Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 683
Filtrar
1.
Nat Commun ; 15(1): 3856, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719852

RESUMEN

The Modified Vaccinia Ankara vaccine developed by Bavarian Nordic (MVA-BN) was widely deployed to prevent mpox during the 2022 global outbreak. This vaccine was initially approved for mpox based on its reported immunogenicity (from phase I/II trials) and effectiveness in animal models, rather than evidence of clinical efficacy. However, no validated correlate of protection after vaccination has been identified. Here we performed a systematic search and meta-analysis of the available data to test whether vaccinia-binding ELISA endpoint titer is predictive of vaccine effectiveness against mpox. We observe a significant correlation between vaccine effectiveness and vaccinia-binding antibody titers, consistent with the existing assumption that antibody levels may be a correlate of protection. Combining this data with analysis of antibody kinetics after vaccination, we predict the durability of protection after vaccination and the impact of dose spacing. We find that delaying the second dose of MVA-BN vaccination will provide more durable protection and may be optimal in an outbreak with limited vaccine stock. Although further work is required to validate this correlate, this study provides a quantitative evidence-based approach for using antibody measurements to predict the effectiveness of mpox vaccination.


Asunto(s)
Anticuerpos Antivirales , Eficacia de las Vacunas , Virus Vaccinia , Humanos , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Virus Vaccinia/inmunología , Vacunación/métodos , Ensayo de Inmunoadsorción Enzimática , Vaccinia/inmunología , Vaccinia/prevención & control , Vacuna contra Viruela/inmunología , Vacuna contra Viruela/administración & dosificación , Animales
2.
J Virol ; 97(12): e0127223, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38009914

RESUMEN

IMPORTANCE: Human poxvirus infections have caused significant public health burdens both historically and recently during the unprecedented global Mpox virus outbreak. Although vaccinia virus (VACV) infection of mice is a commonly used model to explore the anti-poxvirus immune response, little is known about the metabolic changes that occur in vivo during infection. We hypothesized that the metabolome of VACV-infected skin would reflect the increased energetic requirements of both virus-infected cells and immune cells recruited to sites of infection. Therefore, we profiled whole VACV-infected skin using untargeted mass spectrometry to define the metabolome during infection, complementing these experiments with flow cytometry and transcriptomics. We identified specific metabolites, including nucleotides, itaconic acid, and glutamine, that were differentially expressed during VACV infection. Together, this study offers insight into both virus-specific and immune-mediated metabolic pathways that could contribute to the clearance of cutaneous poxvirus infection.


Asunto(s)
Reprogramación Metabólica , Metaboloma , Piel , Virus Vaccinia , Vaccinia , Animales , Ratones , Citometría de Flujo , Perfilación de la Expresión Génica , Glutamina/metabolismo , Espectrometría de Masas , Nucleótidos/metabolismo , Piel/inmunología , Piel/metabolismo , Piel/virología , Vaccinia/inmunología , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/metabolismo , Carga Viral
3.
Front Immunol ; 12: 727046, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691033

RESUMEN

Vaccinia virus (VV) is the most studied member of the poxvirus family, is responsible for the successful elimination of smallpox worldwide, and has been developed as a vaccine vehicle for infectious diseases and cancer immunotherapy. We have previously shown that the unique potency of VV in the activation of CD8+ T cell response is dependent on efficient activation of the innate immune system through Toll-like receptor (TLR)-dependent and -independent pathways. However, it remains incompletely defined what regulate CD8+ T cell response to VV infection. In this study, we showed that γδT cells play an important role in promoting CD8+ T cell response to VV infection. We found that γδT cells can directly present viral antigens in the context of MHC-I for CD8+ T cell activation to VV in vivo, and we further demonstrated that cell-intrinsic MyD88 signaling in γδT cells is required for activation of γδT cells and CD8+ T cells. These results illustrate a critical role for γδT cells in the regulation of adaptive T cell response to viral infection and may shed light on the design of more effective vaccine strategies based on manipulation of γδT cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Intraepiteliales/inmunología , Vaccinia/inmunología , Animales , Presentación de Antígeno , Antígenos Virales/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Virus Vaccinia
4.
Viruses ; 13(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34452494

RESUMEN

Mass vaccination has played a critical role in the global eradication of smallpox. Various vaccinia virus (VACV) strains, whose origin has not been clearly documented in most cases, have been used as live vaccines in different countries. These VACV strains differed in pathogenicity towards various laboratory animals and in reactogenicity exhibited upon vaccination of humans. In this work, we studied the development of humoral and cellular immune responses in BALB/c mice inoculated intranasally (i.n.) or intradermally (i.d.) with the VACV LIVP strain at a dose of 105 PFU/mouse, which was used in Russia as the first generation smallpox vaccine. Active synthesis of VACV-specific IgM in the mice occurred on day 7 after inoculation, reached a maximum on day 14, and decreased by day 29. Synthesis of virus-specific IgG was detected only from day 14, and the level increased significantly by day 29 after infection of the mice. Immunization (i.n.) resulted in significantly higher production of VACV-specific antibodies compared to that upon i.d. inoculation of LIVP. There were no significant differences in the levels of the T cell response in mice after i.n. or i.d. VACV administration at any time point. The maximum level of VACV-specific T-cells was detected on day 14. By day 29 of the experiment, the level of VACV-specific T-lymphocytes in the spleen of mice significantly decreased for both immunization procedures. On day 30 after immunization with LIVP, mice were infected with the cowpox virus at a dose of 46 LD50. The i.n. immunized mice were resistant to this infection, while 33% of i.d. immunized mice died. Our findings indicate that the level of the humoral immune response to vaccination may play a decisive role in protection of animals from orthopoxvirus reinfection.


Asunto(s)
Inmunidad Adaptativa , Virus de la Viruela Vacuna/fisiología , Viruela Vacuna/prevención & control , Reinfección/prevención & control , Virus Vaccinia/inmunología , Vaccinia/inmunología , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Viruela Vacuna/inmunología , Viruela Vacuna/virología , Virus de la Viruela Vacuna/genética , Virus de la Viruela Vacuna/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Reinfección/inmunología , Reinfección/virología , Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vaccinia/virología , Virus Vaccinia/genética , Virus Vaccinia/fisiología , Vacunas Virales/inmunología
5.
Ann Rheum Dis ; 80(12): 1537-1544, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34226189

RESUMEN

OBJECTIVES: The monoclonal anti-CD20 antibody rituximab is frequently applied in the treatment of lymphoma as well as autoimmune diseases and confers efficient depletion of recirculating B cells. Correspondingly, B cell-depleted patients barely mount de novo antibody responses during infections or vaccinations. Therefore, efficient immune responses of B cell-depleted patients largely depend on protective T cell responses. METHODS: CD8+ T cell expansion was studied in rituximab-treated rheumatoid arthritis (RA) patients and B cell-deficient mice on vaccination/infection with different vaccines/pathogens. RESULTS: Rituximab-treated RA patients vaccinated with Influvac showed reduced expansion of influenza-specific CD8+ T cells when compared with healthy controls. Moreover, B cell-deficient JHT mice infected with mouse-adapted Influenza or modified vaccinia virus Ankara showed less vigorous expansion of virus-specific CD8+ T cells than wild type mice. Of note, JHT mice do not have an intrinsic impairment of CD8+ T cell expansion, since infection with vaccinia virus induced similar T cell expansion in JHT and wild type mice. Direct type I interferon receptor signalling of B cells was necessary to induce several chemokines in B cells and to support T cell help by enhancing the expression of MHC-I. CONCLUSIONS: Depending on the stimulus, B cells can modulate CD8+ T cell responses. Thus, B cell depletion causes a deficiency of de novo antibody responses and affects the efficacy of cellular response including cytotoxic T cells. The choice of the appropriate vaccine to vaccinate B cell-depleted patients has to be re-evaluated in order to efficiently induce protective CD8+ T cell responses.


Asunto(s)
Antirreumáticos/efectos adversos , Artritis Reumatoide/tratamiento farmacológico , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunogenicidad Vacunal/inmunología , Vacunas contra la Influenza/inmunología , Interferón Tipo I/inmunología , Rituximab/efectos adversos , Animales , Estudios de Casos y Controles , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Ratones , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/inmunología , Vaccinia/inmunología , Virus Vaccinia/inmunología
6.
Virol J ; 18(1): 124, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107993

RESUMEN

BACKGROUND: The vaccinia virus (VACV) isolates, Guarani P1 virus (GP1V) and Passatempo virus (PSTV), were isolated during zoonotic outbreaks in Brazil. Each one of them belongs to two different VACV clades, defined by biological aspects that include virulence in mice and phylogenetic analysis. Considering that information about how vaccinia viruses from different groups elicit immune responses in animals is scarce, we investigated such responses in mice infected either by GP1V (group 2) or PSTV (group 1), using VACV Western Reserve strain (VACV-WR) as control. METHODS: The severity of the infections was evaluated in BALB/c mice considering diverse clinical signs and defined scores, and the immune responses triggered by GP1V and PSTV infections were analysed by immune cell phenotyping and intra-cytoplasmic cytokines detection. RESULTS: We detected a reduction in total lymphocytes (CD3 +), macrophages (CD14 +), and NK cells (CD3-CD49 +) in animals infected with VACV-WR or GP1V. The VACV-WR and GP1V viruses, belonging to the most virulent group in a murine model, were able to down-modulate the cell immune responses upon mice infection. In contrast, PSTV, a virus considered less virulent in a murine model, showed little ability to down-modulate the mice immune responses. Mice infected with VACV-WR and GP1V viruses presented significant weight loss and developed lesions in their spleens, as well as damage to liver and lungs whereas mice infected with PSTV developed only moderate clinical signs. CONCLUSIONS: Our results suggest that VACV immunomodulation in vivo is clade-related and is proportional to the strain's virulence upon infection. Our data corroborate the classification of the different Brazilian VACV isolates into clades 1 and 2, taking into account not only phylogenetic criteria, but also clinical and immunological data.


Asunto(s)
Inmunomodulación , Virus Vaccinia , Vaccinia , Animales , Modelos Animales de Enfermedad , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C , Filogenia , Vaccinia/inmunología , Vaccinia/virología , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Virulencia
7.
Front Immunol ; 12: 645210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959127

RESUMEN

Vaccination is one of the most efficient public healthcare measures to fight infectious diseases. Nevertheless, the immune mechanisms induced in vivo by vaccination are still unclear. The route of administration, an important vaccination parameter, can substantially modify the quality of the response. How the route of administration affects the generation and profile of immune responses is of major interest. Here, we aimed to extensively characterize the profiles of the innate and adaptive response to vaccination induced after intradermal, subcutaneous, or intramuscular administration with a modified vaccinia virus Ankara model vaccine in non-human primates. The adaptive response following subcutaneous immunization was clearly different from that following intradermal or intramuscular immunization. The subcutaneous route induced a higher level of neutralizing antibodies than the intradermal and intramuscular vaccination routes. In contrast, polyfunctional CD8+ T-cell responses were preferentially induced after intradermal or intramuscular injection. We observed the same dichotomy when analyzing the early molecular and cellular immune events, highlighting the recruitment of cell populations, such as CD8+ T lymphocytes and myeloid-derived suppressive cells, and the activation of key immunomodulatory gene pathways. These results demonstrate that the quality of the vaccine response induced by an attenuated vaccine is shaped by early and subtle modifications of the innate immune response. In this immunization context, the route of administration must be tailored to the desired type of protective immune response. This will be achieved through systems vaccinology and mathematical modeling, which will be critical for predicting the efficacy of the vaccination route for personalized medicine.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Células Supresoras de Origen Mieloide/inmunología , Vacunación , Virus Vaccinia/inmunología , Vaccinia/inmunología , Vacunas Virales/farmacología , Animales , Inyecciones Intradérmicas , Inyecciones Intramusculares , Macaca fascicularis , Masculino , Vacunas Atenuadas/farmacología
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.
Mucosal Immunol ; 14(3): 728-742, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33479479

RESUMEN

Induction of memory CD8 T cells residing in peripheral tissues is of interest for T cell-based vaccines as these cells are located at mucosal and barrier sites and can immediately exert effector functions, thus providing protection in case of local pathogen encounter. Different memory CD8 T cell subsets patrol peripheral tissues, but it is unclear which subset is superior in providing protection upon secondary infections. We used influenza virus to induce predominantly tissue resident memory T cells or cytomegalovirus to elicit a large pool of effector-like memory cells in the lungs and determined their early protective capacity and mechanism of reactivation. Both memory CD8 T cell pools have unique characteristics with respect to their phenotype, localization, and maintenance. However, these distinct features do not translate into different capacities to control a respiratory vaccinia virus challenge in an antigen-specific manner, although differential activation mechanisms are utilized. While influenza-induced memory CD8 T cells respond to antigen by local proliferation, MCMV-induced memory CD8 T cells relocate from the vasculature into the tissue in an antigen-independent and partially chemokine-driven manner. Together these results bear relevance for the development of vaccines aimed at eliciting a protective memory CD8 T cell pool at mucosal sites.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/inmunología , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Pulmón/inmunología , Muromegalovirus/fisiología , Infecciones por Orthomyxoviridae/inmunología , Virus Vaccinia/fisiología , Vaccinia/inmunología , Animales , Proliferación Celular , Células Cultivadas , Humanos , Memoria Inmunológica , Pulmón/virología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Activación Viral , Latencia del Virus
11.
Immunity ; 54(2): 276-290.e5, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33434494

RESUMEN

The oropharyngeal mucosa serves as a perpetual pathogen entry point and a critical site for viral replication and spread. Here, we demonstrate that type 1 innate lymphoid cells (ILC1s) were the major immune force providing early protection during acute oral mucosal viral infection. Using intravital microscopy, we show that ILC1s populated and patrolled the uninfected labial mucosa. ILC1s produced interferon-γ (IFN-γ) in the absence of infection, leading to the upregulation of key antiviral genes, which were downregulated in uninfected animals upon genetic ablation of ILC1s or antibody-based neutralization of IFN-γ. Thus, tonic IFN-γ production generates increased oral mucosal viral resistance even before infection. Our results demonstrate barrier-tissue protection through tissue surveillance in the absence of rearranged-antigen receptors and the induction of an antiviral state during homeostasis. This aspect of ILC1 biology raises the possibility that these cells do not share true functional redundancy with other tissue-resident lymphocytes.


Asunto(s)
Interferón gamma/metabolismo , Linfocitos/inmunología , Orofaringe/inmunología , Mucosa Respiratoria/inmunología , Virus Vaccinia/fisiología , Vaccinia/inmunología , Animales , Células Cultivadas , Resistencia a la Enfermedad , Humanos , Inmunidad Innata , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Células TH1/inmunología
12.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33177193

RESUMEN

The poxviral B1 and B12 proteins are a homologous kinase-pseudokinase pair, which modulates a shared host pathway governing viral DNA replication and antiviral defense. While the molecular mechanisms involved are incompletely understood, B1 and B12 seem to intersect with signaling processes mediated by their cellular homologs termed the vaccinia-related kinases (VRKs). In this study, we expand upon our previous characterization of the B1-B12 signaling axis to gain insights into B12 function. We begin our studies by demonstrating that modulation of B12 repressive activity is a conserved function of B1 orthologs from divergent poxviruses. Next, we characterize the protein interactome of B12 using multiple cell lines and expression systems and discover that the cellular kinase VRK1 is a highly enriched B12 interactor. Using complementary VRK1 knockdown and overexpression assays, we first demonstrate that VRK1 is required for the rescue of a B1-deleted virus upon mutation of B12. Second, we find that VRK1 overexpression is sufficient to overcome repressive B12 activity during B1-deleted virus replication. Interestingly, we also evince that B12 interferes with the ability of VRK1 to phosphoinactivate the host defense protein BAF. Thus, B12 restricts vaccinia virus DNA accumulation in part by repressing the ability of VRK1 to inactivate BAF. Finally, these data establish that a B12-VRK1-BAF signaling axis forms during vaccinia virus infection and is modulated via kinases B1 and/or VRK2. These studies provide novel insights into the complex mechanisms that poxviruses use to hijack homologous cellular signaling pathways during infection.IMPORTANCE Viruses from diverse families encode both positive and negative regulators of viral replication. While their functions can sometimes be enigmatic, investigation of virus-encoded, negative regulators of viral replication has revealed fascinating aspects of virology. Studies of poxvirus-encoded genes have largely concentrated on positive regulators of their replication; however, examples of fitness gains attributed to poxvirus gene loss suggests that negative regulators of poxvirus replication also impact infection dynamics. This study focuses on the vaccinia B12 pseudokinase, a protein capable of inhibiting vaccinia DNA replication. Here, we elucidate the mechanisms by which B12 inhibits vaccinia DNA replication, demonstrating that B12 activates the antiviral protein BAF by inhibiting the activity of VRK1, a cellular modulator of BAF. Combined with previous data, these studies provide evidence that poxviruses govern their replication by employing both positive and negative regulators of viral replication.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Virus Vaccinia/enzimología , Vaccinia/inmunología , Proteínas Virales/metabolismo , Antivirales , Proteínas de Unión al ADN/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Vaccinia/metabolismo , Vaccinia/virología , Proteínas Virales/genética
13.
Front Immunol ; 11: 1458, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765505

RESUMEN

Modified Vaccinia virus Ankara (MVA) is an attenuated strain of vaccinia virus and currently under investigation as a promising vaccine vector against infectious diseases and cancer. MVA acquired mutations in host range and immunomodulatory genes, rendering the virus deficient for replication in most mammalian cells. MVA has a high safety profile and induces robust immune responses. However, the role of innate immune triggers for the induction of cytotoxic T cell responses after vaccination is incompletely understood. Stimulator of interferon genes (STING) is an adaptor protein which integrates signaling downstream of several DNA sensors and therefore mediates the induction of type I interferons and other cytokines or chemokines in response to various dsDNA viruses. Since the type I interferon response was entirely STING-dependent during MVA infection, we studied the effect of STING on primary and secondary cytotoxic T cell responses and memory T cell formation after MVA vaccination in STING KO mice. Moreover, we analyzed the impact of STING on the maturation of bone marrow-derived dendritic cells (BMDCs) and their functionality as antigen presenting cells for cytotoxic T cells during MVA infection in vitro. Our results show that STING has an impact on the antigen processing and presentation capacity of conventionel DCs and played a crucial role for DC maturation and type I interferon production. Importantly, STING was required for the induction of efficient cytotoxic T cell responses in vivo, since we observed significantly decreased short-lived effector and effector memory T cell responses after priming in STING KO mice. These findings indicate that STING probably integrates innate immune signaling downstream of different DNA sensors in DCs and shapes the cytotoxic T cell response via the DC maturation phenotype which strongly depends on type I interferons in this infection model. Understanding the detailed functions of innate immune triggers during MVA infection will contribute to the optimized design of MVA-based vaccines.


Asunto(s)
Células Dendríticas/inmunología , Vectores Genéticos/genética , Proteínas de la Membrana/metabolismo , Linfocitos T Citotóxicos/inmunología , Virus Vaccinia/genética , Vaccinia/inmunología , Animales , Presentación de Antígeno , Células Cultivadas , Femenino , Humanos , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Vacunación
14.
Sci Rep ; 10(1): 8350, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32433493

RESUMEN

Regulatory T cells (Tregs) maintain peripheral self-tolerance and limit immune mediated pathology. Like effector T cells, Tregs can specialize in TH1-dominated immune responses and co-express T-bet together with Foxp3. This allows for expression of CXCR3 and efficient homing to sites of TH1 responses. However, whether such functional specialization is paralleled by memory generation among Tregs is unknown. In this study, we investigated the ability of polyclonal Tregs to form functional memory in response to viral infection. Using adoptive transfer models to compare infection-experienced Tregs generated upon acute Lymphocytic Choriomeningitis Virus (LCMV) WE and Vaccinia Virus (VV) infections with naive Tregs, we observed no differences in their phenotype or their in vivo maintenance. When comparing functional properties of infection-experienced and naive Tregs, we found no differences in in vitro suppressive capacity nor in their ability to limit the effector response upon homologous, systemic or local re-challenge in vivo. Our results suggest that no functional Treg memory is generated in the context of systemic LCMV or VV infection, but we cannot rule out the possibility that the generation of Treg memory may be possible in other contexts.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Memoria Inmunológica , Linfocitos T Reguladores/inmunología , Vaccinia/inmunología , Traslado Adoptivo , Animales , Infecciones por Arenaviridae/virología , Modelos Animales de Enfermedad , Humanos , Virus de la Coriomeningitis Linfocítica , Ratones , Linfocitos T Reguladores/trasplante , Vaccinia/virología , Virus Vaccinia/inmunología
15.
Virology ; 544: 55-63, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32174514

RESUMEN

Historic observations suggest that survivors of smallpox maintained lifelong immunity and protection to subsequent infection compared to vaccinated individuals. Although protective immunity by vaccination using a related virus (vaccinia virus (VACV) strains) was the key for smallpox eradication, it does not uniformly provide long term, or lifelong protective immunity (Heiner et al., 1971). To determine differences in humoral immune responses, mice were inoculated with VACV either systemically, using intranasal inoculation (IN), or locally by an intradermal (ID) route. We hypothesized that sub-lethal IN infections may mimic systemic or naturally occurring infection and lead to an immunodominance reaction, in contrast to localized ID immunization. The results demonstrated systemic immunization through an IN route led to enhanced adaptive immunity to VACV-expressed protein targets both in magnitude and in diversity when compared to an ID route using a VACV protein microarray. In addition, cytokine responses, assessed using a Luminex® mouse cytokine multiplex kit, following IN infection was greater than that stemming from ID infection. Overall, the results suggest that the route of immunization (or infection) influences antibody responses. The greater magnitude and diversity of response in systemic infection provides indirect evidence for anecdotal observations made during the smallpox era that survivors maintain lifelong protection. These findings also suggest that systemic or disseminated host immune induction may result in a superior response, that may influence the magnitude of, as well as duration of protective responses.


Asunto(s)
Inmunidad Humoral , Virus Vaccinia/inmunología , Vaccinia/inmunología , Inmunidad Adaptativa , Administración Intranasal , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vaccinia/virología
16.
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
17.
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
18.
PLoS Pathog ; 15(10): e1007778, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31603920

RESUMEN

Type I interferons (T1-IFN) are critical in the innate immune response, acting upon infected and uninfected cells to initiate an antiviral state by expressing genes that inhibit multiple stages of the lifecycle of many viruses. T1-IFN triggers the production of Interferon-Stimulated Genes (ISGs), activating an antiviral program that reduces virus replication. The importance of the T1-IFN response is highlighted by the evolution of viral evasion strategies to inhibit the production or action of T1-IFN in virus-infected cells. T1-IFN is produced via activation of pathogen sensors within infected cells, a process that is targeted by virus-encoded immunomodulatory molecules. This is probably best exemplified by the prototypic poxvirus, Vaccinia virus (VACV), which uses at least 6 different mechanisms to completely block the production of T1-IFN within infected cells in vitro. Yet, mice lacking aspects of T1-IFN signaling are often more susceptible to infection with many viruses, including VACV, than wild-type mice. How can these opposing findings be rationalized? The cytosolic DNA sensor cGAS has been implicated in immunity to VACV, but has yet to be linked to the production of T1-IFN in response to VACV infection. Indeed, there are two VACV-encoded proteins that effectively prevent cGAS-mediated activation of T1-IFN. We find that the majority of VACV-infected cells in vivo do not produce T1-IFN, but that a small subset of VACV-infected cells in vivo utilize cGAS to sense VACV and produce T1-IFN to protect infected mice. The protective effect of T1-IFN is not mediated via ISG-mediated control of virus replication. Rather, T1-IFN drives increased expression of CCL4, which recruits inflammatory monocytes that constrain the VACV lesion in a virus replication-independent manner by limiting spread within the tissue. Our findings have broad implications in our understanding of pathogen detection and viral evasion in vivo, and highlight a novel immune strategy to protect infected tissue.


Asunto(s)
Quimiocina CCL4/metabolismo , Interferón Tipo I/farmacología , Proteínas de la Membrana/fisiología , Nucleotidiltransferasas/fisiología , Virus Vaccinia/efectos de los fármacos , Vaccinia/prevención & control , Carga Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Quimiocina CCL4/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/virología , Vaccinia/inmunología , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/inmunología , Replicación Viral
19.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31375596

RESUMEN

A variety of strains of vaccinia virus (VACV) have been used as recombinant vaccine vectors with the aim of inducing robust CD8+ T cell immunity. While much of the pioneering work was done with virulent strains, such as Western Reserve (WR), attenuated strains such as modified vaccinia virus Ankara (MVA) are more realistic vectors for clinical use. To unify this literature, side-by-side comparisons of virus strains are required. Here, we compare the form of antigen that supports optimal CD8+ T cell responses for VACV strains WR and MVA using equivalent constructs. We found that for multiple antigens, minimal antigenic constructs (epitope minigenes) that prime CD8+ T cells via the direct presentation pathway elicited optimal responses from both vectors, which was surprising because this finding contradicts the prevailing view in the literature for MVA. We then went on to explore the discrepancy between current and published data for MVA, finding evidence that the expression locus and in some cases the presence of the viral thymidine kinase may influence the ability of this strain to prime optimal responses from antigens that require direct presentation. This extends our knowledge of the design parameters for VACV vectored vaccines, especially those based on MVA.IMPORTANCE Recombinant vaccines based on vaccinia virus and particularly attenuated strains such as MVA are in human clinical trials, but due to the complexity of these large vectors much remains to be understood about the design parameters that alter their immunogenicity. Previous work had found that MVA vectors should be designed to express stable protein in order to induce robust immunity by CD8+ (cytotoxic) T cells. Here, we found that the primacy of stable antigen is not generalizable to all designs of MVA and may depend where a foreign antigen is inserted into the MVA genome. This unexpected finding suggests that there is an interaction between genome location and the best form of antigen for optimal T cell priming in MVA and thus possibly other vaccine vectors. It also highlights that our understanding of antigen presentation by even the best studied of vaccine vectors remains incomplete.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Fragmentos de Péptidos/inmunología , Timidina Quinasa/metabolismo , Virus Vaccinia/inmunología , Vaccinia/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/genética , Linfocitos T CD8-positivos/metabolismo , Femenino , Genoma Viral , Inmunización , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Timidina Quinasa/genética , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/clasificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
20.
J Virol ; 93(19)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31315995

RESUMEN

CD69 is highly expressed on the leukocyte surface upon viral infection, and its regulatory role in the vaccinia virus (VACV) immune response has been recently demonstrated using CD69-/- mice. Here, we show augmented control of VACV infection using the anti-human CD69 monoclonal antibody (MAb) 2.8 as both preventive and therapeutic treatment for mice expressing human CD69. This control was related to increased natural killer (NK) cell reactivity and increased numbers of cytokine-producing T and NK cells in the periphery. Moreover, similarly increased immunity and protection against VACV were reproduced over both long and short periods in anti-mouse CD69 MAb 2.2-treated immunocompetent wild-type (WT) mice and immunodeficient Rag2-/- CD69+/+ mice. This result was not due to synergy between infection and anti-CD69 treatment since, in the absence of infection, anti-human CD69 targeting induced immune activation, which was characterized by mobilization, proliferation, and enhanced survival of immune cells as well as marked production of several innate proinflammatory cytokines by immune cells. Additionally, we showed that the rapid leukocyte effect induced by anti-CD69 MAb treatment was dependent on mTOR signaling. These properties suggest the potential of CD69-targeted therapy as an antiviral adjuvant to prevent derived infections.IMPORTANCE In this study, we demonstrate the influence of human and mouse anti-CD69 therapies on the immune response to VACV infection. We report that targeting CD69 increases the leukocyte numbers in the secondary lymphoid organs during infection and improves the capacity to clear the viral infection. Targeting CD69 increases the numbers of gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing NK and T cells. In mice expressing human CD69, treatment with an anti-CD69 MAb produces increases in cytokine production, survival, and proliferation mediated in part by mTOR signaling. These results, together with the fact that we have mainly worked with a human-CD69 transgenic model, reveal CD69 as a treatment target to enhance vaccine protectiveness.


Asunto(s)
Factores Inmunológicos/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Lectinas Tipo C/antagonistas & inhibidores , Linfocitos T/inmunología , Virus Vaccinia/inmunología , Vaccinia/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/administración & dosificación , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/administración & dosificación , Antígenos de Diferenciación de Linfocitos T/genética , Modelos Animales de Enfermedad , Humanos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/genética , Lectinas Tipo C/administración & dosificación , Lectinas Tipo C/genética , Ratones , Ratones Transgénicos , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Vaccinia/inmunología , Vaccinia/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...