Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 10(6): 968-982, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25683719

RESUMO

Natural killer (NK) cells mediate innate immune responses against hazardous cells and are particularly important for the control of human cytomegalovirus (HCMV). NKG2D is a key NK activating receptor that recognizes a family of stress-induced ligands, including MICA, MICB, and ULBP1-6. Notably, most of these ligands are targeted by HCMV proteins and a miRNA to prevent the killing of infected cells by NK cells. A particular highly prevalent MICA allele, MICA∗008, is considered to be an HCMV-resistant "escape variant" that confers advantage to human NK cells in recognizing infected cells. However, here we show that HCMV uses its viral glycoprotein US9 to specifically target MICA∗008 and thus escapes NKG2D attack. The finding that HCMV evolved a protein dedicated to countering a single host allele illustrates the dynamic co-evolution of host and pathogen.

2.
J Clin Invest ; 124(12): 5305-16, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25384219

RESUMO

Human cytomegalovirus (HCMV) infection is the most common cause of congenital viral infections and a major source of morbidity and mortality after organ transplantation. NK cells are pivotal effector cells in the innate defense against CMV. Recently, hallmarks of adaptive responses, such as memory-like features, have been recognized in NK cells. HCMV infection elicits the expansion of an NK cell subset carrying an activating receptor heterodimer, comprising CD94 and NKG2C (CD94/NKG2C), a response that resembles the clonal expansion of adaptive immune cells. Here, we determined that expansion of this NKG2C(+) subset and general NK cell recovery rely on signals derived from CD14(+) monocytes. In a coculture system, a subset of CD14(+) cells with inflammatory monocyte features produced IL-12 in response to HCMV-infected fibroblasts, and neutralization of IL-12 in this model substantially reduced CD25 upregulation and NKG2C(+) subset expansion. Finally, blockade of CD94/NKG2C on NK cells or silencing of the cognate ligand HLA-E in infected fibroblasts greatly impaired expansion of NKG2C(+) NK cells. Together, our results reveal that IL-12, CD14(+) cells, and the CD94/NKG2C/HLA-E axis are critical for the expansion of NKG2C(+) NK cells in response to HCMV infection. Moreover, strategies targeting the NKG2C(+) NK cell subset have the potential to be exploited in NK cell-based intervention strategies against viral infections and cancer.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Monócitos/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Infecções por Citomegalovirus/patologia , Feminino , Humanos , Imunidade Celular , Subunidade alfa de Receptor de Interleucina-2/imunologia , Células Matadoras Naturais/patologia , Receptores de Lipopolissacarídeos/imunologia , Masculino , Monócitos/patologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Antígenos HLA-E
3.
PLoS Pathog ; 10(5): e1004131, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24830376

RESUMO

Human cytomegalovirus (HCMV) establishes lifelong infection with recurrent episodes of virus production and shedding despite the presence of adaptive immunological memory responses including HCMV immune immunoglobulin G (IgG). Very little is known how HCMV evades from humoral and cellular IgG-dependent immune responses, the latter being executed by cells expressing surface receptors for the Fc domain of IgG (FcγRs). Remarkably, HCMV expresses the RL11-encoded gp34 and UL119-118-encoded gp68 type I transmembrane glycoproteins which bind Fcγ with nanomolar affinity. Using a newly developed FcγR activation assay, we tested if the HCMV-encoded Fcγ binding proteins (HCMV FcγRs) interfere with individual host FcγRs. In absence of gp34 or/and gp68, HCMV elicited a much stronger activation of FcγRIIIA/CD16, FcγRIIA/CD32A and FcγRI/CD64 by polyclonal HCMV-immune IgG as compared to wildtype HCMV. gp34 and gp68 co-expression culminates in the late phase of HCMV replication coinciding with the emergence of surface HCMV antigens triggering FcγRIII/CD16 responses by polyclonal HCMV-immune IgG. The gp34- and gp68-dependent inhibition of HCMV immune IgG was fully reproduced when testing the activation of primary human NK cells. Their broad antagonistic function towards FcγRIIIA, FcγRIIA and FcγRI activation was also recapitulated in a gain-of-function approach based on humanized monoclonal antibodies (trastuzumab, rituximab) and isotypes of different IgG subclasses. Surface immune-precipitation showed that both HCMV-encoded Fcγ binding proteins have the capacity to bind trastuzumab antibody-HER2 antigen complexes demonstrating simultaneous linkage of immune IgG with antigen and the HCMV inhibitors on the plasma membrane. Our studies reveal a novel strategy by which viral FcγRs can compete for immune complexes against various Fc receptors on immune cells, dampening their activation and antiviral immunity.


Assuntos
Proteínas de Transporte/metabolismo , Citomegalovirus/imunologia , Glicoproteínas/metabolismo , Fragmentos Fc das Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de IgG/antagonistas & inibidores , Proteínas Virais/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Citomegalovirus/fisiologia , Células HEK293 , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ligação Proteica , Receptores de IgG/metabolismo , Replicação Viral
4.
J Immunol ; 192(1): 447-58, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24319264

RESUMO

We have previously characterized mouse CMV (MCMV)-encoded immune-evasive IFN signaling inhibition and identified the viral protein pM27 as inducer of proteasomal degradation of STAT2. Extending our analysis to STAT1 and STAT3, we found that MCMV infection neither destabilizes STAT1 protein nor prevents STAT1 tyrosine Y701 phosphorylation, nuclear translocation, or the capability to bind γ-activated sequence DNA-enhancer elements. Unexpectedly, the analysis of STAT3 revealed an induction of STAT3 Y705 phosphorylation by MCMV. In parallel, we found decreasing STAT3 protein amounts upon MCMV infection, although STAT3 expression normally is positive autoregulative. STAT3 phosphorylation depended on the duration of MCMV infection, the infectious dose, and MCMV gene expression but was independent of IFNAR1, IL-10, IL-6, and JAK2. Although STAT3 phosphorylation did not require MCMV immediate early 1, pM27, and late gene expression, it was restricted to MCMV-infected cells and not transmitted to bystander cells. Despite intact STAT1 Y701 phosphorylation, IFN-γ-induced target gene transcription (e.g., IRF1 and suppressor of cytokine signaling [SOCS] 1) was strongly impaired. Likewise, the induction of STAT3 target genes (e.g., SOCS3) by IL-6 was also abolished, indicating that MCMV antagonizes STAT1 and STAT3 despite the occurrence of tyrosine phosphorylation. Consistent with the lack of SOCS1 induction, STAT1 phosphorylation was prolonged upon IFN-γ treatment. We conclude that the inhibition of canonical STAT1 and STAT3 target gene expression abrogates their intrinsic negative feedback loops, leading to accumulation of phospho-tyrosine-STAT3 and prolonged STAT1 phosphorylation. These findings challenge the generalization of tyrosine-phosphorylated STATs necessarily being transcriptional active and document antagonistic effects of MCMV on STAT1/3-dependent target gene expression.


Assuntos
Janus Quinases/metabolismo , Muromegalovirus/fisiologia , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Receptor gp130 de Citocina/metabolismo , Regulação da Expressão Gênica , Regulação Viral da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Interferon gama/farmacologia , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Transporte Proteico , Receptor de Interferon alfa e beta/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo
5.
J Gen Virol ; 95(Pt 3): 659-670, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24337170

RESUMO

Despite a rigorous blockade of interferon-γ (IFN-γ) signalling in infected fibroblasts as a mechanism of immune evasion by human cytomegalovirus (HCMV), IFN-γ induced indoleamine-2,3-dioxygenase (IDO) has been proposed to represent the major antiviral restriction factor limiting HCMV replication in epithelial cells. Here we show that HCMV efficiently blocks transcription of IFN-γ-induced IDO mRNA both in infected fibroblasts and epithelial cells even in the presence of a preexisting IFN-induced antiviral state. This interference results in severe suppression of IDO bioactivity in HCMV-infected cells and restoration of vigorous HCMV replication. Depletion of IDO expression nonetheless substantially alleviated the antiviral impact of IFN-γ treatment in both cell types. These findings highlight the effectiveness of this IFN-γ induced effector gene in restricting HCMV productivity, but also the impact of viral counter-measures.


Assuntos
Infecções por Citomegalovirus/enzimologia , Citomegalovirus/fisiologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Replicação Viral , Linhagem Celular , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo
6.
Science ; 338(6110): 1088-93, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23180859

RESUMO

The human cytomegalovirus (HCMV) genome was sequenced 20 years ago. However, like those of other complex viruses, our understanding of its protein coding potential is far from complete. We used ribosome profiling and transcript analysis to experimentally define the HCMV translation products and follow their temporal expression. We identified hundreds of previously unidentified open reading frames and confirmed a fraction by means of mass spectrometry. We found that regulated use of alternative transcript start sites plays a broad role in enabling tight temporal control of HCMV protein expression and allowing multiple distinct polypeptides to be generated from a single genomic locus. Our results reveal an unanticipated complexity to the HCMV coding capacity and illustrate the role of regulated changes in transcript start sites in generating this complexity.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/genética , Genoma Viral , Fases de Leitura Aberta , Processamento Alternativo , Variação Genética , Humanos , Biossíntese de Proteínas/genética , Proteoma/genética , Análise de Sequência de DNA , Transcrição Gênica
7.
Future Microbiol ; 7(11): 1269-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23075446

RESUMO

Most human individuals are latently infected with human CMV, a prototypic ß-herpesvirus, frequently acquired during early childhood. In the absence of adequate immune control, the otherwise asymptomatic infection causes life-threatening disease. To enable efficient replication and to maintain lifelong latency in immunocompetent hosts, CMVs have evolved numerous molecules mediating immune evasive properties, targeting both innate and adaptive immune responses. Upon infection, cells secrete interferons (IFNs), which initiate an extremely fast signal transduction cascade upon binding to their cognate receptors, culminating in a pronounced change in the cellular gene expression profile. This response leads to the establishment of an intracellular antimicrobial state and to the recruitment, as well as stimulation, of the adaptive immune system. Unfortunately, CMVs impede the IFN system by interfering with its induction, signaling and downstream effector functions. This review aims to present our current understanding of such cytomegaloviral IFN-evasive properties, their pathogenic implications and potential for therapeutic exploitation.


Assuntos
Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/imunologia , Interferons/imunologia , Imunidade Adaptativa/imunologia , Animais , Humanos , Camundongos , Transdução de Sinais/imunologia , Latência Viral/imunologia , Replicação Viral/imunologia
8.
J Virol ; 85(24): 13260-70, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21976655

RESUMO

Human cytomegalovirus is a ubiquitous herpesvirus that establishes lifelong latent infection. Changes in immune homeostasis induce the reactivation of lytic infection, which is mostly inapparent in healthy individuals but often causes overt disease in immunocompromised hosts. Based on discrepant tumor necrosis factor receptor 1 surface disposition between human cytomegalovirus AD169 variants differing in the ULb' region, we identified the latency-associated gene product pUL138, which also is expressed during productive infection, as a selective potentiator of tumor necrosis factor receptor 1, one of the key receptors of innate immunity. Ectopically expressed pUL138 coprecipitated with tumor necrosis factor receptor 1, extended the protein half-life, and enhanced its signaling responses, thus leading to tumor necrosis factor receptor 1 hyperresponsiveness. Conversely, the targeted deletion of UL138 from the human cytomegaloviral genome strongly reduced tumor necrosis factor receptor 1 surface densities of infected cells. Remarkably, the comparison of UL138 deficiency to ULb' deficiency revealed the presence of further positive modulators of tumor necrosis factor alpha signal transduction encoded within the human cytomegalovirus ULb' region, identifying this region as a hub for multilayered tumor necrosis factor alpha signaling regulation.


Assuntos
Citomegalovirus/patogenicidade , Interações Hospedeiro-Patógeno , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/imunologia , Deleção de Genes , Humanos , Imunoprecipitação , Ligação Proteica , Mapeamento de Interação de Proteínas , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Proteínas Virais/imunologia
9.
PLoS Pathog ; 7(6): e1002069, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21698215

RESUMO

The mouse cytomegaloviral (MCMV) protein pM27 represents an indispensable factor for viral fitness in vivo selectively, antagonizing signal transducer and activator of transcription 2 (STAT2)-mediated interferon signal transduction. We wished to explore by which molecular mechanism pM27 accomplishes this effect. We demonstrate that pM27 is essential and sufficient to curtail the protein half-life of STAT2 molecules. Pharmacologic inhibition of the proteasome restored STAT2 amounts, leading to poly-ubiquitin-conjugated STAT2 forms. PM27 was found in complexes with an essential host ubiquitin ligase complex adaptor protein, DNA-damage DNA-binding protein (DDB) 1. Truncation mutants of pM27 showed a strict correlation between DDB1 interaction and their ability to degrade STAT2. SiRNA-mediated knock-down of DDB1 restored STAT2 in the presence of pM27 and strongly impaired viral replication in interferon conditioned cells, thus phenocopying the growth attenuation of M27-deficient virus. In a constructive process, pM27 recruits DDB1 to exploit ubiquitin ligase complexes catalyzing the obstruction of the STAT2-dependent antiviral state of cells to permit viral replication.


Assuntos
Citomegalovirus/fisiologia , Proteínas de Ligação a DNA/fisiologia , Interferon gama/farmacologia , Replicação Viral/genética , Animais , Células Cultivadas , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Células NIH 3T3 , Transfecção
10.
PLoS One ; 6(1): e14532, 2011 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-21264213

RESUMO

Herpesviral entry is a highly elaborated process requiring many proteins to act in precise conjunction. Neutralizing antibodies interfere with this process to abrogate viral infection. Based on promoter transactivation of a reporter gene we established a novel method to quantify herpesvirus entry and neutralization by antibodies. Following infection with mouse and human cytomegalovirus and Herpes simplex virus 1 we observed promoter transactivation resulting in substantial luciferase expression (>1000-fold). No induction was elicited by UV-inactivated viruses. The response was MOI-dependent and immunoblots confirmed a correlation between luciferase induction and pp72-IE1 expression. Monoclonal antibodies, immune sera and purified immunoglobulin preparations decreased virus-dependent luciferase induction dose-dependently, qualifying this approach as surrogate virus neutralization test. Besides the reduced hands-on time, this assay allows analysis of herpesvirus entry in semi-permissive and non-adherent cells, which were previously non-assessable but play significant roles in herpesvirus pathology.


Assuntos
Reações Antígeno-Anticorpo , Herpesvirus Humano 1/genética , Ativação Transcricional , Internalização do Vírus , Animais , Anticorpos Antivirais , Citomegalovirus , Genes Reporter , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Humanos , Camundongos , Muromegalovirus , Testes de Neutralização
11.
J Virol ; 83(8): 3684-95, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19211768

RESUMO

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


Assuntos
Fibroblastos/virologia , Fator Regulador 1 de Interferon/imunologia , Interferon gama/imunologia , Vaccinia virus/imunologia , Vaccinia virus/fisiologia , Replicação Viral , Animais , Células Cultivadas , Replicação do DNA , DNA Viral/biossíntese , Vírus da Ectromelia/fisiologia , Deleção de Genes , Humanos , Fator Regulador 1 de Interferon/genética , Janus Quinase 2/metabolismo , Camundongos , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1/metabolismo , Receptor de Interferon gama
12.
J Gen Virol ; 89(Pt 10): 2416-2426, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18796709

RESUMO

We have investigated the role of signal transducer and activator of transcription (STAT) 2 during human cytomegalovirus (HCMV) replication and found that protein levels of STAT2 are downregulated. STAT2 downregulation was observed in HCMV clinical isolates and laboratory strains with the exception of strain Towne. The HCMV-induced loss of STAT2 protein occurred despite an increased accumulation of STAT2 mRNA; it required HCMV early gene expression. The decrease in STAT2 was sensitive to proteasome inhibition, suggesting degradation of STAT2 via the ubiquitin proteasome pathway. Notably, pUL27, the HCMV homologue of the mouse CMV pM27 protein, which mediates the selective proteolysis of STAT2, did not induce STAT2 downregulation. Moreover, preceding STAT2 degradation, alpha/beta interferon (IFN)-receptor-mediated tyrosine phosphorylation of STAT2 was inhibited in HCMV-infected cells. This effect was paralleled by impaired tyrosine activation of STAT1 and STAT3. Accordingly, IFNs affected the replication efficiency of STAT2 degrading and non-degrading HCMV strains to a similar degree. In summary, HCMV abrogates IFN receptor signalling at multiple checkpoints by independent mechanisms including UL27-independent degradation of STAT2 and a preceding blockade of STAT2 phosphorylation.


Assuntos
Citomegalovirus/patogenicidade , Fator de Transcrição STAT2/metabolismo , Tirosina/metabolismo , Animais , Linhagem Celular , Citomegalovirus/classificação , Regulação para Baixo , Fibroblastos/virologia , Humanos , Interferon Tipo I/metabolismo , Interferon Tipo I/farmacologia , Camundongos , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Transcrição STAT2/química , Especificidade da Espécie , Proteínas Virais/metabolismo
13.
J Gen Virol ; 89(Pt 5): 1131-1141, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18420790

RESUMO

We have investigated beta interferon (IFN-beta) and IFN-alpha4 gene expression and activation of related transcription factors in mouse cytomegalovirus (MCMV)-infected fibroblasts. mRNA analysis demonstrated an initial phase of IFN gene induction upon MCMV infection, which was followed by a sustained MCMV-mediated simultaneous downregulation of IFN-beta and IFN-alpha4 gene expression. The induction of IFN transcription resulted from the activation of the components of the IFN-beta enhanceosome, i.e. IFN regulatory factor (IRF) 3, nuclear factor (NF)-kappaB, activating transcription factor (ATF)-2 and c-Jun. Activation of the transcription factors occurred rapidly and in a sequential order upon infection, but only lasted a while. As a consequence, IFN-alpha/beta gene expression became undetectable 6 h post-infection and throughout the MCMV replication cycle. This effect is based on an active interference since restimulation of IFN gene induction by further external stimuli (e.g. Sendai virus infection) was completely abolished. This inhibition required MCMV gene expression and was not observed in cells infected with UV-inactivated MCMV virions. The efficiency of inhibition is achieved by a concerted blockade of IkappaBalpha degradation and a lack of nuclear accumulation of IRF3 and ATF-2/c-Jun. Using an MCMV mutant lacking pM27, a signal transducer and activator of transcription (STAT) 2-specific inhibitor of Jak/STAT signalling, we found that the initial phase of IFN induction and the subsequent inhibition does not depend on the positive-IFN feedback loop. Our findings indicate that the MCMV-mediated downregulation of IFN transcription in fibroblasts relies on a large arsenal of inhibitory mechanisms targeting each pathway that contributes to the multiprotein enhanceosome complex.


Assuntos
Regulação da Expressão Gênica , Interferon beta/biossíntese , Muromegalovirus/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Núcleo Celular/química , Células Cultivadas , Fibroblastos/virologia , Perfilação da Expressão Gênica , Proteínas I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/biossíntese , Interferon-alfa , Interferon beta/antagonistas & inibidores , Camundongos , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Recombinantes , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...