely in biological research and vaccine development. In recent years vaccinia virus and other poxviruses have been found to express a collection of proteins that block parts of the host response to infection. Some of these proteins are secreted from the infected cell where they bind and neutralise host cytokines, chemokines and interferons (IFN). In this paper three such proteins that bind interleukin (IL)-1 beta, type I IFNs and CC chemokines are described. The study of these immunomodulatory molecules is enhancing our understanding of virus pathogenesis, yielding fundamental information about the immune system, and providing new molecules that have potential application for the treatment of immunological disorders or infectious diseases.


Subject(s)
Vaccinia virus/metabolism , Vaccinia/immunology , Viral Proteins/immunology , Viral Proteins/metabolism , Animals , Chemokines, CC/metabolism , Humans , Interferon Type I/metabolism , Interleukin-1/metabolism , Neutralization Tests , Protein Binding , Vaccinia/virology , Vaccinia virus/growth & development , Vaccinia virus/immunology
17.
J Gen Virol ; 79 ( Pt 5): 1159-67, 1998 May.
Article in English | MEDLINE | ID: mdl-9603331

ABSTRACT

Modified virus Ankara (MVA) is a vaccinia virus (VV) strain that was attenuated by serial passage through chick embryo fibroblasts (CEFs) and contains six large genomic deletions compared with parental virus. MVA replicates well in CEFs, but poorly in most mammalian cells. Recombinant MVA is a promising human vaccine candidate due to its restricted host range, immunogenicity and avirulence in animal models, and excellent safety record as a smallpox vaccine. Here we present a further characterization of MVA and demonstrate that: (i) MVA can replicate, albeit poorly, in transformed human cell lines, but not in primary human fibroblasts although there is limited cell-to-cell spread; (ii) MVA is a potent inducer of type I interferon (IFN) from primary human cells, which may restrict virus spread in vivo; and (iii) unlike other VV strains, MVA does not express soluble receptors for IFN-gamma, IFN-alpha/beta, tumour necrosis factor and CC chemokines, but does express a soluble interleukin-1beta receptor. This provides a plausible and testable explanation for the good immunogenicity of MVA despite its poor replication in mammals. The implications of these findings for the use of MVA as a safe and immunogenic human vaccine candidate are discussed.


Subject(s)
Receptors, Cytokine/metabolism , Vaccinia virus/immunology , Vaccinia virus/physiology , Viral Vaccines , Virus Replication , Amino Acid Sequence , Animals , Cell Line , Cell Line, Transformed , Cell Transformation, Viral , Cells, Cultured , HeLa Cells , Humans , Interferon Type I/immunology , Membrane Proteins , Molecular Sequence Data , Rabbits , Receptor, Interferon alpha-beta , Receptors, Interferon/metabolism , Vaccinia virus/metabolism , Interferon gamma Receptor
18.
J Immunol ; 160(2): 624-33, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9551896

ABSTRACT

Chemokines direct migration of immune cells into sites of inflammation and infection. Chemokine receptors are seven-transmembrane domain proteins that, in contrast to other cytokine receptors, cannot be easily engineered as soluble chemokine inhibitors. Poxviruses encode several soluble cytokine receptors to evade immune surveillance, providing new strategies for immune modulation. Here we show that vaccinia virus and other orthopoxviruses (cowpox and camelpox) express a secreted 35-kDa chemokine binding protein (vCKBP) with no sequence similarity to known cellular chemokine receptors. The vCKBP binds CC, but not CXC or C, chemokines with high affinity (Kd = 0.1-15 nM for different CC chemokines), blocks the interaction of chemokines with cellular receptors, and inhibits chemokine-induced elevation of intracellular calcium levels and cell migration in vitro, thus representing a soluble inhibitor that binds and sequesters chemokines. The potential of vCKBP as a therapeutic agent in vivo was illustrated in a guinea pig skin model by the blockade of eotaxin-induced eosinophil infiltration. a feature of allergic inflammatory reactions. Furthermore, vCKBP may enable the rational design of antagonists to neutralize pathogens that use chemokine receptors to initiate infection, such as HIV or the malarial parasite.


Subject(s)
Carrier Proteins/physiology , Chemokines/antagonists & inhibitors , Chemokines/metabolism , Vaccinia virus/immunology , Viral Proteins/physiology , Animals , Binding Sites/drug effects , Binding, Competitive , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemokine CCL11 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/metabolism , Chemotactic Factors, Eosinophil/antagonists & inhibitors , Cowpox virus/genetics , Cowpox virus/immunology , Cytokines/antagonists & inhibitors , Guinea Pigs , Humans , Molecular Weight , Orthopoxvirus/genetics , Orthopoxvirus/immunology , Proteoglycans/metabolism , Receptors, Chemokine/metabolism , Skin Window Technique , Solubility , Vaccinia virus/genetics , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Virology ; 232(1): 86-90, 1997 May 26.
Article in English | MEDLINE | ID: mdl-9185591

ABSTRACT

The soluble B18R protein coded by vaccinia virus exerts properties of a type I interferon (IFN)-receptor with broad species specificity. We analyzed neutralizing and binding activity of the B18R protein against several recombinant human type I IFNs. The B18R protein inhibited the antiviral potency of IFN-alpha1, IFN-alpha2, IFN-alpha8/1/8, and IFN-omega on human cells. The N-terminal domain of human type I IFN is involved in the high affinity binding to its cellular receptor. To localize the binding domain(s) of IFN with the B18R protein, competition experiments between B18R, and mapped monoclonal antibodies to IFN-alpha1 and IFN-alpha2 were performed. Surprisingly, our data indicated that the contact area between the B18R protein and IFN comprised in addition to the N-terminal region of IFN-molecule also its C-terminal portion. We suggest that this different pattern of interaction with a ligand might determine the ability of B18R protein to bind type I IFNs of different species.


Subject(s)
Interferon-alpha/metabolism , Receptors, Interferon/metabolism , Vaccinia virus/genetics , Viral Proteins/metabolism , Antiviral Agents/antagonists & inhibitors , Cell Line , Humans , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/physiology , Membrane Proteins , Neutralization Tests , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics
20.
J Gen Virol ; 78 ( Pt 3): 677-85, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9049422

ABSTRACT

The vaccinia virus (VV) strain Western Reserve B13R gene encodes a 38.5 kDa intracellular polypeptide that is non-essential for virus replication in vitro and does not affect virus virulence in a murine intranasal model. The protein has 92% amino acid identity with the cowpox virus cytokine response modifier A (crmA) protein which inhibits the interleukin (IL)-1beta converting enzyme (ICE). Here, we show that extracts from THP-1 cells infected with VV strains expressing B13R prevent the cleavage of in vitro transcribed and translated pro-IL-1beta into mature IL-1beta. Similarly, THP-1 cells infected with VVs expressing B13R process pro-IL-1beta into mature IL-1beta inefficiently in situ. Despite its inhibition of ICE, B13R does not prevent fever in infected mice, a systemic effect mediated by IL-1beta. Instead, fever is controlled by the VV IL-1beta receptor, encoded by gene B15R, and deletion of both the B13R and B15R genes did not increase the febrile response compared to deletion of B15R alone. The B13R protein does, however, block apoptosis mediated by anti-Fas antibodies or by tumour necrosis factor (TNF) and cycloheximide. Using DNA fragmentation, chromium release and microscopic analyses it was shown that cells infected with wild-type VV strain WR, or a revertant virus in which the B13R gene had been re-inserted into the B13R deletion mutant, are more resistant than uninfected cells or deletion mutant-infected cells to apoptosis mediated by anti-Fas and TNF.


Subject(s)
Antigens, Viral/immunology , Apoptosis/immunology , Cysteine Endopeptidases/immunology , Fever/immunology , Serpins/immunology , Tumor Necrosis Factor-alpha/pharmacology , Vaccinia virus/immunology , fas Receptor/metabolism , Animals , Antigens, Viral/genetics , Caspase 1 , Cysteine Endopeptidases/metabolism , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Serpins/genetics , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...