Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acta Virol ; 64(3): 307-324, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32985205

RESUMO

Mitochondria are multitasking organelles that play a central role in energy production, survival and primary host defense against viral infections. Therefore, viruses target mitochondria dynamics and functions to benefit their replication and morphogenetic processes. We endeavor to understand the role of mitochondria during infection of ectromelia virus (ECTV), hence our investigations on mitochondria-related genes in non-immune (L929 fibroblasts) and immune (RAW 264.7 macrophages) cells. Our results show that during later stages of infection, ECTV significantly decreases the expression of mitochondria-related genes regulating many aspects of mitochondrial physiology and functions, including mitochondrial transport, small molecule transport, membrane polarization and potential, targeting proteins to mitochondria, inner membrane translocation, and apoptosis. Such down-regulation is cell-specific, since macrophages exhibited a more profound down-regulation of mitochondria-related genes compared to infected L929 fibroblasts. Only L929 cells exhibited up-regulation of two important genes responsible for oxidative phosphorylation and subsequent ATP production: Slc25a23 and Slc25a31. Changes in the expression of mitochondria-related genes are accompanied by altered mitochondria morphology and distribution in both types of cells. In depth Ingenuity Pathway Analysis (IPA) identified the "Sirtuin Signaling Pathway" as the most significant top canonical pathway associated with ECTV infection in both analyzed cell types. Taken together, down-regulation of mitochondria-related genes observed especially in macrophages indicates dysfunctional mitochondria, possibly contributing to energy collapse and induction of intrinsic pathway of apoptosis. Meanwhile, alteration of the expression of several mitochondria-related genes in fibroblasts without apoptosis induction may represent poxviral strategy to control cellular energy metabolism for efficient replication. Keywords: ectromelia virus; mitochondria; fibroblasts; macrophages.


Assuntos
Ectromelia Infecciosa/genética , Fibroblastos , Macrófagos , Mitocôndrias/genética , Transcriptoma , Animais , Vírus da Ectromelia , Ectromelia Infecciosa/imunologia , Camundongos , Células RAW 264.7
2.
Int J Mol Med ; 42(2): 1044-1053, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29749430

RESUMO

The ectromelia virus (ECTV) is a mouse specific Orthopoxvirus that causes lethal infection in some mouse strains. ECTV infection of these mouse strains has been used as a valuable model for understanding the interplay between Orthopoxvirus species and their hosts, including variola virus in humans. Although poxviruses encode numerous proteins required for DNA and RNA synthesis, and are less dependent on host functions than other DNA viruses, a detailed understanding of the host factors required for the replication of poxviruses is lacking. Heat shock protein 70 (Hsp70) isoforms have been reported to serve various roles in the replication cycle of numerous viruses. In the present study, microarray and reverse transcription­quantitative polymerase chain reaction analysis were conducted to investigate the host gene expression profiles following ECTV infection in mice and cell cultures. The results indicated that one Hsp70 isoform, Hsp70 member 1B (Hspa1b), was highly upregulated during ECTV infection in vitro and in vivo. Subsequently, overexpression of Hspa1b protein and small interfering RNA­mediated gene silencing of Hspa1b revealed that Hspa1b is required for efficient replication of ECTV. Furthermore, the results demonstrated that ECTV replication may be significantly suppressed by two chemical Hspa1b inhibitors: Quercetin and VER155008. In conclusion, the present study clearly demonstrated that ECTV infection upregulates the expression of Hspa1b in order to promote its replication. The dependence on Hsp70 may be used as a novel therapeutic target for the treatment of Orthopoxvirus infection.


Assuntos
Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/virologia , Proteínas de Choque Térmico HSP70/genética , Interações Hospedeiro-Patógeno , Camundongos/virologia , Replicação Viral , Animais , Replicação do DNA , Masculino , Camundongos Endogâmicos BALB C , Regulação para Cima
3.
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
4.
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
5.
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
6.
Immunology ; 142(4): 603-13, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24673683

RESUMO

The c-Jun N-terminal kinase (JNK) signalling pathway appears to act as a critical intermediate in the regulation of lymphocyte activation and proliferation. The majority of studies on the importance of JNK are focused on its role in T helper responses, with very few reports addressing the mechanisms of JNK in governing CD8 T-cell-mediated immunity. By using a well-defined mousepox model, we demonstrate that JNK is involved in CD8(+) T-cell-mediated antiviral responses. Deficiency of either JNK1 or JNK2 impaired viral clearance, subsequently resulting in an increased susceptibility to ectromelia virus in resistant mice. The impairment of CD8 responses in JNK-deficient mice was not directly due to an inhibition of effector T-cell expansion, as both JNK1 and JNK2 had limited effect on the activation-induced cell death of CD8(+) T cells, and only JNK2-deficient mice exhibited a significant change in CD8(+) T-cell proliferation after acute ectromelia virus infection. However, optimal activation of CD8(+) T cells and their effector functions require signals from both JNK1 and JNK2. Our results suggest that the JNK pathway acts as a critical intermediate in antiviral immunity through regulation of the activation and effector function of CD8(+) T cells rather than by altering their expansion.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Proteína Quinase 8 Ativada por Mitógeno/imunologia , Proteína Quinase 9 Ativada por Mitógeno/imunologia , Transdução de Sinais/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/patologia , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/genética , Transdução de Sinais/genética
7.
Antiviral Res ; 94(1): 44-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22381921

RESUMO

The human population is currently faced with the potential use of natural or recombinant variola and monkeypox viruses as biological weapons. Furthermore, the emergence of human monkeypox in Africa and its expanding environs poses a significant natural threat. Such occurrences would require therapeutic and prophylactic intervention with antivirals to minimize morbidity and mortality of exposed populations. Two orally-bioavailable antivirals are currently in clinical trials; namely CMX001, an ether-lipid analog of cidofovir with activity at the DNA replication stage and ST-246, a novel viral egress inhibitor. Both of these drugs have previously been evaluated in the ectromelia/mousepox system; however, the trigger for intervention was not linked to a disease biomarker or a specific marker of virus replication. In this study we used lethal, intranasal, ectromelia virus infections of C57BL/6 and hairless SKH1 mice to model human disease and evaluate exanthematous rash (rash) as an indicator to initiate antiviral treatment. We show that significant protection can be provided to C57BL/6 mice by CMX001 or ST-246 when therapy is initiated on day 6 post infection or earlier. We also show that significant protection can be provided to SKH1 mice treated with CMX001 at day 3 post infection or earlier, but this is four or more days before detection of rash (ST-246 not tested). Although in this model rash could not be used as a treatment trigger, viral DNA was detected in blood by day 4 post infection and in the oropharyngeal secretions (saliva) by day 2-3 post infection - thus providing robust and specific markers of virus replication for therapy initiation. These findings are discussed in the context of current respiratory challenge animal models in use for the evaluation of poxvirus antivirals.


Assuntos
Benzamidas/administração & dosagem , Biomarcadores Farmacológicos/análise , Citosina/análogos & derivados , Ectromelia Infecciosa/tratamento farmacológico , Isoindóis/administração & dosagem , Monkeypox virus/efeitos dos fármacos , Organofosfonatos/administração & dosagem , Varíola/tratamento farmacológico , Animais , Linhagem Celular , Citosina/administração & dosagem , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Vírus da Ectromelia/efeitos dos fármacos , Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/virologia , Feminino , Humanos , Camundongos , Camundongos Pelados , Camundongos Endogâmicos C57BL , Monkeypox virus/fisiologia , Varíola/virologia , Vírus da Varíola/efeitos dos fármacos , Vírus da Varíola/genética , Vírus da Varíola/fisiologia , Replicação Viral/efeitos dos fármacos
8.
J Exp Med ; 207(11): 2369-81, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-20876312

RESUMO

Although it is well known that aged hosts are generally more susceptible to viral diseases than the young, specific dysfunctions of the immune system directly responsible for this increased susceptibility have yet to be identified. We show that mice genetically resistant to mousepox (the mouse parallel of human smallpox) lose resistance at mid-age. Surprisingly, this loss of resistance is not a result of intrinsically defective T cell responses. Instead, the primary reason for the loss of resistance results from a decreased number of total and mature natural killer (NK) cells in the blood and an intrinsic impairment in their ability to migrate to the lymph node draining the site of infection, which is essential to curb systemic virus spread. Hence, our work links the age-dependent increase in susceptibility to a viral disease to a specific defect of NK cells, opening the possibility of exploring treatments to improve NK cell function in the aged with the goal of enhancing their resistance to viral diseases.


Assuntos
Envelhecimento/imunologia , Movimento Celular/imunologia , Suscetibilidade a Doenças/imunologia , Ectromelia Infecciosa/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Envelhecimento/genética , Animais , Movimento Celular/genética , Ectromelia Infecciosa/genética , Humanos , Imunidade Inata/genética , Camundongos , Camundongos Mutantes , Varíola/genética , Varíola/imunologia
9.
Mol Biol (Mosk) ; 43(4): 616-22, 2009.
Artigo em Russo | MEDLINE | ID: mdl-19807023

RESUMO

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.


Assuntos
Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/genética , Deleção de Genes , Genes Virais/genética , Proteínas Virais/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus da Ectromelia/metabolismo , Ectromelia Infecciosa/metabolismo , Camundongos
10.
J Immunol ; 183(5): 3324-31, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19657092

RESUMO

A strong cell-mediated immune response is critical for controlling viral infections and is regulated by a number of cytokines, including IL-12 and IL-18. Indeed, some viruses have evolved to specifically target these pathways to counter the host immune response. Orthopoxviruses, including ectromelia virus, encode immune evasion molecules that specifically target IL-18 and IFN-gamma. We hypothesized that IL-12 and IL-18 are pivotal for induction of IFN-gamma production and subsequent generation of an effective host response to ectromelia virus infection. In this study, we demonstrate that absence of both IL-12p40 and IL-18 resulted in increased susceptibility to infection that was associated with skewing of the cytokine response to Th2 and a reduction in NK and CTL responses. The decrease in CTL response correlated with a defect in CD8(+) T cell proliferation and lower numbers of virus-specific CD8(+) T cells. Lack of either IL-12p40 and/or IL-18 was also associated with reduced numbers of CD8(+) T cells at sites of infection and with an increase in the numbers of splenic T regulatory cells. Taken together, our data indicate that IL-12p40 and IL-18 act in concert and play an important antiviral role through the up-regulation of IFN-gamma production and cell-mediated immune responses.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Subunidade p40 da Interleucina-12/fisiologia , Interleucina-18/fisiologia , Animais , Células Cultivadas , Citotoxicidade Imunológica/genética , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/mortalidade , Predisposição Genética para Doença/genética , Imunidade Celular/genética , Imunidade Inata/genética , Subunidade p40 da Interleucina-12/antagonistas & inibidores , Subunidade p40 da Interleucina-12/deficiência , Subunidade p40 da Interleucina-12/genética , Interleucina-18/deficiência , Interleucina-18/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/patologia
11.
PLoS Pathog ; 4(12): e1000249, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19112490

RESUMO

Poxviruses subvert the host immune response by producing immunomodulatory proteins, including a complement regulatory protein. Ectromelia virus provides a mouse model for smallpox where the virus and the host's immune response have co-evolved. Using this model, our study investigated the role of the complement system during a poxvirus infection. By multiple inoculation routes, ectromelia virus caused increased mortality by 7 to 10 days post-infection in C57BL/6 mice that lack C3, the central component of the complement cascade. In C3(-/-) mice, ectromelia virus disseminated earlier to target organs and generated higher peak titers compared to the congenic controls. Also, increased hepatic inflammation and necrosis correlated with these higher tissue titers and likely contributed to the morbidity in the C3(-/-) mice. In vitro, the complement system in naïve C57BL/6 mouse sera neutralized ectromelia virus, primarily through the recognition of the virion by natural antibody and activation of the classical and alternative pathways. Sera deficient in classical or alternative pathway components or antibody had reduced ability to neutralize viral particles, which likely contributed to increased viral dissemination and disease severity in vivo. The increased mortality of C4(-/-) or Factor B(-/-) mice also indicates that these two pathways of complement activation are required for survival. In summary, the complement system acts in the first few minutes, hours, and days to control this poxviral infection until the adaptive immune response can react, and loss of this system results in lethal infection.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/mortalidade , Animais , Complemento C3/genética , Complemento C4/genética , Fator B do Complemento/genética , Proteínas do Sistema Complemento/genética , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/patologia , Hepatite Viral Animal/genética , Hepatite Viral Animal/patologia , Hepatite Viral Animal/virologia , Imunidade Inata/genética , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/genética , Necrose/patologia , Necrose/veterinária , Necrose/virologia , Viremia/mortalidade , Replicação Viral/genética
12.
Novartis Found Symp ; 281: 129-36; discussion 136-40, 208-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17534070

RESUMO

There is increased interest in understanding protective immunity to smallpox for two principal reasons. First, it is the only disease that has been successfully eradicated using a live virus vaccine and, second, there exists a potential threat of intentional or unintentional release of variola virus, the causative agent of smallpox. Although mortality rates associated with smallpox were as high as 40%, a significant subset of those infected recovered. The basis of susceptibility or resistance, and the immune parameters associated with recovery, are still unknown. Animal models of poxvirus infections are being employed to understand what constitutes an effective host response. Ectromelia virus is closely related to variola virus and it causes a disease similar to smallpox in mice. This model is well established, resistant and susceptible strains of mice are defined and four genetic loci associated with resistance have been identified. Susceptibility to infec tion and disease severity is also influenced by virus immune evasion strategies. The outcome of infection is clearly dictated by several factors including host and viral genes, both of which influence the immune response. Here we present data on one virus-encoded immune modifier and its effect on the functions of two host genetic loci associ ated with resistance.


Assuntos
Modelos Animais de Doenças , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Predisposição Genética para Doença/genética , Varíola/imunologia , Animais , Ectromelia Infecciosa/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Varíola/genética
13.
J Virol ; 80(13): 6333-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775321

RESUMO

Renewed interest in smallpox and the need for safer vaccines have highlighted our lack of understanding of the requirements for protective immunity. Since smallpox has been eradicated, surrogate animal models of closely related orthopoxviruses, such as ectromelia virus, have been used to establish critical roles for CD8 T cells in the control of primary infection. To study the requirements for protection against secondary infection, we have used a prime-challenge regime, in which avirulent ectromelia virus was used to prime mice that were then challenged with virulent ectromelia virus. In contrast to primary infection, T cells are not required for recovery from secondary infection, since gene knockout mice deficient in CD8 T-cell function and wild-type mice acutely depleted of CD4, CD8, or both subsets were fully protected. Protection correlated with effective virus control and generation of neutralizing antibody. Notably, primed mice that lacked B cells, major histocompatibility complex class II, or CD40 succumbed to secondary infection. Thus, antibody is essential, but CD4 or CD8 T cells are not required for recovery from secondary poxvirus infection.


Assuntos
Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/genética , Antígenos CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Ectromelia Infecciosa/genética , Feminino , Genes MHC da Classe II/genética , Genes MHC da Classe II/imunologia , Humanos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Varíola/imunologia , Vacinas Atenuadas/imunologia
14.
Antiviral Res ; 66(1): 1-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15781125

RESUMO

Improved vaccines and therapies for virulent poxvirus infection are required, particularly in the light of recent threats of bioterrorism. Cidofovir (HPMPC) is an acyclic nucleoside analog with proven efficacy against poxviruses. Here, we evaluated HPMPC in mice given a recombinant ectromelia virus (ECTV) encoding interleukin-4 (ECTV-IL-4) that is highly immune suppressive. Mousepox-sensitive BALB/c mice given HPMPC for five consecutive days after infection were protected against the lethal effects of a control ECTV recombinant, although they suffered a chronic form of mousepox disease. High doses of the drug resulted in a milder localized disease. In contrast, HPMPC failed to protect mousepox-resistant C57BL/6 mice against ECTV-IL-4, although its lethal effects were delayed by five daily doses of 20 mg/kg or a single dose of 100 mg/kg. Higher daily doses further delayed mortality, although the majority of animals eventually succumbed to infection. It appears that HPMPC inhibited ECTV-IL-4 replication without clearance, with the virus having a lethal effect when the drug was removed. Resistance of ECTV-IL-4 to HPMPC treatment may relate to the virus's ability to inhibit antiviral cell-mediated immunity. Interestingly, ECTV-IL-4-mediated immune suppression was not accompanied by a reduction in systemic IFN-gamma expression, suggestive of an alternative or highly localized suppressive mechanism.


Assuntos
Antivirais/uso terapêutico , Citosina/análogos & derivados , Citosina/uso terapêutico , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Interleucina-4/fisiologia , Organofosfonatos/uso terapêutico , Animais , Antivirais/administração & dosagem , Cidofovir , Citosina/administração & dosagem , Vírus da Ectromelia/efeitos dos fármacos , Ectromelia Infecciosa/tratamento farmacológico , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Organofosfonatos/administração & dosagem
15.
Proc Natl Acad Sci U S A ; 101(24): 9057-62, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15184649

RESUMO

Ectromelia virus (ECTV), a natural mouse pathogen and an orthopoxvirus, has been used to investigate the correlation between polarized type 1 or type 2 immune responses and resistance to disease in poxvirus infections by using well defined resistant and susceptible mouse strains. Our data show that distinct differences exist in the cytokine profiles expressed in resistant and susceptible mice infected with ECTV. Resistant C57BL/6 mice generate a type 1 cytokine response [IFN-gamma, IL-2, and tumor necrosis factor (TNF)], within the first few days of infection, which is associated with strong cytotoxic T lymphocyte response (CTL) and recovery from ECTV infection. Susceptible strains of mice (BALB/c and A/J) on the other hand generate a type 2 cytokine response (IL-4 but little or no IFN-gamma and IL-2), which is associated with a weak or an absent CTL response, resulting in uncontrolled virus replication and death. Although deletion of IL-4 function alone did not change the outcome of infection in susceptible mice, the loss of IFN-gamma function in resistant mice abrogated natural killer (NK) cell and CTL effector functions resulting in fulminant disease and 100% mortality. Therefore, a clear link exists between the early production of specific type 1 cytokines, in particular, IFN-gamma, the nature of the cellular immune response, and disease outcome in this virus model. This finding in the mousepox model raises the possibility that inappropriate cytokine responses may result in increased susceptibility to smallpox in humans.


Assuntos
Citocinas/imunologia , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Citocinas/biossíntese , Ectromelia Infecciosa/genética , Feminino , Predisposição Genética para Doença , Imunidade Celular/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Testes de Neutralização , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Carga Viral
16.
J Virol ; 75(3): 1205-10, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11152493

RESUMO

Genetic resistance to clinical mousepox (ectromelia virus) varies among inbred laboratory mice and is characterized by an effective natural killer (NK) response and the early onset of a strong CD8(+) cytotoxic T-lymphocyte (CTL) response in resistant mice. We have investigated the influence of virus-expressed mouse interleukin-4 (IL-4) on the cell-mediated response during infection. It was observed that expression of IL-4 by a thymidine kinase-positive ectromelia virus suppressed cytolytic responses of NK and CTL and the expression of gamma interferon by the latter. Genetically resistant mice infected with the IL-4-expressing virus developed symptoms of acute mousepox accompanied by high mortality, similar to the disease seen when genetically sensitive mice are infected with the virulent Moscow strain. Strikingly, infection of recently immunized genetically resistant mice with the virus expressing IL-4 also resulted in significant mortality due to fulminant mousepox. These data therefore suggest that virus-encoded IL-4 not only suppresses primary antiviral cell-mediated immune responses but also can inhibit the expression of immune memory responses.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Interleucina-4/fisiologia , Linfócitos T Citotóxicos/imunologia , Animais , Ectromelia Infecciosa/genética , Feminino , Predisposição Genética para Doença , Imunização , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Recombinação Genética
17.
Vet Immunol Immunopathol ; 63(1-2): 105-9, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9656445

RESUMO

Ectromelia virus (EV) is a natural mouse pathogen that causes a generalized infection termed mousepox, which, in the genetically resistant C57BL/6 (B6) mouse, is an inapparent disease. In contrast, BALB/c and A strain mice are highly susceptible; one infectious virus particle can result in 100% mortality. The contribution of cytokines in the induction of protective immune responses and recovery from infection with EV in B6, BALB/c and A strain mice have been. In the spleen and lymph node (LN) of resistant B6 mice, IL-2, IFN-gamma and TNF-alpha were induced rapidly with large numbers of cells producing these cytokines. All three cytokines were virtually absent in BALB/c and A strain mice. No significant differences were found in the numbers of IL-4 producing cells in the spleen or LN of both resistant and susceptible mice. IFN-gamma-producing cells were detected in the spleen but not in the lymph node whereas IL-2-producing cells were detected only in the lymph node of B6 mice. Despite significant increases in the IFN-gamma mRNA levels in the LN of B6 mice, no protein was detected by immunocytochemistry. The mRNA levels of IL-2, TNF-alpha and IL-12 were also rapidly upregulated in LN of B6 mice. The rapid induction of type I cytokines strongly correlated with a potent antiviral CTL response in B6 mice. The absence of these cytokines also correlated with a complete absence or delayed induction of CTL responses to EV in both the BALB/c and A strain mice. IFN-gamma gene knock out mice on a B6 background were as susceptible to EV as the BALB/c and A strain mice.


Assuntos
Citocinas/biossíntese , Vírus da Ectromelia/imunologia , Vírus da Ectromelia/patogenicidade , Animais , Anticorpos Antivirais/biossíntese , Citocinas/genética , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/imunologia , Imunidade Celular , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Linfócitos T Citotóxicos/imunologia , Células Th1/imunologia , Células Th2/imunologia
18.
J Exp Med ; 186(9): 1591-6, 1997 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-9348317

RESUMO

The antiviral nature of tumor necrosis factor (TNF) is generally well accepted. TNF appears to induce multiple antiviral mechanisms, and to synergize with interferon (IFN)-gamma in promoting antiviral activities. We infected TNF receptor (TNFR)-deficient mice with the virulent murine pathogen, ectromelia virus (EV), and observed that otherwise resistant mice were susceptible to lethal infection. To study the molecular basis of the antiviral action of TNF, mice were infected with a recombinant vaccinia virus encoding murine TNF (VV-HA-TNF). In normal mice, the replication of VV-HA-TNF was highly attenuated. In contrast, mice in which the TNFR type 1 (p55) or the TNFR type 2 (p75) were genetically disrupted showed a moderate defect in their capacity to clear the TNF-encoding virus. The contribution of both TNF receptors to the control of VV-HA-TNF was confirmed by the enhanced replication of VV-HA-TNF in mice deficient for both p55 and p75. These observations were corroborated by infecting TNFR-deficient mice with EV. For both infections, the p55 and p75 TNFRs were necessary to maintain normal levels of resistance. Thus, the antiviral activity of TNF is mediated via both TNFRs in vivo. Furthermore, these studies establish that TNF is an important component of the host response to a natural virus infection.


Assuntos
Antivirais/farmacologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recombinação Genética , Fator de Necrose Tumoral alfa/genética , Vacínia/genética , Vacínia/imunologia , Vacínia/virologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética , Vaccinia virus/patogenicidade , Replicação Viral/efeitos dos fármacos
19.
Am J Pathol ; 150(4): 1407-20, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9094996

RESUMO

Innate resistance of C57BL/6 (B6) mice to lethal mousepox is controlled by multiple genes. Previously, four resistance genes were localized to specific subchromosomal regions and transferred onto a susceptible DBA/2 (D2) background by serial backcrossing and intercrossing to produce congenic strains. Intraperitoneally inoculated ectromelia virus was uniformly lethal and achieved similar titers in B6 and D2 mice but elicited differential responses in liver, spleen, and circulating blood leukocytes. The distribution of these response phenotypes in congenic strains linked control of phenotypes with specific subchromosomal regions. D2.R1 mice, which carried a differential segment of chromosome 6, exhibited a B6 liver response and intermediate spleen and circulating leukocyte responses. D2.R2 and D2.R4 mice, which carried differential segments of chromosomes 2 and 1, respectively, exhibited a D2 liver response, a B6 spleen response, and an intermediate circulating leukocyte response. The localization of control of liver response phenotypes to chromosome 6 implicates cells that express natural killer (NK) cell receptor NKR-P1 alloantigens. The localization of control of spleen and circulating leukocyte responses to chromosomes 1, 2, and 6 implicates NK cells, the fifth component of complement, and a gene near the selectin gene complex in recruitment of circulating leukocytes to spleen.


Assuntos
Antígenos de Superfície/toxicidade , Quimiotaxia de Leucócito/imunologia , Complemento C5/toxicidade , Ectromelia Infecciosa/etiologia , Hepatite Animal/etiologia , Células Matadoras Naturais/metabolismo , Lectinas Tipo C , Baço/patologia , Animais , Antígenos de Superfície/genética , Quimiotaxia de Leucócito/genética , Mapeamento Cromossômico , Complemento C5/genética , Cruzamentos Genéticos , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/mortalidade , Ectromelia Infecciosa/patologia , Feminino , Ligação Genética , Hepatite Animal/genética , Hepatite Animal/imunologia , Hepatite Animal/patologia , Imunidade Inata , Células Matadoras Naturais/virologia , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Repetições de Microssatélites , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Necrose
20.
J Virol ; 69(11): 6958-64, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474114

RESUMO

DBA/2 (D2) mice are susceptible and C57BL/6 (B6) mice are resistant to lethal mousepox. A congenic resistant strain, D2.B6-Rmp-4r (D2.R4), was developed by serially backcrossing male mice that survived ectromelia virus infection with D2 mice, beginning with (B6 x D2)F1 mice. Male D2.R4 mice were at least 300-fold more resistant to lethal mousepox than male D2 mice. Female D2.R4 mice were 100-fold more resistant than male D2.R4 mice and 500-fold more resistant than female D2 mice. Neonatal gonadectomy prevented development of resistance in D2.R4 mice of both sexes. Differences in resistance between strains and between sexes correlated with restriction of virus replication in spleen and liver, but gender differences were less evident in liver than in spleen. High-resolution interval mapping of the 19 autosomes of D2.R4 mice using dispersed informative microsatellites as marker loci revealed a segment of distal chromosome 1 to be of B6 origin. Haplotypes for a marker locus, D1Mit57, from the differential segment were determined in (D2.R4 x D2)F1 x D2 backcross mice, which were then infected with ectromelia virus. Significantly more heterozygotes than homozygotes survived ectromelia virus infection in both sexes. Whereas nearly all surviving males were heterozygotes, 44% of surviving females were homozygotes. These results indicate that resistance in D2.R4 mice is determined by a gonad-dependent gene on distal chromosome 1, provisionally named Rmp-4, and by an ovary-dependent factor that is not genetically linked to Rmp-4.


Assuntos
Mapeamento Cromossômico , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/imunologia , Predisposição Genética para Doença , Imunidade Inata/genética , Animais , Animais Recém-Nascidos , Cruzamentos Genéticos , DNA Satélite/análise , DNA Satélite/genética , DNA Viral/análise , Feminino , Marcadores Genéticos , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Orquiectomia , Ovariectomia , Poxviridae/isolamento & purificação , Poxviridae/fisiologia , Caracteres Sexuais , Especificidade da Espécie , Baço/virologia , Fatores de Tempo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA