Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 108(30): 12419-24, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21746921

RESUMO

Inflammasomes are intracellular multiprotein signaling complexes that activate Caspase-1, leading to the cleavage and secretion of IL-1ß and IL-18, and ultimately host cell death. Inflammasome activation is a common cellular response to infection; however, the consequences of inflammasome activation during acute infection and in the development of long-term protective immunity is not well understood. To investigate the role of the inflammasome in vivo, we engineered a strain of Listeria monocytogenes that ectopically expresses Legionella pneumophila flagellin, a potent activator of the Nlrc4 inflammasome. Compared with wild-type L. monocytogenes, strains that ectopically secreted flagellin induced robust host cell death and IL-1ß secretion. These strains were highly attenuated both in bone marrow-derived macrophages and in vivo compared with wild-type L. monocytogenes. Attenuation in vivo was dependent on Nlrc4, but independent of IL-1ß/IL-18 or neutrophil activity. L. monocytogenes strains that activated the inflammasome generated significantly less protective immunity, a phenotype that correlated with decreased induction of antigen-specific T cells. Our data suggest that avoidance of inflammasome activation is a critical virulence strategy for intracellular pathogens, and that activation of the inflammasome leads to decreased long-term protective immunity and diminished T-cell responses.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/imunologia , Animais , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Ligação ao Cálcio/deficiência , Proteínas do Citoesqueleto/imunologia , Feminino , Flagelina/genética , Flagelina/imunologia , Engenharia Genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Imunidade Inata , Imunização , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Listeriose/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Linfócitos T/imunologia , Virulência/genética , Virulência/imunologia
2.
PLoS One ; 14(1): e0209153, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30601871

RESUMO

Dysregulated signaling via the epidermal growth factor receptor (EGFR)-family is believed to contribute to the progression of a diverse array of cancers. The most common variant of EGFR is EGFRvIII, which results from a consistent and tumor-specific in-frame deletion of exons 2-7 of the EGFR gene. This deletion generates a novel glycine at the junction and leads to constitutive ligand-independent activity. This junction forms a novel shared tumor neo-antigen with demonstrated immunogenicity in both mice and humans. A 21-amino acid peptide spanning the junctional region was selected, and then one or five copies of this 21-AA neo-peptide were incorporated into live-attenuated Listeria monocytogenes-based vaccine vector. These vaccine candidates demonstrated efficient secretion of the recombinant protein and potent induction of EGFRvIII-specific CD8+ T cells, which prevented growth of an EGFRvIII-expressing squamous cell carcinoma. These data demonstrate the potency of a novel cancer-specific vaccine candidate that can elicit EGFRvIII-specific cellular immunity, for the purpose of targeting EGFRvIII positive cancers that are resistant to conventional therapies.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Animais , Vacinas Anticâncer/imunologia , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/terapia , Feminino , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
3.
Infect Immun ; 76(8): 3742-53, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18541651

RESUMO

Recombinant vaccines derived from the facultative intracellular bacterium Listeria monocytogenes are presently undergoing early-stage clinical evaluation in oncology treatment settings. This effort has been stimulated in part due to preclinical results that illustrate potent activation of innate and adaptive immune effectors by L. monocytogenes vaccines, combined with efficacy in rigorous animal models of malignant and infectious disease. Here, we evaluated the immunologic potency of a panel of isogenic vaccine strains that varied only in prfA. PrfA is an intracellularly activated transcription factor that induces expression of virulence genes and encoded heterologous antigens (Ags) in appropriately engineered vaccine strains. Mutant strains with PrfA locked into a constitutively active state are known as PrfA* mutants. We assessed the impacts of three PrfA* mutants, G145S, G155S, and Y63C, on the immunologic potencies of live-attenuated and photochemically inactivated nucleotide excision repair mutant (killed but metabolically active [KBMA]) vaccines. While PrfA* substantially increased Ag expression in strains grown in broth culture, Ag expression levels were equivalent in infected macrophage and dendritic cell lines, conditions that more closely parallel those in the immunized host. However, only the prfA(G155S) allele conferred significantly enhanced vaccine potency to KBMA vaccines. In the KBMA vaccine background, we show that PrfA*(G155S) enhanced functional cellular immunity following an intravenous or intramuscular prime-boost immunization regimen. These results form the basis of a rationale for including the prfA(G155S) allele in future live-attenuated or KBMA L. monocytogenes vaccines advanced to the clinical setting.


Assuntos
Antígenos/biossíntese , Antígenos/imunologia , Vacinas Bacterianas/imunologia , Listeria monocytogenes/imunologia , Fatores de Terminação de Peptídeos/genética , Substituição de Aminoácidos/genética , Animais , Antígenos/genética , Antígenos de Bactérias/biossíntese , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/genética , Feminino , Imunização Secundária , Injeções Intramusculares , Injeções Intravenosas , Dose Letal Mediana , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeriose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Regulon , Vacínia/prevenção & controle , Virulência , Fatores de Virulência/biossíntese , Fatores de Virulência/imunologia
4.
Cell Host Microbe ; 7(5): 412-9, 2010 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-20417169

RESUMO

A host defense strategy against pathogens is the induction of cell death, thereby eliminating the pathogen's intracellular niche. Pyroptosis, one such form of cell death, is dependent on inflammasome activation. In a genetic screen to identify Listeria monocytogenes mutants that induced altered levels of host cell death, we identified a mutation in lmo2473 that caused hyperstimulation of IL-1beta secretion and pyroptosis following bacteriolysis in the macrophage cytosol. In addition, strains engineered to lyse in the cytosol by expression of both bacteriophage holin and lysin or induced to lyse by treatment with ampicillin stimulated pyroptosis. Pyroptosis was independent of the Nlrp3 and Nlrc4 inflammasome receptors but dependent on the inflammasome adaptor ASC and the cytosolic DNA sensor AIM2. Importantly, wild-type L. monocytogenes were also found to lyse, albeit at low levels, and trigger AIM2-dependent pyroptosis. These data suggested that pyroptosis is triggered by bacterial DNA released during cytosolic lysis.


Assuntos
Morte Celular , Listeria monocytogenes/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Proteínas Nucleares/imunologia , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Bacteriólise , Bacteriófagos/genética , Citosol/microbiologia , Proteínas de Ligação a DNA , Camundongos , Mucoproteínas/genética , Mucoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
J Immunother ; 32(8): 856-69, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19752748

RESUMO

Immunotherapeutic vaccine is potentially an effective strategy to combat cancer. Essential components of an effective vaccine must include antigens that are processed by the major histocompatibility complex class I pathway, presented by the tumor major histocompatibility complex molecules, and an effective antigen delivery platform that is capable of breaking self-tolerance. In this study, we characterized a set of ovarian cancer-specific T-cell epitopes delivered by live-attenuated recombinant Listeria monocytogenes (Lm DeltaactADeltainlB) as a vaccine vector. We present data that peptide-specific T cells recognize the human monocytic cell line THP-1 infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Furthermore, we demonstrate that recombinant L. monocytogenes (Lm)-infected antigen-presenting cells can prime and expand epitope-specific CD8 T cells in vitro and such CD8 T cells recognize not only peptide-loaded targets but also ovarian and breast tumor cells presenting endogenous epitopes. Finally, peptide-specific T cells generated using peripheral blood mononuclear cell from ovarian cancer patients recognize target cells infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Our results demonstrate that live-attenuated recombinant Lm can be used effectively as a vehicle to deliver cancer peptide antigens singly or as a multiepitope construct. Thus, the use of recombinant live-attenuated Lm strains encoding endogenously processed and presented tumor epitopes/antigens represents an attractive strategy for active cancer immunotherapy in a clinical setting.


Assuntos
Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Listeria monocytogenes/imunologia , Neoplasias Ovarianas/terapia , Apresentação de Antígeno , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Mapeamento de Epitopos , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Feminino , Antígeno HLA-A2/metabolismo , Humanos , Listeriose/imunologia , Ativação Linfocitária , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA