Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
J Cell Biol ; 223(6)2024 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-38709216

RESUMEN

Autophagy is an essential degradation program required for cell homeostasis. Among its functions is the engulfment and destruction of cytosolic pathogens, termed xenophagy. Not surprisingly, many pathogens use various strategies to circumvent or co-opt autophagic degradation. For poxviruses, it is known that infection activates autophagy, which however is not required for successful replication. Even though these complex viruses replicate exclusively in the cytoplasm, autophagy-mediated control of poxvirus infection has not been extensively explored. Using the prototypic poxvirus, vaccinia virus (VACV), we show that overexpression of the xenophagy receptors p62, NDP52, and Tax1Bp1 restricts poxvirus infection. While NDP52 and Tax1Bp1 were degraded, p62 initially targeted cytoplasmic virions before being shunted to the nucleus. Nuclear translocation of p62 was dependent upon p62 NLS2 and correlated with VACV kinase mediated phosphorylation of p62 T269/S272. This suggests that VACV targets p62 during the early stages of infection to avoid destruction and further implies that poxviruses exhibit multi-layered control of autophagy to facilitate cytoplasmic replication.


Asunto(s)
Autofagia , Núcleo Celular , Proteína Sequestosoma-1 , Virus Vaccinia , Humanos , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células HEK293 , Células HeLa , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Fosforilación , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Vaccinia/metabolismo , Vaccinia/virología , Vaccinia/genética , Virus Vaccinia/metabolismo , Virus Vaccinia/genética , Replicación Viral
2.
Int J Mol Sci ; 25(3)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38338659

RESUMEN

Vaccinia virus (Orthopoxvirus) F17 protein is a major virion structural phosphoprotein having a molecular weight of 11 kDa. Recently, it was shown that F17 synthesised in infected cells interacts with mTOR subunits to evade cell immunity and stimulate late viral protein synthesis. Several years back, we purified an 11 kDa protein that inhibited protein synthesis in reticulocyte lysate from virions, and that possesses all physico-chemical properties of F17 protein. To investigate this discrepancy, we used defective vaccinia virus particles devoid of the F17 protein (designated iF17- particles) to assess their ability to inhibit protein synthesis. To this aim, we purified iF17- particles from cells infected with a vaccinia virus mutant which expresses F17 only in the presence of IPTG. The SDS-PAGE protein profiles of iF17- particles or derived particles, obtained by solubilisation of the viral membrane, were similar to that of infectious iF17 particles. As expected, the profiles of full iF17- particles and those lacking the viral membrane were missing the 11 kDa F17 band. The iF17- particles did attach to cells and injected their viral DNA into the cytoplasm. Co-infection of the non-permissive BSC40 cells with a modified vaccinia Ankara (MVA) virus, expressing an mCherry protein, and iF17- particles, induced a strong mCherry fluorescence. Altogether, these experiments confirmed that the iF17- particles can inject their content into cells. We measured the rate of protein synthesis as a function of the multiplicity of infection (MOI), in the presence of puromycin as a label. We showed that iF17- particles did not inhibit protein synthesis at high MOI, by contrast to the infectious iF17 mutant. Furthermore, the measured efficiency to inhibit protein synthesis by the iF17 mutant virus generated in the presence of IPTG, was threefold to eightfold lower than that of the wild-type WR virus. The iF17 mutant contained about threefold less F17 protein than wild-type WR. Altogether these results strongly suggest that virion-associated F17 protein is essential to mediate a stoichiometric inhibition of protein synthesis, in contrast to the late synthesised F17. It is possible that this discrepancy is due to different phosphorylation states of the free and virion-associated F17 protein.


Asunto(s)
Virus Vaccinia , Vaccinia , Humanos , Virus Vaccinia/genética , Vaccinia/genética , Isopropil Tiogalactósido , Línea Celular , Fosfoproteínas , Virión/genética
3.
Microbiol Spectr ; 10(6): e0389322, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36453897

RESUMEN

Interferon-stimulated gene 15 (ISG15) is a 15-kDa ubiquitin-like modifier that binds to target proteins in a process termed ISGylation. ISG15, first described as an antiviral molecule against many viruses, participates in numerous cellular processes, from immune modulation to the regulation of genome stability. Interestingly, the role of ISG15 as a regulator of cell metabolism has recently gained strength. We previously described ISG15 as a regulator of mitochondrial functions in bone marrow-derived macrophages (BMDMs) in the context of Vaccinia virus (VACV) infection. Here, we demonstrate that ISG15 regulates lipid metabolism in BMDMs and that ISG15 is necessary to modulate the impact of VACV infection on lipid metabolism. We show that Isg15-/- BMDMs demonstrate alterations in the levels of several key proteins of lipid metabolism that result in differences in the lipid profile compared with Isg15+/+ (wild-type [WT]) BMDMs. Specifically, Isg15-/- BMDMs present reduced levels of neutral lipids, reflected by decreased lipid droplet number. These alterations are linked to increased levels of lipases and are independent of enhanced fatty acid oxidation (FAO). Moreover, we demonstrate that VACV causes a dysregulation in the proteomes of BMDMs and alterations in the lipid content of these cells, which appear exacerbated in Isg15-/- BMDMs. Such metabolic changes are likely caused by increased expression of the metabolic regulators peroxisome proliferator-activated receptor-γ (PPARγ) and PPARγ coactivator-1α (PGC-1α). In summary, our results highlight that ISG15 controls BMDM lipid metabolism during viral infections, suggesting that ISG15 is an important host factor to restrain VACV impact on cell metabolism. IMPORTANCE The functions of ISG15 are continuously expanding, and growing evidence supports its role as a relevant modulator of cell metabolism. In this work, we highlight how the absence of ISG15 impacts macrophage lipid metabolism in the context of viral infections and how poxviruses modulate metabolism to ensure successful replication. Our results open the door to new advances in the comprehension of macrophage immunometabolism and the interaction between VACV and the host.


Asunto(s)
Citocinas , Metabolismo de los Lípidos , Ubiquitinas , Vaccinia , Citocinas/metabolismo , Interferones , Lípidos , PPAR gamma/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo , Vaccinia/genética , Vaccinia/metabolismo , Virus Vaccinia/genética , Animales
4.
PLoS Pathog ; 18(12): e1010800, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36574441

RESUMEN

Genome-wide genetic screens are powerful tools to identify genes that act as host factors of viruses. We have applied this technique to analyze the infection of HeLa cells by Vaccinia virus, in an attempt to find genes necessary for infection. Infection of cell populations harboring single gene inactivations resulted in no surviving cells, suggesting that no single gene knock-out was able to provide complete resistance to Vaccinia virus and thus allow cells to survive infection. In the absence of an absolute infection blockage, we explored if some gene inactivations could provide partial protection leading to a reduced probability of infection. Multiple experiments using modified screening procedures involving replication restricted viruses led to the identification of multiple genes whose inactivation potentially increase resistance to infection and therefore cell survival. As expected, significant gene hits were related to proteins known to act in virus entry, such as ITGB1 and AXL as well as genes belonging to their downstream related pathways. Additionally, we consistently found ß2-microglobulin, encoded by the B2M gene, among the screening top hits, a novel finding that was further explored. Inactivation of B2M resulted in 54% and 91% reduced VV infection efficiency in HeLa and HAP1 cell lines respectively. In the absence of B2M, while virus binding to the cells was unaffected, virus internalization and early gene expression were significantly diminished. These results point to ß2-microglobulin as a relevant factor in the Vaccinia virus entry process.


Asunto(s)
Virus Vaccinia , Vaccinia , Humanos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Pruebas Genéticas , Células HeLa , Vaccinia/genética , Virus Vaccinia/genética , Replicación Viral/genética , Microglobulina beta-2
5.
Viruses ; 14(2)2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35215908

RESUMEN

The transcription factors IRF3 and NF-κB are crucial in innate immune signalling in response to many viral and bacterial pathogens. However, mechanisms leading to their activation remain incompletely understood. Viral RNA can be detected by RLR receptors, such as RIG-I and MDA5, and the dsRNA receptor TLR3. Alternatively, the DExD-Box RNA helicases DDX1-DDX21-DHX36 activate IRF3/NF-κB in a TRIF-dependent manner independent of RIG-I, MDA5, or TLR3. Here, we describe DDX50, which shares 55.6% amino acid identity with DDX21, as a non-redundant factor that promotes activation of the IRF3 signalling pathway following its stimulation with viral RNA or infection with RNA and DNA viruses. Deletion of DDX50 in mouse and human cells impaired IRF3 phosphorylation and IRF3-dependent endogenous gene expression and cytokine/chemokine production in response to cytoplasmic dsRNA (polyIC transfection), and infection by RNA and DNA viruses. Mechanistically, whilst DDX50 co-immunoprecipitated TRIF, it acted independently to the previously described TRIF-dependent RNA sensor DDX1. Indeed, shRNA-mediated depletion of DDX1 showed DDX1 was dispensable for signalling in response to RNA virus infection. Importantly, loss of DDX50 resulted in a significant increase in replication and dissemination of virus following infection with vaccinia virus, herpes simplex virus, or Zika virus, highlighting its important role as a broad-ranging viral restriction factor.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Herpes Simple/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Simplexvirus/fisiología , Virus Vaccinia/fisiología , Vaccinia/metabolismo , Infección por el Virus Zika/metabolismo , Virus Zika/fisiología , Animales , ARN Helicasas DEAD-box/genética , Herpes Simple/genética , Herpes Simple/virología , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/genética , Ratones , Fosforilación , Transducción de Señal , Simplexvirus/genética , Vaccinia/genética , Vaccinia/virología , Virus Vaccinia/genética , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/virología
6.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33912921

RESUMEN

Viral infection both activates stress signaling pathways and redistributes ribosomes away from host mRNAs to translate viral mRNAs. The intricacies of this ribosome shuffle from host to viral mRNAs are poorly understood. Here, we uncover a role for the ribosome-associated quality control (RQC) factor ZNF598 during vaccinia virus mRNA translation. ZNF598 acts on collided ribosomes to ubiquitylate 40S subunit proteins uS10 (RPS20) and eS10 (RPS10), initiating RQC-dependent nascent chain degradation and ribosome recycling. We show that vaccinia infection enhances uS10 ubiquitylation, indicating an increased burden on RQC pathways during viral propagation. Consistent with an increased RQC demand, we demonstrate that vaccinia virus replication is impaired in cells that either lack ZNF598 or express a ubiquitylation-deficient version of uS10. Using SILAC-based proteomics and concurrent RNA-seq analysis, we determine that translation, but not transcription of vaccinia virus mRNAs is compromised in cells with deficient RQC activity. Additionally, vaccinia virus infection reduces cellular RQC activity, suggesting that co-option of ZNF598 by vaccinia virus plays a critical role in translational reprogramming that is needed for optimal viral propagation.


Asunto(s)
Virus Vaccinia , Vaccinia , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Biosíntesis de Proteínas , Control de Calidad , Ribosomas/metabolismo , Vaccinia/genética , Virus Vaccinia/genética
7.
Virology ; 558: 22-27, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33711560

RESUMEN

Cytomegalovirus (CMV) promoter drives various gene expression and yields sufficient protein for further functional investigation. Receptor binding domain (RBD) on spike protein of the SARS_CoV2 is the most critical portal for virus infection. Thus native conformational RBD protein may facilitate biochemical identification of RBD and provide valuable support of drug and vaccine design for curing COVID-19. We attempted to express RBD under CMV promoter in vitro, but failed. RBD-specific mRNA cannot be detected in cell transfected with recombinant plasmids, in which CMV promoter governs the RBD transcription. Additionally, the pCMV-Tag2B-SARS_CoV2_RBD trans-inactivates CMV promoter transcription activity. Alternatively, we identified that both Chicken ß-actin promoter and Vaccinia virus-specific medium/late (M/L) promoter (pSYN) can highly precede SARS_CoV2 RBD expression. Our findings provided evidence that SARS_CoV2 RBD gene can be driven by Chicken ß-actin promoter or Vaccinia virus-specific medium/late promoter instead of CMV promoter, thus providing valuable information for RBD feature exploration.


Asunto(s)
COVID-19/virología , Citomegalovirus/genética , Regiones Promotoras Genéticas , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Actinas/genética , Animales , Células CHO , Pollos , Chlorocebus aethiops , Clonación Molecular , Cricetulus , Células HEK293 , Humanos , Dominios Proteicos , Transcripción Genética , Vaccinia/genética , Células Vero
8.
PLoS Pathog ; 17(1): e1009215, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33439897

RESUMEN

Poxvirus systems have been extensively used as vaccine vectors. Herein a RNA-Seq analysis of intramuscular injection sites provided detailed insights into host innate immune responses, as well as expression of vector and recombinant immunogen genes, after vaccination with a new multiplication defective, vaccinia-based vector, Sementis Copenhagen Vector. Chikungunya and Zika virus immunogen mRNA and protein expression was associated with necrosing skeletal muscle cells surrounded by mixed cellular infiltrates. The multiple adjuvant signatures at 12 hours post-vaccination were dominated by TLR3, 4 and 9, STING, MAVS, PKR and the inflammasome. Th1 cytokine signatures were dominated by IFNγ, TNF and IL1ß, and chemokine signatures by CCL5 and CXCL12. Multiple signatures associated with dendritic cell stimulation were evident. By day seven, vaccine transcripts were absent, and cell death, neutrophil, macrophage and inflammation annotations had abated. No compelling arthritis signatures were identified. Such injection site vaccinology approaches should inform refinements in poxvirus-based vector design.


Asunto(s)
Vectores Genéticos/administración & dosificación , Inmunidad Innata/inmunología , Reacción en el Punto de Inyección/inmunología , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vaccinia/inmunología , Infección por el Virus Zika/inmunología , Animales , Femenino , Vectores Genéticos/genética , Genoma Viral , Ratones , Ratones Endogámicos C57BL , RNA-Seq , Vacunas Sintéticas/inmunología , Vaccinia/genética , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/aislamiento & purificación , Vacunología , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología
9.
J Immunol ; 206(4): 776-784, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33419767

RESUMEN

There is a paucity of information on dendritic cell (DC) responses to vaccinia virus (VACV), including the traffic of DCs to the draining lymph node (dLN). In this study, using a mouse model of infection, we studied skin DC migration in response to VACV and compared it with the tuberculosis vaccine Mycobacterium bovis bacille Calmette-Guérin (BCG), another live attenuated vaccine administered via the skin. In stark contrast to BCG, skin DCs did not relocate to the dLN in response to VACV. Infection with UV-inactivated VACV or modified VACV Ankara promoted DC movement to the dLN, indicating that interference with skin DC migration requires replication-competent VACV. This suppressive effect of VACV was capable of mitigating responses to a secondary challenge with BCG in the skin, ablating DC migration, reducing BCG transport, and delaying CD4+ T cell priming in the dLN. Expression of inflammatory mediators associated with BCG-triggered DC migration were absent from virus-injected skin, suggesting that other pathways invoke DC movement in response to replication-deficient VACV. Despite adamant suppression of DC migration, VACV was still detected early in the dLN and primed Ag-specific CD4+ T cells. In summary, VACV blocks skin DC mobilization from the site of infection while retaining the ability to access the dLN to prime CD4+ T cells.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Piel/inmunología , Virus Vaccinia/inmunología , Vaccinia/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/genética , Ratones , Ratones Noqueados , Mycobacterium bovis/inmunología , Vaccinia/genética , Virus Vaccinia/genética
10.
PLoS Pathog ; 16(10): e1008660, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33075093

RESUMEN

Mammary carcinoma, including triple-negative breast carcinomas (TNBC) are tumor-types for which human and canine pathologies are closely related at the molecular level. The efficacy of an oncolytic vaccinia virus (VV) was compared in low-passage primary carcinoma cells from TNBC versus non-TNBC. Non-TNBC cells were 28 fold more sensitive to VV than TNBC cells in which VV replication is impaired. Single-cell RNA-seq performed on two different TNBC cell samples, infected or not with VV, highlighted three distinct populations: naïve cells, bystander cells, defined as cells exposed to the virus but not infected and infected cells. The transcriptomes of these three populations showed striking variations in the modulation of pathways regulated by cytokines and growth factors. We hypothesized that the pool of genes expressed in the bystander populations was enriched in antiviral genes. Bioinformatic analysis suggested that the reduced activity of the virus was associated with a higher mesenchymal status of the cells. In addition, we demonstrated experimentally that high expression of one gene, DDIT4, is detrimental to VV production. Considering that DDIT4 is associated with a poor prognosis in various cancers including TNBC, our data highlight DDIT4 as a candidate resistance marker for oncolytic poxvirus therapy. This information could be used to design new generations of oncolytic poxviruses. Beyond the field of gene therapy, this study demonstrates that single-cell transcriptomics can be used to identify cellular factors influencing viral replication.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Viroterapia Oncolítica/métodos , Factores de Transcripción/metabolismo , Transcriptoma , Virus Vaccinia/genética , Vaccinia/metabolismo , Replicación Viral , Animales , Biología Computacional , Perros , Femenino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/terapia , Neoplasias Mamarias Animales/virología , Análisis de la Célula Individual , Factores de Transcripción/genética , Vaccinia/genética , Vaccinia/virología
11.
J Virol ; 94(24)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32999026

RESUMEN

We conducted an exhaustive search for three-dimensional structural homologs to the proteins of 20 key phylogenetically distinct nucleocytoplasmic DNA viruses (NCLDV). Structural matches covered 429 known protein domain superfamilies, with the most highly represented being ankyrin repeat, P-loop NTPase, F-box, protein kinase, and membrane occupation and recognition nexus (MORN) repeat. Domain superfamily diversity correlated with genome size, but a diversity of around 200 superfamilies appeared to correlate with an abrupt switch to paralogization. Extensive structural homology was found across the range of eukaryotic RNA polymerase II subunits and their associated basal transcription factors, with the coordinated gain and loss of clusters of subunits on a virus-by-virus basis. The total number of predicted endonucleases across the 20 NCLDV was nearly quadrupled from 36 to 132, covering much of the structural and functional diversity of endonucleases throughout the biosphere in DNA restriction, repair, and homing. Unexpected findings included capsid protein-transcription factor chimeras; endonuclease chimeras; enzymes for detoxification; antimicrobial peptides and toxin-antitoxin systems associated with symbiosis, immunity, and addiction; and novel proteins for membrane abscission and protein turnover.IMPORTANCE We extended the known annotation space for the NCLDV by 46%, revealing high-probability structural matches for fully 45% of the 9,671 query proteins and confirming up to 98% of existing annotations per virus. The most prevalent protein families included ankyrin repeat- and MORN repeat-containing proteins, many of which included an F-box, suggesting extensive host cell modulation among the NCLDV. Regression suggested a minimum requirement for around 36 protein structural superfamilies for a viable NCLDV, and beyond around 200 superfamilies, genome expansion by the acquisition of new functions was abruptly replaced by paralogization. We found homologs to herpesvirus surface glycoprotein gB in cytoplasmic viruses. This study provided the first prediction of an endonuclease in 10 of the 20 viruses examined; the first report in a virus of a phenolic acid decarboxylase, proteasomal subunit, or cysteine knot (defensin) protein; and the first report of a prokaryotic-type ribosomal protein in a eukaryotic virus.


Asunto(s)
Virus ADN/clasificación , Virus ADN/genética , Virus Gigantes/genética , Filogenia , Proteoma/genética , Proteínas Virales/genética , Ancirinas/genética , Citoplasma/virología , ARN Polimerasas Dirigidas por ADN , Células Eucariotas , Evolución Molecular , Tamaño del Genoma , Genoma Viral , Mimiviridae/genética , Vaccinia/genética
12.
PLoS One ; 15(4): e0230711, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32240193

RESUMEN

Vaccinia virus (VACV) has been used extensively as the vaccine against smallpox and as a viral vector for the development of recombinant vaccines and cancer therapies. Replication-competent, non-attenuated VACVs induce strong, long-lived humoral and cell-mediated immune responses and can be effective oncolytic vectors. However, complications from uncontrolled VACV replication in vaccinees and their close contacts can be severe, particularly in individuals with predisposing conditions. In an effort to develop replication-competent VACV vectors with improved safety, we placed VACV late genes encoding core or virion morphogenesis proteins under the control of tet operon elements to regulate their expression with tetracycline antibiotics. These replication-inducible VACVs would only express the selected genes in the presence of tetracyclines. VACVs inducibly expressing the A3L or A6L genes replicated indistinguishably from wild-type VACV in the presence of tetracyclines, whereas there was no evidence of replication in the absence of antibiotics. These outcomes were reflected in mice, where the VACV inducibly expressing the A6L gene caused weight loss and mortality equivalent to wild-type VACV in the presence of tetracyclines. In the absence of tetracyclines, mice were protected from weight loss and mortality, and viral replication was not detected. These findings indicate that replication-inducible VACVs based on the conditional expression of the A3L or A6L genes can be used for the development of safer, next-generation live VACV vectors and vaccines. The design allows for administration of replication-inducible VACV in the absence of tetracyclines (as a replication-defective vector) or in the presence of tetracyclines (as a replication-competent vector) with enhanced safety.


Asunto(s)
Vectores Genéticos/administración & dosificación , Tetraciclinas/farmacología , Virus Vaccinia/crecimiento & desarrollo , Vaccinia/prevención & control , Virión/crecimiento & desarrollo , Replicación Viral , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Vacunas Sintéticas/administración & dosificación , Vaccinia/genética , Vaccinia/virología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/genética , Proteínas Virales/genética , Virión/efectos de los fármacos
13.
PLoS Pathog ; 16(2): e1008279, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32023327

RESUMEN

IFN-γ is an enigmatic cytokine that shows direct anti-viral effects, confers upregulation of MHC-II and other components relevant for antigen presentation, and that adjusts the composition and balance of complex cytokine responses. It is produced during immune responses by innate as well as adaptive immune cells and can critically affect the course and outcome of infectious diseases, autoimmunity, and cancer. To selectively analyze the function of innate immune cell-derived IFN-γ, we generated conditional IFN-γOFF mice, in which endogenous IFN-γ expression is disrupted by a loxP flanked gene trap cassette inserted into the first intron of the IFN-γ gene. IFN-γOFF mice were intercrossed with Ncr1-Cre or CD4-Cre mice that express Cre mainly in NK cells (IFN-γNcr1-ON mice) or T cells (IFN-γCD4-ON mice), respectively. Rosa26RFP reporter mice intercrossed with Ncr1-Cre mice showed selective RFP expression in more than 80% of the NK cells, while upon intercrossing with CD4-Cre mice abundant RFP expression was detected in T cells, but also to a minor extent in other immune cell subsets. Previous studies showed that IFN-γ expression is needed to promote survival of vaccinia virus (VACV) infection. Interestingly, during VACV infection of wild type and IFN-γCD4-ON mice two waves of serum IFN-γ were induced that peaked on day 1 and day 3/4 after infection. Similarly, VACV infected IFN-γNcr1-ON mice mounted two waves of IFN-γ responses, of which the first one was moderately and the second one profoundly reduced when compared with WT mice. Furthermore, IFN-γNcr1-ON as well as IFN-γCD4-ON mice survived VACV infection, whereas IFN-γOFF mice did not. As expected, ex vivo analysis of splenocytes derived from VACV infected IFN-γNcr1-ON mice showed IFN-γ expression in NK cells, but not T cells, whereas IFN-γOFF mice showed IFN-γ expression neither in NK cells nor T cells. VACV infected IFN-γNcr1-ON mice mounted normal cytokine responses, restored neutrophil accumulation, and showed normal myeloid cell distribution in blood and spleen. Additionally, in these mice normal MHC-II expression was detected on peripheral macrophages, whereas IFN-γOFF mice did not show MHC-II expression on such cells. In conclusion, upon VACV infection Ncr1 positive cells including NK cells mount two waves of early IFN-γ responses that are sufficient to promote the induction of protective anti-viral immunity.


Asunto(s)
Antígenos Ly/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Virus Vaccinia/inmunología , Vaccinia/inmunología , Animales , Antígenos Ly/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interferón gamma/genética , Células Asesinas Naturales/patología , Ratones , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Linfocitos T/inmunología , Linfocitos T/patología , Vaccinia/genética , Vaccinia/patología , Virus Vaccinia/genética
14.
J Leukoc Biol ; 107(6): 941-952, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31985117

RESUMEN

Receptor interacting protein kinase 1 (RIP1) is a critical effector of inflammatory responses and cell death activation. Cell death pathways regulated by RIP1 include caspase-dependent apoptosis and caspase-independent necroptosis. The kinase activity of RIP1 has been associated with a number of inflammatory, neurodegenerative, and oncogenic diseases. In this study, we use the RIP1 kinase inhibitor GNE684 to demonstrate that RIP1 inhibition can effectively block skin inflammation and immune cell infiltrates in livers of Sharpin mutant (Cpdm; chronic proliferative dermatitis) mice in an interventional setting, after disease onset. On the other hand, genetic inactivation of RIP1 (RIP1 KD) or ablation of RIP3 (RIP3 KO) or MLKL (MLKL KO) did not affect testicular pathology of aging male mice. Likewise, infection with vaccinia virus or with mouse gammaherpesvirus MHV68 resulted in similar viral clearance in wild-type, RIP1 KD, and RIP3 KO mice. In summary, this study highlights the benefits of inhibiting RIP1 in skin inflammation, as opposed to its lack of relevance for testicular longevity and the response to certain viral infections.


Asunto(s)
Dermatitis/genética , Infecciones por Herpesviridae/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Piel/inmunología , Vaccinia/genética , Animales , Enfermedad Crónica , Dermatitis/inmunología , Dermatitis/patología , Dermatitis/virología , Modelos Animales de Enfermedad , Gammaherpesvirinae/inmunología , Gammaherpesvirinae/patogenicidad , Regulación de la Expresión Génica , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Inflamación , Hígado/inmunología , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Transducción de Señal , Piel/patología , Piel/virología , Testículo/inmunología , Testículo/patología , Testículo/virología , Vaccinia/inmunología , Vaccinia/patología , Vaccinia/virología , Virus Vaccinia/inmunología , Virus Vaccinia/patogenicidad , Replicación Viral/inmunología
15.
Sci Rep ; 9(1): 15684, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31666569

RESUMEN

After assembly in the cytosol, some Vaccinia virus particles go through a complex process that leads to virus egress and eventually cell-to-cell transmission. Intracellular particles are fully infectious, and therefore virus mutants lacking essential functions in the exit pathway are unable to form plaques but can multiply intracellularly. We isolated virus mutants in which two of the genes required for virus spread (F13L and A27L) were deleted independently or concurrently. The phenotypes of the mutant viruses were consistent with the need of A27L and F13L for intercellular virus transmission, the effect of the ΔA27L mutation being more severe than that of ΔF13L. Despite their defect in spread, ΔA27L mutant viruses could be expanded by infecting cell cultures at high multiplicity of infection, followed by the release of virions from infected cells by physical means. We developed a novel system for the isolation of recombinant Vaccinia virus in which selection is efficiently achieved by recovering plaque formation capacity after re-introduction of A27L into a ΔA27L virus. This system allowed the insertion of foreign DNA into the viral genome without the use of additional genetic markers. Furthermore, starting with a double mutant (ΔA27L-ΔF13L) virus, A27L selection was used in conjunction with F13L selection to mediate simultaneous dual insertions in the viral genome. This selection system facilitates combined expression of multiple foreign proteins from a single recombinant virus.


Asunto(s)
Marcadores Genéticos/genética , Virus Vaccinia/genética , Vaccinia/genética , Proteínas del Envoltorio Viral/genética , Línea Celular , Citosol/metabolismo , Citosol/virología , Humanos , Proteínas Mutantes/genética , Mutación/genética , Vaccinia/virología , Virus Vaccinia/patogenicidad , Virión/genética , Virión/crecimiento & desarrollo
16.
Virol Sin ; 34(5): 583-591, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31240620

RESUMEN

Conventional influenza vaccines need to be designed and manufactured yearly. However, they occasionally provide poor protection owing to antigenic mismatch. Hence, there is an urgent need to develop universal vaccines against influenza virus. Using nucleoprotein (NP) and extracellular domain of matrix protein 2 (M2e) genes from the influenza A virus A/Beijing/30/95 (H3N2), we constructed four recombinant vaccinia virus-based influenza vaccines carrying NP fused with one or four copies of M2e genes in different orders. The recombinant vaccinia viruses were used to immunize BALB/C mice. Humoral and cellular responses were measured, and then the immunized mice were challenged with the influenza A virus A/Puerto Rico/8/34 (PR8). NP-specific humoral response was elicited in mice immunized with recombinant vaccinia viruses carrying full-length NP, while robust M2e-specific humoral response was elicited only in the mice immunized with recombinant vaccinia viruses carrying multiple copies of M2e. All recombinant viruses elicited NP- and M2e-specific cellular immune responses in mice. Only immunization with RVJ-4M2eNP induced remarkably higher levels of IL-2 and IL-10 cytokines specific to M2e. Furthermore, RVJ-4M2eNP immunization provided the highest cross-protection in mice challenged with 20 MLD50 of PR8. Therefore, the cross-protection potentially correlates with both NP and M2e-specific humoral and cellular immune responses induced by RVJ-4M2eNP, which expresses a fusion antigen of full-length NP preceded by four M2e repeats. These results suggest that the rational fusion of NP and multiple M2e antigens is critical toward inducing protective immune responses, and the 4M2eNP fusion antigen may be employed to develop a universal influenza vaccine.


Asunto(s)
Protección Cruzada , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Nucleoproteínas/inmunología , Vaccinia/genética , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Femenino , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunización , Subtipo H3N2 del Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Ratones Endogámicos BALB C , Nucleoproteínas/genética , Infecciones por Orthomyxoviridae/prevención & control , Organismos Libres de Patógenos Específicos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas de la Matriz Viral/genética
17.
J Biol Chem ; 294(16): 6416-6429, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30819806

RESUMEN

BTB-Kelch proteins are substrate-specific adaptors for cullin-3 (Cul3) RING-box-based E3 ubiquitin ligases, mediating protein ubiquitylation for subsequent proteasomal degradation. Vaccinia virus encodes three BTB-Kelch proteins: A55, C2, and F3. Viruses lacking A55 or C2 have altered cytopathic effects in cultured cells and altered pathology in vivo Previous studies have shown that the ectromelia virus orthologue of A55 interacts with Cul3 in cells. We report that the N-terminal BTB-BACK (BB) domain of A55 binds directly to the Cul3 N-terminal domain (Cul3-NTD), forming a 2:2 complex in solution. We solved the structure of an A55BB/Cul3-NTD complex from anisotropic crystals diffracting to 2.3/3.7 Å resolution in the best/worst direction, revealing that the overall interaction and binding interface closely resemble the structures of cellular BTB/Cul3-NTD complexes, despite low sequence identity between A55 and cellular BTB domains. Surprisingly, despite this structural similarity, the affinity of Cul3-NTD for A55BB was stronger than for cellular BTB proteins. Glutamate substitution of the A55 residue Ile-48, adjacent to the canonical φX(D/E) Cul3-binding motif, reduced affinity of A55BB for Cul3-NTD by at least 2 orders of magnitude. Moreover, Ile-48 and the φX(D/E) motif are conserved in A55 orthologues from other poxviruses, but not in the vaccinia virus proteins C2 or F3. The high-affinity interaction between A55BB and Cul3-NTD suggests that, in addition to directing the Cul3-RING E3 ligase complex to degrade cellular/viral target proteins that are normally unaffected, A55 may also sequester Cul3 from cellular adaptor proteins, thereby protecting substrates of these cellular adaptors from ubiquitylation and degradation.


Asunto(s)
Proteínas Cullin/química , Complejos Multiproteicos/química , Virus Vaccinia/química , Proteínas Virales/química , Sustitución de Aminoácidos , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación Missense , Dominios Proteicos , Estructura Cuaternaria de Proteína , Proteolisis , Ubiquitinación/genética , Vaccinia/genética , Vaccinia/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Proteínas Virales/genética
18.
J Virol ; 93(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30918073

RESUMEN

In this article we report that the M2 protein encoded by the vaccinia virus is secreted as a homo-oligomer by infected cells and binds two central costimulation molecules, CD80 (B7-1) and CD86 (B7-2). These interactions block the ligation of the two B7 proteins to both soluble CD28 and soluble cytotoxic T-lymphocyte associated protein 4 (CTLA4) but favor the binding of soluble PD-L1 to soluble CD80. M2L gene orthologues are found in several other poxviruses, and the B7-CD28/CTLA4 blocking activity has been identified for several culture supernatants of orthopoxvirus-infected cells and for a recombinant myxoma virus M2 protein homolog (i.e., Gp120-like protein, or Gp120LP). Overall, these data indicate that the M2 poxvirus family of proteins may be involved in immunosuppressive activities broader than the NF-κB inhibition already reported (R. Gedey, X. L. Jin, O. Hinthong, and J. L. Shisler, J Virol 80:8676-8685, 2006, https://doi.org/10.1128/JVI.00935-06). A Copenhagen vaccinia virus with a deletion of the nonessential M2L locus was generated and compared with its parental virus. This M2L-deleted vaccinia virus, unlike the parental virus, does not generate interference with the B7-CD28/CTLA4/PD-L1 interactions. Moreover, this deletion did not affect any key features of the virus (in vitro replication, oncolytic activities in vitro and in vivo, and intratumoral expression of a transgene in an immunocompetent murine model). Altogether, these first results suggest that the M2 protein has the potential to be used as a new immunosuppressive biotherapeutic and that the M2L-deleted vaccinia virus represents an attractive new oncolytic platform with an improved immunological profile.IMPORTANCE The vaccinia virus harbors in its genome several genes dedicated to the inhibition of the host immune response. Among them, M2L was reported to inhibit the intracellular NF-κB pathway. We report here several new putative immunosuppressive activities of M2 protein. M2 protein is secreted and binds cornerstone costimulatory molecules (CD80/CD86). M2 binding to CD80/CD86 blocks their interaction with soluble CD28/CTLA4 but also favors the soluble PD-L1-CD80 association. These findings open the way for new investigations deciphering the immune system effects of soluble M2 protein. Moreover, a vaccinia virus with a deletion of its M2L has been generated and characterized as a new oncolytic platform. The replication and oncolytic activities of the M2L-deleted vaccinia virus are indistinguishable from those of the parental virus. More investigations are needed to characterize in detail the immune response triggered against both the tumor and the virus by this M2-defective vaccinia virus.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Virus Vaccinia/metabolismo , Animales , Antígenos CD/metabolismo , Antígeno B7-1/genética , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Moléculas de Adhesión Celular , Línea Celular , Embrión de Pollo , Humanos , Inmunoconjugados , Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , FN-kappa B/metabolismo , Vaccinia/genética , Vaccinia/metabolismo , Virus Vaccinia/genética , Proteínas Virales/metabolismo
19.
Sci Rep ; 9(1): 2211, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30778113

RESUMEN

Aurora A is a serine/threonine kinase whose role in cell cycle progression and tumour generation has been widely studied. Recent work has revealed an unexpected function for Aurora A during CD4+ T cell activation and, also, in graft versus host disease development. However, it remains unknown whether Aurora A is involved in CD8+ T cell effector function and in cytotoxic T lymphocyte-mediated antiviral response. Here, we show that Aurora A chemical inhibition leads to an impairment of both the peptide-specific cytotoxicity and the degranulation activity of CD8+ T cells. This finding was similarly proven for both mice and human CD8+ CTL activity. As a result of Aurora A blockade, we detected a reduction in the expression induced by T cell activation of genes classically related to the effector function of cytotoxic T lymphocytes such as granzyme B or perforin1. Finally, we have found that Aurora A is necessary for CD8+ T cell-mediated antiviral response, in an in vivo model of vaccinia virus infection. Thus, we can conclude that Aurora A activity is, indeed, needed for the proper effector function of cytotoxic T lymphocytes and for their activity against viral threats.


Asunto(s)
Aurora Quinasa A/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Virosis/genética , Virosis/inmunología , Animales , Aurora Quinasa A/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Inmunomodulación/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Vaccinia/genética , Vaccinia/inmunología , Vaccinia/virología , Virus Vaccinia/inmunología , Virosis/virología
20.
J Virol ; 92(23)2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30209174

RESUMEN

Replication of vaccinia virus in human cells depends on the viral C7 or K1 protein. A previous human genome-wide short interfering RNA (siRNA) screen with a C7/K1 double deletion mutant revealed SAMD9 as a principal host range restriction factor along with additional candidates, including WDR6 and FTSJ1. To compare their abilities to restrict replication, the cellular genes were individually inactivated by CRISPR/Cas9 mutagenesis. The C7/K1 deletion mutant exhibited enhanced replication in each knockout (KO) cell line but reached wild-type levels only in SAMD9 KO cells. SAMD9 was not depleted in either WDR6 or FTSJ1 KO cells, suggesting less efficient alternative rescue mechanisms. Using the SAMD9 KO cells as controls, we verified a specific block in host and viral intermediate and late protein synthesis in HeLa cells and demonstrated that the inhibition could be triggered by events preceding viral DNA replication. Inhibition of cap-dependent and -independent protein synthesis occurred primarily at the translational level, as supported by DNA and mRNA transfection experiments. Concurrent with collapse of polyribosomes, viral mRNA was predominantly in 80S and lighter ribonucleoprotein fractions. We confirmed the accumulation of cytoplasmic granules in HeLa cells infected with the C7/K1 deletion mutant and further showed that viral mRNA was sequestered with SAMD9. RNA granules were still detected in G3BP KO U2OS cells, which remained nonpermissive for the C7/K1 deletion mutant. Inhibition of cap-dependent and internal ribosome entry site-mediated translation, sequestration of viral mRNA, and failure of PKR, RNase L, or G3BP KO cells to restore protein synthesis support an unusual mechanism of host restriction.IMPORTANCE A dynamic relationship exists between viruses and their hosts in which each ostensibly attempts to exploit the other's vulnerabilities. A window is opened into the established condition, which evolved over millennia, if loss-of-function mutations occur in either the virus or host. Thus, the inability of viral host range mutants to replicate in specific cells can be overcome by identifying and inactivating the opposing cellular gene. Here, we investigated a C7/K1 host range mutant of vaccinia virus in which the cellular gene SAMD9 serves as the principal host restriction factor. Host restriction was triggered early in infection and manifested as a block in translation of viral mRNAs. Features of the block include inhibition of cap-dependent and internal ribosome entry site-mediated translation, sequestration of viral RNA, and inability to overcome the inhibition by inactivation of protein kinase R, ribonuclease L, or G3 binding proteins, suggesting a novel mechanism of host restriction.


Asunto(s)
Proteínas de la Membrana/metabolismo , Metiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Biosíntesis de Proteínas , Proteínas/metabolismo , Eliminación de Secuencia , Vaccinia/genética , Replicación Viral , Secuencia de Aminoácidos , Secuencia de Bases , Sistemas CRISPR-Cas , Células HeLa , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas/antagonistas & inhibidores , Proteínas/genética , ARN Viral , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/genética , Virus Vaccinia/aislamiento & purificación , Virus Vaccinia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA