Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 15(12): e1008239, 2019 12.
Article in English | MEDLINE | ID: mdl-31877196

ABSTRACT

Cells sensing infection produce Type I interferons (IFN-I) to stimulate Interferon Stimulated Genes (ISGs) that confer resistance to viruses. During lympho-hematogenous spread of the mouse pathogen ectromelia virus (ECTV), the adaptor STING and the transcription factor IRF7 are required for IFN-I and ISG induction and resistance to ECTV. However, it is unknown which cells sense ECTV and which pathogen recognition receptor (PRR) upstream of STING is required for IFN-I and ISG induction. We found that cyclic-GMP-AMP (cGAMP) synthase (cGAS), a DNA-sensing PRR, is required in bone marrow-derived (BMD) but not in other cells for IFN-I and ISG induction and for resistance to lethal mousepox. Also, local administration of cGAMP, the product of cGAS that activates STING, rescues cGAS but not IRF7 or IFN-I receptor deficient mice from mousepox. Thus, sensing of infection by BMD cells via cGAS and IRF7 is critical for resistance to a lethal viral disease in a natural host.


Subject(s)
Bone Marrow/virology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/virology , Nucleotides, Cyclic/metabolism , Animals , Bone Marrow/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Interferon Type I/metabolism , Mice, Transgenic , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism
2.
Microb Pathog ; 109: 99-109, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28554653

ABSTRACT

Dendritic cells (DCs) are effector cells linking the innate immune system with the adaptive immune response. Many viruses eliminate DCs to prevent host response, induce immunosuppression and to maintain chronic infection. In this study, we examined apoptotic response of dendritic cells during in vitro and in vivo infection with ectromelia virus (ECTV), the causative agent of mousepox. ECTV-infected bone marrow dendritic cells (BMDCs) from BALB/c mice underwent apoptosis through mitochondrial pathway at 48 h post infection, up-regulated FasL and decreased expression of anti-apoptotic Bcl-2 and pro-apoptotic Fas. Similar pattern of Bcl-2, Fas and FasL expression was observed for DCs early during in vivo infection of BALB/c mice. Both BMDCs and DCs from BALB/c mice showed no maturation upon ECTV infection. We conclude that ECTV-infected DCs from BALB/c mouse strain help the virus to spread and to maintain infection.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Ectromelia virus/physiology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , Adaptive Immunity , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 3 , Chlorocebus aethiops , Dendritic Cells/pathology , Dendritic Cells/physiology , Dendritic Cells/virology , Disease Models, Animal , Ectromelia, Infectious/virology , Fas Ligand Protein/metabolism , Gene Expression Regulation , Immunity, Innate , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Up-Regulation , Vero Cells
3.
J Virol ; 89(19): 9974-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26202250

ABSTRACT

UNLABELLED: Viruses that spread systemically from a peripheral site of infection cause morbidity and mortality in the human population. Innate myeloid cells, including monocytes, macrophages, monocyte-derived dendritic cells (mo-DC), and dendritic cells (DC), respond early during viral infection to control viral replication, reducing virus spread from the peripheral site. Ectromelia virus (ECTV), an orthopoxvirus that naturally infects the mouse, spreads systemically from the peripheral site of infection and results in death of susceptible mice. While phagocytic cells have a requisite role in the response to ECTV, the requirement for individual myeloid cell populations during acute immune responses to peripheral viral infection is unclear. In this study, a variety of myeloid-specific depletion methods were used to dissect the roles of individual myeloid cell subsets in the survival of ECTV infection. We showed that DC are the primary producers of type I interferons (T1-IFN), requisite cytokines for survival, following ECTV infection. DC, but not macrophages, monocytes, or granulocytes, were required for control of the virus and survival of mice following ECTV infection. Depletion of either plasmacytoid DC (pDC) alone or the lymphoid-resident DC subset (CD8α(+) DC) alone did not confer lethal susceptibility to ECTV. However, the function of at least one of the pDC or CD8α(+) DC subsets is required for survival of ECTV infection, as mice depleted of both populations were susceptible to ECTV challenge. The presence of at least one of these DC subsets is sufficient for cytokine production that reduces ECTV replication and virus spread, facilitating survival following infection. IMPORTANCE: Prior to the eradication of variola virus, the orthopoxvirus that causes smallpox, one-third of infected people succumbed to the disease. Following successful eradication of smallpox, vaccination rates with the smallpox vaccine have significantly dropped. There is now an increasing incidence of zoonotic orthopoxvirus infections for which there are no effective treatments. Moreover, the safety of the smallpox vaccine is of great concern, as complications may arise, resulting in morbidity. Like many viruses that cause significant human diseases, orthopoxviruses spread from a peripheral site of infection to become systemic. This study elucidates the early requirement for innate immune cells in controlling a peripheral infection with ECTV, the causative agent of mousepox. We report that there is redundancy in the function of two innate immune cell subsets in controlling virus spread early during infection. The viral control mediated by these cell subsets presents a potential target for therapies and rational vaccine design.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cytokines/biosynthesis , Dendritic Cells/classification , Ectromelia virus/physiology , Ectromelia, Infectious/transmission , Ectromelia, Infectious/virology , Granulocytes/immunology , Humans , Immunity, Innate , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/immunology , Virus Replication , Zoonoses/immunology , Zoonoses/transmission , Zoonoses/virology
4.
PLoS Pathog ; 10(8): e1004326, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25122471

ABSTRACT

Poxviruses contain large dsDNA genomes encoding numerous open reading frames that manipulate cellular signalling pathways and interfere with the host immune response. The NF-κB signalling cascade is an important mediator of innate immunity and inflammation, and is tightly regulated by ubiquitination at several key points. A critical step in NF-κB activation is the ubiquitination and degradation of the inhibitor of kappaB (IκBα), by the cellular SCFß-TRCP ubiquitin ligase complex. We show here that upon stimulation with TNFα or IL-1ß, Orthopoxvirus-infected cells displayed an accumulation of phosphorylated IκBα, indicating that NF-κB activation was inhibited during poxvirus infection. Ectromelia virus is the causative agent of lethal mousepox, a natural disease that is fatal in mice. Previously, we identified a family of four ectromelia virus genes (EVM002, EVM005, EVM154 and EVM165) that contain N-terminal ankyrin repeats and C-terminal F-box domains that interact with the cellular SCF ubiquitin ligase complex. Since degradation of IκBα is catalyzed by the SCFß-TRCP ubiquitin ligase, we investigated the role of the ectromelia virus ankyrin/F-box protein, EVM005, in the regulation of NF-κB. Expression of Flag-EVM005 inhibited both TNFα- and IL-1ß-stimulated IκBα degradation and p65 nuclear translocation. Inhibition of the NF-κB pathway by EVM005 was dependent on the F-box domain, and interaction with the SCF complex. Additionally, ectromelia virus devoid of EVM005 was shown to inhibit NF-κB activation, despite lacking the EVM005 open reading frame. Finally, ectromelia virus devoid of EVM005 was attenuated in both A/NCR and C57BL/6 mouse models, indicating that EVM005 is required for virulence and immune regulation in vivo.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/metabolism , NF-kappa B/metabolism , Viral Proteins/metabolism , Animals , Ectromelia virus/immunology , Ectromelia virus/metabolism , Ectromelia, Infectious/immunology , Flow Cytometry , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , NF-kappa B/immunology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/immunology , Virulence/physiology
5.
Article in Russian | MEDLINE | ID: mdl-25842954

ABSTRACT

AIM: Study pharmacodynamic parameters of anti-viral effectiveness of a chemical compound NIOC-14 in experiments in mice infected with ectromelia virus (EV). MATERIALS AND METHODS: EV (K-1 strain) was obtained from the State Collection of Viral Infections and Rickettsioses Causative Agents of the State Scientific Centre of Virology and Biotechnology "Vector". Outbred ICR mice were intranasally infected with EV at a dose of 10 LD50 per animal (10 x 50% lethal doses/animal) and per orally received NIOC-14 or ST-246 as a positive control. Chemical compound NIOC-14 (7-[N'-(4-trifluoromethylbenzoyl)-hidrazincarbonyl]-tricyclo[3.2.2.0(2,4)]non-8-en-6-carbonic acid) was synthesized in Novosibirsk Institute of Organic Chemistry (NIOC). Anti-pox preparation ST-246, developed by SIGA Technologies Inc. (USA), was synthesized in NIOC using the technique described by the authors. RESULTS: 50% effective doses against EV in vivo were shown not to differ significantly between the preparations NIOC-14 (3.59 µg/g mouse mass) and ST-246 (5.08 µg/g mouse mass). During determination of therapeutic window, administration of NIOC-14 to mice 1 day or 1 hour before EV infection, as well as 1, 2 and 4 days after EV infection and then for 9 days was found to ensure 100% animal survival. Administration of NIOC-14 as well as ST-246 resulted in the decrease relative to control of EV titers in lungs, nasal cavity, brains, liver, spleen, kidneys and pancreas. CONCLUSION: Anti-viral effectiveness of NIOC-14 against EV in vivo was thus comparable by all the studied pharmacodynamic parameters with anti-viral activity of anti-pox-virus preparation ST-246.


Subject(s)
Alkenes/administration & dosage , Antiviral Agents/administration & dosage , Ectromelia virus/drug effects , Ectromelia, Infectious/drug therapy , Hydrazines/administration & dosage , Animals , Benzamides/administration & dosage , Ectromelia virus/pathogenicity , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Humans , Isoindoles/administration & dosage , Liver/drug effects , Liver/virology , Mice , Spleen/drug effects , Spleen/virology
6.
J Virol ; 87(12): 7046-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596297

ABSTRACT

Orthopoxviruses (OPVs), which include the agent of smallpox (variola virus), the zoonotic monkeypox virus, the vaccine and zoonotic species vaccinia virus, and the mouse pathogen ectromelia virus (ECTV), form two types of infectious viral particles: the mature virus (MV), which is cytosolic, and the enveloped virus (EV), which is extracellular. It is believed that MVs are required for viral entry into the host, while EVs are responsible for spread within the host. Following footpad infection of susceptible mice, ECTV spreads lymphohematogenously, entering the liver at 3 to 4 days postinfection (dpi). Afterwards, ECTV spreads intrahepatically, killing the host. We found that antibodies to an MV protein were highly effective at curing mice from ECTV infection when administered after the virus reached the liver. Moreover, a mutant ECTV that does not make EV was able to spread intrahepatically and kill immunodeficient mice. Together, these findings indicate that MVs are sufficient for the spread of ECTV within the liver and could have implications regarding the pathogenesis of other OPVs, the treatment of emerging OPV infections, as well as strategies for preparedness in case of accidental or intentional release of pathogenic OPVs.


Subject(s)
Cytosol/virology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/therapy , Liver/virology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Viral/administration & dosage , Antibodies, Viral/immunology , Ectromelia virus/immunology , Ectromelia virus/metabolism , Ectromelia, Infectious/immunology , Ectromelia, Infectious/mortality , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Liver/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Virion/metabolism
7.
PLoS Pathog ; 8(1): e1002475, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22241999

ABSTRACT

Type 1 interferons (T1-IFNs) play a major role in antiviral defense, but when or how they protect during infections that spread through the lympho-hematogenous route is not known. Orthopoxviruses, including those that produce smallpox and mousepox, spread lympho-hematogenously. They also encode a decoy receptor for T1-IFN, the T1-IFN binding protein (T1-IFNbp), which is essential for virulence. We demonstrate that during mousepox, T1-IFNs protect the liver locally rather than systemically, and that the T1-IFNbp attaches to uninfected cells surrounding infected foci in the liver and the spleen to impair their ability to receive T1-IFN signaling, thus facilitating virus spread. Remarkably, this process can be reversed and mousepox cured late in infection by treating with antibodies that block the biological function of the T1-IFNbp. Thus, our findings provide insights on how T1-IFNs function and are evaded during a viral infection in vivo, and unveil a novel mechanism for antibody-mediated antiviral therapy.


Subject(s)
Antibodies, Viral/pharmacology , Ectromelia virus/metabolism , Ectromelia, Infectious/immunology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors , Animals , Antibodies, Viral/immunology , Cell Line , Cricetinae , Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/drug therapy , Ectromelia, Infectious/metabolism , Female , Liver/immunology , Liver/metabolism , Liver/virology , Mice , Mice, Inbred BALB C , Mice, SCID , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Spleen/immunology , Spleen/metabolism , Spleen/virology , Variola virus/immunology , Variola virus/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , Virulence Factors/immunology , Virulence Factors/metabolism , Virus Attachment/drug effects
8.
J Virol ; 86(13): 7298-309, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532670

ABSTRACT

Vaccinia virus (VACV) stimulates long-term immunity against highly pathogenic orthopoxvirus infection of humans (smallpox) and mice (mousepox [ectromelia virus {ECTV}]) despite the lack of a natural host-pathogen relationship with either of these species. Previous research revealed that VACV is able to induce polyfunctional CD8(+) T-cell responses after immunization of humans. However, the degree to which the functional profile of T cells induced by VACV is similar to that generated during natural poxvirus infection remains unknown. In this study, we monitored virus-specific T-cell responses following the dermal infection of C57BL/6 mice with ECTV or VACV. Using polychromatic flow cytometry, we measured levels of degranulation, cytokine expression (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and interleukin-2 [IL-2]), and the cytolytic mediator granzyme B. We observed that the functional capacities of T cells induced by VACV and ECTV were of a similar quality in spite of the markedly different replication abilities and pathogenic outcomes of these viruses. In general, a significant fraction (≥50%) of all T-cell responses were positive for at least three functions both during acute infection and into the memory phase. In vivo killing assays revealed that CD8(+) T cells specific for both viruses were equally cytolytic (∼80% target cell lysis after 4 h), consistent with the similar levels of granzyme B and degranulation detected among these cells. Collectively, these data provide a mechanism to explain the ability of VACV to induce protective T-cell responses against pathogenic poxviruses in their natural hosts and provide further support for the use of VACV as a vaccine platform able to induce polyfunctional T cells.


Subject(s)
Ectromelia virus/immunology , T-Lymphocytes/immunology , Vaccinia virus/immunology , Animals , Cell Degranulation , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Disease Models, Animal , Ectromelia virus/pathogenicity , Ectromelia virus/physiology , Ectromelia, Infectious/immunology , Female , Flow Cytometry , Granzymes/biosynthesis , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Vaccinia/immunology , Vaccinia virus/pathogenicity , Vaccinia virus/physiology
9.
J Virol ; 86(13): 7427-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532690

ABSTRACT

Egress of wrapped virus (WV) to the cell periphery following vaccinia virus (VACV) replication is dependent on interactions with the microtubule motor complex kinesin-1 and is mediated by the viral envelope protein A36. Here we report that ectromelia virus (ECTV), a related orthopoxvirus and the causative agent of mousepox, encodes an A36 homologue (ECTV-Mos-142) that is highly conserved despite a large truncation at the C terminus. Deleting the ECTV A36R gene leads to a reduction in the number of extracellular viruses formed and to a reduced plaque size, consistent with a role in microtubule transport. We also observed a complete loss of virus-associated actin comets, another phenotype dependent on A36 expression during VACV infection. ECTV ΔA36R was severely attenuated when used to infect the normally susceptible BALB/c mouse strain. ECTV ΔA36R replication and spread from the draining lymph nodes to the liver and spleen were significantly reduced in BALB/c mice and in Rag-1-deficient mice, which lack T and B lymphocytes. The dramatic reduction in ECTV ΔA36R titers early during the course of infection was not associated with an augmented immune response. Taken together, these findings demonstrate the critical role that subcellular transport pathways play not only in orthopoxvirus infection in an in vitro context but also during orthopoxvirus pathogenesis in a natural host. Furthermore, despite the attenuation of the mutant virus, we found that infection nonetheless induced protective immunity in mice, suggesting that orthopoxvirus vectors with A36 deletions may be considered another safe vaccine alternative.


Subject(s)
Cytoskeletal Proteins/metabolism , Ectromelia virus/pathogenicity , Ectromelia, Infectious/virology , Host-Pathogen Interactions , Viral Proteins/metabolism , Virus Release , Animals , Ectromelia virus/genetics , Female , Gene Deletion , Liver/virology , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Protein Transport , Spleen/virology , Viral Load , Viral Plaque Assay , Viral Proteins/genetics , Virulence
10.
J Virol ; 85(7): 3557-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270149

ABSTRACT

The emergence of zoonotic orthopoxvirus infections and the threat of possible intentional release of pathogenic orthopoxviruses have stimulated renewed interest in understanding orthopoxvirus infections and the resulting diseases. Ectromelia virus (ECTV), the causative agent of mousepox, offers an excellent model system to study an orthopoxvirus infection in its natural host. Here, we investigated the role of the vaccinia virus ortholog N1L in ECTV infection. Respiratory infection of mice with an N1L deletion mutant virus (ECTVΔN1L) demonstrated profound attenuation of the mutant virus, confirming N1 as an orthopoxvirus virulence factor. Upon analysis of virus dissemination in vivo, we observed a striking deficiency of ECTVΔN1L spreading from the lungs to the livers or spleens of infected mice. Investigating the immunological mechanism controlling ECTVΔN1L infection, we found the attenuated phenotype to be unaltered in mice deficient in Toll-like receptor (TLR) or RIG-I-like RNA helicase (RLH) signaling as well as in those missing the type I interferon receptor or lacking B cells. However, in RAG-1(-/-) mice lacking mature B and T cells, ECTVΔN1L regained virulence, as shown by increasing morbidity and virus spread to the liver and spleen. Moreover, T cell depletion experiments revealed that ECTVΔN1L attenuation was reversed only by removing both CD4(+) and CD8(+) T cells, so the presence of either cell subset was still sufficient to control the infection. Thus, the orthopoxvirus virulence factor N1 may allow efficient ECTV infection in mice by interfering with host T cell function.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/pathology , Ectromelia, Infectious/virology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Viral Proteins/physiology , Virulence Factors/physiology , Animals , Body Weight , Female , Gene Deletion , Histocytochemistry , Immune Tolerance , Liver/pathology , Liver/virology , Lung/pathology , Lung/virology , Mice , Mice, Inbred C57BL , Spleen/virology , Survival Analysis , T-Lymphocytes/immunology , Viral Load , Viral Proteins/genetics , Virulence Factors/genetics
11.
J Virol ; 85(21): 11170-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21849445

ABSTRACT

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/mortality , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Serpins/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Ectromelia virus/genetics , Ectromelia virus/immunology , Ectromelia, Infectious/virology , Gene Deletion , Interferon-gamma/metabolism , Interleukin-18/metabolism , Liver/virology , Lymphocyte Subsets/chemistry , Lymphocyte Subsets/immunology , Mice , Serpins/genetics , Spleen/pathology , Survival Analysis , Viral Load , Viral Proteins/genetics , Virus Replication
12.
Virol J ; 9: 119, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22709563

ABSTRACT

BACKGROUND: In an event of a smallpox outbreak in humans, the window for efficacious treatment by vaccination with vaccinia viruses (VACV) is believed to be limited to the first few days post-exposure (p.e.). We recently demonstrated in a mouse model for human smallpox, that active immunization 2-3 days p.e. with either VACV-Lister or modified VACV Ankara (MVA) vaccines, can rescue animals from lethal challenge of ectromelia virus (ECTV), the causative agent of mousepox. The present study was carried out in order to determine whether a single dose of the anti-viral cidofovir (CDV), administered at different times and doses p.e. either alone or in conjunction with active vaccination, can rescue ECTV infected mice. METHODS: Animals were infected intranasally with ECTV, treated on different days with various single CDV doses and monitored for morbidity, mortality and humoral response. In addition, in order to determine the influence of CDV on the immune response following vaccination, both the "clinical take", IFN-gamma and IgG Ab levels in the serum were evaluated as well as the ability of the mice to withstand a lethal challenge of ECTV. Finally the efficacy of a combined treatment regime of CDV and vaccination p.e. was determined. RESULTS: A single p.e. CDV treatment is sufficient for protection depending on the initiation time and dose (2.5 - 100 mg/kg) of treatment. Solid protection was achieved by a low dose (5 mg/kg) CDV treatment even if given at day 6 p.e., approximately 4 days before death of the control infected untreated mice (mean time to death (MTTD) 10.2). At the same time point complete protection was achieved by single treatment with higher doses of CDV (25 or 100 mg/kg). Irrespective of treatment dose, all surviving animals developed a protective immune response even when the CDV treatment was initiated one day p.e.. After seven days post treatment with the highest dose (100 mg/kg), virus was still detected in some organs (e.g. lung and liver) yet all animals survived, suggesting that efficacious single CDV treatment requires a potent immune system. The combination of CDV and vaccination provided no additional protection over CDV alone. Yet, combining CDV and vaccination maintained vaccination efficacy. CONCLUSIONS: Altogether, our data substantiate the feasibility of single post-exposure antiviral treatment to face orthopoxvirus infection.


Subject(s)
Antiviral Agents/administration & dosage , Cytosine/analogs & derivatives , Ectromelia virus/drug effects , Ectromelia, Infectious/drug therapy , Organophosphonates/administration & dosage , Animals , Antibodies, Viral/blood , Cidofovir , Cytosine/administration & dosage , Disease Models, Animal , Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , Ectromelia, Infectious/pathology , Female , Immunoglobulin G/blood , Interferon-gamma/blood , Mice , Mice, Inbred BALB C , Survival Analysis
13.
J Clin Invest ; 118(5): 1776-84, 2008 May.
Article in English | MEDLINE | ID: mdl-18398511

ABSTRACT

Poxviruses such as the causative agent of smallpox have developed multiple strategies to suppress immune responses, including the suppression of DC activation. Since poxviruses are large DNA viruses, we hypothesized that their detection by DCs may involve the endosomal DNA recognition receptor TLR9. Indeed, we have shown here that DC recognition of ectromelia virus (ECTV), the causative agent of mousepox, completely depended on TLR9. The importance of TLR9 was highlighted by the fact that mice lacking TLR9 showed drastically increased susceptibility to infection with ECTV. In contrast, we found that the strongly attenuated poxvirus modified vaccinia virus Ankara (MVA) activated DCs by both TLR9-dependent and -independent pathways. We therefore tested whether we could use the broader induction of immune responses by MVA to protect mice from a lethal infection with ECTV. Indeed, MVA given at the same time as a lethal dose of ECTV protected mice from death. Importantly, MVA also rescued TLR9-deficient mice if administered 2 full days after an otherwise lethal infection with ECTV. Therefore, these data suggest an essential role for TLR9 in the defense against poxviruses. In addition, postexposure application of MVA may protect against lethal poxvirus infection.


Subject(s)
Dendritic Cells/immunology , Poxviridae Infections , Toll-Like Receptor 9/immunology , Vaccination , Animals , Dendritic Cells/cytology , Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Humans , Immunity/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poxviridae Infections/immunology , Poxviridae Infections/mortality , Poxviridae Infections/prevention & control , Survival Rate , Toll-Like Receptor 9/genetics , Vaccinia virus/immunology
14.
J Virol ; 84(18): 9128-39, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610727

ABSTRACT

Poxviruses produce complement regulatory proteins to subvert the host's immune response. Similar to the human pathogen variola virus, ectromelia virus has a limited host range and provides a mouse model where the virus and the host's immune response have coevolved. We previously demonstrated that multiple components (C3, C4, and factor B) of the classical and alternative pathways are required to survive ectromelia virus infection. Complement's role in the innate and adaptive immune responses likely drove the evolution of a virus-encoded virulence factor that regulates complement activation. In this study, we characterized the ectromelia virus inhibitor of complement enzymes (EMICE). Recombinant EMICE regulated complement activation on the surface of CHO cells, and it protected complement-sensitive intracellular mature virions (IMV) from neutralization in vitro. It accomplished this by serving as a cofactor for the inactivation of C3b and C4b and by dissociating the catalytic domain of the classical pathway C3 convertase. Infected murine cells initiated synthesis of EMICE within 4 to 6 h postinoculation. The levels were sufficient in the supernatant to protect the IMV, upon release, from complement-mediated neutralization. EMICE on the surface of infected murine cells also reduced complement activation by the alternative pathway. In contrast, classical pathway activation by high-titer antibody overwhelmed EMICE's regulatory capacity. These results suggest that EMICE's role is early during infection when it counteracts the innate immune response. In summary, ectromelia virus produced EMICE within a few hours of an infection, and EMICE in turn decreased complement activation on IMV and infected cells.


Subject(s)
Complement Inactivating Agents/immunology , Complement System Proteins/immunology , Ectromelia virus/immunology , Immune Evasion , Viral Proteins/immunology , Virulence Factors/immunology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Ectromelia virus/pathogenicity , Mice , Mice, Inbred C57BL , Neutralization Tests
15.
Viruses ; 13(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34203773

ABSTRACT

Ectromelia virus (ECTV), the causative agent of mousepox, has threatened laboratory mouse colonies worldwide for almost a century. Mousepox has been valuable for the understanding of poxvirus pathogenesis and immune evasion. Here, we have monitored in parallel the pathogenesis of nine ECTVs in BALB/cJ mice and report the full-length genome sequence of eight novel ECTV isolates or strains, including the first ECTV isolated from a field mouse, ECTV-MouKre. This approach allowed us to identify several genes, absent in strains attenuated through serial passages in culture, that may play a role in virulence and a set of putative genes that may be involved in enhancing viral growth in vitro. We identified a putative strong inhibitor of the host inflammatory response in ECTV-MouKre, an isolate that did not cause local foot swelling and developed a moderate virulence. Most of the ECTVs, except ECTV-Hampstead, encode a truncated version of the P4c protein that impairs the recruitment of virions into the A-type inclusion bodies, and our data suggest that P4c may play a role in viral dissemination and transmission. This is the first comprehensive report that sheds light into the phylogenetic and geographic relationship of the worldwide outbreak dynamics for the ECTV species.


Subject(s)
Ectromelia virus/genetics , Ectromelia virus/pathogenicity , Ectromelia, Infectious/pathology , Ectromelia, Infectious/virology , Genomics , Phylogeny , Animals , Disease Models, Animal , Ectromelia virus/classification , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Female , Immune Evasion , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Phylogeography , Viral Proteins/genetics , Virulence
16.
Acta Virol ; 54(1): 41-8, 2010.
Article in English | MEDLINE | ID: mdl-20201613

ABSTRACT

We describe here a contribution of virus-induced actin tails and filopodia in transmission of Ectromelia virus (ECTV) infection in permissive cells detected by the immunofluorescence and confocal microscopy. Immunoblot analysis revealed profoundly decreased beta-actin levels during ECTV replicative cycle in the infected cells 24 hrs post infection (p.i.). These results provided a basis for the further analysis of ECTV motion in the infected cells as well as for impact of ECTV infection on the cytoskeletal proteins.


Subject(s)
Actins/metabolism , Ectromelia virus/pathogenicity , Actins/ultrastructure , Animals , BALB 3T3 Cells/ultrastructure , BALB 3T3 Cells/virology , Chlorocebus aethiops , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/ultrastructure , Ectromelia virus/physiology , Ectromelia, Infectious/virology , Foot/virology , HeLa Cells/ultrastructure , HeLa Cells/virology , Humans , Immunoblotting , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Fluorescence , Vero Cells/ultrastructure , Vero Cells/virology
17.
Sci Rep ; 10(1): 13167, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32759969

ABSTRACT

Vaccination with vaccinia virus (VACV) elicits heterotypic immunity to smallpox, monkeypox, and mousepox, the mechanistic basis for which is poorly understood. It is generally assumed that heterotypic immunity arises from the presentation of a wide array of VACV-derived, CD8+ T cell epitopes that share homology with other poxviruses. Herein this assumption was tested using a large panel of VACV-derived peptides presented by HLA-B*07:02 (B7.2) molecules in a mousepox/ectromelia virus (ECTV)-infection, B7.2 transgenic mouse model. Most dominant epitopes recognized by ECTV- and VACV-reactive CD8+ T cells overlapped significantly without altering immunodominance hierarchy. Further, several epitopes recognized by ECTV-reactive CD8+ T cells were not recognized by VACV-reactive CD8+ T cells, and vice versa. In one instance, the lack of recognition owed to a N72K variation in the ECTV C4R70-78 variant of the dominant VACV B8R70-78 epitope. C4R70-78 does not bind to B7.2 and, hence, it was neither immunogenic nor antigenic. These findings provide a mechanistic basis for VACV vaccination-induced heterotypic immunity which can protect against Variola and Monkeypox disease. The understanding of how cross-reactive responses develop is essential for the rational design of a subunit-based vaccine that would be safe, and effectively protect against heterologous infection.


Subject(s)
Ectromelia, Infectious/prevention & control , HLA-B7 Antigen/genetics , Peptides/immunology , Vaccinia virus/immunology , Viral Proteins/chemistry , Animals , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , HLA-B7 Antigen/metabolism , Immunodominant Epitopes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
18.
Mol Biol (Mosk) ; 43(4): 616-22, 2009.
Article in Russian | MEDLINE | ID: mdl-19807023

ABSTRACT

Mousepox (ectromelia) virus genome contains four genes encoding for kelch-like proteins EVM018, EVM027, EVM150 and EVM167. A complete set of insertion plasmids was constructed to allow the production of recombinant ectromelia viruses with targeted deletions of one to four genes of kelch family both individually (single mutants) and in different combinations (double, triple and quadruple mutants). It was shown that deletion of any of the three genes EVMO18, EVM027 or EVM167 resulted in reduction of 50% lethal dose (LD50) by five and more orders in outbred white mice infected intraperitoneally. Deletion of mousepox kelch-gene EVM150 did not influence the virus virulence. Two or more kelch-genes deletion also resulted in high level of attenuation, which could evidently be due to the lack of three genes EVM167, EVM018 and/or EVM027 identified as virulence factors. The local inflammatory process on the model of intradermal injection of mouse ear pinnae (vasodilatation level, hyperemia, cutaneous edema, arterial thrombosis) was significantly more intensive for wild type virus and virulent mutant deltaEVM150 in comparison with avirulent mutant AEVM167.


Subject(s)
Ectromelia virus/genetics , Ectromelia virus/pathogenicity , Ectromelia, Infectious/genetics , Gene Deletion , Genes, Viral/genetics , Viral Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Ectromelia virus/metabolism , Ectromelia, Infectious/metabolism , Mice
19.
Antivir Ther ; 13(7): 863-73, 2008.
Article in English | MEDLINE | ID: mdl-19043920

ABSTRACT

BACKGROUND: The emergence of human monkeypox and the potential use of recombinant variola and monkeypox viruses as biological terrorist agents have necessitated the development of therapeutic and prophylactic therapies. The primary, or index, cases of smallpox and/or human monkeypox will likely be identified by a characteristic rash. Effective biomarkers will be required to monitor disease progression, guide the choice of therapeutic intervention strategies and evaluate their efficacies. To address this we have evaluated several biomarkers of disease in a lethal mousepox model. METHODS: The efficacy of a single dose of a hexadecyloxypropyl ester of cidofovir (CMX001) at 20, 25 and 30 mg/kg doses administered on days 4, 5, 6 and 7 post-infection was evaluated in A/Ncr mice intranasally infected with low doses of ectromelia virus (<20 plaque-forming units). Mice were monitored for weight loss, blood interferon-gamma levels, alanine aminotransferase (ALT), aspartate aminotransferase, viral DNA copies and neutrophilia levels to stage disease progression. RESULTS: We have used these biomarkers to establish the optimal dosing regimen for treatment and reveal that a single dose of 25 mg/kg of CMX001 can be efficacious at treating lethal mousepox when administered on days 4 or 5 post-infection. This dose significantly reduces ALT, interferon-gamma and DNA copies found in the blood of infected animals. CONCLUSIONS: A single dose regimen of CMX001 is efficacious at treating mousepox. Disease progression and antiviral efficacy can be monitored using several biomarkers that could readily be used in the case of a human monkeypox or smallpox outbreak.


Subject(s)
Antiviral Agents/therapeutic use , Cytosine/analogs & derivatives , Ectromelia virus/pathogenicity , Ectromelia, Infectious/drug therapy , Ectromelia, Infectious/physiopathology , Organophosphonates/therapeutic use , Alanine Transaminase/blood , Animals , Antiviral Agents/administration & dosage , Aspartate Aminotransferases/blood , Biomarkers/analysis , Cell Line , Cytosine/administration & dosage , Cytosine/therapeutic use , DNA, Viral/blood , Disease Models, Animal , Disease Progression , Ectromelia, Infectious/virology , Female , Humans , Interferon-gamma/blood , Mice , Organophosphonates/administration & dosage , Treatment Outcome , Weight Loss
20.
Virology ; 518: 335-348, 2018 05.
Article in English | MEDLINE | ID: mdl-29602068

ABSTRACT

All known orthopoxviruses, including ectromelia virus (ECTV), contain a gene in the E3L family. The protein product of this gene, E3, is a double-stranded RNA-binding protein. It can impact host range and is used by orthopoxviruses to combat cellular defense pathways, such as PKR and RNase L. In this work, we constructed an ECTV mutant with a targeted disruption of the E3L open reading frame (ECTVΔE3L). Infection with this virus resulted in an abortive replication cycle in all cell lines tested. We detected limited transcription of late genes but no significant translation of these mRNAs. Notably, the replication defects of ECTVΔE3L were rescued in human and mouse cells lacking PKR. ECTVΔE3L was nonpathogenic in BALB/c mice, a strain susceptible to lethal mousepox disease. However, infection with ECTVΔE3L induced protective immunity upon subsequent challenge with wild-type virus. In summary, E3L is an essential gene for ECTV.


Subject(s)
Ectromelia virus/immunology , Ectromelia virus/physiology , Ectromelia, Infectious/prevention & control , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/immunology , Virus Replication , Animals , Cell Line , Ectromelia virus/genetics , Ectromelia virus/pathogenicity , Gene Knockout Techniques , Humans , Mice, Inbred BALB C , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL