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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(43): 26885-26894, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33046647

RESUMEN

Ectromelia virus (ECTV) causes mousepox, a surrogate mouse model for smallpox caused by variola virus in humans. Both orthopoxviruses encode tumor necrosis factor receptor (TNFR) homologs or viral TNFR (vTNFR). These homologs are termed cytokine response modifier (Crm) proteins, containing a TNF-binding domain and a chemokine-binding domain called smallpox virus-encoded chemokine receptor (SECRET) domain. ECTV encodes one vTNFR known as CrmD. Infection of ECTV-resistant C57BL/6 mice with a CrmD deletion mutant virus resulted in uniform mortality due to excessive TNF secretion and dysregulated inflammatory cytokine production. CrmD dampened pathology, leukocyte recruitment, and inflammatory cytokine production in lungs including TNF, IL-6, IL-10, and IFN-γ. Blockade of TNF, IL-6, or IL-10R function with monoclonal antibodies reduced lung pathology and provided 60 to 100% protection from otherwise lethal infection. IFN-γ caused lung pathology only when both the TNF-binding and SECRET domains were absent. Presence of the SECRET domain alone induced significantly higher levels of IL-1ß, IL-6, and IL-10, likely overcoming any protective effects that might have been afforded by anti-IFN-γ treatment. The use of TNF-deficient mice and those that express only membrane-associated but not secreted TNF revealed that CrmD is critically dependent on host TNF for its function. In vitro, recombinant Crm proteins from different orthopoxviruses bound to membrane-associated TNF and dampened inflammatory gene expression through reverse signaling. CrmD does not affect virus replication; however, it provides the host advantage by enabling survival. Host survival would facilitate virus spread, which would also provide an advantage to the virus.


Asunto(s)
Virus de la Ectromelia/fisiología , Interacciones Huésped-Patógeno , Receptores del Factor de Necrosis Tumoral/metabolismo , Infecciones del Sistema Respiratorio/virología , Proteínas Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones del Sistema Respiratorio/patología , Carga Viral
2.
J Virol ; 95(19): e0056621, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34260270

RESUMEN

Cytotoxic CD4 T lymphocytes (CD4-CTL) are important in antiviral immunity. For example, we have previously shown that in mice, CD4-CTL are important to control ectromelia virus (ECTV) infection. How viral infections induce CD4-CTL responses remains incompletely understood. We demonstrate here that not only ECTV but also vaccinia virus and lymphocytic choriomeningitis virus induce CD4-CTL, though the response to ECTV is stronger. Using ECTV, we also demonstrate that in contrast to CD8-CTL, CD4-CTL differentiation requires constant virus replication and ceases once the virus is controlled. We also show that major histocompatibility complex class II molecules on CD11c+ cells are required for CD4-CTL differentiation and for mousepox resistance. Transcriptional analysis indicated that antiviral CD4-CTL and noncytolytic T helper 1 (Th1) CD4 T cells have similar transcriptional profiles, suggesting that CD4-CTL are terminally differentiated classical Th1 cells. Interestingly, CD4-CTL and classical Th1 cells expressed similar mRNA levels of the transcription factors ThPOK and GATA-3, necessary for CD4 T cell linage commitment, and Runx3, required for CD8 T cell development and effector function. However, at the protein level, CD4-CTL had higher levels of the three transcription factors, suggesting that further posttranscriptional regulation is required for CD4-CTL differentiation. Finally, CRISPR/Cas9-mediated deletion of Runx3 in CD4 T cells inhibited CD4-CTL but not classical Th1 cell differentiation in response to ECTV infection. These results further our understanding of the mechanisms of CD4-CTL differentiation during viral infection and the role of posttranscriptionally regulated Runx3 in this process. IMPORTANCE While it is well established that cytotoxic CD4 T cells (CD4-CTLs) directly contribute to viral clearance, it remains unclear how CD4-CTL are induced. We now show that CD4-CTLs require sustained antigen presentation and are induced by CD11c-expressing antigen-presenting cells. Moreover, we show that CD4-CTLs are derived from the terminal differentiation of classical T helper 1 (Th1) subset of CD4 cells. Compared to Th1 cells, CD4-CTLs upregulate protein levels of the transcription factors ThPOK, Runx3, and GATA-3 posttranscriptionally. Deletion of Runx3 in differentiated CD4 T cells prevents induction of CD4-CTLs but not classical Th1 cells. These results advance our knowledge of how CD4-CTLs are induced during viral infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ectromelia Infecciosa/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Virosis/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD11/análisis , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Citotoxicidad Inmunológica , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/virología , Antígenos de Histocompatibilidad Clase II/análisis , Hígado/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo , Células TH1/metabolismo , Transcriptoma , Replicación Viral
3.
Immunol Invest ; 49(3): 232-248, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31240969

RESUMEN

Ectromelia virus (ECTV), an orthopoxvirus, undergoes productive replication in conventional dendritic cells (cDCs), resulting in the inhibition of their innate and adaptive immune functions. ECTV replication rate in cDCs is increased due to downregulation of the expression of cathepsins - cystein proteases that orchestrate several steps during DC maturation. Therefore, this study was aimed to determine if downregulation of cathepsins, such as B, L or S, disrupts cDC capacity to induce activating signals in T cells or whether infection of cDCs with ECTV further weakens their functions as antigen-presenting cells. Our results showed that cDCs treated with siRNA against cathepsin B, L and S synthesize similar amounts of pro-inflammatory cytokines and exhibit comparable ability to mature and stimulate alloreactive CD4+ T cells, as untreated wild type (WT) cells. Moreover, ECTV inhibitory effect on cDC innate and adaptive immune functions, observed especially after LPS treatment, was comparable in both cathepsin-silenced and WT cells. Taken together, the absence of cathepsins B, L and S has minimal, if any, impact on the inhibitory effect of ECTV on cDC immune functions. We assume that the virus-mediated inhibition of cathepsin expression in cDCs represents more a survival mechanism than an immune evasion strategy.


Asunto(s)
Catepsinas/deficiencia , Células Dendríticas/inmunología , Virus de la Ectromelia/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Catepsinas/genética , Catepsinas/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Balance Th1 - Th2
4.
BMC Microbiol ; 19(1): 92, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-31077130

RESUMEN

BACKGROUND: Cathepsins are a group of endosomal proteases present in many cells including dendritic cells (DCs). The activity of cathepsins is regulated by their endogenous inhibitors - cystatins. Cathepsins are crucial to antigen processing during viral and bacterial infections, and as such are a prerequisite to antigen presentation in the context of major histocompatibility complex class I and II molecules. Due to the involvement of DCs in both innate and adaptive immune responses, and the quest to understand the impact of poxvirus infection on host cells, we investigated the influence of ectromelia virus (ECTV) infection on cathepsin and cystatin levels in murine conventional DCs (cDCs). ECTV is a poxvirus that has evolved many mechanisms to avoid host immune response and is able to replicate productively in DCs. RESULTS: Our results showed that ECTV-infection of JAWS II DCs and primary murine GM-CSF-derived bone marrow cells down-regulated both mRNA and protein of cathepsin B, L and S, and cystatin B and C, particularly during the later stages of infection. Moreover, the activity of cathepsin B, L and S was confirmed to be diminished especially at later stages of infection in JAWS II cells. Consequently, ECTV-infected DCs had diminished ability to endocytose and process a soluble antigen. Close examination of cellular protein distribution showed that beginning from early stages of infection, the remnants of cathepsin L and cystatin B co-localized and partially co-localized with viral replication centers (viral factories), respectively. Moreover, viral yield increased in cDCs treated with siRNA against cathepsin B, L or S and subsequently infected with ECTV. CONCLUSIONS: Taken together, our results indicate that infection of cDCs with ECTV suppresses cathepsins and cystatins, and alters their cellular distribution which impairs the cDC function. We propose this as an additional viral strategy to escape immune responses, enabling the virus to replicate effectively in infected cells.


Asunto(s)
Catepsinas/genética , Cistatinas/genética , Células Dendríticas/virología , Virus de la Ectromelia/fisiología , Animales , Células Dendríticas/inmunología , Regulación hacia Abajo , Endosomas/inmunología , Endosomas/virología , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Replicación Viral
5.
Arch Virol ; 164(2): 559-565, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30374707

RESUMEN

Ectromelia virus (ECTV) is an orthopoxvirus that productively replicates in dendritic cells (DCs), but its influence on the microtubule (MT) cytoskeleton in DCs is not known. Here, we show that ECTV infection of primary murine granulocyte-macrophage colony stimulating factor-derived bone marrow cells (GM-BM) downregulates numerous genes engaged in MT cytoskeleton organization and dynamics. In infected cells, the MT cytoskeleton undergoes dramatic rearrangement and relaxation, accompanied by disappearance of the microtubule organizing centre (MTOC) and increased acetylation and stabilization of MTs, which are exploited by progeny virions for intracellular transport. This indicates a strong ability of ECTV to subvert the MT cytoskeleton of highly specialized immune cells.


Asunto(s)
Citoesqueleto/metabolismo , Células Dendríticas/metabolismo , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/metabolismo , Macrófagos/metabolismo , Centro Organizador de los Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Animales , Línea Celular , Ectromelia Infecciosa/virología , Interacciones Huésped-Patógeno , Ratones , Ratones Endogámicos BALB C , Microtúbulos/metabolismo
6.
Microb Pathog ; 109: 99-109, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28554653

RESUMEN

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.


Asunto(s)
Apoptosis , Células Dendríticas/inmunología , Virus de la Ectromelia/fisiología , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/inmunología , Inmunidad Adaptativa , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3 , Chlorocebus aethiops , Células Dendríticas/patología , Células Dendríticas/fisiología , Células Dendríticas/virología , Modelos Animales de Enfermedad , Ectromelia Infecciosa/virología , Proteína Ligando Fas/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Regulación hacia Arriba , Células Vero
7.
J Virol ; 89(19): 9974-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26202250

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Virus de la Ectromelia/inmunología , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Citocinas/biosíntesis , Células Dendríticas/clasificación , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/transmisión , Ectromelia Infecciosa/virología , Granulocitos/inmunología , Humanos , Inmunidad Innata , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Replicación Viral , Zoonosis/inmunología , Zoonosis/transmisión , Zoonosis/virología
8.
J Virol ; 88(18): 10946-57, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008931

RESUMEN

UNLABELLED: Immunization with modified vaccinia virus Ankara (MVA) can rapidly protect mice against lethal ectromelia virus (ECTV) infection, serving as an experimental model for severe systemic infections. Importantly, this early protective capacity of MVA vaccination completely depends on virus-specific cytotoxic CD8(+) T cell responses. We used MVA vaccination in the mousepox challenge model using ECTV infection to investigate the previously unknown factors required to elicit rapid protective T cell immunity in normal C57BL/6 mice and in mice lacking the interferon alpha/beta receptor (IFNAR(-/-)). We found a minimal dose of 10(5) PFU of MVA vaccine fully sufficient to allow robust protection against lethal mousepox, as assessed by the absence of disease symptoms and failure to detect ECTV in organs from vaccinated animals. Moreover, MVA immunization at low dosage also protected IFNAR(-/-) mice, indicating efficient activation of cellular immunity even in the absence of type I interferon signaling. When monitoring for virus-specific CD8(+) T cell responses in mice vaccinated with the minimal protective dose of MVA, we found significantly enhanced levels of antigen-specific T cells in animals that were MVA vaccinated and ECTV challenged compared to mice that were only vaccinated. The initial priming of naive CD8(+) T cells by MVA immunization appears to be highly efficient and, even at low doses, mediates a rapid in vivo burst of pathogen-specific T cells upon challenge. Our findings define striking requirements for protective emergency immunization against severe systemic infections with orthopoxviruses. IMPORTANCE: We demonstrate that single-shot low-dose immunizations with vaccinia virus MVA can rapidly induce T cell-mediated protective immunity against lethal orthopoxvirus infections. Our data provide new evidence for an efficient protective capacity of vaccination with replication-deficient MVA. These data are of important practical relevance for public health, as the effectiveness of a safety-tested, next-generation smallpox vaccine based on MVA is still debated. Furthermore, producing sufficient amounts of vaccine is expected to be a major challenge should an outbreak occur. Moreover, prevention of other infections may require rapidly protective immunization; hence, MVA could be an extremely useful vaccine for delivering heterologous T cell antigens, particularly for infectious diseases that fit a scenario of emergency vaccination.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Protección Cruzada , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/inmunología , Receptor de Interferón alfa y beta/deficiencia , Virus Vaccinia/inmunología , Animales , Linfocitos T CD8-positivos/virología , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/virología , Femenino , Inmunidad Celular , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Vaccinia/inmunología , Vaccinia/virología , Virus Vaccinia/genética
9.
Microb Pathog ; 87: 59-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26232502

RESUMEN

Nuclear factor κB (NF-κB) is a pleiotropic transcription factor that regulates the expression of immune response genes. NF-κB signaling can be disrupted by pathogens that prevent host immune response. In this work, we examined the influence of ectromelia (mousepox) virus (ECTV) on NF-κB signaling in murine BALB/3T3 fibroblasts. Activation of NF-κB via tumor necrosis factor (TNF) receptor 1 (TNFR1) in these cells induces proinflammatory cytokine secretion. We show that ECTV does not recruit NF-κB to viral factories or induce NF-κB nuclear translocation in BALB/3T3 cells. Additionally, ECTV counteracts TNF-α-induced p65 NF-κB nuclear translocation during the course of infection. Inhibition of TNF-α-induced p65 nuclear translocation was also observed in neighboring cells that underwent fusion with ECTV-infected cells. ECTV inhibits the key step of NF-κB activation, i.e. Ser32 phosphorylation and degradation of inhibitor κBα (IκBα) induced by TNF-α. We also observed that ECTV prevents TNF-α-induced Ser536 of p65 phosphorylation in BALB/3T3 cells. Studying TNFR1 signaling provides information about regulation of inflammatory response and cell survival. Unraveling poxviral immunomodulatory strategies may be helpful in drug target identification as well as in vaccine development.


Asunto(s)
Virus de la Ectromelia/fisiología , Fibroblastos/inmunología , Fibroblastos/virología , Interacciones Huésped-Patógeno , Evasión Inmune , FN-kappa B/metabolismo , Transducción de Señal , Animales , Células 3T3 BALB , Proteínas I-kappa B/metabolismo , Ratones , Inhibidor NF-kappaB alfa , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Virol ; 86(13): 7298-309, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22532670

RESUMEN

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.


Asunto(s)
Virus de la Ectromelia/inmunología , Linfocitos T/inmunología , Virus Vaccinia/inmunología , Animales , Degranulación de la Célula , Citocinas/biosíntesis , Pruebas Inmunológicas de Citotoxicidad , Modelos Animales de Enfermedad , Virus de la Ectromelia/patogenicidad , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/inmunología , Femenino , Citometría de Flujo , Granzimas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología , Vaccinia/inmunología , Virus Vaccinia/patogenicidad , Virus Vaccinia/fisiología
11.
Acta Virol ; 57(4): 467-70, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24294965

RESUMEN

Induction of autophagy by ectromelia virus (ECTV) in primary cultures of bone marrow-derived macrophages (BMDMs) was investigated. The results showed that ECTV infection of BMDMs resulted in increased formation of autophagosomes, increased level of LC3-II protein present in aggregates and extensive cytoplasmic vacuolization. These data indicate an increased autophagic activity in BMDMs during ECTV infection.


Asunto(s)
Autofagia , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/fisiopatología , Macrófagos/citología , Animales , Línea Celular , Células Cultivadas , Ectromelia Infecciosa/metabolismo , Ectromelia Infecciosa/virología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo
12.
Immunol Cell Biol ; 89(6): 706-15, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21173782

RESUMEN

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.


Asunto(s)
Citocinas/genética , Factor 88 de Diferenciación Mieloide/genética , Receptores Toll-Like/genética , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Chlorocebus aethiops , Citocinas/metabolismo , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/prevención & control , Femenino , Regulación Viral de la Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-12/metabolismo , Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Replicación Viral/inmunología
13.
J Exp Med ; 218(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33765134

RESUMEN

Natural killer (NK) cell activation depends on the signaling balance of activating and inhibitory receptors. CD94 forms inhibitory receptors with NKG2A and activating receptors with NKG2E or NKG2C. We previously demonstrated that CD94-NKG2 on NK cells and its ligand Qa-1b are important for the resistance of C57BL/6 mice to lethal ectromelia virus (ECTV) infection. We now show that NKG2C or NKG2E deficiency does not increase susceptibility to lethal ECTV infection, but overexpression of Qa-1b in infected cells does. We also demonstrate that Qa-1b is down-regulated in infected and up-regulated in bystander inflammatory monocytes and B cells. Moreover, NK cells activated by ECTV infection kill Qa-1b-deficient cells in vitro and in vivo. Thus, during viral infection, recognition of Qa-1b by activating CD94/NKG2 receptors is not critical. Instead, the levels of Qa-1b expression are down-regulated in infected cells but increased in some bystander immune cells to respectively promote or inhibit their killing by activated NK cells.


Asunto(s)
Linfocitos B/inmunología , Citotoxicidad Inmunológica/inmunología , Virus de la Ectromelia/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Virosis/inmunología , Animales , Linfocitos B/metabolismo , Linfocitos B/virología , Efecto Espectador/inmunología , Citotoxicidad Inmunológica/genética , Virus de la Ectromelia/fisiología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Subfamília D de Receptores Similares a Lectina de las Células NK/genética , Subfamília D de Receptores Similares a Lectina de las Células NK/inmunología , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Virosis/virología
14.
J Virol ; 83(8): 3684-95, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19211768

RESUMEN

Vaccinia virus (VACV) replicates in mouse and human fibroblasts with comparable kinetics and efficiency, yielding similar titers of infectious progeny. Here we demonstrate that gamma interferon (IFN-gamma) but not IFN-alpha or IFN-beta pretreatment of mouse fibroblasts prior to VACV infection induces a long-lasting antiviral state blocking VACV replication. In contrast, high doses of IFN-gamma failed to establish an antiviral state in human fibroblasts. In mouse fibroblasts, IFN-gamma impeded the viral replication cycle at the level of late gene transcription and blocked the multiplication of VACV genomes. The IFN-gamma-induced antiviral state invariably prevented the growth of different VACV strains but was not effective against the replication of ectromelia virus. The IFN-gamma effect required intact IFN-gamma receptor signaling prior to VACV infection through Janus kinase 2 (Jak2) and signal transducer and activator of transcription 1 (STAT1). The permissive state of IFN-gamma-treated human cells was unrelated to the VACV-encoded IFN decoy receptors B8 and B18 and associated with a complete disruption of STAT1 homodimer formation and DNA binding. Unlike human fibroblasts, mouse cells responded with long-lasting STAT1 activation which was preserved after VACV infection. The deletion of the IFN regulatory factor 1 (IRF-1) gene from mouse cells rescued efficient VACV replication, demonstrating that IRF-1 target genes have a critical role in VACV control. These data have implications for the understanding of VACV pathogenesis and identify an incongruent IFN-gamma response between the human host and the mouse model.


Asunto(s)
Fibroblastos/virología , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/inmunología , Virus Vaccinia/inmunología , Virus Vaccinia/fisiología , Replicación Viral , Animales , Células Cultivadas , Replicación del ADN , ADN Viral/biosíntesis , Virus de la Ectromelia/fisiología , Eliminación de Gen , Humanos , Factor 1 Regulador del Interferón/genética , Janus Quinasa 2/metabolismo , Ratones , Receptores de Interferón/metabolismo , Factor de Transcripción STAT1/metabolismo , Receptor de Interferón gamma
15.
Acta Virol ; 54(1): 41-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20201613

RESUMEN

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


Asunto(s)
Actinas/metabolismo , Virus de la Ectromelia/patogenicidad , Actinas/ultraestructura , Animales , Células 3T3 BALB/ultraestructura , Células 3T3 BALB/virología , Chlorocebus aethiops , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/ultraestructura , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/virología , Pie/virología , Células HeLa/ultraestructura , Células HeLa/virología , Humanos , Immunoblotting , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Fluorescente , Células Vero/ultraestructura , Células Vero/virología
16.
J Virol ; 82(2): 917-26, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18003726

RESUMEN

Poxviruses encode a number of secreted virulence factors that modulate the host immune response. The vaccinia virus A41 protein is an immunomodulatory protein with amino acid sequence similarity to the 35-kDa chemokine binding protein, but the host immune molecules targeted by A41 have not been identified. We report here that the vaccinia virus A41 ortholog encoded by ectromelia virus, a poxvirus pathogen of mice, named E163 in the ectromelia virus Naval strain, is a secreted 31-kDa glycoprotein that selectively binds a limited number of CC and CXC chemokines with high affinity. A detailed characterization of the interaction of ectromelia virus E163 with mutant forms of the chemokines CXCL10 and CXCL12alpha indicated that E163 binds to the glycosaminoglycan binding site of the chemokines. This suggests that E163 inhibits the interaction of chemokines with glycosaminoglycans and provides a mechanism by which E163 prevents chemokine-induced leukocyte migration to the sites of infection. In addition to interacting with chemokines, E163 can interact with high affinity with glycosaminoglycan molecules, enabling E163 to attach to cell surfaces and to remain in the vicinity of the sites of viral infection. These findings identify E163 as a new chemokine binding protein in poxviruses and provide a molecular mechanism for the immunomodulatory activity previously reported for the vaccinia virus A41 ortholog. The results reported here also suggest that the cell surface and extracellular matrix are important targeting sites for secreted poxvirus immune modulators.


Asunto(s)
Quimiocinas/metabolismo , Virus de la Ectromelia/fisiología , Glicoproteínas/metabolismo , Glicosaminoglicanos/metabolismo , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Quimiocinas/genética , Humanos , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica
17.
Virol J ; 6: 151, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19785778

RESUMEN

BACKGROUND: Ectromelia virus, a member of the Orthopox genus, is the causative agent of the highly infectious mousepox disease. Previous studies have shown that different poxviruses induce cell-cell fusion which is manifested by the formation of multinucleated-giant cells (polykaryocytes). This phenomenon has been widely studied with vaccinia virus in conditions which require artificial acidification of the medium. RESULTS: We show that Ectromelia virus induces cell-cell fusion under neutral pH conditions and requires the presence of a sufficient amount of viral particles on the plasma membrane of infected cells. This could be achieved by infection with a replicating virus and its propagation in infected cells (fusion "from within") or by infection with a high amount of virus particles per cell (fusion "from without"). Inhibition of virus maturation or inhibition of virus transport on microtubules towards the plasma membrane resulted in a complete inhibition of syncytia formation. We show that in contrast to vaccinia virus, Ectromelia virus induces cell-cell fusion irrespectively of its hemagglutination properties and cell-surface expression of the orthologs of the fusion inhibitory complex, A56 and K2. Additionally, cell-cell fusion was also detected in mice lungs following lethal respiratory infection. CONCLUSION: Ectromelia virus induces spontaneous cell-cell fusion in-vitro and in-vivo although expressing an A56/K2 fusion inhibitory complex. This syncytia formation property cannot be attributed to the 37 amino acid deletion in ECTV A56.


Asunto(s)
Fusión Celular , Virus de la Ectromelia/fisiología , Proteínas Virales/fisiología , Animales , Línea Celular , Humanos , Pulmón/patología , Pulmón/virología , Ratones
18.
Science ; 261(5127): 1445-8, 1993 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-7690156

RESUMEN

Interferons (IFNs) induce antiviral activity in many cell types. The ability of IFN-gamma to inhibit replication of ectromelia, vaccinia, and herpes simplex-1 viruses in mouse macrophages correlated with the cells' production of nitric oxide (NO). Viral replication was restored in IFN-gamma-treated macrophages exposed to inhibitors of NO synthase. Conversely, epithelial cells with no detectable NO synthesis restricted viral replication when transfected with a complementary DNA encoding inducible NO synthase or treated with organic compounds that generate NO. In mice, an inhibitor of NO synthase converted resolving ectromelia virus infection into fulminant mousepox. Thus, induction of NO synthase can be necessary and sufficient for a substantial antiviral effect of IFN-gamma.


Asunto(s)
Aminoácido Oxidorreductasas/biosíntesis , Virus de la Ectromelia/fisiología , Interferón gamma/farmacología , Macrófagos/microbiología , Replicación Viral , Aminoácido Oxidorreductasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Línea Celular , Células Cultivadas , Virus de la Ectromelia/efectos de los fármacos , Ectromelia Infecciosa/microbiología , Inducción Enzimática , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacología , Óxido Nítrico Sintasa , Simplexvirus/efectos de los fármacos , Simplexvirus/fisiología , Transfección , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/fisiología , Replicación Viral/efectos de los fármacos , omega-N-Metilarginina
19.
Arch Immunol Ther Exp (Warsz) ; 67(6): 401-414, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31324924

RESUMEN

Poxviruses utilize multiple strategies to prevent activation of extrinsic and intrinsic apoptotic pathways for successful replication. Mitochondrial heat shock proteins (mtHsps), especially Hsp60 and its cofactor Hsp10, are engaged in apoptosis regulation; however, until now, the influence of poxviruses on mtHsps has never been studied. We used highly infectious Moscow strain of ectromelia virus (ECTV) to investigate the mitochondrial heat shock response and apoptotic potential in permissive L929 fibroblasts. Our results show that ECTV-infected cells exhibit mostly mitochondrial localization of Hsp60 and Hsp10, and show overexpression of both proteins during later stages of infection. ECTV infection has only moderate effect on the electron transport chain subunit expression. Moreover, increase of mtHsp amounts is accompanied by lack of apoptosis, and confirmed by reduced level of pro-apoptotic Bax protein and elevated levels of anti-apoptotic Bcl-2 and Bcl-xL proteins. Taken together, we show a positive relationship between increased levels of Hsp60 and Hsp10 and decreased apoptotic potential of L929 fibroblasts, and further hypothesize that Hsp60 and/or its cofactor play important roles in maintaining protein homeostasis in mitochondria for promotion of cell survival allowing efficient replication of ECTV.


Asunto(s)
Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/inmunología , Fibroblastos/fisiología , Respuesta al Choque Térmico/inmunología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Apoptosis , Línea Celular , Fibroblastos/virología , Regulación de la Expresión Génica , Evasión Inmune , Ratones , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Virulencia , Replicación Viral
20.
Int J Mol Med ; 42(2): 1044-1053, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29749430

RESUMEN

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.


Asunto(s)
Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/virología , Proteínas HSP70 de Choque Térmico/genética , Interacciones Huésped-Patógeno , Ratones/virología , Replicación Viral , Animales , Replicación del ADN , Masculino , Ratones Endogámicos BALB C , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA