Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO J ; 31(1): 201-13, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22036947

RESUMO

Early during Gram-negative sepsis, excessive release of pro-inflammatory cytokines can cause septic shock that is often followed by a state of immune paralysis characterized by the failure to mount adaptive immunity towards secondary microbial infections. Especially, the early mechanisms responsible for such immune hypo-responsiveness are unclear. Here, we show that TLR4 is the key immune sensing receptor to initiate paralysis of T-cell immunity after bacterial sepsis. Downstream of TLR4, signalling through TRIF but not MyD88 impaired the development of specific T-cell immunity against secondary infections. We identified type I interferon (IFN) released from splenic macrophages as the critical factor causing T-cell immune paralysis. Early during sepsis, type I IFN acted selectively on dendritic cells (DCs) by impairing antigen presentation and secretion of pro-inflammatory cytokines. Our results reveal a novel immune regulatory role for type I IFN in the initiation of septic immune paralysis, which is distinct from its well-known immune stimulatory effects. Moreover, we identify potential molecular targets for therapeutic intervention to overcome impairment of T-cell immunity after sepsis.


Assuntos
Imunidade Adaptativa , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Sepse/imunologia , Baço/metabolismo , Animais , Células Dendríticas/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Sepse/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
2.
PLoS Pathog ; 9(8): e1003536, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935506

RESUMO

Infections with high-risk human papillomaviruses (HPVs) are causally involved in the development of anogenital cancer. HPVs apparently evade the innate immune response of their host cells by dysregulating immunomodulatory factors such as cytokines and chemokines, thereby creating a microenvironment that favors malignancy. One central key player in the immune surveillance interactome is interleukin-1 beta (IL-1ß) which not only mediates inflammation, but also links innate and adaptive immunity. Because of its pleiotropic physiological effects, IL-1ß production is tightly controlled on transcriptional, post-translational and secretory levels. Here, we describe a novel mechanism how the high-risk HPV16 E6 oncoprotein abrogates IL-1ß processing and secretion in a NALP3 inflammasome-independent manner. We analyzed IL-1ß regulation in immortalized keratinocytes that harbor the HPV16 E6 and/or E7 oncogenes as well as HPV-positive cervical tumor cells. While in primary and in E7-immortalized human keratinocytes the secretion of IL-1ß was highly inducible upon inflammasome activation, E6-positive cells did not respond. Western blot analyses revealed a strong reduction of basal intracellular levels of pro-IL-1ß that was independent of dysregulation of the NALP3 inflammasome, autophagy or lysosomal activity. Instead, we demonstrate that pro-IL-1ß is degraded in a proteasome-dependent manner in E6-positive cells which is mediated via the ubiquitin ligase E6-AP and p53. Conversely, in E6- and E6/E7-immortalized cells pro-IL-1ß levels were restored by siRNA knock-down of E6-AP and simultaneous recovery of functional p53. In the context of HPV-induced carcinogenesis, these data suggest a novel post-translational mechanism of pro-IL-1ß regulation which ultimately inhibits the secretion of IL-1ß in virus-infected keratinocytes. The clinical relevance of our results was further confirmed in HPV-positive tissue samples, where a gradual decrease of IL-1ß towards cervical cancer could be discerned. Hence, attenuation of IL-1ß by the HPV16 E6 oncoprotein in immortalized cells is apparently a crucial step in viral immune evasion and initiation of malignancy.


Assuntos
Transformação Celular Viral/imunologia , Papillomavirus Humano 16/imunologia , Imunidade Inata , Interleucina-1beta/imunologia , Proteínas Oncogênicas Virais/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Proteólise , Proteínas Repressoras/imunologia , Evasão Tumoral/imunologia , Proteína Supressora de Tumor p53/imunologia , Ubiquitina-Proteína Ligases/imunologia , Linhagem Celular Transformada , Transformação Celular Viral/genética , Feminino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Processamento de Proteína Pós-Traducional/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Evasão Tumoral/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
3.
Gastroenterology ; 138(1): 336-46, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19737567

RESUMO

BACKGROUND & AIMS: Dendritic cell activation through ligation of pattern recognition receptors leading to full functional maturation causes induction of CD8(+) T-cell immunity through increased delivery of costimulatory signals instead of tolerance. Here we investigate whether organ-resident antigen-presenting cells, such as liver sinusoidal endothelial cells (LSECs), also switch from tolerogenic to immunogenic CD8(+) T-cell activation upon such stimulation. METHODS: Murine LSECs were isolated by immunomagnetic separation and analyzed for functional maturation upon triggering pattern recognition receptors or viral infection employing gene expression analysis and T cell coculture assays. In vivo relevance of the findings was confirmed with bone-marrow chimeric animals. RESULTS: LSECs expressed numerous pattern recognition receptors that allowed for sentinel function, but ligand-induced activation of these receptors was not sufficient to overcome tolerance induction of CD8(+) T cells. Importantly, viral infection with murine cytomegalovirus caused functional maturation of antigen-presenting LSECs and was sufficient to promote antigen-specific differentiation into effector CD8(+) T cells in the absence of dendritic cells and independent of CD80/86. CONCLUSIONS: These results shed new light on the generation of organ-specific immunity and may contribute to overcoming tolerance in relevant situations, such as cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Infecções por Herpesviridae/imunologia , Tolerância Imunológica/imunologia , Fígado/virologia , Muromegalovirus , Transferência Adotiva , Animais , Medula Óssea , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Quimera , Células Endoteliais/citologia , Células Endoteliais/virologia , Ligantes , Fígado/citologia , Fígado/imunologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo
4.
J Virol ; 84(1): 516-31, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864388

RESUMO

Parvovirus minute virus of mice (MVMp) is endowed with oncotropic properties so far ascribed only to the dependency of the virus life cycle on cellular factors expressed during S phase and/or modulated by malignant transformation. For other viruses oncotropism relies on their inability to circumvent type I interferon (IFN)-induced innate antiviral mechanisms, the first line of defense triggered by normal cells against viral infections. These agents propagate, therefore, preferentially in transformed/tumor cells, which often lack functional antiviral mechanisms. The present study aimed at investigating whether antiviral processes also contribute to MVMp oncotropism. Our results demonstrate that in contrast to MVMp-permissive transformed mouse A9 fibroblasts, freshly isolated normal counterparts (mouse embryonic fibroblasts [MEFs]) mount, through production and release of type I IFNs upon their infection, an antiviral response against MVMp lytic multiplication. Pretreatment of MEFs with a type I IFN-beta-neutralizing antibody, prior to MVMp infection, inhibits the virus-triggered antiviral response and improves the fulfillment of the MVMp life cycle. Our results also show that part of the A9 permissiveness to MVMp relies on the inability to produce type I IFNs upon parvovirus infection, a feature related either to an A9 intrinsic deficiency of this process or to an MVMp-triggered inhibitory mechanism, since stimulation of these cells by exogenous IFN-beta strongly inhibits the parvovirus life cycle. Taken together, our results demonstrate for the first time that parvovirus infection triggers an innate antiviral response in normal cells and suggest that the MVMp oncotropism depends at least in part on the failure of infected transformed cells to mount such a response.


Assuntos
Imunidade Inata , Vírus Miúdo do Camundongo/imunologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Fibroblastos/imunologia , Fibroblastos/virologia , Humanos , Interferon Tipo I/biossíntese , Interferon beta/farmacologia , Camundongos , Replicação Viral/efeitos dos fármacos
5.
Psychiatry Res ; 185(1-2): 243-7, 2011 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-20580843

RESUMO

Interferon (IFN) alpha proteins are proinflammatory cytokines having immunomodulating and antiviral properties. States during which cytokine systems are activated (e.g., during viral infection or during treatment of chronic hepatitis C and various malignancies with IFN alpha, etc.) can be associated with depression-like syndromes or even full-blown depressive episodes. Therefore, the role of IFN alpha and other cytokines in the pathogenesis of depressive disorder ("cytokine hypothesis of depression") has been assessed for many years with contradictory results. We have investigated whether intraperitoneal administration of high doses (up to 600 µg/kg body weight) of pegylated, recombinant human IFN alpha 2a in mice induces changes known to be associated with depression using three different readouts: behavior in a model of despair (Porsolt swim test), presence of anhedonia (sucrose preference test), and sensitivity of the hypothalamic-pituitary-adrenal system (dexamethasone suppression test). We also assessed potential IFN-induced changes in gene expression in the liver. In none of the performed experiments, depression-associated effects could be found despite very high serum levels of IFN-induced antiviral activity compared to levels measured in hepatitis C virus (HCV) patients treated routinely with pegylated recombinant human IFN alpha 2a. The lack of such expected effects is probably due to the fact that pegylated human recombinant IFN alpha 2a does not activate the murine class I IFN receptor. Our results do not support the hypothesis that administration of recombinant pegylated human IFN alpha to mice produces a robust model of depression.


Assuntos
Antivirais/efeitos adversos , Depressão/induzido quimicamente , Interferon-alfa/efeitos adversos , Polietilenoglicóis/efeitos adversos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Interferon alfa-2 , Masculino , Camundongos , Proteínas Recombinantes , Fatores de Tempo
6.
Blood ; 112(2): 264-76, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18469200

RESUMO

Inflammatory responses represent a hallmark of numerous pathologies including sepsis, bacterial infection, insulin resistance, and malign obesity. Here we describe an unexpected coactivator function for the nuclear receptor interacting protein 140 (RIP140) for nuclear factor kappaB (NFkappaB), a master transcriptional regulator of inflammation in multiple tissues. Previous work has shown that RIP140 suppresses the expression of metabolic gene networks, but we have found that genetic as well as acute deficiency of RIP140 leads to the inhibition of the proinflammatory program in macrophages. The ability of RIP140 to function as a coactivator for cytokine gene promoter activity relies on direct protein-protein interactions with the NFkappaB subunit RelA and histone acetylase cAMP-responsive element binding protein (CREB)-binding protein (CBP). RIP140-dependent control of proinflammatory gene expression via RelA/CBP may, therefore, represent a molecular rational for the cellular integration of metabolic and inflammatory pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína de Ligação a CREB/metabolismo , Citocinas/genética , Regulação da Expressão Gênica/imunologia , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Humanos , Inflamação/genética , Camundongos , Camundongos Knockout , NF-kappa B/imunologia , Proteína 1 de Interação com Receptor Nuclear , Ligação Proteica/imunologia , Fator de Transcrição RelA/imunologia
7.
J Immunol ; 181(6): 3823-33, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768836

RESUMO

Type I IFNs represent a major antimicrobial defense mechanism due to their property of enhancing immune responses by priming both innate and adaptive immune cells. Plasmacytoid dendritic cells (pDC) are the major source of type I IFN in the human body and represent innate immune cells involved in first-line defense against invading pathogens. Although pDC activation has been extensively studied upon stimulation with synthetic TLR ligands, viruses, and intracellular bacteria, there is only scarce information on extracellular bacteria. In this study we show that the triggering of human pDC-derived IFN-alpha secretion by Staphylococcus aureus is independent of TLR2 and specific for coagulase-positive staphylococci. Specificity of the pDC response to S. aureus is independent of the bacterial virulence factors protein A and alpha-toxin but is mediated by Ag-specific IgG and CD32. S. aureus-induced pDC activation can be blocked by inhibitory DNA oligonucleotides and chloroquine, suggesting that engagement of TLR7/9 by bacterial nucleic acids after CD32-mediated uptake of these compounds may play a central role in this process. Altogether, we propose that in marked contrast to nonselective TLR2-dependent activation of most innate immune cells, pDC activation by S. aureus represents an Ag-specific memory response since it requires the presence of class-switched immunoglobulins.


Assuntos
Anticorpos Antibacterianos/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Imunoglobulina G/fisiologia , Memória Imunológica , Staphylococcus aureus/imunologia , Anticorpos Antibacterianos/metabolismo , Linhagem Celular , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Interferon-alfa/metabolismo , Lipoproteínas/fisiologia , Receptores de IgG/metabolismo , Receptores de IgG/fisiologia , Receptor 2 Toll-Like/fisiologia
8.
Int J Oncol ; 34(4): 971-82, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19287954

RESUMO

Newcastle Disease Virus (NDV) is an avian paramyxovirus with anti-neoplastic and immune-stimulatory properties which has raised considerable interest for cancer therapy. To better understand the molecular nature of the tumor selective replication of NDV, we investigated the cellular responses of murine normal and tumor cells after infection by NDV. To this end, we compared the basal expression of different antiviral proteins as well as the expression induced by the addition of NDV to the cells in vitro and in vivo. Primary macrophages were found to be resistant to NDV infection and exhibited a high basal and induced expression of various antiviral genes. In contrast, macrophage-derived RAW tumor cells were highly susceptible to NDV infection and displayed a low expression of several antiviral genes. Macrophage-derived J774 tumor cells were intermediate with regard to NDV replication and antiviral gene expression. The responsiveness to exogenously added IFN-alpha was found highest in normal macrophages, lowest in the RAW cells, and intermediate in the J774 cells. We also analysed dendritic cells as well as additional normal and tumor cell types. A strong inverse correlation was obeserved between the susceptibility to infection and the basal expression of the antiviral genes RIG-I, IRF3, IRF7 and IFN-beta. A strong expression of these genes can explain the resistance of normal cells to NDV infection and a weak antiviral gene expression the broad susceptibility of tumor cells.


Assuntos
Regulação da Expressão Gênica , Fator Regulador 3 de Interferon/biossíntese , Fator Regulador 7 de Interferon/biossíntese , Interferon beta/biossíntese , Vírus da Doença de Newcastle/metabolismo , Animais , Linhagem Celular Tumoral , Interferon-alfa/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Modelos Genéticos , RNA/metabolismo
9.
PLoS One ; 14(4): e0215062, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30990863

RESUMO

Interferon α (IFNα) counteracts viral infections by activating various IFNα-stimulated genes (ISGs). These genes encode proteins that block viral transport into the host cell and inhibit viral replication, gene transcription and translation. Due to the existence of 14 different, highly homologous isoforms of mouse IFNα, an IFNα knockout mouse has not yet been established by genetic knockout strategies. An scFv intrabody for holding back IFNα isoforms in the endoplasmic reticulum (ER) and thus counteracting IFNα secretion is reported. The intrabody was constructed from the variable domains of the anti-mouse IFNα rat monoclonal antibody 4EA1 recognizing the 5 isoforms IFNα1, IFNα2, IFNα4, IFNα5, IFNα6. A soluble form of the intrabody had a KD of 39 nM to IFNα4. It could be demonstrated that the anti-IFNα intrabody inhibits clearly recombinant IFNα4 secretion by HEK293T cells. In addition, the secretion of IFNα4 was effectively inhibited in stably transfected intrabody expressing RAW 264.7 macrophages and dendritic D1 cells. Colocalization of the intrabody with IFNα4 and the ER marker calnexin in HEK293T cells indicated complex formation of intrabody and IFNα4 inside the ER. Intracellular binding of intrabody and antigen was confirmed by co-immunoprecipitation. Complexes of endogenous IFNα and intrabody could be visualized in the ER of Poly (I:C) stimulated RAW 264.7 macrophages and D1 dendritic cells. Infection of macrophages and dendritic cells with the vesicular stomatitis virus VSV-AV2 is attenuated by IFNα and IFNß. The intrabody increased virus proliferation in RAW 264.7 macrophages and D1 dendritic cells under IFNß-neutralizing conditions. To analyze if all IFNα isoforms are recognized by the intrabody was not in the focus of this study. Provided that binding of the intrabody to all isoforms was confirmed, the establishment of transgenic intrabody mice would be promising for studying the function of IFNα during viral infection and autoimmune diseases.


Assuntos
Células Dendríticas/imunologia , Retículo Endoplasmático/imunologia , Interferon-alfa/antagonistas & inibidores , Macrófagos/imunologia , Anticorpos de Cadeia Única/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Interferon-alfa/efeitos dos fármacos , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Células RAW 264.7
10.
Vet Immunol Immunopathol ; 104(1-2): 105-15, 2005 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-15661336

RESUMO

Interferon-gamma (IFN-gamma) is a key cytokine in cell-mediated immunity. To measure IFN-gamma production of equine lymphocytes (eqIFN-gamma), we developed a quantitative ELISA. Monoclonal antibodies (mAb) were produced against bacterially derived eqIFN-gamma. The mAbs recognised recombinant and lymphocyte-derived eqIFN-gamma in ELISA, Western blotting, as well as flow cytometric and microscopic analysis. In contrast to bacterially derived material, mammalian and insect cell-derived eqIFN-gamma was biologically active but could be neutralised by one of the monoclonal antibodies. Unexpectedly, glycosylation seemed to be required for antiviral activity of eqIFN-gamma.


Assuntos
Ensaio de Imunoadsorção Enzimática/veterinária , Cavalos/imunologia , Interferon gama/imunologia , Linfócitos/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Western Blotting/veterinária , Ensaio de Imunoadsorção Enzimática/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Citometria de Fluxo/veterinária , Interferon gama/biossíntese , Interferon gama/genética , Interferon gama/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos
11.
PLoS One ; 8(1): e55086, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383065

RESUMO

The oncotropism of Minute Virus of Mice (MVMp) is partially related to the stimulation of an antiviral response mediated by type-I interferons (IFNs) in normal but not in transformed mouse cells. The present work was undertaken to assess whether the oncotropism displayed against human cells by MVMp and its rat homolog H-1PV also depends on antiviral mechanisms and to identify the pattern recognition receptor (PRR) involved. Despite their low proliferation rate which represents a drawback for parvovirus multiplication, we used human peripheral blood mononuclear cells (hPBMCs) as normal model specifically because all known PRRs are functional in this mixed cell population and moreover because some of its subsets are among the main IFN producers upon infections in mammals. Human transformed models consisted in lines and tumor cells more or less permissive to both parvoviruses. Our results show that irrespective of their permissiveness, transformed cells do not produce IFNs nor develop an antiviral response upon parvovirus infection. However, MVMp- or H-1PV-infected hPBMCs trigger such defense mechanisms despite an absence of parvovirus replication and protein expression, pointing to the viral genome as the activating element. Substantial reduction of an inhibitory oligodeoxynucleotide (iODN) of the latter IFN production identified TLR-9 as a potential PRR for parvoviruses in hPBMCs. However, neither the iODN treatment nor an antibody-induced neutralization of the IFN-triggered effects restored parvovirus multiplication in these cells as expected by their weak proliferation in culture. Finally, given that a TLR-9 activation could also not be observed in parvovirus-infected human lines reported to be endowed with a functional TLR-9 pathway (Namalwa, Raji, and HEK293-TLR9(+/+)), our data suggest that transformed human cells do not sense MVMp or H-1PV either because of an absence of PRR expression or an intrinsic, or virus-driven defect in the endosomal sensing of the parvovirus genomes by TLR-9.


Assuntos
Imunidade Inata , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Vírus Miúdo do Camundongo/fisiologia , Receptor Toll-Like 9/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Viral/imunologia , Genoma Viral/genética , Células HEK293 , Humanos , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/metabolismo , Camundongos , Vírus Miúdo do Camundongo/genética , Ratos , Transdução de Sinais/imunologia
12.
Cancer Res ; 71(20): 6410-8, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21900395

RESUMO

IFN-γ promotes tumoral immune surveillance, but its involvement in controlling metastases is less clear. Using a mouse model of pulmonary metastases, we show that local IFN-γ treatment inhibits formation of metastases through its regulation of IRF-1 in tumor cells. IRF-1 is an IFN-γ-induced transcription factor pivotal in the regulation of infection and inflammation. IRF-1 blockade abolished the inhibitory effect of IFN-γ on tumor metastases, whereas ectopic expression of IRF-1 phenocopied the inhibitory effects of IFN-γ. IRF-1 did not affect the survival of tumor cells in the circulation or their infiltration into lungs, but it was essential to support the pulmonary attraction and activation of natural killer (NK) cells. Depleting NK cells from mice abolished the protective effect of IFN-γ or IRF-1 on metastases. In addition, cytotoxicity assays revealed that tumor cells expressing IRF-1 were targeted more effectively by NK cells than IRF-1 nonexpressing tumor cells. Moreover, NK cells isolated from lungs inoculated with IRF-1-expressing tumor cells exhibit a greater cytotoxic activity. Mechanistic investigations revealed that IRF-1-induced NK cell cytotoxicity was independent of perforin and granzyme B but dependent on the NK cell activating receptor DNAM-1. Taken together, our findings establish IRF-1 as an essential mediator of the cross-talk between tumor cells and NK cells that mediate immune surveillance in the metastatic niche.


Assuntos
Carcinoma/imunologia , Neoplasias do Colo/imunologia , Fator Regulador 1 de Interferon/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/imunologia , Carcinoma/tratamento farmacológico , Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Granzimas/farmacologia , Fator Regulador 1 de Interferon/antagonistas & inibidores , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Perforina/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Receptores Virais/imunologia , Regulação para Cima
13.
Cancer Res ; 69(22): 8718-25, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19887612

RESUMO

We have investigated interferon-kappa (IFN-kappa) regulation in the context of human papillomavirus (HPV)-induced carcinogenesis using primary human foreskin keratinocytes (HFK), immortalized HFKs encoding individual oncoproteins of HPV16 (E6, E7, and E6/E7), and cervical carcinoma cells. Here, IFN-kappa was suppressed in the presence of E6, whereas its expression was not affected in HFKs or E7-immortalized HFKs. Transcription could be reactivated after DNA demethylation but was decreased again upon drug removal. Partial reactivation could also be accomplished when E6 was knocked down, suggesting a contribution of E6 in IFN-kappa de novo methylation. We identified a single CpG island near the transcriptional start site as being involved in selective IFN-kappa expression. To prove the functional relevance of IFN-kappa in building up an antiviral response, IFN-kappa was ectopically expressed in cervical carcinoma cells where protection against vesicular stomatitis virus-mediated cytolysis could be achieved. Reconstitution of IFN-kappa was accompanied by an increase of p53, MxA, and IFN-regulatory factors, which was reversed by knocking down either IFN-kappa or p53 by small interfering RNA. This suggests the existence of a positive feedback loop between IFN-kappa, p53, and components of IFN signaling pathway to maintain an antiviral state. Our in vitro findings were further corroborated in biopsy samples of cervical cancer patients, in which IFN-kappa was also downregulated when compared with normal donor tissue. This is the first report showing an epigenetic silencing of type I IFN after HPV16 oncogene expression and revealing a novel strategy on how high-risk HPVs can abolish the innate immune response in their genuine host cells.


Assuntos
Regulação da Expressão Gênica , Inativação Gênica/fisiologia , Interferon Tipo I/genética , Infecções por Papillomavirus/genética , Transdução de Sinais/fisiologia , Western Blotting , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Regulação para Baixo , Feminino , Expressão Gênica , Papillomavirus Humano 16/imunologia , Humanos , Interferon Tipo I/biossíntese , Queratinócitos/virologia , Proteínas Oncogênicas Virais/biossíntese , Proteínas Oncogênicas Virais/genética , Infecções por Papillomavirus/complicações , RNA Interferente Pequeno , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia
14.
Int J Cancer ; 120(10): 2119-26, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17266037

RESUMO

Although it is generally assumed that cancer arises from a singular cell, a tumor must be considered as a dynamic and emergent biological structure, whose organizing principle is determined by genetic and epigenetic modifications, occurring variably in response to microenvironmental selection conditions. As previously shown, HPV-positive cervical carcinoma cells have lost their ability to induce IFN-beta upon TNF-alpha treatment. However, regarding cancer as a non-linear system, which may, even in the absence of an apparent selection pressure, fluctuate between different "metastable" phenotypes, we demonstrate that TNF-alpha mediated IFN-beta induction is not irreversibly disturbed in all cells. Using the IFN-beta sensitive Encephalomyocarditis virus (EMCV) as a tool to monitor antiviral activity in long-term established malignant HeLa cells, rare IFN-beta expressing clones were rescued from a population of non-responsive and EMCV-sensitive cells. Antiviral activity was mediated by the re-expression of IRF-1 and p48 (IRF-9), both key regulatory molecules normally found to be suppressed in cervical carcinoma cells. Upon inoculating of selected clones into immunocompromised animals, a reduced or even an absence of tumorigenicity of initially highly malignant cells could be discerned. These data indicate that both the absence of interferon signaling and the ability to form tumors were reversed in a minority of cells. We provide a paradigm for the existence of innate genetic redundancy mechanisms, where a particular phenotype persists and can be isolated without application of drugs generally changing the epigenetic context.


Assuntos
Interferon beta/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/imunologia , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Butiratos/farmacologia , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Vírus da Encefalomiocardite/crescimento & desenvolvimento , Vírus da Encefalomiocardite/imunologia , Feminino , Células HeLa , Inibidores de Histona Desacetilases , Papillomavirus Humano 18/imunologia , Humanos , Hospedeiro Imunocomprometido , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/biossíntese , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon beta/genética , Interferon beta/imunologia , Camundongos , Camundongos Nus , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Neoplasias do Colo do Útero/virologia
15.
Immunol Cell Biol ; 85(5): 383-90, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17404592

RESUMO

Plasmacytoid dendritic cells (PDC) in human blood are the main source of virus-induced interferon (IFN)-alpha. They exhibit a lineage-negative phenotype but all express BDCA-4, which is homologous to the neuronal receptor neuropilin-1. Specific staining with anti-BDCA-4 antibody is used for positive isolation of PDC from blood by magnetic cells sorting. Here, it is demonstrated that these positively selected PDC showed reduced or completely abolished IFN-alpha release compared to unstained PDC, which were negatively selected by magnetic depletion of lineage-positive blood mononuclear cells. In addition, treatment of these unstained PDC with anti-BDCA-4 mAb also resulted in at least two-fold lower or reduced virus-induced IFN-alpha production. It is shown that the antibody not only affects cell survival or block virus attachment but also reduces IFN-alpha release induced by non-viral CpG oligodeoxynucleotides. In conclusion, data suggest an immunoregulatory role for BDCA-4 on PDC as demonstrated for IFN-alpha response to virus.


Assuntos
Anticorpos Monoclonais/farmacologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Interferon gama/biossíntese , Neuropilina-1/imunologia , Vírus do Sarcoma de Rous/efeitos dos fármacos , Vírus do Sarcoma de Rous/fisiologia , Separação Celular , Células Dendríticas/efeitos dos fármacos , Citometria de Fluxo , Humanos , Interferon gama/genética , Fenótipo , Transcrição Gênica/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
16.
Eur J Immunol ; 37(6): 1702-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17506029

RESUMO

Systemic lupus erythematosus (SLE), an autoimmune disease characterized by chronic nephritis, arthritis and dermatitis, and the presence of antinuclear autoantibodies, is associated with complement factor deficiencies in the classical activation pathway. In addition, IFN-alpha seems to be a key cytokine in SLE as an activated IFN-alpha system is regularly observed in patients with SLE. Here, we demonstrate that in lupus-susceptible, complement C4-deficient mice the lack of complement results in elevated intravascular levels of apoptotic DNA. The apoptotic DNA is targeted to the splenic marginal zone where it accumulates and induces IFN-alpha. As such, we present here a unifying hypothesis for the induction of SLE that incorporates the role of complement deficiency and elevated levels of IFN-alpha.


Assuntos
Apoptose/imunologia , Complemento C4/genética , DNA/metabolismo , Interferon-alfa/genética , Lúpus Eritematoso Sistêmico/imunologia , Animais , Anticorpos Antinucleares/imunologia , Anticorpos Antinucleares/metabolismo , Anticorpos Antinucleares/farmacologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Complexo Antígeno-Anticorpo/análise , Complexo Antígeno-Anticorpo/imunologia , Apoptose/genética , Antígeno CD11b/análise , Complemento C4/deficiência , DNA/imunologia , DNA/farmacologia , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Imunoglobulina M/farmacologia , Marcação In Situ das Extremidades Cortadas , Interferon-alfa/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Antígenos Comuns de Leucócito/análise , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Baço/metabolismo
17.
Biochem Biophys Res Commun ; 335(2): 584-9, 2005 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-16091286

RESUMO

Type I interferons (IFNs) have been shown to be involved in many immune defence and inflammatory responses. We here show that IFN-beta plays an absolute essential role in the efficient induction of all type I IFNs after infection of primary embryonic as well as primary adult fibroblasts with Sendai virus. In contrast, after immortalization of such fibroblasts with SV40 large T antigen, IFN-alpha4 can be induced independently of IFN-beta. However, efficient secretion of type I IFNs even in immortalized fibroblasts is only found when the complete signalling loop is induced by IFN-beta.


Assuntos
Transformação Celular Viral , Fibroblastos/metabolismo , Interferon Tipo I/biossíntese , Animais , Bioensaio , Células Cultivadas , Fibroblastos/virologia , Inflamação , Interferons/metabolismo , Cinética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus Sendai/metabolismo , Transdução de Sinais , Fatores de Tempo
18.
J Virol ; 76(1): 280-91, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11739693

RESUMO

In the present study we show that malignant human papillomavirus (HPV)-positive cells lost their ability to synthesize endogenous beta interferon (IFN-beta) upon tumor necrosis factor alpha (TNF-alpha) treatment. IFN-beta transcription, however, was reinducible in nonmalignant HPV-positive cells, which was confirmed in functional protection assays against encephalomyocarditis virus or vesicular stomatitis virus infections. Addition of neutralizing antibodies against IFN-beta blocked the antiviral effect, excluding the possibility that other IFN types were involved. Conversely, both malignant and immortalized cells could be protected against viral cytolysis when either IFN-beta, IFN-alpha, or IFN-gamma was added exogenously. This indicates that only the cross talk between TNF-alpha and the IFN-beta pathways, and not IFN-alpha/beta and IFN-gamma signaling in general, is perturbed in cervical carcinoma cells. Notably, full virus protection was restricted exclusively to nonmalignant cells, indicating that the antiviral effect correlates with the growth-inhibitory and virus-suppressive properties of TNF-alpha. The IFN-regulatory factors IRF-1 and p48 (ISGF3gamma) emerged as key regulatory molecules in the differential IFN-beta response, since their transcription was either absent or only inefficiently enhanced in tumorigenic cells upon treatment with TNF-alpha. Inducibility of both genes, however, became reestablished in cervical carcinoma cells, which were complemented to nontumorigenicity after somatic cell hybridization. Complementation was paralleled by the entire reconstitution of cytokine-mediated IFN-beta expression and the ability of TNF-alpha to exert an antiviral state. In contrast, under conditions where tumor suppression was not accomplished upon somatic cell hybridization, neither expression of IRF-1, p48, and IFN-beta nor antiviral activity could be restored.


Assuntos
Interferon beta/metabolismo , Infecções por Papillomavirus/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Neoplasias do Colo do Útero/metabolismo , Antineoplásicos/farmacologia , Antivirais/farmacologia , Feminino , Células HeLa , Humanos , Células Híbridas , Interferon beta/biossíntese , Interferon beta/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/virologia
19.
J Virol ; 78(12): 6143-50, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15163707

RESUMO

Hantaviruses represent important human pathogens and can induce hemorrhagic fever with renal syndrome (HFRS), which is characterized by endothelial dysfunction. Both pathogenic and nonpathogenic hantaviruses replicate without causing any apparent cytopathic effect, suggesting that immunopathological mechanisms play an important role in pathogenesis. We compared the antiviral responses triggered by Hantaan virus (HTNV), a pathogenic hantavirus associated with HFRS, and Tula virus (TULV), a rather nonpathogenic hantavirus, in human umbilical vein endothelial cells (HUVECs). Both HTNV- and TULV-infected cells showed increased levels of molecules involved in antigen presentation. However, TULV-infected HUVECs upregulated HLA class I molecules more rapidly. Interestingly, HTNV clearly induced the production of beta interferon (IFN-beta), whereas expression of this cytokine was barely detectable in the supernatant or in extracts from TULV-infected HUVECs. Nevertheless, the upregulation of HLA class I on both TULV- and HTNV-infected cells could be blocked by neutralizing anti-IFN-beta antibodies. Most strikingly, the antiviral MxA protein, which interferes with hantavirus replication, was already induced 16 h after infection with TULV. In contrast, HTNV-infected HUVECs showed no expression of MxA until 48 h postinfection. In accordance with the kinetics of MxA expression, TULV replicated only inefficiently in HUVECs, whereas HTNV-infected cells produced high titers of virus particles that decreased after 48 h postinfection. Both hantavirus species, however, could replicate equally well in Vero E6 cells, which lack an IFN-induced MxA response. Thus, delayed induction of antiviral MxA in endothelial cells after infection with HTNV could allow viral dissemination and contribute to the pathogenesis leading to HFRS.


Assuntos
Células Endoteliais/imunologia , Células Endoteliais/virologia , Vírus Hantaan/patogenicidade , Orthohantavírus/patogenicidade , Animais , Chlorocebus aethiops , Proteínas de Ligação ao GTP/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon beta/metabolismo , Proteínas de Resistência a Myxovirus , Veias Umbilicais , Células Vero , Replicação Viral
20.
J Immunol ; 168(3): 1226-34, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11801659

RESUMO

The CD95 (also called APO-1/Fas) system plays a major role in the induction of apoptosis in lymphoid and nonlymphoid tissues. The CD95 ligand (CD95L) is induced in response to a variety of signals, including IFN-gamma and TCR/CD3 stimulation. Here we report the identification of two positive regulatory IFN-regulatory factor-dependent domains (PRIDDs) in the CD95L promoter and its 5' untranslated region, respectively. EMSAs demonstrate specific binding of IFN-gamma-induced IFN-regulatory factor 1 (IRF-1) to the PRIDD sequences. Ectopic IRF-1 expression induces CD95L promoter activity. Furthermore, we demonstrate that PRIDDs play an important role in TCR/CD3-mediated CD95L induction. Most interestingly, viral IRFs of human herpes virus 8 (HHV8) totally abolish IRF-1-mediated and strongly reduce TCR/CD3-mediated CD95L induction. We demonstrate here for the first time that viral IRFs inhibit activation-induced cell death. Thus, these results demonstrate an important mechanism of HHV8 to modulate the immune response by down-regulation of CD95L expression. Inhibition of CD95-dependent T cell function might contribute to the immune escape of HHV8.


Assuntos
Apoptose/imunologia , Proteínas de Ligação a DNA/fisiologia , Imunossupressores/farmacologia , Glicoproteínas de Membrana/genética , Fosfoproteínas/fisiologia , Regiões Promotoras Genéticas/imunologia , Fatores de Transcrição/fisiologia , Proteínas Virais/fisiologia , Receptor fas/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Proteína Ligante Fas , Regulação da Expressão Gênica/imunologia , Vetores Genéticos/biossíntese , Vetores Genéticos/imunologia , Células HeLa , Herpesvirus Humano 8/imunologia , Humanos , Fator Regulador 1 de Interferon , Fatores Reguladores de Interferon , Interferon gama/farmacologia , Células Jurkat , Ligantes , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/metabolismo , Mutagênese Sítio-Dirigida , Mutação Puntual , Estrutura Terciária de Proteína/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA