Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 13(6)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34203773

RESUMO

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.


Assuntos
Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/patologia , Ectromelia Infecciosa/virologia , Genômica , Filogenia , Animais , Modelos Animais de Doenças , Vírus da Ectromelia/classificação , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Feminino , Evasão da Resposta Imune , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Filogeografia , Proteínas Virais/genética , Virulência
2.
Sci Rep ; 10(1): 13167, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32759969

RESUMO

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.


Assuntos
Ectromelia Infecciosa/prevenção & controle , Antígeno HLA-B7/genética , Peptídeos/imunologia , Vaccinia virus/imunologia , Proteínas Virais/química , Animais , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Antígeno HLA-B7/metabolismo , Epitopos Imunodominantes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
3.
PLoS Pathog ; 15(12): e1008239, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31877196

RESUMO

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.


Assuntos
Medula Óssea/virologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/virologia , Nucleotídeos Cíclicos/metabolismo , Animais , Medula Óssea/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/metabolismo , Camundongos Transgênicos , Nucleotidiltransferases/genética , Proteínas Serina-Treonina Quinases/metabolismo
4.
Virology ; 518: 335-348, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29602068

RESUMO

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.


Assuntos
Vírus da Ectromelia/imunologia , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/prevenção & controle , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vacinas Virais/imunologia , Replicação Viral , Animais , Linhagem Celular , Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Técnicas de Inativação de Genes , Humanos , Camundongos Endogâmicos BALB C , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
5.
Vaccine ; 35(33): 4245-4254, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28625523

RESUMO

Eczema vaccinatum is a severe and occasionally lethal complication of smallpox vaccine, characterized by systemic viral dissemination, distant from the initial inoculation site of the vaccine. A major risk factor for eczema vaccinatum is a background of atopic dermatitis, a chronic, common allergic, relapsing disorder, manifested by dry and inflamed skin, itchy rash, Th2 biased immune response and hypersensitivity to various antigens. Unlike the severe manifestations of eczema vaccinatum in humans, current models present only mild symptoms that limits examination of potential therapeutics for eczema vaccinatum. The atopic dermatitis and eczema vaccinatum models we present here, are the first to simulate the severity of the diseases in humans. Indeed, dermatitic mice display persistent severe dermatitis, characterized by dry and inflamed skin with barrier dysfunction, epidermal hyperplasia and significant elevation of serum IgE. By exposing atopic dermatitis mice to ectromelia virus, we generated eczema vaccinatum that mimic the human disease better than known eczema vaccinatum models. Similarly to humans, eczematous mice displayed enlarged and disseminated skin lesions, which correlated with elevated viral load. Cidofovir and antiviral antibodies conferred protection even when treatment started at a late eczematous stage. Moreover, we are the first to demonstrate that despite a severe background of atopic dermatitis, modified vaccinia Ankara virus (MVA) vaccination protects against lethal ectromelia virus exposure. We finally show that protection by MVA vaccination is dependent on CD4+ T cells and is associated with significant activation of CD8+ cytotoxic T cells and induction of humoral immunity.


Assuntos
Dermatite Atópica/complicações , Modelos Animais de Doenças , Vírus da Ectromelia/imunologia , Erupção Variceliforme de Kaposi/tratamento farmacológico , Erupção Variceliforme de Kaposi/prevenção & controle , Vacina Antivariólica/administração & dosagem , Vacina Antivariólica/efeitos adversos , Animais , Anticorpos Antivirais/administração & dosagem , Antivirais/administração & dosagem , Linfócitos T CD4-Positivos , Cidofovir , Citosina/administração & dosagem , Citosina/análogos & derivados , Vírus da Ectromelia/patogenicidade , Feminino , Humanos , Erupção Variceliforme de Kaposi/patologia , Camundongos , Organofosfonatos/administração & dosagem
6.
Microb Pathog ; 109: 99-109, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28554653

RESUMO

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.


Assuntos
Apoptose , Células Dendríticas/imunologia , Vírus da Ectromelia/fisiologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Imunidade Adaptativa , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3 , Chlorocebus aethiops , Células Dendríticas/patologia , Células Dendríticas/fisiologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Ectromelia Infecciosa/virologia , Proteína Ligante Fas/metabolismo , Regulação da Expressão Gênica , Imunidade Inata , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Regulação para Cima , Células Vero
7.
PLoS One ; 12(3): e0173697, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28282449

RESUMO

It is often not possible to demonstrate causality within the context of gut microbiota dysbiosis-linked diseases. Thus, we need a better understanding of the mechanisms whereby an altered host immunophysiology shapes its resident microbiota. In this regard, immune-modulating poxvirus strains and mutants could differentially alter gut mucosal immunity in the context of a natural immune response, providing a controlled natural in vivo setting to deepen our understanding of the immune determinants of microbiome composition. This study represents a proof-of-concept that the use of an existing collection of different immune-modulating poxviruses may represent an innovative tool in gut microbiome research. To this end, 16S rRNA amplicon sequencing and RNAseq transcriptome profiling were employed as proxies for microbiota composition and gut immunophysiological status in the analysis of caecal samples from control mice and mice infected with various poxvirus types. Our results show that different poxvirus species and mutants elicit different shifts in the mice mucosa-associated microbiota and, in some instances, significant concomitant shifts in gut transcriptome profiles, thus providing an initial validation to the proposed model.


Assuntos
Microbioma Gastrointestinal/fisiologia , Infecções por Poxviridae/imunologia , Poxviridae/patogenicidade , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Feminino , Microbioma Gastrointestinal/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos Endogâmicos BALB C , Mutação , Poxviridae/genética , Poxviridae/imunologia , Infecções por Poxviridae/microbiologia , Infecções por Poxviridae/fisiopatologia , RNA Ribossômico 16S , Vaccinia virus/genética , Vaccinia virus/patogenicidade
8.
Biomed Res Int ; 2017: 6456180, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29430463

RESUMO

Ectromelia virus (ECTV), the causative agent of mousepox, has emerged as a valuable model for investigating the host-Orthopoxvirus relationship as it relates to pathogenesis and the immune response. ECTV is a mouse-specific virus and causes high mortality in susceptible mice strains, including BALB/c and C3H, whereas C57BL/6 and 129 strains are resistant to the disease. To understand the host genetic factors in different mouse strains during the ECTV infection, we carried out a microarray analysis of spleen tissues derived from BALB/c and C57BL/6 mice, respectively, at 3 and 10 days after ECTV infection. Differential Expression of Genes (DEGs) analyses revealed distinct differences in the gene profiles of susceptible and resistant mice. The susceptible BALB/c mice generated more DEGs than the resistant C57BL/6 mice. Additionally, gene ontology and KEGG pathway analysis showed the DEGs of susceptible mice were involved in innate immunity, apoptosis, metabolism, and cancer-related pathways, while the DEGs of resistant mice were largely involved in MAPK signaling and leukocyte transendothelial migration. Furthermore, the BALB/c mice showed a strong induction of interferon-induced genes, which, however, were weaker in the C57BL/6 mice. Collectively, the differential transcriptome profiles of susceptible and resistant mouse strains with ECTV infection will be crucial for further uncovering the molecular mechanisms of the host-Orthopoxvirus interaction.


Assuntos
Resistência à Doença/genética , Ectromelia Infecciosa/genética , Interações Hospedeiro-Parasita/genética , Transcriptoma/genética , Animais , Suscetibilidade a Doenças/virologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/patologia , Ectromelia Infecciosa/virologia , Regulação da Expressão Gênica , Imunidade Inata/genética , Interferons/genética , Camundongos , Baço/metabolismo , Baço/virologia
9.
Virology ; 501: 107-114, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27898336

RESUMO

Most poxviruses encode a homolog of a ~200,000-kDa membrane protein originally identified in variola virus. We investigated the importance of the ectromelia virus (ECTV) homolog C15 in a natural infection model. In cultured mouse cells, the replication of a mutant virus with stop codons near the N-terminus (ECTV-C15Stop) was indistinguishable from a control virus (ECTV-C15Rev). However, for a range of doses injected into the footpads of BALB/c mice there was less mortality with the mutant. Similar virus loads were present at the site of infection with mutant or control virus whereas there was less ECTV-C15Stop in popliteal and inguinal lymph nodes, spleen and liver indicating decreased virus spread and replication. The latter results were supported by immunohistochemical analyses. Decreased spread was evidently due to immune modulatory activity of C15, rather than to an intrinsic viral function, as the survival of infected mice depended on CD4+ and CD8+ T cells.


Assuntos
Vírus da Ectromelia/metabolismo , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/metabolismo , Ectromelia Infecciosa/virologia , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Animais , Modelos Animais de Doenças , Vírus da Ectromelia/genética , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/patologia , Feminino , Humanos , Fígado/patologia , Fígado/virologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Baço/patologia , Baço/virologia , Vírus da Varíola/genética , Vírus da Varíola/metabolismo , Proteínas Virais/genética , Virulência
10.
Adv Immunol ; 129: 251-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26791861

RESUMO

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.


Assuntos
Imunidade Adaptativa , Resistência à Doença/imunologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Varíola/imunologia , Vaccinia virus/imunologia , Animais , Modelos Animais de Doenças , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/prevenção & controle , Ectromelia Infecciosa/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Proteínas Virais/imunologia , Virulência
11.
J Virol ; 89(19): 9974-85, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26202250

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Citocinas/biossíntese , Células Dendríticas/classificação , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/transmissão , Ectromelia Infecciosa/virologia , Granulócitos/imunologia , Humanos , Imunidade Inata , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Replicação Viral , Zoonoses/imunologia , Zoonoses/transmissão , Zoonoses/virologia
12.
Artigo em Russo | MEDLINE | ID: mdl-25842954

RESUMO

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.


Assuntos
Alcenos/administração & dosagem , Antivirais/administração & dosagem , Vírus da Ectromelia/efeitos dos fármacos , Ectromelia Infecciosa/tratamento farmacológico , Hidrazinas/administração & dosagem , Animais , Benzamidas/administração & dosagem , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/prevenção & controle , Ectromelia Infecciosa/virologia , Humanos , Isoindóis/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/virologia , Camundongos , Baço/efeitos dos fármacos , Baço/virologia
13.
PLoS One ; 10(3): e0118685, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25751266

RESUMO

Ectromelia virus (ECTV) causes mousepox in mice, a disease very similar to smallpox in humans. ECTV and variola virus (VARV), the agent of smallpox, are closely related orthopoxviruses. Mousepox is an excellent small animal model to study the genetic and immunologic basis for resistance and susceptibility of humans to smallpox. Resistance to mousepox is dependent on a strong polarized type 1 immune response, associated with robust natural killer (NK) cell, cytotoxic T lymphocyte (CTL) and gamma interferon (IFN-γ) responses. In contrast, ECTV-susceptible mice generate a type 2 response, associated with weak NK cell, CTL and IFN-γ responses but robust IL-4 responses. Nonetheless, susceptible strains infected with mutant ECTV lacking virus-encoded IFN-γ binding protein (vIFN-γbp) (ECTV-IFN-γbpΔ) control virus replication through generation of type 1 response. Since the IL-4/IL-13/STAT-6 signaling pathways polarize type 2/T helper 2 (Th2) responses with a corresponding suppression of IFN-γ production, we investigated whether the combined absence of vIFN-γbp, and one or more host genes involved in Th2 response development, influence generation of protective immunity. Most mutant mouse strains infected with wild-type (WT) virus succumbed to disease more rapidly than WT animals. Conversely, the disease outcome was significantly improved in WT mice infected with ECTV-IFN-γbpΔ but absence of IL-4/IL-13/STAT-6 signaling pathways did not provide any added advantage. Deficiency in IL-13 or STAT-6 resulted in defective CTL responses, higher mortality rates and accelerated deaths. Deficiencies in IL-4/IL-13/STAT-6 signaling pathways significantly reduced the numbers of IFN-γ producing CD4 and CD8 T cells, indicating an absence of a switch to a Th1-like response. Factors contributing to susceptibility or resistance to mousepox are far more complex than a balance between Th1 and Th2 responses.


Assuntos
Citocinas/genética , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Células Th2/metabolismo , Proteínas Virais/genética , Animais , Linhagem Celular , Citocinas/metabolismo , Suscetibilidade a Doenças , Vírus da Ectromelia/genética , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/mortalidade , Ectromelia Infecciosa/virologia , Técnicas de Silenciamento de Genes , Interferon gama/genética , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais , Proteínas Virais/imunologia
14.
PLoS Pathog ; 10(8): e1004326, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25122471

RESUMO

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.


Assuntos
Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/metabolismo , NF-kappa B/metabolismo , Proteínas Virais/metabolismo , Animais , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/metabolismo , Ectromelia Infecciosa/imunologia , Citometria de Fluxo , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , NF-kappa B/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/imunologia , Virulência/fisiologia
15.
Virology ; 456-457: 108-20, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24889230

RESUMO

Currently, little is known about the ankyrin/F-box protein B4. Here, we report that B4R-null viruses exhibited reduced plaque size in tissue culture, and decreased ability to spread, as assessed by multiple-step growth analysis. Electron microscopy indicated that B4R-null viruses still formed mature and extracellular virions; however, there was a slight decrease of virions released into the media following deletion of B4R. Deletion of B4R did not affect the ability of the virus to rearrange actin; however, VACV811, a large vaccinia virus deletion mutant missing 55 open reading frames, had decreased ability to produce actin tails. Using ectromelia virus, a natural mouse pathogen, we demonstrated that virus devoid of EVM154, the B4R homolog, showed decreased spread to organs and was attenuated during infection. This initial characterization suggests that B4 may play a role in virus spread, and that other unidentified mediators of actin tail formation may exist in vaccinia virus.


Assuntos
Vaccinia virus/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Estruturas Animais/virologia , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/patologia , Ectromelia Infecciosa/virologia , Feminino , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Vaccinia virus/genética , Vaccinia virus/ultraestrutura , Carga Viral , Ensaio de Placa Viral , Proteínas Virais/genética , Vírion/ultraestrutura , Virulência
16.
Acta Biochim Pol ; 61(1): 171-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24660173

RESUMO

Mitochondria are extremely important organelles in the life of a cell. Recent studies indicate that mitochondria also play a fundamental role in the cellular innate immune mechanisms against viral infections. Moreover, mitochondria are able to alter their shape continuously through fusion and fission. These tightly regulated processes are activated or inhibited under physiological or pathological (e.g. viral infection) conditions to help restore homeostasis. However, many types of viruses, such as orthopoxviruses, have developed various strategies to evade the mitochondrial-mediated antiviral innate immune responses. Moreover, orthopoxviruses exploit the mitochondria for their survival. Such viral activity has been reported during vaccinia virus (VACV) infection. Our study shows that the Moscow strain of ectromelia virus (ECTV-MOS), an orthopoxvirus, alters the mitochondrial network in permissive L929 cells. Upon infection, the branching structure of the mitochondrial network collapses and becomes disorganized followed by destruction of mitochondrial tubules during the late stage of infection. Small, discrete mitochondria co-localize with progeny virions, close to the cell membrane. Furthermore, clustering of mitochondria is observed around viral factories, particularly between the nucleus and viroplasm. Our findings suggest that ECTV-MOS modulates mitochondrial cellular distribution during later stages of the replication cycle, probably enabling viral replication and/or assembly as well as transport of progeny virions inside the cell. However, this requires further investigation.


Assuntos
Vírus da Ectromelia/genética , Imunidade Inata/genética , Mitocôndrias/genética , Animais , Linhagem Celular , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/virologia , Camundongos , Mitocôndrias/virologia , Vaccinia virus/genética
17.
Viral Immunol ; 26(5): 322-35, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24116707

RESUMO

Several studies have provided evidence that complex relationships between autophagic and apoptotic cell death pathways occur in cancer and virus-infected cells. Previously, we demonstrated that infection of macrophages with Moscow strain of ectromelia virus (ECTV-MOS) induces apoptosis under in vitro and in vivo conditions. Here, we found that autophagy was induced in RAW 264.7 cells during infection with ECTV-MOS. Silencing of beclin 1, an autophagy-related gene, reduced the percentage of late apoptotic cells in virus-infected RAW 264.7 macrophages. Pharmacological modulation of autophagy by wortmannin (inhibitor) or rapamycin (inductor) did not affect or cause increased apoptosis in ECTV-MOS-infected RAW 264.7 cells, respectively. Meantime, blocking apoptosis by a pan-caspase inhibitor, Z-VAD-FMK, increased the formation of autophagosomes in infected macrophages. Taken together, three important points arise from our study. First, autophagy may co-occur with apoptosis in RAW 264.7 cells exposed to ECTV-MOS. Second, at later stages of infection, autophagy may partially participate in the execution of macrophage cell death by enhancing apoptosis. Third, when apoptosis is blocked infected macrophages undergo increased autophagy. Our results provide new information about the relationship between autophagy and apoptosis in ECTV-MOS-infected macrophages.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/imunologia , Autofagia/imunologia , Vírus da Ectromelia/imunologia , Macrófagos/imunologia , Clorometilcetonas de Aminoácidos/farmacologia , Androstadienos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína Beclina-1 , Inibidores de Caspase/farmacologia , Linhagem Celular , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Imunossupressores/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Sirolimo/farmacologia , Wortmanina
18.
J Virol ; 87(12): 7046-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23596297

RESUMO

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.


Assuntos
Citosol/virologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/terapia , Fígado/virologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/imunologia , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/metabolismo , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/mortalidade , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Fígado/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Vírion/metabolismo
19.
Virol J ; 9: 119, 2012 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-22709563

RESUMO

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.


Assuntos
Antivirais/administração & dosagem , Citosina/análogos & derivados , Vírus da Ectromelia/efeitos dos fármacos , Ectromelia Infecciosa/tratamento farmacológico , Organofosfonatos/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Cidofovir , Citosina/administração & dosagem , Modelos Animais de Doenças , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/patologia , Feminino , Imunoglobulina G/sangue , Interferon gama/sangue , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida
20.
J Virol ; 86(13): 7298-309, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22532670

RESUMO

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.


Assuntos
Vírus da Ectromelia/imunologia , Linfócitos T/imunologia , Vaccinia virus/imunologia , Animais , Degranulação Celular , Citocinas/biossíntese , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Vírus da Ectromelia/patogenicidade , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/imunologia , Feminino , Citometria de Fluxo , Granzimas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Vacínia/imunologia , Vaccinia virus/patogenicidade , Vaccinia virus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...