Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
mSphere ; 8(2): e0006223, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36920219

ABSTRACT

The ongoing worldwide monkeypox outbreak is caused by viral lineages (globally referred to as hMPXV1) that are related to but distinct from clade IIb MPXV viruses transmitted within Nigeria. Analysis of the genetic differences has indicated that APOBEC-mediated editing might be responsible for the unexpectedly high number of mutations observed in hMPXV1 genomes. Here, using 1,624 publicly available hMPXV1 sequences, we analyzed the mutations that accrued between 2017 and the emergence of the current predominant variant (B.1), as well as those that that have been accumulating during the 2022 outbreak. We confirmed an overwhelming prevalence of C-to-T and G-to-A mutations, with a sequence context (5'-TC-3') consistent with the preferences of several human APOBEC3 enzymes. We also found that mutations preferentially occur in highly expressed viral genes, although no transcriptional asymmetry was observed. A comparison of the mutation spectrum and context was also performed against the human-specific variola virus (VARV) and the zoonotic cowpox virus (CPXV), as well as fowlpox virus (FWPV). The results indicated that in VARV genomes, C-to-T and G-to-A changes were more common than the opposite substitutions, although the effect was less marked than for hMPXV1. Conversely, no preference toward C-to-T and G-to-A changes was observed in CPXV and FWPV. Consistently, the sequence context of C-to-T changes confirmed a preference for a T in the -1 position for VARV, but not for CPXV or FWPV. Overall, our results strongly support the view that, irrespective of the transmission route, orthopoxviruses infecting humans are edited by the host APOBEC3 enzymes. IMPORTANCE Analysis of the viral lineages responsible for the 2022 monkeypox outbreak suggested that APOBEC enzymes are driving hMPXV1 evolution. Using 1,624 public sequences, we analyzed the mutations that accumulated between 2017 and the emergence of the predominant variant and those that characterize the last outbreak. We found that the mutation spectrum of hMPXV1 has been dominated by TC-to-TT and GA-to-AA changes, consistent with the editing activity of human APOBEC3 proteins. We also found that mutations preferentially affect highly expressed viral genes, possibly because transcription exposes single-stranded DNA (ssDNA), a target of APOBEC3 editing. Notably, analysis of the human-specific variola virus (VARV) and the zoonotic cowpox virus (CPXV) indicated that in VARV genomes, TC-to-TT and GA-to-AA changes are likewise extremely frequent. Conversely, no preference toward TC-to-TT and GA-to-AA changes is observed in CPXV. These results suggest that APOBEC3 proteins have an impact on the evolution of different human-infecting orthopoxviruses.


Subject(s)
Mpox (monkeypox) , Orthopoxvirus , Smallpox , Variola virus , Animals , Humans , Orthopoxvirus/genetics , Cowpox virus/genetics , Cowpox virus/metabolism , Mutation , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism
2.
Int J Mol Sci ; 24(2)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36675269

ABSTRACT

Cowpox is caused by a DNA virus known as the cowpox virus (CPXV) belonging to the Orthopoxvirus genus in the family Poxviridae. Cowpox is a zoonotic disease with the broadest host range among the known poxviruses. The natural reservoir hosts of CPXV are wild rodents. Recently, the cases of orthopoxviral infections have been increasing worldwide, and cowpox is considered the most common orthopoxviral infection in Europe. Cowpox is often a self-limiting disease, although cidofovir or anti-vaccinia gammaglobulin can be used in severe and disseminated cases of human cowpox. In this computational study, a molecular docking analysis of thymine- and arabinofuranosyl-thymine-related structures (1-21) on two cowpox-encoded proteins was performed with respect to the cidofovir standard and a 3D ligand-based pharmacophore model was generated. Three chemical structures (PubChem IDs: 123370001, 154137224, and 90413364) were identified as potential candidates for anti-cowpox agents. Further studies combining in vitro and in silico molecular dynamics simulations to test the stability of these promising compounds could effectively improve the future design of cowpox virus inhibitors, as molecular docking studies are not sufficient to consider a ligand a potential drug.


Subject(s)
Cowpox virus , Cowpox , Animals , Humans , Cowpox virus/genetics , Cowpox virus/metabolism , Thymine/metabolism , Cidofovir/pharmacology , Ligands , Molecular Docking Simulation , Rodentia
3.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31645446

ABSTRACT

Cowpox virus (CPXV) is a zoonotic orthopoxvirus (OPV) that causes spillover infections from its animal hosts to humans. In 2009, several human CPXV cases occurred through transmission from pet rats. An isolate from a diseased rat, RatPox09, exhibited significantly increased virulence in Wistar rats and caused high mortality compared to that caused by the mildly virulent laboratory strain Brighton Red (BR). The RatPox09 genome encodes four genes which are absent in the BR genome. We hypothesized that their gene products could be major factors influencing the high virulence of RatPox09. To address this hypothesis, we employed several BR-RatPox09 chimeric viruses. Using Red-mediated mutagenesis, we generated BR-based knock-in mutants with single or multiple insertions of the respective RatPox09 genes. High-throughput sequencing was used to verify the genomic integrity of all recombinant viruses, and transcriptomic analyses confirmed that the expression profiles of the genes that were adjacent to the modified ones were unaltered. While the in vitro growth kinetics were comparable to those of BR and RatPox09, we discovered that a knock-in BR mutant containing the four RatPox09-specific genes was as virulent as the RatPox09 isolate, causing death in over 75% of infected Wistar rats. Unexpectedly, the insertion of gCPXV0030 (g7tGP) alone into the BR genome resulted in significantly higher clinical scores and lower survival rates matching the rate for rats infected with RatPox09. The insertion of gCPXV0284, encoding the BTB (broad-complex, tramtrack, and bric-à-brac) domain protein D7L, also increased the virulence of BR, while the other two open reading frames failed to rescue virulence independently. In summary, our results confirmed our hypothesis that a relatively small set of four genes can contribute significantly to CPXV virulence in the natural rat animal model.IMPORTANCE With the cessation of vaccination against smallpox and its assumed cross-protectivity against other OPV infections, waning immunity could open up new niches for related poxviruses. Therefore, the identification of virulence mechanisms in CPXV is of general interest. Here, we aimed to identify virulence markers in an experimental rodent CPXV infection model using bacterial artificial chromosome (BAC)-based virus recombineering. We focused our work on the recent zoonotic CPXV isolate RatPox09, which is highly pathogenic in Wistar rats, unlike the avirulent BR reference strain. In several animal studies, we were able to identify a novel set of CPXV virulence genes. Two of the identified virulence genes, encoding a putative BTB/POZ protein (CPXVD7L) and a B22R-family protein (CPXV7tGP), respectively, have not yet been described to be involved in CPXV virulence. Our results also show that single genes can significantly affect virulence, thus facilitating adaptation to other hosts.


Subject(s)
Cowpox virus , Genome, Viral , Mutation , Animals , Chlorocebus aethiops , Cowpox/genetics , Cowpox/metabolism , Cowpox virus/genetics , Cowpox virus/metabolism , Cowpox virus/pathogenicity , Humans , Mutagenesis , Rats , Rats, Wistar , Vero Cells
4.
Biochem J ; 476(9): 1335-1357, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30992316

ABSTRACT

Poxviruses encode many proteins that enable them to evade host anti-viral defense mechanisms. Spi-2 proteins, including Cowpox virus CrmA, suppress anti-viral immune responses and contribute to poxviral pathogenesis and lethality. These proteins are 'serpin' protease inhibitors, which function via a pseudosubstrate mechanism involving initial interactions between the protease and a cleavage site within the serpin. A conformational change within the serpin interrupts the cleavage reaction, deforming the protease active site and preventing dissociation. Spi-2 proteins like CrmA potently inhibit caspases-1, -4 and -5, which produce proinflammatory cytokines, and caspase-8, which facilitates cytotoxic lymphocyte-mediated target cell death. It is not clear whether both of these functions are equally perilous for the virus, or whether only one must be suppressed for poxviral infectivity and spread but the other is coincidently inhibited merely because these caspases are biochemically similar. We compared the caspase specificity of CrmA to three orthologs from orthopoxviruses and four from more distant chordopoxviruses. All potently blocked caspases-1, -4, -5 and -8 activity but exhibited negligible inhibition of caspases-2, -3 and -6. The orthologs differed markedly in their propensity to inhibit non-mammalian caspases. We determined the specificity of CrmA mutants bearing various residues in positions P4, P3 and P2 of the cleavage site. Almost all variants retained the ability to inhibit caspase-1, but many lacked caspase-8 inhibitory activity. The retention of Spi-2 proteins' caspase-8 specificity during chordopoxvirus evolution, despite this function being readily lost through cleavage site mutagenesis, suggests that caspase-8 inhibition is crucial for poxviral pathogenesis and spread.


Subject(s)
Caspase 1 , Caspase 8 , Cowpox virus , Proteolysis , Serpins , Viral Proteins , Caspase 1/chemistry , Caspase 1/genetics , Caspase 1/metabolism , Caspase 8/chemistry , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Cowpox virus/chemistry , Cowpox virus/genetics , Cowpox virus/metabolism , Humans , Mutagenesis, Site-Directed , Serpins/chemistry , Serpins/genetics , Serpins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Immunol Cell Biol ; 96(2): 137-148, 2018 02.
Article in English | MEDLINE | ID: mdl-29363167

ABSTRACT

Viruses may interfere with the MHC class I antigen presentation pathway in order to avoid CD8+ T cell-mediated immunity. A key target within this pathway is the peptide transporter TAP. This transporter plays a central role in MHC class I-mediated peptide presentation of endogenous antigens. In addition, TAP plays a role in antigen cross-presentation of exogenously derived antigens by dendritic cells (DCs). In this study, a soluble form of the cowpox virus TAP inhibitor CPXV012 is synthesized for exogenous delivery into the antigen cross-presentation route of human monocyte-derived (mo)DCs. We show that soluble CPXV012 localizes to TAP+ compartments that carry internalized antigen and is a potent inhibitor of antigen cross-presentation. CPXV012 stimulates the prolonged deposition of antigen fragments in storage compartments of moDCs, as a result of reduced endosomal acidification and reduced antigen proteolysis when soluble CPXV012 is present. Thus, a dual function can be proposed for CPXV012: inhibition of TAP-mediated peptide transport and inhibition of endosomal antigen degradation. We propose this second function for soluble CPXV012 can serve to interfere with antigen cross-presentation in a peptide transport-independent manner.


Subject(s)
Antigen Presentation/immunology , Cowpox virus/metabolism , Cross-Priming/immunology , Dendritic Cells/immunology , Endocytosis , Monocytes/cytology , Viral Proteins/metabolism , Amino Acid Sequence , Endosomes/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Peptides/metabolism , Protein Domains , Solubility , Viral Proteins/chemistry
6.
J Biol Chem ; 290(26): 15973-84, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940088

ABSTRACT

The blockade of tumor necrosis factor (TNF) by etanercept, a soluble version of the human TNF receptor 2 (hTNFR2), is a well established strategy to inhibit adverse TNF-mediated inflammatory responses in the clinic. A similar strategy is employed by poxviruses, encoding four viral TNF decoy receptor homologues (vTNFRs) named cytokine response modifier B (CrmB), CrmC, CrmD, and CrmE. These vTNFRs are differentially expressed by poxviral species, suggesting distinct immunomodulatory properties. Whereas the human variola virus and mouse ectromelia virus encode one vTNFR, the broad host range cowpox virus encodes all vTNFRs. We report the first comprehensive study of the functional and binding properties of these four vTNFRs, providing an explanation for their expression profile among different poxviruses. In addition, the vTNFRs activities were compared with the hTNFR2 used in the clinic. Interestingly, CrmB from variola virus, the causative agent of smallpox, is the most potent TNFR of those tested here including hTNFR2. Furthermore, we demonstrate a new immunomodulatory activity of vTNFRs, showing that CrmB and CrmD also inhibit the activity of lymphotoxin ß. Similarly, we report for the first time that the hTNFR2 blocks the biological activity of lymphotoxin ß. The characterization of vTNFRs optimized during virus-host evolution to modulate the host immune response provides relevant information about their potential role in pathogenesis and may be used to improve anti-inflammatory therapies based on soluble decoy TNFRs.


Subject(s)
Cowpox virus/metabolism , Poxviridae/metabolism , Receptors, Tumor Necrosis Factor, Type II/chemistry , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor Decoy Receptors/chemistry , Tumor Necrosis Factor Decoy Receptors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cowpox virus/chemistry , Cowpox virus/genetics , Humans , Lymphotoxin-beta/metabolism , Mice , Molecular Sequence Data , Poxviridae/chemistry , Poxviridae/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Sequence Alignment , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factors/metabolism , Viral Proteins/genetics
7.
Mol Immunol ; 55(2): 139-42, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23141382

ABSTRACT

Following primary infection, herpesviruses persist for life in their hosts, even when vigorous anti-viral immunity has been induced. Failure of the host immune system to eliminate infected cells is facilitated by highly effective immune evasion strategies acquired by these herpesviruses during millions of years of co-evolution with their hosts. Here, we review the mechanisms of action of viral gene products that lead to cytotoxic T cell evasion through interference with the function of the transporter associated with antigen processing, TAP. The viral TAP inhibitors impede transport of peptides from the cytosol into the ER lumen, thereby preventing peptide loading onto MHC class I complexes. Recent insights have revealed a pattern of functional convergent evolution. In every herpesvirus subfamily, inhibitors of TAP function have been identified that are, surprisingly, unrelated in genome location, structure, and mechanism of action. Recently, cowpox virus has also been found to encode a TAP inhibitor. Expanding our knowledge on how viruses perturb antigen presentation, in particular by targeting TAP, not only provides information on viral pathogenesis, but also reveals novel aspects of the cellular processes corrupted by these viruses, notably the translocation of peptides by the ATP-binding cassette (ABC) transporter TAP. As the various TAP inhibitors are anticipated to impede discrete conformational transitions it is expected that crystal structures of TAP-inhibitor complexes will reveal valuable structural information on the actual mechanism of peptide translocation by TAP. Viral TAP inhibitors are also used for various (clinical) applications, for example, as effective tools in antigen presentation studies and as immunomodulators in immunotherapy for cancer, heterologous vaccination, and transplant protection.


Subject(s)
Antigen Presentation , Herpesviridae/immunology , Herpesviridae/pathogenicity , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Cowpox virus/genetics , Cowpox virus/immunology , Cowpox virus/metabolism , Herpesviridae/genetics , Herpesviridae/metabolism , Humans , Immune Evasion , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/immunology , Viral Proteins/metabolism
8.
IDrugs ; 13(10): 680-2, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20878585

ABSTRACT

The World Congress of Virus and Infections, held in Busan, South Korea, included topics reviewing the field of zoonoses. This conference report highlights selected presentations on surveillance, epidemiology and measures for the control and prevention of zoonotic diseases. Topics discussed include human factors influencing zoonoses, the molecular epidemiology of Crimean-Congo hemorrhagic fever, the emerging Nipah virus, and the re-emergence of cowpox virus.


Subject(s)
Communicable Diseases, Emerging , Zoonoses , Animals , Communicable Diseases, Emerging/epidemiology , Cowpox virus/metabolism , Cowpox virus/pathogenicity , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean/epidemiology , Hemorrhagic Fever, Crimean/prevention & control , Hemorrhagic Fever, Crimean/virology , Humans , Nipah Virus/genetics , Nipah Virus/immunology , Nipah Virus/metabolism , Risk Factors , Zoonoses/epidemiology , Zoonoses/transmission , Zoonoses/virology
9.
Mol Biol (Mosk) ; 44(6): 1054-63, 2010.
Article in Russian | MEDLINE | ID: mdl-21290827

ABSTRACT

Orthopoxviruses bear in their genomes several genes coding for homologous secreted proteins able to bind tumor necrosis factor. Different species of the genus possess different sets of these tumor necrosis factor-binding proteins. Viriola virus encodes the only one of them named CrmB. Despite sharing high sequence identity, CrmB proteins belonging to distinct orthopoxviral species were shown to significantly differ by their physico-chemical and biological properties. We modeled spatial structures of tumor necrosis factor receptor domains of variola and cowpox virus CrmB proteins bound to either murine, or human or mutated human tumor necrosis factor. In the sequence of last the arginine residue at position 31 is substituted with glutamine that is characteristic for murine tumor necrosis factor. Theoretical analysis of modeled ligand-receptor complexes revealed that the least stable should be the complex of cowpox virus CrmB with human tumor necrosis factor, and that arginine to glutamine substitution at position 31 should significantly stabilize binding of corresponding human tumor necrosis factor mutant to cowpox virus CrmB. Experimental evaluation of recombinant variola and cowpox virus CrmB efficiencies in inhibiting cytotoxic effect of all these tumor necrosis factors have approved our predictions.


Subject(s)
Cowpox virus/metabolism , Models, Molecular , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factors/metabolism , Variola virus/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Arginine/genetics , Cowpox virus/genetics , Glutamine/genetics , Humans , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/chemistry , Variola virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
10.
PLoS One ; 4(9): e6883, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19727399

ABSTRACT

BACKGROUND: Often described as an extremely rare zoonosis, cowpox virus (CPXV) infections are on the increase in Germany. CPXV is rodent-borne with a broad host range and contains the largest and most complete genome of all poxviruses, including parts with high homology to variola virus (smallpox). So far, most CPXV cases have occurred individually in unvaccinated animals and humans and were caused by genetically distinguishable virus strains. METHODOLOGY/PRINCIPAL FINDINGS: Generalized CPXV infections in banded mongooses (Mungos mungo) and jaguarundis (Herpailurus yagouaroundi) at a Zoological Garden were observed with a prevalence of the affected animal group of 100% and a mortality of 30%. A subsequent serological investigation of other exotic animal species provided evidence of subclinical cases before the onset of the outbreak. Moreover, a time-delayed human cowpox virus infection caused by the identical virus strain occurred in a different geographical area indicating that handling/feeding food rats might be the common source of infection. CONCLUSIONS/SIGNIFICANCE: Reports on the increased zoonotic transmission of orthopoxviruses have renewed interest in understanding interactions between these viruses and their hosts. The list of animals known to be susceptible to CPXV is still growing. Thus, the likely existence of unknown CPXV hosts and their distribution may present a risk for other exotic animals but also for the general public, as was shown in this outbreak. Animal breeders and suppliers of food rats represent potential multipliers and distributors of CPXV, in the context of increasingly pan-European trading. Taking the cessation of vaccination against smallpox into account, this situation contributes to the increased incidence of CPXV infections in man, particularly in younger age groups, with more complicated courses of clinical infections.


Subject(s)
Cowpox virus/metabolism , Cowpox/epidemiology , Animals , Cowpox/transmission , Disease Outbreaks , Felidae , Female , Herpestidae , Humans , Male , Microscopy, Electron , Phylogeny , Polymerase Chain Reaction , Rats , Risk , Time Factors , Zoonoses/epidemiology , Zoonoses/transmission
11.
Antivir Ther ; 13(3): 357-68, 2008.
Article in English | MEDLINE | ID: mdl-18572748

ABSTRACT

BACKGROUND: Concerns about the potential use of smallpox in bioterrorism have stimulated interest in the development of novel antiviral treatments. Currently, there are no effective therapies against smallpox and new treatment strategies are greatly needed. METHODS: In this study, specifically designed small interfering RNAs (siRNAs), targeting five proteins essential for orthopoxvirus replication, were investigated for their ability to inhibit vaccinia virus strain Western Reserve (VACVWR) replication. RESULTS: Among these siRNAs, 100 nM siD5R-2, an siRNA targeting the D5 protein, decreased VACVWR replication up to 90% when used either prophylactically or therapeutically in human lung carcinoma A549 cells. This siRNA induced a striking concentration-dependent inhibition of VACVWR replication and a prolonged prophylactic antiviral effect that lasted for 72 h, at a concentration of 100 nM. Confocal microscopy of Alexa-siD5R-2-treated VACVWR-infected cells confirmed a decrease in viral replication. Furthermore, siD5R-2 was shown to specifically reduce the D5R mRNA and protein expression using real-time reverse transcriptase-PCR and western blotting analysis, without inducing interferon-13 in A549 cells. We also demonstrated the antiviral potency of siD5R-2 against different pathogenic orthopoxviruses, such as cowpox and monkeypox viruses, which were inhibited up to 70% at the lowest concentration (1 nM) tested. Finally, siD5R-2 showed antiviral effects in VACVWR-infected human keratinocyte and fibroblast cell cultures. CONCLUSIONS: These results suggest that siD5R-2 could be a potential candidate to treat poxvirus infections.


Subject(s)
Gene Expression Regulation, Viral , Genetic Therapy/methods , Orthopoxvirus/genetics , Poxviridae Infections/therapy , RNA Interference , RNA, Small Interfering/metabolism , Viral Proteins/genetics , Virus Replication/genetics , Blotting, Western , Cell Line, Tumor , Cowpox virus/genetics , Cowpox virus/metabolism , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/virology , Fluorescent Antibody Technique , Humans , Interferon-beta/metabolism , Keratinocytes/metabolism , Keratinocytes/virology , Monkeypox virus/genetics , Monkeypox virus/metabolism , Orthopoxvirus/metabolism , Poxviridae Infections/metabolism , Poxviridae Infections/virology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/metabolism
12.
Bull Exp Biol Med ; 141(4): 448-52, 2006 Apr.
Article in English | MEDLINE | ID: mdl-17152368

ABSTRACT

The mechanisms of infection development in intraperitoneal inoculation of mice by ectromelia virus strain K-1 and cowpox strain EP-2 were studied. Ultrastructural parameters of virus assembly and maturation are described. Differences in the types of cells replicating the viruses and in the type of visceral injuries were detected. The studies showed a local type of strain EP-2 cowpox infection and dissemination of ectromelia strain K-1.


Subject(s)
Orthopoxvirus/metabolism , Viscera/pathology , Viscera/virology , Animals , Apoptosis , Cowpox virus/metabolism , Cytoplasm/metabolism , Fibroblasts/ultrastructure , Lymphocytes/pathology , Lymphocytes/ultrastructure , Macrophages/virology , Male , Mice , Peritoneum/ultrastructure , Poxviridae Infections/virology , Spleen/ultrastructure
13.
Biochim Biophys Acta ; 1764(11): 1710-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17070121

ABSTRACT

Tumor necrosis factor (TNF), a potent proinflammatory and antiviral cytokine, is a critical extracellular immune regulator targeted by poxviruses through the activity of virus-encoded family of TNF-binding proteins (CrmB, CrmC, CrmD, and CrmE). The only TNF-binding protein from variola virus (VARV), the causative agent of smallpox, infecting exclusively humans, is CrmB. Here we have aligned the amino acid sequences of CrmB proteins from 10 VARV, 14 cowpox virus (CPXV), and 22 monkeypox virus (MPXV) strains. Sequence analyses demonstrated a high homology of these proteins. The regions homologous to cd00185 domain of the TNF receptor family, determining the specificity of ligand-receptor binding, were found in the sequences of CrmB proteins. In addition, a comparative analysis of the C-terminal SECRET domain sequences of CrmB proteins was performed. The differences in the amino acid sequences of these domains characteristic of each particular orthopoxvirus species were detected. It was assumed that the species-specific distinctions between the CrmB proteins might underlie the differences in these physicochemical and biological properties. The individual recombinant proteins VARV-CrmB, MPXV-CrmB, and CPXV-CrmB were synthesized in a baculovirus expression system in insect cells and isolated. Purified VARV-CrmB was detectable as a dimer with a molecular weight of 90 kDa, while MPXV- and CPXV-CrmBs, as monomers when fractioned by non-reducing SDS-PAGE. The CrmB proteins of VARV, MPXV, and CPXV differed in the efficiencies of inhibition of the cytotoxic effects of human, mouse, or rabbit TNFs in L929 mouse fibroblast cell line. Testing of CrmBs in the experimental model of LPS-induced shock using SPF BALB/c mice detected a pronounced protective effect of VARV-CrmB. Thus, our data demonstrated the difference in anti-TNF activities of VARV-, MPXV-, and CPXV-CrmBs and efficiency of VARV-CrmB rather than CPXV- or MPXV-CrmBs against LPS-induced mortality in mice.


Subject(s)
Cowpox virus/metabolism , Monkeypox virus/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Variola virus/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Mice , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spodoptera , Viral Proteins/chemistry
14.
Virology ; 329(1): 199-212, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15476887

ABSTRACT

Host restriction of vaccinia virus has been previously described in CHO and RK13 cells in which a cowpox virus CP77 gene rescues vaccinia virus growth at the viral protein translation level. Here we investigate the restrictive stage of vaccinia virus in HeLa cells using a vaccinia mutant virus (VV-hr) that contains a deletion of 18-kb genome sequences resulting in no growth in HeLa cells. Insertion of CP77 gene into VV-hr generated a recombinant virus (VV-36hr) that multiplied well in HeLa cells. Both viruses could enter cells, initiate viral DNA replication and intermediate gene transcription. However, translation of viral intermediate gene was only detected in cells infected with VV-36hr, indicating that CP77 relieves host restriction at the intermediate gene translation stage in HeLa cells. Caspase-2 and -3 activation was observed in HeLa cells infected with VV-hr coupled with dramatic morphological alterations and cleavage of the translation initiation factor eIF4G. Caspase activation was reduced in HeLa cells infected with VV-36hr, indicating that CP77 acts upstream of caspase activation. Enhanced phosphorylation of PKR and eIF2alpha was also observed in cells infected with VV-hr and was suppressed by CP77. Suppression of eIF4G cleavage with the caspase inhibitor ZVAD did not rescue virus translation, whereas expression of a mutant eIF2alpha protein with an alanine substitution of serine at amino acid position 51 (eIF2alphaS51A) partially restored viral translation and moderately increased virus growth in HeLa cells.


Subject(s)
Cowpox virus/pathogenicity , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Viral , Protein Biosynthesis , Vaccinia virus/metabolism , Viral Proteins/genetics , Animals , Apoptosis , CHO Cells , Cowpox virus/genetics , Cowpox virus/metabolism , Cricetinae , HeLa Cells , Humans , Mutation , Phosphorylation , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Proteins/metabolism
15.
J Virol ; 76(22): 11216-25, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388681

ABSTRACT

The orthopoxvirus gene p4c has been identified in the genome of the vaccinia virus strain Western Reserve. This gene encodes the 58-kDa structural protein P4c present on the surfaces of the intracellular mature virus (IMV) particles. The gene is disrupted in the genome of cowpox virus Brighton Red (BR), demonstrating that although the P4c protein may be advantageous for virus replication in vivo, it is not essential for virus replication in vitro. Complementation and recombination analyses with the p4c gene have shown that the P4c protein is required to direct the IMV into the A-type inclusions (ATIs) produced by cowpox virus BR. The p4c gene is highly conserved among most members of the orthopoxvirus genus, including viruses that produce ATIs, such as cowpox, ectromelia, and raccoonpox viruses, as well as those such as variola, monkeypox, vaccinia, and camelpox viruses, which do not. The conservation of the p4c gene among the orthopoxviruses, irrespective of their capacities to produce ATIs, suggests that the P4c protein provides functions in addition to that of directing IMV into ATIs. These findings, and the presence of the P4c protein in IMV but not extracellular enveloped virus (D. Ulaeto, D. Grosenbach, and D. E. Hruby, J. Virol. 70:3372-3377, 1996), suggest a model in which the P4c protein may play a role in the retrograde movement of IMV particles, thereby contributing to the retention of IMV particles within the cytoplasm and within ATIs when they are present. In this way, the P4c protein may affect both viral morphogenesis and processes of virus dissemination.


Subject(s)
Gene Expression Regulation, Viral , Inclusion Bodies, Viral/metabolism , Orthopoxvirus/metabolism , Viral Structural Proteins/genetics , Virion/metabolism , Amino Acid Sequence , Animals , Cell Line/ultrastructure , Cowpox virus/genetics , Cowpox virus/metabolism , Cowpox virus/ultrastructure , Genetic Complementation Test , HeLa Cells/ultrastructure , Humans , Mice , Microscopy, Electron , Molecular Sequence Data , Orthopoxvirus/genetics , Orthopoxvirus/ultrastructure , Recombination, Genetic , Vaccinia virus/genetics , Vaccinia virus/metabolism , Vaccinia virus/ultrastructure , Viral Proteins/metabolism , Viral Structural Proteins/chemistry , Viral Structural Proteins/metabolism
16.
Antiviral Res ; 54(3): 129-42, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12062386

ABSTRACT

The Brighton strain of cowpox virus causes lethal bronchopneumonia when delivered as a small-particle (1 microm) aerosol to weanling BALB/c mice. We showed previously that this disease can be prevented or cured with one subcutaneous injection of cidofovir (HPMPC, Vistide). To determine whether even better results could be obtained by delivering the drug directly to the respiratory tract, we administered cidofovir by small-particle aqueous aerosol before or after aerosolized cowpox infection. In a series of five experiments, aerosol doses of 0.5-5 mg/kg were always more effective than 25 mg/kg and sometimes more effective than 100 mg/kg injected subcutaneously, as measured by changes in body and lung weight, lung viral titers, pulmonary pathology and survival. A cyclic analog ((1-[(S)-2-hydroxy-2-oxo-1,4,2-dioxaphosphorinan-5-yl)methyl] cytosine) (cHPMPC) was less protective. The results suggest that aerosolized cidofovir would be effective for prophylaxis or early post-exposure therapy of human smallpox or monkeypox virus infection.


Subject(s)
Antiviral Agents/pharmacology , Cowpox virus/growth & development , Cowpox/drug therapy , Cytosine/pharmacology , Organophosphonates , Organophosphorus Compounds/pharmacology , Administration, Inhalation , Aerosols , Animals , Antiviral Agents/administration & dosage , Body Weight , Bronchopneumonia/drug therapy , Bronchopneumonia/prevention & control , Bronchopneumonia/virology , Cidofovir , Cowpox/prevention & control , Cowpox/virology , Cowpox virus/metabolism , Cytosine/administration & dosage , Cytosine/analogs & derivatives , Female , Injections, Subcutaneous , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Organ Size , Organophosphorus Compounds/administration & dosage , Statistics, Nonparametric
17.
Antiviral Res ; 54(2): 113-20, 2002 May.
Article in English | MEDLINE | ID: mdl-12062396

ABSTRACT

The acyclic purine nucleoside analog, 2-amino-7-[(1,3-dihydroxy-2-propoxy)methyl]purine (S2242) and its orally active diacetate ester prodrug (HOE961) were reported to be potent inhibitors of vaccinia virus replication in cell culture and in infected mice. These compounds were evaluated further, using infections with the related cowpox virus. Against a wild-type (WT) cowpox virus strain in mouse C127I cell culture, 50% effective concentrations (EC(50), determined by plaque reduction assays) of S2242 and cidofovir (a positive control) were 3.5 and 1.0 microM, respectively. EC(50) values obtained against a cidofovir-resistant strain of the virus were 33 and 230 microM, respectively. Compounds were at least ten-fold less potent against WT virus in Vero cells than C127I cells. S2242 and cidofovir were 50% inhibitory to the proliferation of uninfected C127I cells at 340 and 180 microM, respectively, but neither compound inhibited Vero cell growth at 1000 microM. Mice were lethally infected with cowpox virus by intranasal inoculation, followed 24 h later by antiviral treatment for 5 consecutive days. Once or twice daily intraperitoneal (i.p.) treatments with either S2242 or HOE961 at 100 mg/kg per day resulted in > or = 70 survival compared with no survivors in the placebo group. Lower doses of these compounds (10 and 30 mg/kg per day) were not protective, however. Cidofovir was 100% protective at 30 mg/kg per day. A 10-day course of treatment gave comparable survival results and demonstrated the oral efficacy of HOE961. Treatments with S2242 (100 mg/kg per day) and cidofovir (30 mg/kg per day) each reduced lung and nasal virus titers by approximately ten-fold, whereas, HOE961 (100 mg/kg per day) was less active. Overall, S2242 and HOE961 were found to be effective against cowpox virus infections in mice but were less potent than cidofovir. Since, HOE961 was orally active, it may have advantages over the other parenterally administered compounds for treating orthopoxvirus infections.


Subject(s)
Antiviral Agents/pharmacology , Cowpox virus/growth & development , Cowpox/drug therapy , Organophosphonates , Prodrugs/pharmacology , Purines/pharmacology , Animals , Area Under Curve , Body Weight , Chlorocebus aethiops , Cidofovir , Cowpox virus/metabolism , Cytosine/analogs & derivatives , Cytosine/pharmacology , Disease Models, Animal , Female , Lung/virology , Mice , Mice, Inbred BALB C , Nasal Mucosa/virology , Organophosphorus Compounds/pharmacology , Statistics, Nonparametric , Vero Cells
18.
Virology ; 279(1): 22-6, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145885

ABSTRACT

The affinities of purified recombinant human IL-18 binding protein (BP) and ectromelia and cowpox virus homologs for human and murine IL-18 were compared by plasmon resonance. The dissociation constants of human IL-18BP were similar for murine and human IL-18. By contrast, the dissociation constants of the viral proteins for murine IL-18 were 12- to 50-fold lower than that for human IL-18. The ectromelia and cowpox virus proteins were biologically active, as judged by their ability to inhibit induction of interferon-gamma by murine and human IL-18. The relative affinities of the orthopoxvirus IL-18BPs are consistent with the rodent host range of the viruses.


Subject(s)
Glycoproteins/metabolism , Orthopoxvirus/metabolism , Animals , Cell Line , Cowpox virus/genetics , Cowpox virus/metabolism , Ectromelia virus/genetics , Ectromelia virus/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Interleukin-18/antagonists & inhibitors , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-18/pharmacology , Lipopolysaccharides/pharmacology , Mice , Orthopoxvirus/genetics , Poxviridae Infections/virology , Recombinant Proteins/metabolism , Spleen/cytology , Spleen/immunology , Surface Plasmon Resonance , Tumor Necrosis Factor-alpha/pharmacology
19.
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
20.
Virus Res ; 60(2): 147-57, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10392723

ABSTRACT

Myristylation is one of several post-translational modifications that occur on vaccinia virus (VV) proteins. Previously, time course labeling of VV-infected cells with myristic acid had indicated that five late proteins (17, 25, 36, 38 and 92 kDa) are myristylated. Four of these proteins were mapped to the E7R, L1R, AI6L and G9R open-reading frames, respectively, because of the predicted presence of the N-myristyltransferase recognition sequence (M-G-X-X-X-S/T/A) at their amino termini. In contrast, computer analyses of large (80-100 kDa) VV open reading frames did not reveal any predicted species with this N-terminal motif. By immunoprecipitation with monospecific sera and transient expression of cloned gene products, the myristylated 92-kDa protein has been demonstrated to be the A-type inclusion protein encoded by the Western Reserve (WR) strain of VV. Labeling of cowpox virus (CPV) infected cells with myristic acid indicated that the 160-kDa A-type inclusion protein appears to be myristylated as well. Both the VV 92-kDa and the CPV 160-kDa A-type inclusion proteins labeled with myristic acid were stable to hydroxylamine treatment, suggesting an amide linkage between the fatty acid and the acceptor protein. HPLC analysis confirmed that the 92-kDa protein was in fact myristylated. This data suggests that poxvirus ATI proteins may be subject to a novel type of internal myristylation modification, and the roles such modifications may play in the replication cycles of these viruses is discussed.


Subject(s)
Cowpox virus/metabolism , Viral Proteins/metabolism , Acylation , Amino Acid Sequence , Animals , Cell Line , Chromatography, High Pressure Liquid , Consensus Sequence , Cowpox virus/genetics , Myristic Acid/analysis , Myristic Acid/metabolism , Protein Processing, Post-Translational , Sequence Alignment , Sequence Homology, Amino Acid , Sulfur Radioisotopes , Tritium , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...