Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
1.
Viruses ; 13(6)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203773

RESUMEN

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.


Asunto(s)
Virus de la Ectromelia/genética , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/patología , Ectromelia Infecciosa/virología , Genómica , Filogenia , Animales , Modelos Animales de Enfermedad , Virus de la Ectromelia/clasificación , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Femenino , Evasión Inmune , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Filogeografía , Proteínas Virales/genética , Virulencia
2.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33992803

RESUMEN

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


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/prevención & control , Proteínas Virales/genética , Vacunas de ARNm/administración & dosificación , Animales , Composición de Medicamentos , Ectromelia Infecciosa/inmunología , Inmunización Secundaria , Memoria Inmunológica , Liposomas , Masculino , Ratones , Nanopartículas , Péptidos/química , Péptidos/genética , Péptidos/inmunología , Seudouridina/análogos & derivados , Seudouridina/química , Proteínas Virales/química , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/química , Vacunas Virales/farmacología , Vacunas de ARNm/química , Vacunas de ARNm/farmacología
3.
PLoS Pathog ; 17(5): e1009593, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34015056

RESUMEN

Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Animales , Citocinas/inmunología , Resistencia a la Enfermedad , Ectromelia Infecciosa/virología , Femenino , Hepatocitos/inmunología , Hepatocitos/virología , Inmunidad Innata , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/virología , Receptor de Interferón alfa y beta/genética
4.
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
5.
PLoS Pathog ; 16(8): e1008685, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32745153

RESUMEN

Smallpox and monkeypox pose severe threats to human health. Other orthopoxviruses are comparably virulent in their natural hosts, including ectromelia, the cause of mousepox. Disease severity is linked to an array of immunomodulatory proteins including the B22 family, which has homologs in all pathogenic orthopoxviruses but not attenuated vaccine strains. We demonstrate that the ectromelia B22 member, C15, is necessary and sufficient for selective inhibition of CD4+ but not CD8+ T cell activation by immunogenic peptide and superantigen. Inhibition is achieved not by down-regulation of surface MHC- II or co-stimulatory protein surface expression but rather by interference with antigen presentation. The appreciable outcome is interference with CD4+ T cell synapse formation as determined by imaging studies and lipid raft disruption. Consequently, CD4+ T cell activating stimulus shifts to uninfected antigen-presenting cells that have received antigen from infected cells. This work provides insight into the immunomodulatory strategies of orthopoxviruses by elucidating a mechanism for specific targeting of CD4+ T cell activation, reflecting the importance of this cell type in control of the virus.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas Virales/inmunología , Animales , Ectromelia Infecciosa/metabolismo , Ectromelia Infecciosa/virología , Femenino , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Virales/metabolismo , Virulencia
6.
Aging Cell ; 19(7): e13170, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32657004

RESUMEN

It is known that aging decreases natural resistance to viral diseases due to dysfunctional innate and adaptive immune responses, but the nature of these dysfunctions, particularly in regard to innate immunity, is not well understood. We have previously shown that C57BL/6J (B6) mice lose their natural resistance to footpad infection with ectromelia virus (ECTV) due to impaired maturation and recruitment of natural killer (NK) cells to the draining popliteal lymph node (dLN). More recently, we have also shown that in young B6 mice infected with ECTV, the recruitment of NK cells is dependent on a complex cascade whereby migratory dendritic cells (mDCs) traffic from the skin to the dLN, where they produce CCL2 and CCL7 to recruit inflammatory monocytes (iMOs). In the dLN, mDCs also upregulate NKG2D ligands to induce interferon gamma (IFN-γ) expression by group 1 innate lymphoid cells (G1-ILCs), mostly NK in cells but also some ILC1. In response to the IFN-γ, the incoming uninfected iMOs secret CXCL9 to recruit the critical NK cells. Here, we show that in aged B6 mice, the trafficking of mDCs to the dLN in response to ECTV is decreased, resulting in impaired IFN-γ expression by G1-ILCs, reduced accumulation of iMOs, and attenuated CXCL9 production by iMOs, which likely contributes to decrease in NK cell recruitment. Together, these data indicate that defects in the mDC response to viral infection during aging result in a reduced innate immune response in the dLN and contribute to increased susceptibility to viral disease in the aged.


Asunto(s)
Células Dendríticas/metabolismo , Virus de la Ectromelia/inmunología , Inmunidad Innata/inmunología , Ganglios Linfáticos/metabolismo , Envejecimiento , Animales , Ratones
7.
J Immunol ; 204(6): 1582-1591, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32015010

RESUMEN

NK cells play an important role in antiviral resistance. The integrin α2, which dimerizes with integrin ß1, distinguishes NK cells from innate lymphoid cells 1 and other leukocytes. Despite its use as an NK cell marker, little is known about the role of α2ß1 in NK cell biology. In this study, we show that in mice α2ß1 deficiency does not alter the balance of NK cell/ innate lymphoid cell 1 generation and slightly decreases the number of NK cells in the bone marrow and spleen without affecting NK cell maturation. NK cells deficient in α2ß1 had no impairment at entering or distributing within the draining lymph node of ectromelia virus (ECTV)-infected mice or at becoming effectors but proliferated poorly in response to ECTV and did not increase in numbers following infection with mouse CMV (MCMV). Still, α2ß1-deficient NK cells efficiently protected from lethal mousepox and controlled MCMV titers in the spleen. Thus, α2ß1 is required for optimal NK cell proliferation but is dispensable for protection against ECTV and MCMV, two well-established models of viral infection in which NK cells are known to be important.


Asunto(s)
Ectromelia Infecciosa/inmunología , Infecciones por Herpesviridae/inmunología , Integrina alfa2beta1/metabolismo , Células Asesinas Naturales/inmunología , Animales , Recuento de Células , Proliferación Celular , Modelos Animales de Enfermedad , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/sangre , Ectromelia Infecciosa/virología , Femenino , Infecciones por Herpesviridae/sangre , Infecciones por Herpesviridae/virología , Humanos , Inmunidad Innata , Integrina alfa2beta1/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Muromegalovirus/inmunología , Replicación Viral/inmunología
8.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31776282

RESUMEN

Chronic viral infections. like those of humans with cytomegalovirus, human immunodeficiency virus (even when under antiretroviral therapy), and hepatitis C virus or those of mice with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13), result in immune dysfunction that predisposes the host to severe infections with unrelated pathogens. It is known that C57BL/6 (B6) mice are resistant to mousepox, a lethal disease caused by the orthopoxvirus ectromelia virus (ECTV), and that this resistance requires natural killer (NK) cells and other immune cells. We show that most B6 mice chronically infected with CL13 succumb to mousepox but that most of those that recovered from acute infection with the LCMV Armstrong (Arm) strain survive. We also show that B6 mice chronically infected with CL13 and those that recovered from Arm infection have a reduced frequency and a reduced number of NK cells. However, at steady state, NK cells in mice that have recovered from Arm infection mature normally and, in response to ECTV, get activated, become more mature, proliferate, and increase their cytotoxicity in vivo Conversely, in mice chronically infected with CL13, NK cells are immature and residually activated, and following ECTV infection, they do not mature, proliferate, or increase their cytotoxicity. Given the well-established importance of NK cells in resistance to mousepox, these data suggest that the NK cell dysfunction caused by CL13 persistence may contribute to the susceptibility of CL13-infected mice to mousepox. Whether chronic infections similarly affect NK cells in humans should be explored.IMPORTANCE Infection of adult mice with the clone 13 (CL13) strain of lymphocytic choriomeningitis virus (LCMV) is extensively used as a model of chronic infection. In this paper, we show that mice chronically infected with CL13 succumb to challenge with ectromelia virus (ECTV; the agent of mousepox) and that natural killer (NK) cells in CL13-infected mice are reduced in numbers and have an immature and partially activated phenotype but do respond to ECTV. These data may provide additional clues why humans chronically infected with certain pathogens are less resistant to viral diseases.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2/genética , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31826990

RESUMEN

It is well established that chronic viral infections can cause immune suppression, resulting in increased susceptibility to other infectious diseases. However, the effects of chronic viral infection on T-cell responses and vaccination against highly pathogenic viruses are not well understood. We have recently shown that C57BL/6 (B6) mice lose their natural resistance to wild-type (WT) ectromelia virus (ECTV) when chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13). Here we compared the T-cell response to ECTV in previously immunologically naive mice that were chronically infected with CL13 or that were convalescent from acute infection with the Armstrong (Arm) strain of LCMV. Our results show that mice that were chronically infected with CL13 but not those that had recovered from Arm infection have highly defective ECTV-specific CD8+ and CD4+ T-cell responses to WT ECTV. These defects are at least partly due to the chronic infection environment. In contrast to mice infected with WT ECTV, mice chronically infected with CL13 survived without signs of disease when infected with ECTV-Δ036, a mutant ECTV strain that is highly attenuated. Strikingly, mice chronically infected with CL13 mounted a strong CD8+ T-cell response to ECTV-Δ036 and survived without signs of disease after a subsequent challenge with WT ECTV. Our work suggests that enhanced susceptibility to acute viral infections in chronically infected individuals can be partly due to poor T-cell responses but that sufficient T-cell function can be recovered and resistance to acute infection can be restored by immunization with highly attenuated vaccines.IMPORTANCE Chronic viral infections may result in immunosuppression and enhanced susceptibility to infections with other pathogens. For example, we have recently shown that mice chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13) are highly susceptible to mousepox, a disease that is caused by ectromelia virus and that is the mouse homolog of human smallpox. Here we show chronic CL13 infection severely disrupts the expansion, proliferation, activation, and cytotoxicity of T cells in response due at least in part to the suppressive effects of the chronic infection milieu. Notably, despite this profound immunodeficiency, mice chronically infected with CL13 could be protected by vaccination with a highly attenuated variant of ECTV. These results demonstrate that protective vaccination of immunosuppressed individuals is possible, provided that proper immunization tools are used.


Asunto(s)
Ectromelia Infecciosa/inmunología , Inmunidad Innata/inmunología , Inmunización , Coriomeningitis Linfocítica/inmunología , Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Virus de la Ectromelia/inmunología , Femenino , Humanos , Tolerancia Inmunológica , Memoria Inmunológica , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación
10.
J Biol Chem ; 294(13): 5214-5227, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30723161

RESUMEN

Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.


Asunto(s)
Virus de la Ectromelia/inmunología , Factores Inmunológicos/inmunología , Linfotoxina-alfa/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Señuelo del Factor de Necrosis Tumoral/inmunología , Proteínas Virales/inmunología , Secuencia de Aminoácidos , Animales , Virus de la Ectromelia/química , Ectromelia Infecciosa/virología , Humanos , Factores Inmunológicos/química , Ratones , Modelos Moleculares , Dominios Proteicos , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Virales/química
11.
Viruses ; 10(12)2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30563103

RESUMEN

The poxviruses are large, linear, double-stranded DNA viruses about 130 to 230 kbp, that have an animal origin and evolved to infect a wide host range. Variola virus (VARV), the causative agent of smallpox, is a poxvirus that infects only humans, but other poxviruses such as monkey poxvirus and cowpox virus (CPXV) have crossed over from animals to infect humans. Therefore understanding the biology of poxviruses can devise antiviral strategies to prevent these human infections. In this study we used a system-based approach to examine the host responses to three orthopoxviruses, CPXV, vaccinia virus (VACV), and ectromelia virus (ECTV) in the murine macrophage RAW 264.7 cell line. Overall, we observed a significant down-regulation of gene expressions for pro-inflammatory cytokines, chemokines, and related receptors. There were also common and virus-specific changes in the immune-regulated gene expressions for each poxvirus-infected RAW cells. Collectively our results showed that the murine macrophage RAW 264.7 cell line is a suitable cell-based model system to study poxvirus host response.


Asunto(s)
Virus de la Viruela Vacuna/inmunología , Citocinas/inmunología , Virus de la Ectromelia/inmunología , Macrófagos/inmunología , Virus Vaccinia/inmunología , Animales , Quimiocinas/genética , Quimiocinas/inmunología , Citocinas/genética , Regulación hacia Abajo , Expresión Génica , Macrófagos/virología , Ratones , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa , Células RAW 264.7 , Regulación hacia Arriba
12.
Front Immunol ; 9: 1297, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29963044

RESUMEN

Activation of the DNA-dependent innate immune pathway plays a pivotal role in the host defense against poxvirus. Cyclic GMP-AMP synthase (cGAS) is a key cytosolic DNA sensor that produces the cyclic dinucleotide cGMP-AMP (cGAMP) upon activation, which triggers stimulator of interferon genes (STING), leading to type I Interferons (IFNs) production and an antiviral response. Ectromelia virus (ECTV) has emerged as a valuable model for investigating the host-Orthopoxvirus relationship. However, the role of cGas-Sting pathway in response to ECTV is not clearly understood. Here, we showed that murine cells (L929 and RAW264.7) mount type I IFN responses to ECTV that are dependent upon cGas, Sting, TANK binding kinase 1 (Tbk1), and interferon regulatory factor 3 (Irf3) signaling. Disruption of cGas or Sting expression in mouse macrophages blocked the type I IFN production and facilitated ECTV replication. Consistently, mice deficient in cGas or Sting exhibited lower type I IFN levels and higher viral loads, and are more susceptible to mousepox. Collectively, our study indicates that the cGas-Sting pathway is critical for sensing of ECTV infection, inducing the type I IFN production, and controlling ECTV replication.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Animales , Chlorocebus aethiops , Ectromelia Infecciosa/virología , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células 3T3 NIH , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Células RAW 264.7 , Células Vero , Replicación Viral
13.
Virology ; 518: 335-348, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29602068

RESUMEN

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.


Asunto(s)
Virus de la Ectromelia/inmunología , Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/prevención & control , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Replicación Viral , Animales , Línea Celular , Virus de la Ectromelia/genética , Virus de la Ectromelia/patogenicidad , Técnicas de Inactivación de Genes , Humanos , Ratones Endogámicos BALB C , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
14.
Antiviral Res ; 152: 18-25, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29427674

RESUMEN

In this study, five phage display antibodies (pdAbs) against ectromelia virus (ECTV) were selected from vaccinia virus (VACV)-immune phage-display library of human single chain variable fragments (scFv). ELISA demonstrated that selected pdAbs could recognize ECTV, VACV, and cowpox virus (CPXV). Atomic force microscopy visualized binding of the pdAbs to VACV. Three of the selected pdAbs neutralized variola virus (VARV) in the plaque reduction neutralization test. Western blot analysis of ECTV, VARV, VACV, and CPXV proteins indicated that neutralizing pdAbs bound orthopoxvirus 35 kDa proteins, which are encoded by the open reading frames orthologous to the ORF H3L in VACV. The fully human antibody fh1A was constructed on the base of the VH and VL domains of pdAb, which demonstrated a dose-dependent inhibition of plaque formation after infection with VARV, VACV, and CPXV. To determine the p35 region responsible for binding to neutralizing pdAbs, a panel of truncated p35 proteins was designed and expressed in Escherichia coli cells, and a minimal p35 fragment recognized by selected neutralizing pdAbs was identified. In addition, peptide phage-display combinatorial libraries were applied to localize the epitope. The obtained data indicated that the epitope responsible for recognition by the neutralizing pdAbs is discontinuous and amino acid residues located within two p35 regions, 15-19 aa and 232-237 aa, are involved in binding with neutralizing anti-p35 antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Ectromelia/inmunología , Virus de la Viruela/inmunología , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Virus de la Ectromelia/genética , Mapeo Epitopo , Humanos , Pruebas de Neutralización , Biblioteca de Péptidos , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Viruela/inmunología , Viruela/virología , Virus de la Viruela/química , Virus de la Viruela/genética , Virus de la Viruela/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
15.
Vaccine ; 35(33): 4245-4254, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28625523

RESUMEN

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.


Asunto(s)
Dermatitis Atópica/complicaciones , Modelos Animales de Enfermedad , Virus de la Ectromelia/inmunología , Erupción Variceliforme de Kaposi/tratamiento farmacológico , Erupción Variceliforme de Kaposi/prevención & control , Vacuna contra Viruela/administración & dosificación , Vacuna contra Viruela/efectos adversos , Animales , Anticuerpos Antivirales/administración & dosificación , Antivirales/administración & dosificación , Linfocitos T CD4-Positivos , Cidofovir , Citosina/administración & dosificación , Citosina/análogos & derivados , Virus de la Ectromelia/patogenicidad , Femenino , Humanos , Erupción Variceliforme de Kaposi/patología , Ratones , Organofosfonatos/administración & dosificación
16.
Immunol Cell Biol ; 95(8): 676-683, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28428612

RESUMEN

The biological role of granzyme K, a serine protease of cytotoxic T lymphocytes (CTL), is controversial. It has been reported to induce perforin-mediated cell death in vitro, but is also reported to be non-cytotoxic and to operate in inflammatory processes. To elucidate the biological role of this protease, we have deleted the granzyme K gene in mice (mutant allele: Gzmktm1.1Pib; MGI:5636646). Gzmk -/- mice are healthy, anatomically normal, fecund and show normal hematopoietic development. Gzmk -/- mice readily recover from lymphocytic choriomeningitis virus and mouse pox Ectromelia virus infection. Ex vivo, virus-specific granzyme K-deficient CTL are indistinguishable from those of wild-type mice in apoptosis induction of target cells. These data suggest that granzyme K does not play an essential role in viral immunity or cytotoxicity. Our granzyme K knockout line completes the collection of mouse models for the human granzymes, and will further our understanding of their biological roles and relationships.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Granzimas/genética , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos Virales/inmunología , Células Cultivadas , Citotoxicidad Inmunológica , Granzimas/metabolismo , Hematopoyesis/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Viral Immunol ; 30(5): 315-329, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28453414

RESUMEN

Poxviruses have evolved numerous mechanisms to avoid the immune response of the infected host, and many of these mechanisms have not been fully described. Here, we studied the transcriptional response of innate immune genes in BALB/c and C57BL/6 peritoneal macrophages following infection with the Moscow strain of ectromelia virus (ECTV-Mos) with the aim of delineating innate immune genes that contribute to the difference between susceptibility and resistance to lethal infection. We show a generalized downregulation of many genes in four categories (toll-like receptor signaling, NOD-like receptor signaling, RIG-I-like receptor signaling, and type I interferon signaling) of antiviral innate immune receptors, downstream signaling pathways, and responsive components. Two important observations were made. First, 14 innate antiviral genes were differentially expressed with fold change upregulation of two and above occurring in C57BL/6 mice, known to be resistant to ECTV-Mos infection, whereas the same genes were downregulated in BALB/c mice with fold change of two and below. Second, the cathepsin group of genes was downregulated in both strains of mice but with profound fold changes of 17, 38, and 62 downregulation for CtsL, CtsB, and CtsS, respectively, in C57BL/6 mice. We show that a poxvirus profoundly downregulates both the mRNA and protein expression of these three cathepsins and this change appears to support virus replication. Based on these data we propose that the variations in gene expression observed may contribute to the difference in resistance/susceptibility between BALB/c and C57BL/6 mice to lethal infection by ECTV-Mos.


Asunto(s)
Resistencia a la Enfermedad , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Perfilación de la Expresión Génica , Inmunidad Innata , Macrófagos Peritoneales/inmunología , Animales , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
18.
Adv Immunol ; 129: 251-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26791861

RESUMEN

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.


Asunto(s)
Inmunidad Adaptativa , Resistencia a la Enfermedad/inmunología , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/inmunología , Viruela/inmunología , Virus Vaccinia/inmunología , Animales , Modelos Animales de Enfermedad , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/prevención & control , Ectromelia Infecciosa/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Vacunación , Proteínas Virales/inmunología , Virulencia
19.
Cell Mol Immunol ; 13(1): 65-72, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25531393

RESUMEN

Viral infection triggers activation of the transcription factors NF-κB and IRF3, which collaborate to induce the expression of type I interferons (IFNs) and elicit innate antiviral response. In this report, we identified Krüppel-like factor 4 (KLF4) as a negative regulator of virus-triggered signaling. Overexpression of KLF4 inhibited virus-induced activation of ISRE and IFN-ß promoter in various types of cells, while knockdown of KLF4 potentiated viral infection-triggered induction of IFNB1 and downstream genes and attenuated viral replication. In addition, KLF4 was found to be localized in the cytosol and nucleus, and viral infection promoted the translocation of KLF4 from cytosol to nucleus. Upon virus infection, KLF4 was bound to the promoter of IFNB gene and inhibited the recruitment of IRF3 to the IFNB promoter. Our study thus suggests that KLF4 negatively regulates cellular antiviral response.


Asunto(s)
Virus de la Ectromelia/inmunología , Interacciones Huésped-Patógeno , Interferón beta/inmunología , Factores de Transcripción de Tipo Kruppel/inmunología , Virus Sendai/inmunología , Fraccionamiento Celular , Núcleo Celular/inmunología , Núcleo Celular/virología , Inmunoprecipitación de Cromatina , Citosol/inmunología , Citosol/virología , Virus de la Ectromelia/crecimiento & desarrollo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/genética , Luciferasas/genética , Luciferasas/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , Regiones Promotoras Genéticas , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Virus Sendai/crecimiento & desarrollo , Transducción de Señal , Replicación Viral
20.
PLoS Pathog ; 11(12): e1005342, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26700306

RESUMEN

Orthopoxviruses (OPV), including variola, vaccinia, monkeypox, cowpox and ectromelia viruses cause acute infections in their hosts. With the exception of variola virus (VARV), the etiological agent of smallpox, other OPV have been reported to persist in a variety of animal species following natural or experimental infection. Despite the implications and significance for the ecology and epidemiology of diseases these viruses cause, those reports have never been thoroughly investigated. We used the mouse pathogen ectromelia virus (ECTV), the agent of mousepox and a close relative of VARV to investigate virus persistence in inbred mice. We provide evidence that ECTV causes a persistent infection in some susceptible strains of mice in which low levels of virus genomes were detected in various tissues late in infection. The bone marrow (BM) and blood appeared to be key sites of persistence. Contemporaneous with virus persistence, antiviral CD8 T cell responses were demonstrable over the entire 25-week study period, with a change in the immunodominance hierarchy evident during the first 3 weeks. Some virus-encoded host response modifiers were found to modulate virus persistence whereas host genes encoded by the NKC and MHC class I reduced the potential for persistence. When susceptible strains of mice that had apparently recovered from infection were subjected to sustained immunosuppression with cyclophosphamide (CTX), animals succumbed to mousepox with high titers of infectious virus in various organs. CTX treated index mice transmitted virus to, and caused disease in, co-housed naïve mice. The most surprising but significant finding was that immunosuppression of disease-resistant C57BL/6 mice several weeks after recovery from primary infection generated high titers of virus in multiple tissues. Resistant mice showed no evidence of a persistent infection. This is the strongest evidence that ECTV can persist in inbred mice, regardless of their resistance status.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/transmisión , Animales , Terapia de Inmunosupresión , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Recurrencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...