Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Vopr Virusol ; 68(4): 277-282, 2023 Sep 21.
Article in Russian | MEDLINE | ID: mdl-38156584

ABSTRACT

INTRODUCTION: The mouse-specific orthopoxvirus, ectromelia virus, is one of the best models that can be used to study key issues of pathogenesis, prevention, and treatment of smallpox, and to develop measures to increase virulence, transmissibility, or the ability to overcome vaccine immunity. The aim of the work is to screen the antiviral activity of samples from Inonotus obliquus chaga and humic acid from brown coal in vitro against ectromelia virus. MATERIALS AND METHODS: We used ectromelia virus, strain K-1 (reg. No V-142), obtained from the State Collection of Pathogens of Viral Infections and Rickettsioses of the State Scientific Center of Virology and Biotechnology "Vector"; Vero Е6 cell culture (No 70) from the Collection of cell cultures of the State Scientific Center of Virology and Biotechnology "Vector". Nine samples from chaga I. obliquus and humic acid from brown coal were used to evaluate the changes in the infectivity of the ectromelia virus on cell culture using 2 schemes of application of drugs and virus (preventive and therapeutic schemes), and to assess their cytotoxicity and antiviral activity. RESULTS: 50% cytotoxic concentration, 50% virus-inhibiting concentrations and selectivity index were determined for all samples. The studied samples were shown to be non-toxic to the monolayer of Vero cell culture in a dilution of 300 and more micrograms/ml, while demonstrated high antiviral activity against strain K-1 of ectromelia virus in two application schemes - preventive and curative. CONCLUSION: All samples tested for ectromelia virus in vitro can be considered promising for further development of drugs against diseases caused by orthopoxviruses.


Subject(s)
Antiviral Agents , Ectromelia virus , Ectromelia, Infectious , Animals , Antiviral Agents/pharmacology , Cell Culture Techniques , Coal , Ectromelia virus/drug effects , Ectromelia, Infectious/prevention & control , Humic Substances , Vero Cells , Chlorocebus aethiops , Inonotus/chemistry
2.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33992803

ABSTRACT

It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 µg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Viral Proteins/genetics , mRNA Vaccines/administration & dosage , Animals , Drug Compounding , Ectromelia, Infectious/immunology , Immunization, Secondary , Immunologic Memory , Liposomes , Male , Mice , Nanoparticles , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/pharmacology , mRNA Vaccines/chemistry , mRNA Vaccines/pharmacology
3.
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
4.
Nat Commun ; 9(1): 1790, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29724993

ABSTRACT

The role of cytokines and chemokines in anti-viral defense has been demonstrated, but their relative contribution to protective anti-viral responses in vivo is not fully understood. Cytokine response modifier D (CrmD) is a secreted receptor for TNF and lymphotoxin containing the smallpox virus-encoded chemokine receptor (SECRET) domain and is expressed by ectromelia virus, the causative agent of the smallpox-like disease mousepox. Here we show that CrmD is an essential virulence factor that controls natural killer cell activation and allows progression of fatal mousepox, and demonstrate that both SECRET and TNF binding domains are required for full CrmD activity. Vaccination with recombinant CrmD protects animals from lethal mousepox. These results indicate that a specific set of chemokines enhance the inflammatory and protective anti-viral responses mediated by TNF and lymphotoxin, and illustrate how viruses optimize anti-TNF strategies with the addition of a chemokine binding domain as soluble decoy receptors.


Subject(s)
Chemokines/physiology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/prevention & control , Inflammation/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Female , Inflammation/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Poxviridae/pathogenicity , Virulence Factors/physiology , Virus Replication
5.
Vaccine ; 36(18): 2427-2434, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29599088

ABSTRACT

Newborns are considered difficult to protect against infections shortly after birth, due to their ineffective immune system that shows quantitative and qualitative differences compared to adults. However, here we show that a single vaccination of mice at birth with a replication-deficient live vaccine Modified Vaccinia Ankara [MVA] efficiently induces antigen-specific B- and T-cells that fully protect against a lethal Ectromelia virus challenge. Protection was induced within 2 weeks and using genetically modified mice we show that this protection was mainly T-cell dependent. Persisting immunological T-cell memory and neutralizing antibodies were obtained with the single vaccination. Thus, MVA administered as early as at birth induced immediate and long-term protection against an otherwise fatal disease and appears attractive as a new generation smallpox vaccine that is effective also in children. Moreover, it may have the potential to serve as platform for childhood vaccines as indicated by measles specific T- and B-cell responses induced in newborn mice vaccinated with recombinant MVA expressing measles antigens.


Subject(s)
Immunization Schedule , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Ectromelia, Infectious/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
6.
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
7.
Adv Immunol ; 129: 251-76, 2016.
Article in English | MEDLINE | ID: mdl-26791861

ABSTRACT

Ectromelia virus is a mouse-specific orthopoxvirus that, following footpad infection or natural transmission, causes mousepox in most strains of mice, while a few strains, such as C57BL/6, are resistant to the disease but not to the infection. Mousepox is an acute, systemic, highly lethal disease of remarkable semblance to smallpox, caused by the human-specific variola virus. Starting in 1929 with its discovery by Marchal, work with ECTV has provided essential information for our current understanding on how viruses spread lympho-hematogenously, the genetic control of antiviral resistance, the role of different components of the innate and adaptive immune system in the control of primary and secondary infections with acute viruses, and how the mechanisms of immune evasion deployed by the virus affect virulence in vivo. Here, I review the literature on the pathogenesis and immunobiology of ECTV infection in vivo.


Subject(s)
Adaptive Immunity , Disease Resistance/immunology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , Smallpox/immunology , Vaccinia virus/immunology , Animals , Disease Models, Animal , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Humans , Mice , Mice, Inbred C57BL , Vaccination , Viral Proteins/immunology , Virulence
8.
Pathog Dis ; 73(9): ftv088, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26474845

ABSTRACT

Eradication of smallpox has led to cessation of vaccination programs. This has rendered the human population increasingly susceptible not only to variola virus infection but also to infections with other representatives of Poxviridae family that cause zoonotic variola-like diseases. Thus, new approaches for designing improved vaccine against smallpox are required. Discovering that orthopoxviruses, e.g. variola virus, vaccinia virus, ectromelia virus, share common immunodominant antigen, may result in the development of such a vaccine. In our study, the generation of antigen-specific CD8(+) T cells in mice during the acute and memory phase of the immune response was induced using the vaccinia virus immunodominant TSYKFESV epitope and CpG oligodeoxynucleotides as adjuvants. The role of the generated TSYKFESV-specific CD8(+) T cells was evaluated in mice during ectromelia virus infection using systemic and mucosal model. Moreover, the involvement of dendritic cells subsets in the adaptive immune response stimulation was assessed. Our results indicate that the TSYKFESV epitope/TLR9 agonist approach, delivered systemically or mucosally, generated strong CD8(+) T-cell response when measured 10 days after immunization. Furthermore, the TSYKFESV-specific cell population remained functionally active 2 months post-immunization, and gave cross-protection in virally challenged mice, even though the numbers of detectable antigen-specific T cells decreased.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Protection , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Epitopes, T-Lymphocyte/immunology , Vaccinia virus/immunology , Animals , Antigens, Viral/administration & dosage , Epitopes, T-Lymphocyte/administration & dosage , Male , Mice, Inbred C57BL , Oligodeoxyribonucleotides
9.
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
10.
PLoS One ; 9(10): e110545, 2014.
Article in English | MEDLINE | ID: mdl-25350003

ABSTRACT

Eradication of smallpox and discontinuation of the vaccination campaign resulted in an increase in the percentage of unvaccinated individuals, highlighting the need for postexposure efficient countermeasures in case of accidental or deliberate viral release. Intranasal infection of mice with ectromelia virus (ECTV), a model for human smallpox, is curable by vaccination with a high vaccine dose given up to 3 days postexposure. To further extend this protective window and to reduce morbidity, mice were vaccinated postexposure with Vaccinia-Lister, the conventional smallpox vaccine or Modified Vaccinia Ankara, a highly attenuated vaccine in conjunction with TLR3 or TLR9 agonists. We show that co-administration of the TLR3 agonist poly(I:C) even 5 days postexposure conferred protection, avoiding the need to increase the vaccination dose. Efficacious treatments prevented death, ameliorated disease symptoms, reduced viral load and maintained tissue integrity of target organs. Protection was associated with significant elevation of serum IFNα and anti-vaccinia IgM antibodies, modulation of IFNγ response, and balanced activation of NK and T cells. TLR9 agonists (CpG ODNs) were less protective than the TLR3 agonist poly(I:C). We show that activation of type 1 IFN by poly(I:C) and protection is achievable even without co-vaccination, requiring sufficient amount of the viral antigens of the infective agent or the vaccine. This study demonstrated the therapeutic potential of postexposure immune modulation by TLR activation, allowing to alleviate the disease symptoms and to further extend the protective window of postexposure vaccination.


Subject(s)
Smallpox Vaccine/immunology , Smallpox/prevention & control , Toll-Like Receptor 3/agonists , Toll-Like Receptor 9/agonists , Adaptive Immunity/drug effects , Animals , Disease Models, Animal , Ectromelia virus/immunology , Ectromelia, Infectious/metabolism , Ectromelia, Infectious/mortality , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Female , Immunomodulation/drug effects , Interferon-gamma/blood , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Poly I-C/administration & dosage , Poly I-C/pharmacology , Smallpox/metabolism , Smallpox Vaccine/administration & dosage , Vaccination , Vaccines, Attenuated , Viral Load
11.
Antiviral Res ; 111: 42-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25128688

ABSTRACT

Natural orthopoxvirus outbreaks such as vaccinia, cowpox, cattlepox and buffalopox continue to cause morbidity in the human population. Monkeypox virus remains a significant agent of morbidity and mortality in Africa. Furthermore, monkeypox virus's broad host-range and expanding environs make it of particular concern as an emerging human pathogen. Monkeypox virus and variola virus (the etiological agent of smallpox) are both potential agents of bioterrorism. The first line response to orthopoxvirus disease is through vaccination with first-generation and second-generation vaccines, such as Dryvax and ACAM2000. Although these vaccines provide excellent protection, their widespread use is impeded by the high level of adverse events associated with vaccination using live, attenuated virus. It is possible that vaccines could be used in combination with antiviral drugs to reduce the incidence and severity of vaccine-associated adverse events, or as a preventive in individuals with uncertain exposure status or contraindication to vaccination. We have used the intranasal mousepox (ectromelia) model to evaluate the efficacy of vaccination with Dryvax or ACAM2000 in conjunction with treatment using the broad spectrum antiviral, brincidofovir (BCV, CMX001). We found that co-treatment with BCV reduced the severity of vaccination-associated lesion development. Although the immune response to vaccination was quantifiably attenuated, vaccination combined with BCV treatment did not alter the development of full protective immunity, even when administered two days following ectromelia challenge. Studies with a non-replicating vaccine, ACAM3000 (MVA), confirmed that BCV's mechanism of attenuating the immune response following vaccination with live virus was, as expected, by limiting viral replication and not through inhibition of the immune system. These studies suggest that, in the setting of post-exposure prophylaxis, co-administration of BCV with vaccination should be considered a first response to a smallpox emergency in subjects of uncertain exposure status or as a means of reduction of the incidence and severity of vaccine-associated adverse events.


Subject(s)
Antiviral Agents/administration & dosage , Cytosine/analogs & derivatives , Ectromelia virus/physiology , Ectromelia, Infectious/prevention & control , Organophosphonates/administration & dosage , Smallpox Vaccine/administration & dosage , Animals , Cytosine/administration & dosage , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/virology , Female , Humans , Immunity , Mice , Mice, Inbred C57BL , Smallpox Vaccine/immunology , Vaccination , Virus Replication
12.
Virol J ; 10: 229, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23842430

ABSTRACT

Vaccinia virus protein A33 (A33VACV) plays an important role in protection against orthopoxviruses, and hence is included in experimental multi-subunit smallpox vaccines. In this study we show that single-dose vaccination with recombinant Sindbis virus expressing A33VACV, is sufficient to protect mice against lethal challenge with vaccinia virus WR (VACV-WR) and ectromelia virus (ECTV) but not against cowpox virus (CPXV), a closely related orthopoxvirus. Moreover, a subunit vaccine based on the cowpox virus A33 ortholog (A33CPXV) failed to protect against cowpox and only partially protected mice against VACV-WR challenge. We mapped regions of sequence variation between A33VACV and A33CPXVand analyzed the role of such variations in protection. We identified a single protective region located between residues 104-120 that harbors a putative H-2Kd T cell epitope as well as a B cell epitope - a target for the neutralizing antibody MAb-1G10 that blocks spreading of extracellular virions. Both epitopes in A33CPXV are mutated and predicted to be non-functional. Whereas vaccination with A33VACV did not induce in-vivo CTL activity to the predicted epitope, inhibition of virus spread in-vitro, and protection from lethal VACV challenge pointed to the B cell epitope highlighting the critical role of residue L118 and of adjacent compensatory residues in protection. This epitope's critical role in protection, as well as its modifications within the orthopoxvirus genus should be taken in context with the failure of A33 to protect against CPXV as demonstrated here. These findings should be considered when developing new subunit vaccines and monoclonal antibody based therapeutics against orthopoxviruses, especially variola virus, the etiologic agent of smallpox.


Subject(s)
Cowpox virus/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Membrane Glycoproteins/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Adaptive Immunity , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Drug Carriers , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Variation , Genetic Vectors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Sindbis Virus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage
13.
J Virol ; 86(18): 9748-59, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740418

ABSTRACT

The antigens recognized by individual CD8(+) T cells are small peptides bound to major histocompatibility complex (MHC) class I molecules. The CD8(+) T cell response to a virus is restricted to several peptides, and the magnitudes of the effector as well as memory phases of the response to the individual peptides are generally hierarchical. The peptide eliciting a stronger response is called immunodominant (ID), and those with smaller-magnitude responses are termed subdominant (SD). The relative importance of ID and SD determinants in protective immunity remains to be fully elucidated. We previously showed that multispecific memory CD8(+) T cells can protect susceptible mice from mousepox, an acute lethal viral disease. It remained unknown, however, whether CD8(+) T cells specific for single ID or SD peptides could be protective. Here, we demonstrate that immunization with dendritic cells pulsed with ID and some but not all SD peptides induces memory CD8(+) T cells that are fully capable of protecting susceptible mice from mousepox. Additionally, while natural killer (NK) cells are essential for the natural resistance of nonimmune C57BL/6 (B6) to mousepox, we show that memory CD8(+) T cells of single specificity also protect B6 mice depleted of NK cells. This suggests it is feasible to produce effective antiviral CD8(+) T cell vaccines using single CD8(+) T cell determinants and that NK cells are no longer essential when memory CD8(+) T cells are present.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Amino Acid Sequence , Animals , Ectromelia virus/genetics , Ectromelia, Infectious/immunology , Ectromelia, Infectious/virology , H-2 Antigens/genetics , H-2 Antigens/immunology , Immunization , Immunodominant Epitopes/genetics , Immunologic Memory , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
14.
Vaccine ; 29(52): 9691-6, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-21983358

ABSTRACT

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


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Gene Deletion , RNA-Binding Proteins/genetics , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Viral Proteins/genetics , Virulence Factors/genetics , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Body Weight , Cricetinae , Ectromelia, Infectious/immunology , Ectromelia, Infectious/mortality , Ectromelia, Infectious/pathology , Female , Mice , Pregnancy , Smallpox Vaccine/administration & dosage , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viremia/prevention & control
15.
Immunol Cell Biol ; 89(6): 706-15, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21173782

ABSTRACT

Using Toll-like receptor (TLR) and MyD88 gene knock-out (GKO) mice the effect of TLRs and MyD88 on virus replication, interferon (IFN)-ß production, natural killer (NK) cell and CD8T cell responses were assessed following ectromelia virus (ECTV) and recombinant vaccinia virus (rVV) infection. The capacity for rVVs encoding cytokines to restore immune function in MyD88(-/-) mice was clearly demonstrated. Results showed that TLR2(-/-), TLR4(-/-)and TLR7(-/-) mice survived ECTV infection whereas MyD88(-/-) and TLR9(-/-)mice, in contrast, were highly susceptible. Next, following infection with rVV, MyD88(-/-) mice elicited reduced serum IFN-ß, NK cell and CD8T cell responses compared with wild-type mice, whereas TLR9(-/-) mice showed elevated CD8T cell responses. When MyD88(-/-)mice were infected with rVV co-expressing IFN-ß these mice were able to restore IFN-ß levels and CD8T cell responses but not NK cell activation. Interestingly, even though rVV co-expressing interleukin (IL)-2 enhanced NK cell activation in MyD88(-/-) mice, this was not associated with an antiviral effect, as observed in normal mice. Surprisingly, co-infection with rVV IL-2/rVV IL-12, but not rVV IL-2/rVV IFN-ß, restored the attenuated phenotype of rVV IL-2 in MyD88(-/-) mice indicating that the IL-2/IL-12 combination promotes antiviral responses. Our results clearly show that the CD8T cell defect observed in MyD88(-/-) mice to vaccinia virus infection can be restored by rVV-encoding IFN-ß demonstrating the critical role of this cytokine in T cell mediated immunity and illustrates that the model can provide an effective platform for the elucidation of cytokine immunobiology.


Subject(s)
Cytokines/genetics , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptors/genetics , Vaccinia virus/genetics , Vaccinia virus/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Chlorocebus aethiops , Cytokines/metabolism , Ectromelia virus/physiology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/prevention & control , Female , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-12/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Virus Replication/immunology
16.
Virology ; 385(1): 11-21, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19100593

ABSTRACT

The intranasal lethal mousepox model employing the A/Ncr mouse strain is used to evaluate anti-orthopoxvirus therapies. These infections mimic large droplet transmission and result in 100% mortality within 7-10 days with as little as 1 PFU of ectromelia virus. Unlike the A/Ncr model, humans are less susceptible to lethal respiratory infections with variola virus and monkeypox virus as demonstrated by their lower mortality rates. In this study we show that a low dose intranasal infection of C57BL/6 mice results in 60-80% mortality and better models smallpox. Comparing CMX001 (HDP-cidofovir) efficacy in the A/Ncr strain and the C57BL/6 strain revealed that delayed treatment with CMX001 is more efficacious at preventing severe disease in the C57BL/6 strain. The increased efficacy of CMX001 in C57BL/6 over A/Ncr following an intranasal infection with ectromelia appears to be mediated by a stronger Th1 cell mediated response. Following footpad infection we show that the C57BL/6 strain has earlier and more robust transcriptional activity, Th1 cytokine secretions, antigen presenting activity and IFNgamma splenic CD8+ T cell responses as compared to the A/Ncr strain. As a result of the enhanced immune response in the C57BL/6 strain, non-lethal intradermal ectromelia infections can therapeutically protect up to 3 days following a homologous, lethal intranasal infection - much like how smallpox vaccination can protect humans for up to 4 days following intranasal variola infection.


Subject(s)
Antiviral Agents/therapeutic use , Cytosine/analogs & derivatives , Disease Models, Animal , Ectromelia virus/physiology , Ectromelia, Infectious/prevention & control , Organophosphonates/therapeutic use , Animals , Cell Line , Chlorocebus aethiops , Cytokines/immunology , Cytosine/therapeutic use , Ectromelia, Infectious/immunology , Ectromelia, Infectious/mortality , Female , Mice , Mice, Inbred C57BL
17.
J Immunol ; 180(11): 7158-66, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490714

ABSTRACT

The type I IFNs exert a range of activities that include antiviral, antiproliferative, and immunomodulatory effects. To study this further, we have constructed recombinant vaccinia viruses expressing HIV or hemagglutinin (HA) Ags along with murine type I IFNs, IFN-alpha(4) (HA-VV-IFN-alpha(4)), IFN-beta (HA-VV-IFN-beta), or IFN-epsilon (HIV-VV-IFN-epsilon), a recently discovered member of this family. Our aims were to characterize IFN-epsilon functionality as a type I IFN and also to study the biological properties of these factors toward the development of safer and more effective vector-based vaccines. HIV-VV-IFN-epsilon and HA-VV-IFN-beta grew to lower titers than did their parental controls in murine cell lines. In vivo, however, HIV-VV-IFN-epsilon growth was not attenuated, while IFN-beta demonstrated potent local antiviral activity with no replication of HA-VV-IFN-beta detected. Flow cytofluorometric analysis of B lymphocytes incubated with virally encoded IFN-epsilon showed up-regulation of activation markers CD69 and CD86, while RT-PCR of IFN-epsilon-treated cells revealed that gene expression levels of antiviral proteins were elevated, indicating the induction of an antiviral state. The use of these constructs in a poxvirus prime-boost immunization regime led to robust humoral and cellular immune responses against the encoded Ags, despite the lack of replication in the case of HA-VV-IFN-beta. Thus, coexpression of these factors may be beneficial in the design of safer vector-based vaccines. Our data also indicate that while IFN-epsilon exhibits certain biological traits similar to other type I IFNs, it may also have a specific role in mucosal immune regulation that is quite distinct.


Subject(s)
Interferon Type I/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Animals , Cell Line , Ectromelia virus/pathogenicity , Ectromelia, Infectious/immunology , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Female , Genetic Vectors , HIV/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization, Secondary , Interferon Type I/metabolism , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Up-Regulation , Vaccinia/immunology , Vaccinia virus/growth & development , Vaccinia virus/pathogenicity
18.
J Exp Med ; 205(4): 981-92, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18391063

ABSTRACT

Nonliving antiviral vaccines traditionally target proteins expressed at the surface of the virion with the hope of inducing neutralizing antibodies. Orthopoxviruses (OPVs), such as the human smallpox virus and the mouse-equivalent ectromelia virus (ECTV; an agent of mousepox), encode immune response modifiers (IRMs) that can increase virulence by decreasing the host immune response. We show that one of these IRMs, the type I interferon (IFN) binding protein (bp) of ECTV, is essential for ECTV virulence and is a natural target of the antibody response. More strikingly, we demonstrate that immunization with recombinant type I IFN bp protects mice from lethal mousepox. Collectively, our experiments have important implications for our understanding of the role of IRMs in OPV virulence and of type I IFNs in OPV infections. Furthermore, our work provides proof of concept that effective antiviral vaccines can be made to prevent disease by targeting virulence factors as an alternative to the traditional approach that attempts to prevent infection by virus neutralization.


Subject(s)
Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Vaccination , Viral Proteins/immunology , Animals , Antibody Formation/immunology , Ectromelia virus/isolation & purification , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Genetic Complementation Test , Immunity, Innate/immunology , Immunocompetence , Interferon Type I/immunology , Mice , Open Reading Frames/genetics , Protein Binding , Receptors, Interferon/deficiency , Recombinant Proteins , Tissue Culture Techniques , Virulence
19.
Antiviral Res ; 77(1): 39-49, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17904231

ABSTRACT

In the 21st century we are faced with the potential use of natural or recombinant VARV and MPXV as biological weapons, and the emergence of human MPXV. Such an occurrences would require therapeutic and prophylactic intervention with antivirals. Cidofovir, an antiviral approved for the treatment of cytomegalovirus retinitis in AIDS patients, has activity against poxviruses, but must be administered intravenously and is associated with nephrotoxicity. An ether-lipid analogue of CDV, CMX001 (HDP-CDV), has potent antiviral activity against a range of DNA viruses including poxviruses, excellent oral bioavailability and minimal nephrotoxicity. CMX001 and CDV are equally efficacious at protecting mice from mortality following high ectromelia virus doses (10,000 x LD(50)) introduced by the intra-nasal route or small particle aerosol. Using CMX001 at a 10mg/kg dose followed by 2.5mg/kg doses every other-day for 14 days provided solid protection against mortality and weight loss following an intra-nasal challenge of (100-200) x LD(50) of ectromelia virus. Furthermore, complete protection against mortality was achieved when administration was delayed until as late as 5 days post-infection, which is 3-4 days prior to the death of the untreated controls. This therapeutic window would be equivalent to intervening during the rash stage of ordinary smallpox.


Subject(s)
Antiviral Agents/therapeutic use , Cytosine/analogs & derivatives , Ectromelia virus/drug effects , Ectromelia, Infectious/drug therapy , Organophosphonates/therapeutic use , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Cidofovir , Cytosine/administration & dosage , Cytosine/metabolism , Cytosine/therapeutic use , Ectromelia, Infectious/prevention & control , Ectromelia, Infectious/virology , Ether , Female , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred A , Organophosphonates/administration & dosage , Organophosphonates/metabolism
20.
Vaccine ; 25(7): 1214-24, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17098336

ABSTRACT

The heightened concern about the intentional release of variola virus has led to the need to develop safer smallpox vaccines. While subunit vaccine strategies are safer than live virus vaccines, subunit vaccines have been hampered by the need for multiple boosts to confer optimal protection. Here we developed a protein-based subunit vaccine strategy that provides rapid protection in mouse models of orthopoxvirus infections after a prime and single boost. Mice vaccinated with vaccinia virus envelope proteins from the mature virus (MV) and extracellular virus (EV) adjuvanted with CpG ODN and alum were protected from lethal intranasal challenge with vaccinia virus and the mouse-specific ectromelia virus. Organs from mice vaccinated with three proteins (A33, B5 and L1) and then sacrificed after challenge contained significantly lower titers of virus when compared to control groups of mice that were not vaccinated or that received sub-optimal formulations of the vaccine. Sera from groups of mice obtained prior to challenge had neutralizing activity against the MV and also inhibited comet formation indicating anti-EV activity. Long-term partial protection was also seen in mice challenged with vaccinia virus 6 months after initial vaccinations. Thus, this work represents a step toward the development of a practical subunit smallpox vaccine.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/prevention & control , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Vaccinia/prevention & control , Viral Proteins/immunology , Adjuvants, Immunologic/pharmacology , Animals , Chemistry, Pharmaceutical , Comet Assay , Female , Immunization Schedule , Immunization, Secondary , Mice , Mice, Inbred BALB C , Neutralization Tests , Reverse Transcriptase Polymerase Chain Reaction , Survival , Viral Proteins/biosynthesis , Viral Proteins/genetics , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...