Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Sci Immunol ; 5(43)2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980485

RESUMO

Detection of intracellular DNA by the cGAS-STING pathway activates a type I interferon-mediated innate immune response that protects from virus infection. Whether there are additional DNA sensing pathways, and how such pathways might function, remains controversial. We show here that humans-but not laboratory mice-have a second, potent, STING-independent DNA sensing pathway (SIDSP). We identify human DNA-dependent protein kinase (DNA-PK) as the sensor of this pathway and demonstrate that DNA-PK activity drives a robust and broad antiviral response. We show that the E1A oncoprotein of human adenovirus 5 and the ICP0 protein of herpes simplex virus 1 block this response. We found heat shock protein HSPA8/HSC70 as a target for inducible phosphorylation in the DNA-PK antiviral pathway. Last, we demonstrate that DNA damage and detection of foreign DNA trigger distinct modalities of DNA-PK activity. These findings reveal the existence, sensor, a specific downstream target, and viral antagonists of a SIDSP in human cells.


Assuntos
Proteína Quinase Ativada por DNA/imunologia , Adenoviridae , Proteínas E1A de Adenovirus/imunologia , Animais , Linhagem Celular , Herpes Simples/imunologia , Herpesvirus Humano 1 , Humanos , Proteínas Imediatamente Precoces/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Ubiquitina-Proteína Ligases/imunologia
2.
Virus Res ; 220: 172-8, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27157859

RESUMO

Multigene-armed oncolytic adenoviruses are capable of efficiently generating a productive antitumor immune response. The chemokine (C-C motif) ligand 21 (CCL21) binds to CCR7 on naïve T cells and dendritic cells (DCs) to promote their chemoattraction to the tumor and resultant antitumor activity. Interleukin 21 (IL21) promotes survival of naïve T cells while maintaining their CCR7 surface expression, which increases their capacity to transmigrate in response to CCL21 chemoattraction. IL21 is also involved in NK cell differentiation and B cell activation and proliferation. The generation of effective antitumor immune responses is a complex process dependent upon coordinated interactions of various subsets of effector cells. Using the AdEasy system, we aimed to construct an oncolytic adenovirus co-expressing CCL21 and IL21 that could selectively replicate in TERTp-positive tumor cells (Ad-CCL21-IL21 virus). The E1A promoter of these oncolytic adenoviruses was replaced by telomerase reverse transcriptase promoter (TERTp). Ad-CCL21-IL21 was constructed from three plasmids, pGTE-IL21, pShuttle-CMV-CCL21 and AdEasy-1 and was homologously recombined and propagated in the Escherichia coli strain BJ5183 and the packaging cell line HEK-293, respectively. Our results showed that our targeted and armed oncolytic adenoviruses Ad-CCL21-IL21 can induce apoptosis in TERTp-positive tumor cells to give rise to viral propagation, in a dose-dependent manner. Importantly, we confirm that these modified oncolytic adenoviruses do not replicate efficiently in normal cells even under high viral loads. Additionally, we investigate the role of Ad-CCL21-IL21 in inducing antitumor activity and tumor specific cytotoxicity of CTLs in vitro. This study suggests that Ad-CCL21-IL21 is a promising targeted tumor-specific oncolytic adenovirus.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Quimiocina CCL21/genética , Interleucinas/genética , Vírus Oncolíticos/genética , Telomerase/genética , Adenoviridae/imunologia , Proteínas E1A de Adenovirus/imunologia , Células CACO-2 , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiocina CCL21/imunologia , Técnicas de Cocultura , Células Epiteliais/citologia , Células Epiteliais/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Células HT29 , Células HeLa , Recombinação Homóloga , Humanos , Interleucinas/imunologia , Masculino , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Plasmídeos/química , Plasmídeos/imunologia , Próstata/citologia , Próstata/imunologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Células THP-1 , Telomerase/imunologia
3.
PLoS Pathog ; 9(6): e1003411, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23785282

RESUMO

Upon infection, human adenovirus (HAdV) must activate the expression of its early genes to reprogram the cellular environment to support virus replication. This activation is orchestrated in large part by the first HAdV gene expressed during infection, early region 1A (E1A). E1A binds and appropriates components of the cellular transcriptional machinery to modulate cellular gene transcription and activate viral early genes transcription. Previously, we identified hBre1/RNF20 as a target for E1A. The interaction between E1A and hBre1 antagonizes the innate antiviral response by blocking H2B monoubiquitination, a chromatin modification necessary for the interferon (IFN) response. Here, we describe a second distinct role for the interaction of E1A with hBre1 in transcriptional activation of HAdV early genes. Furthermore, we show that E1A changes the function of hBre1 from a ubiquitin ligase involved in substrate selection to a scaffold which recruits hPaf1 as a means to stimulate transcription and transcription-coupled histone modifications. By using hBre1 to recruit hPaf1, E1A is able to optimally activate viral early transcription and begin the cycle of viral replication. The ability of E1A to target hBre1 to simultaneously repress cellular IFN dependent transcription while activating viral transcription, represents an elegant example of the incredible economy of action accomplished by a viral regulatory protein through a single protein interaction.


Assuntos
Infecções por Adenoviridae/metabolismo , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/metabolismo , Imunidade Inata , Proteínas Nucleares/metabolismo , Ativação Transcricional , Ubiquitina-Proteína Ligases/metabolismo , Replicação Viral/fisiologia , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/imunologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Linhagem Celular Tumoral , Histonas/genética , Histonas/imunologia , Histonas/metabolismo , Humanos , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/imunologia , Fatores de Transcrição , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
4.
Hum Gene Ther ; 23(6): 623-34, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22475378

RESUMO

Oncolytic viruses represent a novel therapeutic approach for aggressive tumors, such as glioblastoma multiforme, which are resistant to available treatments. Autophagy has been observed in cells infected with oncolytic viruses; however, its role in cell death/survival is unclear. To elucidate the potential therapeutic use of autophagy modulators in association with viral therapy, we analyzed autophagy induction in human glioma cell lines U373MG and U87MG infected with the oncolytic adenovirus dl922-947. dl922-947 infection triggered an autophagic cellular response, as shown by the development of acidic vesicular organelles, LC3-I→LC3-II conversion, and reduction of p62 levels. However, on infection, the Akt/mTOR/p70s6k pathway, which negatively regulates autophagy, was activated, whereas the ERK1/2 pathway, a positive regulator of autophagy, was inhibited. Accordingly, MEK inhibition by PD98059 sensitized glioma cells to dl922-947 effects, whereas autophagy induction by rapamycin protected cells from dl922-947-induced death. Treatment with two inhibitors of autophagy, chloroquine and 3-methyladenine, increased the cytotoxic effects of dl922-947 in vitro. In vivo, the growth of U87MG-induced xenografts was further reduced by adding chloroquine to the dl922-947 treatment. In conclusion, autophagy acts as a survival response in glioma cells infected with dl922-947, thus suggesting autophagy inhibitors as adjuvant/neoadjuvant drugs in oncolytic virus-based treatments.


Assuntos
Adenoviridae/genética , Adjuvantes Imunológicos/farmacologia , Autofagia/efeitos dos fármacos , Vetores Genéticos/uso terapêutico , Glioma/terapia , Vírus Oncolíticos/genética , Adenina/análogos & derivados , Adenina/farmacologia , Adenoviridae/imunologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Animais , Linhagem Celular Tumoral , Cloroquina/farmacologia , Imunofluorescência , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Vírus Oncolíticos/imunologia , Reação em Cadeia da Polimerase , Transdução de Sinais
5.
J Virol ; 86(8): 4222-33, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301148

RESUMO

Early region 1A (E1A) of human adenovirus (HAdV) has been the focus of over 30 years of investigation and is required for the oncogenic capacity of HAdV in rodents. Alternative splicing of the E1A transcript generates mRNAs encoding multiple E1A proteins. The 55-residue (55R) E1A protein, which is encoded by the 9S mRNA, is particularly interesting due to the unique properties it displays relative to all other E1A isoforms. 55R E1A does not contain any of the conserved regions (CRs) present in the other E1A isoforms. The C-terminal region of the 55R E1A protein contains a unique sequence compared to all other E1A isoforms, which results from a frameshift generated by alternative splicing. The 55R E1A protein is thought to be produced preferentially at the late stages of infection. Here we report the first study to directly investigate the function of the species C HAdV 55R E1A protein during infection. Polyclonal rabbit antibodies (Abs) have been generated that are capable of immunoprecipitating HAdV-2 55R E1A. These Abs can also detect HAdV-2 55R E1A by immunoblotting and indirect immunofluorescence assay. These studies indicate that 55R E1A is expressed late and is localized to the cytoplasm and to the nucleus. 55R E1A was able to activate the expression of viral genes during infection and could also promote productive replication of species C HAdV. 55R E1A was also found to interact with the S8 component of the proteasome, and knockdown of S8 was detrimental to viral replication dependent on 55R E1A.


Assuntos
Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/genética , RNA Mensageiro/química , RNA Viral/química , Adenosina Trifosfatases/metabolismo , Proteínas E1A de Adenovirus/imunologia , Adenovírus Humanos/imunologia , Sequência de Aminoácidos , Anticorpos Antivirais/imunologia , Linhagem Celular , Núcleo Celular/metabolismo , Inibição de Contato , Citoplasma/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Transcrição Gênica , Replicação Viral/genética
6.
APMIS ; 117(11): 791-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19845529

RESUMO

E1AF is associated with malignant aggressiveness via regulation of matrix metalloproteinases (MMPs), which play pivotal roles in invasion through the degradation of extracellular matrix of tissues surrounding tumors. However, the clinical significance of E1AF and MMPs in patients with prostate cancer is not fully understood. We reviewed 50 tissue samples from patients with T2-3N0M0 prostate cancer who had undergone radical operation. Expression levels of E1AF, MMP-1, -3, -7, -9 and -14 were determined semiquantitatively by immunohistochemistry. The mean +/- SD percentage of E1AF-stained cancer cells was 8.56 +/- 5.22, and it was significantly higher (p < 0.001) than the E1AF-immunostaining index of normal cells (1.17 +/- 0.61). E1AF immunostaining index in pT3 (12.74 +/- 4.80) was significantly higher (p < 0.001) than that in pT2 (5.78 +/- 3.31). Although E1AF expression correlated with that of MMP-7 and MMP-9 (r = 0.47, p < 0.001 and r = 0.41, p = 0.004, respectively), multivariate analysis showed that E1AF correlated with only MMP-7 expression (OR = 5.81, 95% CI = 1.27-26.59, p = 0.023). Our results demonstrated that increased expression of E1AF is involved in tumor aggression of prostate cancer. This finding may be influenced by regulation of MMP-7. We speculate that E1AF is a possible target in treatment and prevention of tumor growth in prostate cancer.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Metaloproteinases da Matriz/imunologia , Neoplasias da Próstata/imunologia , Proteínas Proto-Oncogênicas/imunologia , Idoso , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/enzimologia , Proteínas Proto-Oncogênicas c-ets , Estudos Retrospectivos
7.
Cancer Gene Ther ; 15(1): 40-50, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18034197

RESUMO

The majority of clinical trials evaluating replication-selective oncolytic adenoviruses utilized mutants with immunomodulatory E3B genes deleted, likely contributing to the attenuated efficacy. We investigated whether an intact immune response could contribute to the observed improved efficacy in response to combinations with chemotherapeutics. Seven carcinoma cell lines were evaluated by combining viral mutants; dl309 (DeltaE3B), dl704 (DeltaE3gp19K), dl312 (DeltaE1A) or wild-type Ad5 with the commonly used clinical drugs cisplatin and paclitaxel. Synergistic effects on cell death were determined by generation of combination indexes in cultured cells. In vivo tumor growth inhibition was achieved by virotherapy alone and was most efficacious with wild-type virus and least with the DeltaE3B mutant. Significantly higher efficacy was observed when the viruses were combined with drugs. The greatest enhancement of tumor inhibition was in combination with the DeltaE3B mutant restoring potency to that of Ad5 wild-type levels, observed only in animals with intact immune response. Increases in infectivity, viral gene expression and replication were identified as potential mechanisms contributing to the synergistic effects. Our results suggest that the attenuation of DeltaE3B mutants can be overcome by low doses of chemotherapeutics only in the presence of an intact immune response indicating a role for T-cell-mediated functions.


Assuntos
Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Cisplatino/farmacologia , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo , Paclitaxel/farmacologia , Adenoviridae/genética , Adenoviridae/imunologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Proteínas E3 de Adenovirus/genética , Animais , Antineoplásicos Fitogênicos/agonistas , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Morte Celular/imunologia , Linhagem Celular Tumoral , Cisplatino/agonistas , Deleção de Genes , Humanos , Imunidade Celular , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Paclitaxel/agonistas , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Virology ; 352(2): 338-44, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16780916

RESUMO

In adenovirus-12 tumorigenic cells, the viral E1A-12 protein mediates transcriptional down-regulation of the major histocompatibility complex (MHC) class I genes by targeting the class I enhancer. Here, we demonstrate by a combination of antisense and chromatin immunoprecipitation (ChIP) analysis that E1A-12 is a physical component of the class I enhancer repression complex, known to comprise COUP-TFII and histone deacetylase 1 (HDAC1). Significantly, E1A antisense was shown to co-eliminate E1A-12 as well as HDAC1 and HDAC8, but not HDAC3, from the enhancer repression complex. Consistent with elimination of HDAC1 and HDAC8, E1A antisense also resulted in a dramatic increase in histone acetylation, a hallmark of transcriptionally active chromatin. Importantly, MHC class I antigen expression was restored on the surface of E1A antisense-transfected cells. These results demonstrate that E1A-12 is associated with the MHC class I complex and apparently mediates class I transcriptional down-regulation by enacting chromatin repression through HDAC1 and HDAC8.


Assuntos
Adenoviridae/genética , Adenoviridae/patogenicidade , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/fisiologia , Genes MHC Classe I , Histona Desacetilases/genética , Adenoviridae/imunologia , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/imunologia , Animais , Sequência de Bases , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cromatina/genética , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Antígenos de Histocompatibilidade Classe I/metabolismo , Camundongos , Oligodesoxirribonucleotídeos Antissenso/genética
9.
J Exp Med ; 202(11): 1477-82, 2005 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-16314433

RESUMO

The expression of the Adenovirus serotype 5 (Ad5) E1A oncogene sensitizes tumor cells to natural killer (NK) cell-mediated killing and tumor rejection in vivo. These effects are dependent on the ability of E1A to bind the transcriptional coadaptor protein p300. To test the hypothesis that E1A up-regulates ligands recognized by the NKG2D-activating receptor, we stably transfected the highly tumorigenic mouse fibrosarcoma cell line MCA-205 with Ad5-E1A or a mutant form of E1A that does not interact with p300 (E1A-Deltap300). Ad5-E1A, but not E1A-Deltap300, up-regulated the expression of the NKG2D ligand retinoic acid early inducible (RAE)-1, but not murine ULBP-like transcript 1, another NKG2D ligand, in four independently derived MCA-205 transfectants. The up-regulation of RAE-1 by E1A targeted MCA-205 tumor cells to lysis by NK cells, resulting in NKG2D-dependent tumor rejection in vivo. Moreover, the up-regulation of NKG2D ligands by E1A was not limited to mouse tumor cells, as E1A also increased the expression of NKG2D ligands on primary baby mouse kidney cells, human MB435S breast cancer cells, and human H4 fibrosarcoma cells.


Assuntos
Adenoviridae , Fibrossarcoma/imunologia , Células Matadoras Naturais/imunologia , Receptores Imunológicos/imunologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/imunologia , Vetores Genéticos , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Humanos , Rim/imunologia , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Receptores Imunológicos/genética , Receptores de Células Matadoras Naturais , Tretinoína/administração & dosagem , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
10.
J Immunol ; 175(2): 855-63, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16002683

RESUMO

Immature dendritic cells (DC), in contrast to their mature counterparts, are incapable of mobilizing a CD8+ CTL response, and, instead, have been reported to induce CTL tolerance. We directly addressed the impact of immature vs mature DC on CTL responses by infusing adenovirus peptide-loaded DC (of the D1 cell line) into mice that had received adenovirus-specific naive TCR-transgenic CD8+ T cells. Whereas i.v. injection of mature DC triggered vigorous CTL expansion, immature DC elicited little proliferation involving only a minority of the TCR-transgenic CTL. Even though the latter CTL developed effector functions, including cytolytic activity and proinflammatory cytokine secretion, these cells differed significantly from CTL primed by mature DC in that they did not exhibit down-regulation of CD62L and CCR7, receptors involved in trapping of T cells in the lymphoid organs. Interestingly, adoptive transfer of CTL effector cells harvested after priming by either mature or immature DC into naive recipient mice, followed by exposure to adenovirus, yielded quantitatively and qualitatively indistinguishable CTL memory responses. Therefore, in vivo priming of naive CD8+ T cells by immature DC, although failing to induce a full-blown, systemic CTL response, resulted in the formation of central memory-like T cells that were able to expand and produce IFN-gamma upon secondary antigenic stimulation.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Deleção Clonal/imunologia , Células Dendríticas/imunologia , Memória Imunológica , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Animais , Apresentação de Antígeno/genética , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/genética , Linhagem Celular , Movimento Celular/genética , Movimento Celular/imunologia , Deleção Clonal/genética , Citotoxicidade Imunológica/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Regulação para Baixo/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Memória Imunológica/genética , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/virologia
11.
Hum Gene Ther ; 16(12): 1439-51, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16390275

RESUMO

Improvement of the therapeutic index of adenoviral gene transfer requires the development of strategies to abrogate adenoviral capsid-induced inflammation and cytokine production. The effect of monomethoxypolyethylene glycol (MPEG) conjugation to adenoviral vectors and of methylprednisolone (MP) on innate immunity, liver inflammation, and thrombocyte counts was evaluated after transfer of 1011 particles of E1/E3/E4- deleted adenoviral vector expressing human apolipoprotein A-I (apoA-I). Gene transfer with unPEGylated vectors induced peak interleukin-6 (IL-6) plasma levels that were 66-fold above baseline levels in C57BL/6 mice. PEGylation combined with 4 mg of MP 6 hr before and at the time of gene transfer suppressed IL-6 plasma levels to baseline values at all time points. This combination resulted in 24-, 28-, 5.9-, 42-, 26-, and 2.5- fold reduced mRNA expression in the liver of monocyte chemoattractant protein-1, macrophage inflammatory protein-2, interferon-inducible protein-10, macrophage inflammatory protein-1 beta, lipopolysaccharide-induced CXC chemokine, and keratinocyte-derived chemokine, respectively; abrogated neutrophil infiltration in the liver; and reduced alanine aminotransferase levels. PEGylation reduced vector uptake in the spleen and in nonparenchymal liver cells. PEGylation also inhibited the development of thrombocytopenia. In conclusion, PEGylation of adenoviral vectors combined with MP administration improves the therapeutic index of adenoviral gene transfer.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/imunologia , Interleucina-6/metabolismo , Fígado/imunologia , Metilprednisolona/farmacologia , Polietilenoglicóis/metabolismo , Animais , Conjugação Genética , Citocinas/metabolismo , Feminino , Imunidade Inata/efeitos dos fármacos , Inflamação/tratamento farmacológico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
J Immunol ; 173(11): 6753-9, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15557168

RESUMO

The fate of naive CD8(+) T cells is determined by the environment in which they encounter MHC class I presented peptide Ags. The manner in which tumor Ags are presented is a longstanding matter of debate. Ag presentation might be mediated by tumor cells in tumor draining lymph nodes or via cross-presentation by professional APC. Either pathway is insufficient to elicit protective antitumor immunity. We now demonstrate using a syngeneic mouse tumor model, expressing an Ag derived from the early region 1A of human adenovirus type 5, that the inadequate nature of the antitumor CTL response is not due to direct Ag presentation by the tumor cells, but results from presentation of tumor-derived Ag by nonactivated CD11c(+) APC. Although this event results in division of naive CTL in tumor draining lymph nodes, it does not establish a productive immune response. Treatment of tumor-bearing mice with dendritic cell-stimulating agonistic anti-CD40 mAb resulted in systemic efflux of CTL with robust effector function capable to eradicate established tumors. For efficacy of anti-CD40 treatment, CD40 ligation of host APC is required because adoptive transfer of CD40-proficient tumor-specific TCR transgenic CTL into CD40-deficient tumor-bearing mice did not lead to productive antitumor immunity after CD40 triggering in vivo. CpG and detoxified LPS (MPL) acted similarly as agonistic anti-CD40 mAb with respect to CD8(+) CTL efflux and tumor eradication. Together these results indicate that dendritic cells, depending on their activation state, orchestrate the outcome of CTL-mediated immunity against tumors, leading either to an ineffective immune response or potent antitumor immunity.


Assuntos
Antígenos Virais de Tumores/metabolismo , Apresentação Cruzada/imunologia , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Proteínas E1A de Adenovirus/imunologia , Proteínas E1A de Adenovirus/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos Virais de Tumores/imunologia , Antígeno CD11c/biossíntese , Antígenos CD40/biossíntese , Antígenos CD40/imunologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Injeções Intralesionais , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas
13.
J Immunol ; 172(10): 6074-9, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15128791

RESUMO

Naive T cells can be tolerized in the periphery by diverse mechanisms. However, the extent to which memory T cells are susceptible to tolerance induction is less well defined. Vaccination of mice with a minimal CTL epitope derived from human adenovirus type 5 E1A in IFA s.c. readily tolerizes naive as well as recently activated CD8(+) T cells due to the overwhelming systemic and persistent presence of the peptide. We have now studied the effect of this peptide on established memory cells, which were induced at least 50 days before by virus vaccination. Memory cells did not undergo peripheral deletion and kept their ability to produce IFN-gamma as well as their cytolytic activity in response to Ag directly ex vivo. However, memory CTL responses in virus vaccinated mice injected with peptide ceased to control tumor outgrowth. Interestingly, functional capacities were regained when T cells were transferred to an Ag-free environment in vivo as determined by their ability to reject an otherwise lethal tumor challenge. Together, these findings indicate that memory CTL responses can be functionally incapacitated, but are not, in contrast to naive or recently activated T cells, irreversibly tolerized by persistent systemic Ag, as memory T cells quickly regain effector function upon disappearance of the Ag.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Proteínas E1A de Adenovirus/metabolismo , Apresentação de Antígeno , Citotoxicidade Imunológica , Memória Imunológica , Linfócitos T Citotóxicos/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/terapia , Proteínas E1A de Adenovirus/administração & dosagem , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Divisão Celular/imunologia , Células Cultivadas , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Tolerância Imunológica/imunologia , Injeções Subcutâneas , Interfase/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/transplante , Infecções Tumorais por Vírus/patologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
14.
J Virol ; 78(9): 4646-54, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15078947

RESUMO

Adenovirus (Ad) E1A and human papillomavirus (HPV) E7 express homologous conserved regions (CRs) that mediate their shared biological functions. Despite their similarities, the expression of E1A sensitizes tumor cells to killing by NK cells and macrophages but the expression of E7 does not, a factor that may contribute to the dissimilar oncogenicities of Ad and HPV. This study was undertaken to define molecular differences between E1A and E7 that are responsible for the ability of E1A and the inability of E7 to sensitize cells to killing by NK cells and macrophages. Genetic mapping studies using human fibrosarcoma cells (H4) that stably expressed mutant forms of E1A showed that only those forms of E1A that interacted with the transcriptional coadaptor protein p300 sensitized cells to killing by NK cells and macrophages. E7 lacks the N-terminal p300-binding region present in E1A. Therefore, a chimeric E1A/E7 gene was constructed that included the N terminus and the CR1 (p300-binding) domain of E1A fused to CR2 and the C-terminal sequences of E7. The E1A/E7 protein interacted with p300 and pRb and immortalized primary mouse embryo fibroblasts (MEF). The expression of E1A/E7 sensitized H4 and MEF cells to killing by activated macrophages but not to killing by NK cells. Therefore, N-terminal differences between E1A and E7 that map to the E1A-p300 binding region accounted for differences in their abilities to sensitize cells to killing by macrophages. However, regions in addition to the E1A-p300 binding region are required to sensitize cells to killing by NK cells.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas E1A de Adenovirus/imunologia , Animais , Citotoxicidade Imunológica , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas Virais/imunologia , Ratos , Proteínas Recombinantes de Fusão/imunologia , Células Tumorais Cultivadas
15.
J Immunol ; 172(3): 1567-74, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734736

RESUMO

Ocular immune privilege is considered essential in the protection against sight-threatening immune responses, as illustrated by the ability of the ocular environment to permit the growth of tumors that are rejected when implanted at other sites. Although several studies indicate that soluble Ag can drain directly into the spleen when injected into the anterior chamber, the primary site of intraocular tumor Ag presentation to tumor-specific CTLs has not been studied. To gain a better understanding of the mechanism involved in ocular immune privilege, we examined to which lymphoid organs anterior chamber tumor Ags primarily drain. Our data show that intraocular tumor Ag drains exclusively to the submandibular lymph nodes, resulting in activation of tumor-specific CTLs, whereas no Ag drainage was found in spleen. However, these tumor-specific CTLs do not distribute systemically and, as a consequence, intraocular tumor growth is unhampered. A similar lack of CTL efficacy has been observed in mice bearing s.c. tumors, which is converted to a systemic tumoricidal CTL response by administration of agonistic anti-CD40 mAb. In contrast, systemic anti-CD40 treatment of eye tumor-bearing mice did not result in mobilizing tumor-specific CTLs or tumor eradication. Together, these results show that intraocular tumor Ag drains to regional lymph nodes for activation of tumor-specific CTLs. However, the induced tumor-specific immunity is insufficient for tumor clearance, even combined with otherwise highly effective immune intervention protocols.


Assuntos
Antígenos de Neoplasias/imunologia , Citotoxicidade Imunológica , Neoplasias Oculares/imunologia , Rejeição de Enxerto/imunologia , Linfonodos/imunologia , Glândula Submandibular/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas E1A de Adenovirus/biossíntese , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/imunologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Animais , Câmara Anterior/imunologia , Câmara Anterior/patologia , Câmara Anterior/virologia , Antígenos de Neoplasias/genética , Antígenos CD40/fisiologia , Divisão Celular/genética , Divisão Celular/imunologia , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/imunologia , Citotoxicidade Imunológica/genética , Epitopos de Linfócito T/imunologia , Neoplasias Oculares/patologia , Neoplasias Oculares/virologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/virologia , Linfonodos/patologia , Linfonodos/virologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/virologia , Glândula Submandibular/patologia , Glândula Submandibular/virologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia
16.
Curr Top Microbiol Immunol ; 273: 245-88, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14674604

RESUMO

A broad spectrum of genetic and molecular investigations carried out with group C, Ad2 and Ad5, and with group A, Ad12, have shown that early region1 (E1) gene products are sufficient for complete transformation of rodent cells in vitro by these viruses. During the past quarter century, the processes by which E1A proteins, in cooperation with E1B proteins, perturb the cell cycle and induce the transformed phenotype, have become well defined. Somewhat less understood is the basis for the differential oncogenicity of these two groups of viruses, and the processes by which the E1A proteins of Ad12 induce a tumorigenic phenotype in transformants resulting from infection of cells in vivo and in vitro. In this chapter we review previous findings and present new evidence which demonstrates that Ad12 E1A possesses two or more independent functions enabling it to induce tumors. One of these functions lies in its capacity to repress transcription of MHC class I genes, allowing the tumor cells to avoid lysis by cytotoxic T lymphocytes. We have shown that class I repression is mediated through increased binding of repressor COUP-TF and decreased binding of NF-kB to the class I enhancer. In addition to mediating immune escape, E1A also determines the susceptibility of transformants to Natural Killer (NK) cell lysis, and in this case, also, Ad12 transformants are not susceptible. By using Ad12 mutants containing chimeric E1A Ad12-Ad5 genes, point mutations, or a specific deletion, we have shown that the unique spacer region of Ad12 E1A is an oncogenic determinant, but is not required for transformation in vitro. Given that the E1A regions responsible for class I repression are first exon encoded, we have examined a set of cell lines transformed by these altered viruses, and have found that while they display greatly reduced tumorigenicity, they maintain a wildtype capacity to repress class I transcription. Whether the spacer contributes to NK evasion remains unresolved. Lastly, we discuss the properties of the Ad2/Ad5 E1A C-terminal negative modulator of tumorigenicity, and examine the effects on transformation, tumor induction and transformant tumorigenicity, when the Ad5 negative modulator is placed by chimeric construction in Ad12 E1A.


Assuntos
Proteínas E1A de Adenovirus/fisiologia , Adenovírus Humanos/fisiologia , Transformação Celular Neoplásica/genética , Antígenos de Histocompatibilidade Classe I/biossíntese , Proteínas E1A de Adenovirus/imunologia , Adenovírus Humanos/classificação , Adenovírus Humanos/genética , Adenovírus Humanos/patogenicidade , Animais
17.
J Immunol ; 170(8): 4119-26, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12682242

RESUMO

Expression of adenovirus (Ad) serotype 2 or 5 (Ad2/5) E1A or human papillomavirus (HPV)16 E7 reportedly sensitizes cells to lysis by macrophages. Macrophages possess several mechanisms to kill tumor cells including TNF-alpha, NO, reactive oxygen intermediates (ROI), and Fas ligand (FasL). E1A sensitizes cells to apoptosis by TNF-alpha, and macrophages kill E1A-expressing cells, in part through the elaboration of TNF-alpha. However, E1A also up-regulates the expression of 70-kDa heat shock protein, a protein that inhibits killing by TNF-alpha and NO, thereby protecting cells from lysis by macrophages. Unlike E1A, E7 does not sensitize cells to killing by TNF-alpha, and the effector mechanism(s) used by macrophages to kill E7-expressing cells remain undefined. The purpose of this study was to further define the capacity of and the effector mechanisms used by macrophages to kill tumor cells that express Ad5 E1A or HPV16 E7. We found that Ad5 E1A, but not HPV16 E7, sensitized tumor cells to lysis by macrophages. Using macrophages derived from mice unable to make TNF-alpha, NO, ROI, or FasL, we determined that macrophages used NO, and to a lesser extent TNF-alpha, but not FasL or ROI, to kill E1A-expressing cells. Through the use of S-nitroso-N-acetylpenicillamine, which releases NO upon exposure to an aqueous environment, E1A was shown to directly sensitize tumor cells to NO-induced death. E1A sensitized tumor cells to lysis by macrophages despite up-regulating the expression of 70-kDa heat shock protein. In summary, E1A, but not E7, sensitized tumor cells to lysis by macrophages. Macrophages killed E1A-expressing cells through NO- and TNF-alpha-dependent mechanisms.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Testes Imunológicos de Citotoxicidade/métodos , Proteínas de Choque Térmico HSP70/biossíntese , Macrófagos/imunologia , Óxido Nítrico/toxicidade , Proteínas Oncogênicas Virais/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Regulação para Cima/imunologia , Células 3T3 , Proteínas E1A de Adenovirus/biossíntese , Adenovírus Humanos/imunologia , Animais , Linhagem Celular Transformada , Proteína Ligante Fas , Proteínas de Choque Térmico HSP70/fisiologia , Humanos , Imunização , Ligantes , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus , Sarcoma Experimental/imunologia , Sarcoma Experimental/prevenção & controle , Sarcoma Experimental/virologia , Superóxidos/metabolismo , Células Tumorais Cultivadas , Receptor fas/metabolismo
18.
Int Immunol ; 13(10): 1301-7, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11581175

RESUMO

Expression of adenovirus E1A gene products in tumor cells enhances NK cell lysis in vitro and NK-mediated rejection in vivo, despite increasing class I molecules on tumor cells. It is unclear why the increased expression of MHC class I molecules does not appear to confer resistance to killing by NK cells. One possibility is the unique capacity of E1A to sensitize cells to multiple NK cell killing mechanisms including perforin/granzyme, Fas ligand, tumor necrosis factor-alpha and TRAIL. To examine this issue, MCA-102-E1A tumor cells (H-2(b)) that express E1A and are NK sensitive were transfected with H-2D(d), the ligand for the NK inhibitory receptor, Ly49A. Expression of H-2D(d) molecules by MCA-102-E1A cells protected them from lysis by a Ly49A(+) NK cell clone and Ly49A(+) NK cells isolated from C57BL/6 nude mice. In contrast, NK cell-mediated rejection of MCA-102-E1A tumor cells was not inhibited by the expression of H-2D(d) molecules, nor was killing by polyclonal populations of NK cells isolated from C57BL/6-nude mice. H-2D(d) interacts with several inhibitory Ly49 receptors that are non-clonally expressed on NK cells in C57BL/6 mice: Ly49A (20% of NK cells), Ly49G2 (54% of NK cells) and Ly49C/I (47% of NK cells). Our data indicate that while E1A sensitizes cells to NK cell killing, it does not interfere with signal transduction by inhibitory NK receptors. Therefore, a small population of NK cells that do not express Ly49A, Ly49G2 or Ly49C/I inhibitory receptors are likely responsible for the rejection of MCA-102-E1A-D(d) tumor cells in vivo.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Antígenos Ly , Antígenos de Neoplasias/imunologia , Citotoxicidade Imunológica/imunologia , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/imunologia , Animais , Antígenos H-2 , Lectinas Tipo C , Glicoproteínas de Membrana , Camundongos , Subfamília A de Receptores Semelhantes a Lectina de Células NK , Ratos , Receptores Semelhantes a Lectina de Células NK , Sarcoma Experimental , Células Tumorais Cultivadas
19.
J Immunol ; 167(5): 2522-8, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11509591

RESUMO

Encounter of Ag by naive T cells can lead to T cell priming as well as tolerance. The balance between immunity and tolerance is controlled by the conditions of Ag encounter and the activation status of the APC. We have investigated the rules that govern this balance in case an environment that normally induces tolerance is reverted into a milieu that promotes T cell priming, using a minimal CTL epitope derived from human adenovirus type 5 E1A. Vaccination of mice s.c. with E1A peptide in IFA readily induces CTL tolerance, resulting in the inability to control E1A-expressing tumors. The present study shows that efficient CTL priming is achieved when this peptide vaccine is combined with systemic administration of APC-activating compounds like agonistic anti-CD40 mAb or polyriboinosinate-polyribocytidylate. Surprisingly, this CTL response is not long-lasting and therefore fails to protect against tumor outgrowth. Disappearance of CTL reactivity was strongly associated with systemic persistence of the peptide for >200 days. In contrast, peptide administered in PBS does not persist and generates long term CTL immunity capable of rejecting Ad5E1A-positive tumors, when combined with CD40 triggering. Thus, presentation of CTL epitopes in an appropriate costimulatory setting by activated APC, although being essential and sufficient for CTL priming, eventually results in tolerance when the Ag persists systemically for prolonged times. These observations are important for the development of immune intervention schemes in autoimmunity and cancer.


Assuntos
Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Tolerância Imunológica , Linfócitos T Citotóxicos/imunologia , Proteínas E1A de Adenovirus/administração & dosagem , Proteínas E1A de Adenovirus/imunologia , Animais , Antígenos CD40/metabolismo , Humanos , Cinética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo
20.
J Immunol ; 166(12): 7151-7, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11390461

RESUMO

Vaccination with a peptide representing a CTL epitope from the human papillomavirus (HPV)16 E7 protein induces a specific CTL response that prevents the outgrowth of HPV16 E7-expressing tumors. In contrast, vaccination with a peptide encoding an adenovirus type 5 (Ad5) E1A CTL epitope results in CTL tolerance and enhanced growth of an Ad5 E1A-expressing tumor. It is unclear why these peptides induce such opposite effects. To determine whether a difference in pharmacokinetics can explain the functional contrasts, tritiated Ad5 E1A and HPV16 E7 peptides were injected into mice. Results show that the tolerizing peptide spread through the body 16 times faster than the activating peptide and was cleared at least 2 times faster. The HPV16 E7 peptide kinetics correlated with the kinetics of HPV16 E7-specific CTL induction. In contrast, Ad5 E1A peptide injection resulted in physical deletion of preexisting Ad5 E1A-specific CTLs within 24 h after injection. This tolerization occurred at the time when the peptide reached its maximum peptide concentration in the organs. These data suggest that ubiquitous expression of the tolerizing Ad5 E1A peptide within a short period of time causes activation-induced cell death of Ad5 E1A-specific CTLs. Therefore, information on the pharmacokinetics of peptides is vital for the safety and efficacy of peptide-based vaccines.


Assuntos
Proteínas E1A de Adenovirus/imunologia , Proteínas E1A de Adenovirus/farmacocinética , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Proteínas Oncogênicas Virais/imunologia , Proteínas Oncogênicas Virais/farmacocinética , Papillomaviridae/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas E1A de Adenovirus/administração & dosagem , Animais , Deleção Clonal , Difusão , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Injeções Subcutâneas , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas Virais/administração & dosagem , Especificidade de Órgãos/imunologia , Proteínas E7 de Papillomavirus , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacocinética , Ligação Proteica/imunologia , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo , Trítio/metabolismo , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Vacinas Virais/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA