Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1360140, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38711513

RESUMO

Introduction: Modified Vaccinia Virus Ankara (MVA) is a safe vaccine vector inducing long- lasting and potent immune responses. MVA-mediated CD8+T cell responses are optimally induced, if both, direct- and cross-presentation of viral or recombinant antigens by dendritic cells are contributing. Methods: To improve the adaptive immune responses, we investigated the role of the purinergic receptor P2X7 (P2RX7) in MVA-infected feeder cells as a modulator of cross-presentation by non-infected dendritic cells. The infected feeder cells serve as source of antigen and provide signals that help to attract dendritic cells for antigen take up and to license these cells for cross-presentation. Results: We demonstrate that presence of an active P2RX7 in major histocompatibility complex (MHC) class I (MHCI) mismatched feeder cells significantly enhanced MVA-mediated antigen cross-presentation. This was partly regulated by P2RX7-specific processes, such as the increased availability of extracellular particles as well as the altered cellular energy metabolism by mitochondria in the feeder cells. Furthermore, functional P2RX7 in feeder cells resulted in a delayed but also prolonged antigen expression after infection. Discussion: We conclude that a combination of the above mentioned P2RX7-depending processes leads to significantly increased T cell activation via cross- presentation of MVA-derived antigens. To this day, P2RX7 has been mostly investigated in regards to neuroinflammatory diseases and cancer progression. However, we report for the first time the crucial role of P2RX7 for antigen- specific T cell immunity in a viral infection model.


Assuntos
Apresentação Cruzada , Células Dendríticas , Vetores Genéticos , Receptores Purinérgicos P2X7 , Vaccinia virus , Animais , Humanos , Camundongos , Apresentação de Antígeno/imunologia , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2X7/imunologia , Receptores Purinérgicos P2X7/metabolismo , Vaccinia virus/imunologia
2.
Cell Rep ; 42(11): 113277, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37864791

RESUMO

Sensing of human immunodeficiency virus type 1 (HIV-1) DNA is mediated by the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling axis. Signal transduction and regulation of this cascade is achieved by post-translational modifications. Here we show that cGAS-STING-dependent HIV-1 sensing requires interferon-stimulated gene 15 (ISG15). ISG15 deficiency inhibits STING-dependent sensing of HIV-1 and STING agonist-induced antiviral response. Upon external stimuli, STING undergoes ISGylation at residues K224, K236, K289, K347, K338, and K370. Inhibition of STING ISGylation at K289 suppresses STING-mediated type Ⅰ interferon induction by inhibiting its oligomerization. Of note, removal of STING ISGylation alleviates gain-of-function phenotype in STING-associated vasculopathy with onset in infancy (SAVI). Molecular modeling suggests that ISGylation of K289 is an important regulator of oligomerization. Taken together, our data demonstrate that ISGylation at K289 is crucial for STING activation and represents an important regulatory step in DNA sensing of viruses and autoimmune responses.


Assuntos
DNA , Interferon Tipo I , Humanos , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Transdução de Sinais/genética , Imunidade Inata , Ubiquitinas , Citocinas
3.
J Infect Dis ; 228(9): 1227-1230, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37129073

RESUMO

The spread of nonzoonotic monkeypox virus (MPXV) infections necessitates the reevaluation of hygiene measures. To date, only limited data are available on MPXV surface stability. Here, we evaluate the stability of infectious MPXV on stainless steel stored at different temperatures, while using different interfering substances to mimic environmental contamination. MPXV persistence increased with decreasing temperature. Additionally, we were able to show that MPXV could efficiently be inactivated by alcohol- and aldehyde-based surface disinfectants. These findings underline the stability of MPXV on inanimate surfaces and support the recommendations to use alcohol-based disinfectants as prevention measures or in outbreak situations.


Assuntos
Desinfetantes , Monkeypox virus , Desinfetantes/farmacologia , Etanol , Temperatura , Aldeídos
4.
Emerg Infect Dis ; 29(1): 189-192, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36394568

RESUMO

Increasing nonzoonotic human monkeypox virus (MPXV) infections urge reevaluation of inactivation strategies. We demonstrate efficient inactivation of MPXV by 2 World Health Organization‒recommended alcohol-based hand rub solutions. When compared with other (re)emerging enveloped viruses, MPXV displayed the greatest stability. Our results support rigorous adherence to use of alcohol-based disinfectants.


Assuntos
Desinfetantes , Mpox , Vírus , Humanos , Monkeypox virus , Desinfetantes/farmacologia , Etanol , Mpox/epidemiologia , Mpox/prevenção & controle , 2-Propanol , Organização Mundial da Saúde
5.
Front Immunol ; 11: 1458, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32765505

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Vetores Genéticos/genética , Proteínas de Membrana/metabolismo , Linfócitos T Citotóxicos/imunologia , Vaccinia virus/genética , Vacínia/imunologia , Animais , Apresentação de Antígeno , Células Cultivadas , Feminino , Humanos , Interferon Tipo I/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Vacinação
6.
Front Immunol ; 10: 2850, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31867011

RESUMO

Cytotoxic CD8+ T cell (CTL) responses play an essential role in antiviral immunity. Here, we focused on the activation of CTL which recognize epitopes derived from viral or recombinant antigens with either early or late expression kinetics after infection with Modified Vaccinia Virus Ankara (MVA). Late antigens but not early antigens failed to efficiently stimulate murine CTL lines in vitro and were unable to activate and expand protective memory T cell responses in mice in vivo. The reduced or absent presentation of late antigens was not due to impaired antigen presentation or delayed protein synthesis, but was caused by sequestration of late antigens within viral factories (VFs). Additionally, the trapping of late antigens in VFs conflicts with antigen processing and presentation as proteasomal activity was strongly reduced or absent in VFs, suggesting inefficient antigen degradation. This study gives for the first time a mechanistic explanation for the weak immunogenicity of late viral antigens for memory CTL activation. Since MVA is preferentially used as a boost vector in heterologous prime/boost vaccinations, this is an important information for future vaccine design.


Assuntos
Antígenos Virais/imunologia , Memória Imunológica , Linfócitos T Citotóxicos/imunologia , Vaccinia virus/imunologia , Animais , Apresentação de Antígeno , Vetores Genéticos , Células HeLa , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo
7.
J Immunol ; 199(5): 1737-1747, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768725

RESUMO

Experimental CMV-based vaccine vectors expressing a single MHC class I-restricted high-avidity epitope provided strong, T cell-dependent protection against viruses or tumors. In this study we tested the low-avidity epitope KCSRNRQYL, and show that a mouse CMV (MCMV) vector provides complete immune control of recombinant vaccinia virus expressing the same epitope if KCSRNRQYL is expressed within the immediate-early MCMV gene ie2 The same epitope expressed within the early M45 gene provided no protection, although MCMV vectors expressing the high-avidity epitope SSIEFARL induced protective immunity irrespective of gene expression context. Immune protection was matched by Ag-induced, long-term expansion of effector memory CD8 T cells, regardless of epitope avidity. We explained this pattern by observing regularities in Ag competition, where responses to high-avidity epitopes outcompeted weaker ones expressed later in the replicative cycle of the virus. Conversely, robust and early expression of a low-avidity epitope compensated its weak intrinsic antigenicity, resulting in strong and sustained immunity and immune protection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos/imunologia , Proteínas Imediatamente Precoces/imunologia , Transativadores/imunologia , Vacinas Virais/imunologia , Animais , Infecções por Citomegalovirus/prevenção & controle , Epitopos de Linfócito T/genética , Regulação Viral da Expressão Gênica , Vetores Genéticos/genética , Humanos , Proteínas Imediatamente Precoces/genética , Imunização , Memória Imunológica , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Transativadores/genética , Vacinas de DNA , Replicação Viral
8.
J Exp Med ; 213(13): 3075-3086, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27899444

RESUMO

Tissue-resident memory CD8+ T cells (TRM) constitute a major component of the immune-surveillance system in nonlymphoid organs. Local, noncognate factors are both necessary and sufficient to support the programming of TRM cell fate in tissue-infiltrating T cells. Recent evidence suggests that TCR signals received in infected nonlymphoid tissues additionally contribute to TRM cell formation. Here, we asked how antigen-dependent pathways influence the generation of skin-resident memory T cells that arise from a polyclonal repertoire of cells induced by infection with an antigenically complex virus and recombinant vaccine vector. We found that CD8+ T cells of different specificities underwent antigen-dependent competition in the infected tissue, which shaped the composition of the local pool of TRM cells. This local cross-competition was active for T cells recognizing antigens that are coexpressed by infected cells. In contrast, TRM cell development remained largely undisturbed by the presence of potential competitors when antigens expressed in the same tissue were segregated through infection with antigenically distinct viral quasispecies. Functionally, local cross-competition might serve as a gatekeeping mechanism to regulate access to the resident memory niche and to fine-tune the local repertoire of antiviral TRM cells.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD8-Positivos/patologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais/genética , Vacínia/genética , Vacínia/patologia , Vaccinia virus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...