Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Immunity ; 44(6): 1406-21, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27287410

RESUMO

Monobenzone is a pro-hapten that is exclusively metabolized by melanocytes, thereby haptenizing melanocyte-specific antigens, which results in cytotoxic autoimmunity specifically against pigmented cells. Studying monobenzone in a setting of contact hypersensitivity (CHS), we observed that monobenzone induced a long-lasting, melanocyte-specific immune response that was dependent on NK cells, yet fully intact in the absence of T- and B cells. Consistent with the concept of "memory NK cells," monobenzone-induced NK cells resided in the liver and transfer of these cells conferred melanocyte-specific immunity to naive animals. Monobenzone-exposed skin displayed macrophage infiltration and cutaneous lymph nodes showed an inflammasome-dependent influx of macrophages with a tissue-resident phenotype, coinciding with local NK cell activation. Indeed, macrophage depletion or the absence of the NLRP3 inflammasome, the adaptor protein ASC or interleukin-18 (IL-18) abolished monobenzone CHS, thereby establishing a non-redundant role for the NLRP3 inflammasome as a critical proinflammatory checkpoint in the induction of hapten-dependent memory NK cells.


Assuntos
Dermatite de Contato/imunologia , Memória Imunológica , Inflamassomos/imunologia , Células Matadoras Naturais/imunologia , Macrófagos/fisiologia , Melanócitos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Imunidade Adaptativa , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Células Cultivadas , Hidroquinonas , Interleucina-18/genética , Interleucina-18/metabolismo , Fígado/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
2.
J Med Virol ; 94(1): 388-392, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34415572

RESUMO

In the current COVID-19 pandemic, a better understanding of the relationship between merely binding and functionally neutralizing antibodies is necessary to characterize protective antiviral immunity following infection or vaccination. This study analyzes the level of correlation between the novel quantitative EUROIMMUN Anti-SARS-CoV-2 QuantiVac ELISA (IgG) and a microneutralization assay. A panel of 123 plasma samples from a COVID-19 outbreak study population, preselected by semiquantitative anti-SARS-CoV-2 IgG testing, was used to assess the relationship between the novel quantitative ELISA (IgG) and a microneutralization assay. Binding IgG targeting the S1 antigen was detected in 106 (86.2%) samples using the QuantiVac ELISA, while 89 (72.4%) samples showed neutralizing antibody activity. Spearman's correlation analysis demonstrated a strong positive relationship between anti-S1 IgG levels and neutralizing antibody titers (rs = 0.819, p < 0.0001). High and low anti-S1 IgG levels were associated with a positive predictive value of 72.0% for high-titer neutralizing antibodies and a negative predictive value of 90.8% for low-titer neutralizing antibodies, respectively. These results substantiate the implementation of the QuantiVac ELISA to assess protective immunity following infection or vaccination.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Imunoglobulina G/sangue , SARS-CoV-2/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/imunologia , COVID-19/patologia , Teste Sorológico para COVID-19/métodos , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Neutralização/métodos , Sensibilidade e Especificidade , Glicoproteína da Espícula de Coronavírus/imunologia , Adulto Jovem
3.
Nature ; 490(7420): 412-6, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23051752

RESUMO

Adoptive cell transfer therapies (ACTs) with cytotoxic T cells that target melanocytic antigens can achieve remissions in patients with metastatic melanomas, but tumours frequently relapse. Hypotheses explaining the acquired resistance to ACTs include the selection of antigen-deficient tumour cell variants and the induction of T-cell tolerance. However, the lack of appropriate experimental melanoma models has so far impeded clear insights into the underlying mechanisms. Here we establish an effective ACT protocol in a genetically engineered mouse melanoma model that recapitulates tumour regression, remission and relapse as seen in patients. We report the unexpected observation that melanomas acquire ACT resistance through an inflammation-induced reversible loss of melanocytic antigens. In serial transplantation experiments, melanoma cells switch between a differentiated and a dedifferentiated phenotype in response to T-cell-driven inflammatory stimuli. We identified the proinflammatory cytokine tumour necrosis factor (TNF)-α as a crucial factor that directly caused reversible dedifferentiation of mouse and human melanoma cells. Tumour cells exposed to TNF-α were poorly recognized by T cells specific for melanocytic antigens, whereas recognition by T cells specific for non-melanocytic antigens was unaffected or even increased. Our results demonstrate that the phenotypic plasticity of melanoma cells in an inflammatory microenvironment contributes to tumour relapse after initially successful T-cell immunotherapy. On the basis of our work, we propose that future ACT protocols should simultaneously target melanocytic and non-melanocytic antigens to ensure broad recognition of both differentiated and dedifferentiated melanoma cells, and include strategies to sustain T-cell effector functions by blocking immune-inhibitory mechanisms in the tumour microenvironment.


Assuntos
Desdiferenciação Celular , Imunoterapia , Inflamação/patologia , Melanoma/patologia , Melanoma/terapia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/transplante , Transferência Adotiva , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Microambiente Tumoral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Antígeno gp100 de Melanoma/metabolismo
4.
J Mol Cell Biol ; 15(1)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36626927

RESUMO

Radiotherapy induces DNA damage, resulting in cell-cycle arrest and activation of cell-intrinsic death pathways. However, the radioresistance of some tumour entities such as malignant melanoma limits its clinical application. The innate immune sensing receptor retinoic acid-inducible gene I (RIG-I) is ubiquitously expressed and upon activation triggers an immunogenic form of cell death in a variety of tumour cell types including melanoma. To date, the potential of RIG-I ligands to overcome radioresistance of tumour cells has not been investigated. Here, we demonstrate that RIG-I activation enhanced the extent and immunogenicity of irradiation-induced tumour cell death in human and murine melanoma cells in vitro and improved survival in the murine B16 melanoma model in vivo. Transcriptome analysis pointed to a central role for p53, which was confirmed using p53-/- B16 cells. In vivo, the additional effect of RIG-I in combination with irradiation on tumour growth was absent in mice carrying p53-/- B16 tumours, while the antitumoural response to RIG-I stimulation alone was maintained. Our results identify p53 as a pivotal checkpoint that is triggered by RIG-I resulting in enhanced irradiation-induced tumour cell death. Thus, the combined administration of RIG-I ligands and radiotherapy is a promising approach to treating radioresistant tumours with a functional p53 pathway, such as melanoma.


Assuntos
Melanoma Experimental , Proteína Supressora de Tumor p53 , Animais , Camundongos , Humanos , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , Ligantes , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Imunoterapia/métodos , Melanoma Maligno Cutâneo
5.
Mol Ther Nucleic Acids ; 27: 1225-1234, 2022 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-35186439

RESUMO

The SARS-CoV-2 pandemic has underscored the need for rapidly usable prophylactic and antiviral treatments against emerging viruses. The targeted stimulation of antiviral innate immune receptors can trigger a broad antiviral response that also acts against new, unknown viruses. Here, we used the K18-hACE2 mouse model of COVID-19 to examine whether activation of the antiviral RNA receptor RIG-I protects mice from lethal SARS-CoV-2 infection and reduces disease severity. We found that prophylactic, systemic treatment of mice with the specific RIG-I ligand 3pRNA, but not type I interferon, 1-7 days before viral challenge, improved survival of mice by up to 50%. Survival was also improved with therapeutic 3pRNA treatment starting 1 day after viral challenge. This improved outcome was associated with lower viral load in oropharyngeal swabs and in the lungs and brains of 3pRNA-treated mice. Moreover, 3pRNA-treated mice exhibited reduced lung inflammation and developed a SARS-CoV-2-specific neutralizing antibody response. These results demonstrate that systemic RIG-I activation by therapeutic RNA oligonucleotide agonists is a promising strategy to convey effective, short-term antiviral protection against SARS-CoV-2 infection, and it has great potential as a broad-spectrum approach to constrain the spread of newly emerging viruses until virus-specific therapies and vaccines become available.

6.
Pharmacol Ther ; 228: 107931, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34171328

RESUMO

Infections of the Coronavirus SARS-CoV-2 continue to spread around the globe, causing Coronavirus Disease (COVID)-19. Infected people are at risk of developing acute interstitial pneumonia, which can result in lethal complications, particularly in patients with pre-existing co-morbidities. Novel prophylactic and therapeutic interventions are urgently needed to limit the infection-associated health risk for the population and to contain the pandemic. Animal models are indispensable to assessing the efficacy and safety of potential new antivirals, vaccines, and other innovative therapies, such as nucleic acid agonists of innate immune sensing receptors. In this review, we provide an overview of the commonly used animal models to study SARS-CoV-2 and COVID-19, including a summary of their susceptibility to infection, the spectrum of symptoms elicited, and the potential for drug development in each model. We hope that this review will help researchers to decide on the right model organism to quickly address their specific scientific questions.


Assuntos
Tratamento Farmacológico da COVID-19 , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Animais
7.
Nat Commun ; 6: 8755, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26530832

RESUMO

Inflammation promotes phenotypic plasticity in melanoma, a source of non-genetic heterogeneity, but the molecular framework is poorly understood. Here we use functional genomic approaches and identify a reciprocal antagonism between the melanocyte lineage transcription factor MITF and c-Jun, which interconnects inflammation-induced dedifferentiation with pro-inflammatory cytokine responsiveness of melanoma cells favouring myeloid cell recruitment. We show that pro-inflammatory cytokines such as TNF-α instigate gradual suppression of MITF expression through c-Jun. MITF itself binds to the c-Jun regulatory genomic region and its reduction increases c-Jun expression that in turn amplifies TNF-stimulated cytokine expression with further MITF suppression. This feed-forward mechanism turns poor peak-like transcriptional responses to TNF-α into progressive and persistent cytokine and chemokine induction. Consistently, inflammatory MITF(low)/c-Jun(high) syngeneic mouse melanomas recruit myeloid immune cells into the tumour microenvironment as recapitulated by their human counterparts. Our study suggests myeloid cell-directed therapies may be useful for MITF(low)/c-Jun(high) melanomas to counteract their growth-promoting and immunosuppressive functions.


Assuntos
Desdiferenciação Celular/genética , Citocinas/imunologia , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Células Mieloides/imunologia , Neoplasias Cutâneas/genética , Animais , Desdiferenciação Celular/imunologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Immunoblotting , Imuno-Histoquímica , Inflamação , Melanoma/imunologia , Camundongos , Fator de Transcrição Associado à Microftalmia/imunologia , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-jun , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Cutâneas/imunologia , Fator de Necrose Tumoral alfa/imunologia
8.
Cancer Discov ; 4(6): 674-87, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24589924

RESUMO

UNLABELLED: Infiltration of human melanomas with cytotoxic immune cells correlates with spontaneous type I IFN activation and a favorable prognosis. Therapeutic blockade of immune-inhibitory receptors in patients with preexisting lymphocytic infiltrates prolongs survival, but new complementary strategies are needed to activate cellular antitumor immunity in immune cell-poor melanomas. Here, we show that primary melanomas in Hgf-Cdk4(R24C) mice, which imitate human immune cell-poor melanomas with a poor outcome, escape IFN-induced immune surveillance and editing. Peritumoral injections of immunostimulatory RNA initiated a cytotoxic inflammatory response in the tumor microenvironment and significantly impaired tumor growth. This critically required the coordinated induction of type I IFN responses by dendritic, myeloid, natural killer, and T cells. Importantly, antibody-mediated blockade of the IFN-induced immune-inhibitory interaction between PD-L1 and PD-1 receptors further prolonged the survival. These results highlight important interconnections between type I IFNs and immune-inhibitory receptors in melanoma pathogenesis, which serve as targets for combination immunotherapies. SIGNIFICANCE: Using a genetically engineered mouse melanoma model, we demonstrate that targeted activation of the type I IFN system with immunostimulatory RNA in combination with blockade of immune-inhibitory receptors is a rational strategy to expose immune cell-poor tumors to cellular immune surveillance.


Assuntos
Interferon Tipo I/imunologia , Melanoma Experimental/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Antígeno B7-H1/imunologia , Carcinógenos , Imunoglobulina G/administração & dosagem , Fatores Imunológicos/administração & dosagem , Melanoma Experimental/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poli I-C/administração & dosagem , Receptor de Morte Celular Programada 1/imunologia , RNA/administração & dosagem , Microambiente Tumoral/imunologia
9.
PLoS One ; 8(8): e71057, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23940691

RESUMO

Therapeutic oligonucleotides including siRNA and immunostimulatory ligands of Toll-like receptors (TLR) or RIG-I like helicases (RLH) are a promising novel class of drugs. They are in clinical development for a broad spectrum of applications, e.g. as adjuvants in vaccines and for the immunotherapy of cancer. Species-specific immune activation leading to cytokine release is characteristic for therapeutic oligonucleotides either as an unwanted side effect or intended pharmacology. Reliable in vitro tests designed for therapeutic oligonucleotides are therefore urgently needed in order to predict clinical efficacy and to prevent unexpected harmful effects in clinical development. To serve this purpose, we here established a human whole blood assay (WBA) that is fast and easy to perform. Its response to synthetic TLR ligands (R848: TLR7/8, LPS: TLR4) was on a comparable threshold to the more time consuming peripheral blood mononuclear cell (PBMC) based assay. By contrast, the type I IFN profile provoked by intravenous CpG-DNA (TLR9 ligand) in humans in vivo was more precisely replicated in the WBA than in stimulated PBMC. Since Heparin and EDTA, but not Hirudin, displaced oligonucleotides from their delivery agent, only Hirudin qualified as the anticoagulant to be used in the WBA. The Hirudin WBA exhibited a similar capacity as the PBMC assay to distinguish between TLR7-activating and modified non-stimulatory siRNA sequences. RNA-based immunoactivating TLR7/8- and RIG-I-ligands induced substantial amounts of IFN-α in the Hirudin-WBA dependent on delivery agent used. In conclusion, we present a human Hirudin WBA to determine therapeutic oligonucleotide-induced cytokine release during preclinical development that can readily be performed and offers a close reflection of human cytokine response in vivo.


Assuntos
Citocinas/sangue , Oligodesoxirribonucleotídeos/farmacologia , RNA Interferente Pequeno/farmacologia , Anticoagulantes/farmacologia , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ácido Edético/farmacologia , Heparina/farmacologia , Humanos , Imidazóis/farmacologia , Interferon-alfa/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/farmacologia , Receptores Imunológicos , Receptor 4 Toll-Like/agonistas , Receptor 7 Toll-Like/agonistas , Transfecção
10.
Pigment Cell Melanoma Res ; 23(5): 649-60, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20649939

RESUMO

Genetically engineered mouse models offer new opportunities to investigate the role of cell-mediated immunity in the natural progression of melanoma in an immunocompetent host. Here we report that Hgf-Cdk4(R24C) mice spontaneously develop a spectrum of primary melanomas with high penetrance during their first year of life. Malignant transformation proceeds in a stepwise manner from multiple melanocytic nevi to single nodular melanomas and disseminated metastases in most mice. Migrating melanoma cells invade the draining lymph nodes without activating the immune system. Autochthonous primary tumors are destroyed following experimental introduction of immune surveillance using an adoptive lymphocyte transfer approach. However, some tumor cells are able to survive, evade immune cell control, and recur both locally and systemically. Immune tolerance in recurring tumors may be supported by immunosuppressive Gr1(+) myeloid cells. Taken together, our results demonstrate that primary and metastatic melanomas developing spontaneously in Hgf-Cdk4(R24C) mice effectively evade cellular immune surveillance.


Assuntos
Transformação Celular Neoplásica , Quinase 4 Dependente de Ciclina/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Vigilância Imunológica/imunologia , Melanoma , Neoplasias Cutâneas , Linfócitos T/imunologia , Animais , Quinase 4 Dependente de Ciclina/genética , Fator de Crescimento de Hepatócito/genética , Humanos , Linfonodos/imunologia , Linfonodos/patologia , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica , Nevo/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
11.
Cancer Res ; 69(15): 6265-74, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19622767

RESUMO

The development of therapeutic strategies which induce effective cellular antitumor immunity represents an important goal in cancer immunology. Here, we used the unique features of the genetically engineered Hgf-Cdk4(R24C) mouse model to identify a combination chemoimmunotherapy for melanoma. These mice develop primary cutaneous melanomas which grow progressively and metastasize in the absence of immunogenic foreign proteins such as oncogenes or antigens. Primary and metastatic tumors evade innate and adaptive immune defenses, although they naturally express melanocytic antigens which can be recognized by antigen-specific T cells. We found that primary melanomas continued to grow despite infiltration with adoptively transferred, in vivo-activated, tumor-specific CD8(+) T cells. To promote tumor immune defense, we developed a treatment protocol consisting of four complementary components: (a) chemotherapeutic preconditioning prior to (b) adoptive lymphocyte transfer and (c) viral vaccination followed by (d) adjuvant peritumoral injections of immunostimulatory nucleic acids. Lymphocyte ablation and innate antiviral immune stimulation cooperatively enhanced the expansion and the effector cell differentiation of adoptively transferred lymphocytes. The efficacy of the different treatment approaches converged in the tumor microenvironment and induced a strong cytotoxic inflammatory response enabling preferential recognition and destruction of melanoma cells. This combination chemoimmunotherapy caused complete regression of advanced primary melanomas in the skin and metastases in the lung with minimal autoimmune side effects. Our results in a clinically highly relevant experimental model provide a scientific rationale to evaluate similar strategies which unleash the power of innate and adaptive immune defense in future clinical trials.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/farmacologia , Ciclofosfamida/farmacologia , Imunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Linfócitos T/imunologia , Animais , Vacinas Anticâncer/imunologia , Terapia Combinada , Humanos , Imunoterapia Adotiva , Interferon Tipo I/farmacologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligonucleotídeos/imunologia , Oligonucleotídeos/farmacologia , Poli I-C/farmacologia , Proteínas Recombinantes , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia
12.
Nat Med ; 14(11): 1256-63, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18978796

RESUMO

Genetic and epigenetic plasticity allows tumors to evade single-targeted treatments. Here we direct Bcl2-specific short interfering RNA (siRNA) with 5'-triphosphate ends (3p-siRNA) against melanoma. Recognition of 5'-triphosphate by the cytosolic antiviral helicase retinoic acid-induced protein I (Rig-I, encoded by Ddx58) activated innate immune cells such as dendritic cells and directly induced expression of interferons (IFNs) and apoptosis in tumor cells. These Rig-I-mediated activities synergized with siRNA-mediated Bcl2 silencing to provoke massive apoptosis of tumor cells in lung metastases in vivo. The therapeutic activity required natural killer cells and IFN, as well as silencing of Bcl2, as evidenced by rescue with a mutated Bcl2 target, by site-specific cleavage of Bcl2 messenger RNA in lung metastases and downregulation of Bcl-2 protein in tumor cells in vivo. Together, 3p-siRNA represents a single molecule-based approach in which Rig-I activation on both the immune- and tumor cell level corrects immune ignorance and in which gene silencing corrects key molecular events that govern tumor cell survival.


Assuntos
RNA Helicases DEAD-box/metabolismo , Inativação Gênica , Melanoma/genética , Melanoma/metabolismo , Fosfatos/metabolismo , RNA Interferente Pequeno/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose/imunologia , Linhagem Celular Tumoral , Transplante de Células , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Melanoma/imunologia , Melanoma/patologia , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptores Imunológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA