Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
NPJ Vaccines ; 5(1): 5, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31969991

RESUMO

Glioblastoma is the most prevalent and aggressive brain cancer. With a median overall survival of ~15-20 months under standard therapy, novel treatment approaches are desperately needed. A recent phase II clinical trial with a personalized immunotherapy based on tumor lysate-charged dendritic cell (DC) vaccination, however, failed to prolong survival. Here, we investigated tumor tissue from trial patients to explore glioblastoma survival-related factors. We followed an innovative approach of combining mass spectrometry-based quantitative proteomics (n = 36) with microRNA sequencing plus RT-qPCR (n = 38). Protein quantification identified, e.g., huntingtin interacting protein 1 (HIP1), retinol-binding protein 1 (RBP1), ferritin heavy chain (FTH1) and focal adhesion kinase 2 (FAK2) as factor candidates correlated with a dismal prognosis. MicroRNA analysis identified miR-216b, miR-216a, miR-708 and let-7i as molecules potentially associated with favorable tissue characteristics as they were enriched in patients with a comparably longer survival. To illustrate the utility of integrated miRNomics and proteomics findings, focal adhesion was studied further as one example for a pathway of potential general interest. Taken together, we here mapped possible drivers of glioblastoma outcome under immunotherapy in one of the largest DC vaccination tissue analysis cohorts so far-demonstrating usefulness and feasibility of combined proteomics/miRNomics approaches. Future research should investigate agents that sensitize glioblastoma to (immuno)therapy-potentially building on insights generated here.

2.
Acta Neuropathol Commun ; 6(1): 135, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518425

RESUMO

Audencel is a dendritic cell (DC)-based cellular cancer immunotherapy against glioblastoma multiforme (GBM). It is characterized by loading of DCs with autologous whole tumor lysate and in vitro maturation via "danger signals". The recent phase II "GBM-Vax" trial showed no clinical efficacy for Audencel as assessed with progression-free and overall survival in all patients. Here we present immunological research accompanying the trial with a focus on immune system factors related to outcome and Audencel's effect on the immune system. Methodologically, peripheral blood samples (from apheresis before Audencel or venipuncture during Audencel) were subjected to functional characterization via enzyme-linked immunospot (ELISPOT) assays connected with cytokine bead assays (CBAs) as well as phenotypical characterization via flow cytometry and mRNA quantification. GBM tissue samples (from surgery) were subjected to T cell receptor sequencing and immunohistochemistry. As results we found: Patients with favorable pre-existing anti-tumor characteristics lived longer under Audencel than Audencel patients without them. Pre-vaccination blood CD8+ T cell count and ELISPOT Granzyme B production capacity in vitro upon tumor antigen exposure were significantly correlated with overall survival. Despite Audencel's general failure to induce a significant clinical response, it nevertheless seemed to have an effect on the immune system. For instance, Audencel led to a significant up-regulation of the Th1-related immunovariables ELISPOT IFNγ, the transcription factor T-bet in the blood and ELISPOT IL-2 in a dose-dependent manner upon vaccination. Post-vaccination levels of ELISPOT IFNγ and CD8+ cells in the blood were indicative of a significantly better survival. In summary, Audencel failed to reach an improvement of survival in the recent phase II clinical trial. No clinical efficacy was registered. Our concomitant immunological work presented here indicates that outcome under Audencel was influenced by the state of the immune system. On the other hand, Audencel also seemed to have stimulated the immune system. Overall, these immunological considerations suggest that DC immunotherapy against glioblastoma should be studied further - with the goal of translating an apparent immunological response into a clinical response. Future research should concentrate on investigating augmentation of immune reactions through combination therapies or on developing meaningful biomarkers.


Assuntos
Neoplasias Encefálicas/terapia , Linfócitos T CD8-Positivos/fisiologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/fisiologia , Glioblastoma/terapia , Antígenos CD/metabolismo , Compostos de Boro/metabolismo , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Feminino , Glioblastoma/sangue , Glioblastoma/imunologia , Humanos , Estimativa de Kaplan-Meier , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Estudos Longitudinais , Masculino , Resultado do Tratamento , Regulação para Cima
3.
Cell Immunol ; 325: 33-40, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29402391

RESUMO

Gangliosides shed by tumors into their microenvironment (TME) are immunoinhibitory. Interferon-γ (IFN-γ) may boost antitumor immune responses. Thus we wondered whether IFN-γ would counteract tumor ganglioside-mediated immune suppression. To test this hypothesis, we exposed human monocyte-derived LPS-activated dendritic cells (DC) to IFN-γ and to a highly purified ganglioside, GD1a. DC ganglioside exposure decreased TLR-dependent p38 signaling, explaining the previously observed ganglioside-induced down-modulation of pro-inflammatory surface markers and cytokines. Strikingly, while increasing LPS-dependent DC responses, IFN-γ unexpectedly did not counteract the inhibitory effects of GD1a. Rather, induction of indoleamine 2,3-dioxygenase (IDO1), and expression of STAT1/IRF-1 and programmed cell death ligand (PD-L1), indicated that the immunoinhibitory, not an immune stimulatory, IFN-γ-signaling axis, was active. The combination, IFN-γ and DC ganglioside enrichment, markedly impaired DC stimulatory potential of CD8+ T-cells. We suggest that gangliosides and IFN-γ may act in concert as immunosuppressive mediators in the TME, possibly promoting tumor progression.


Assuntos
Gangliosídeos/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Apoptose/imunologia , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Gangliosídeos/metabolismo , Voluntários Saudáveis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Neoplasias/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/metabolismo , Microambiente Tumoral/imunologia
4.
Sci Rep ; 7(1): 11746, 2017 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-28924177

RESUMO

Maintaining dendritic cells (DC) in a state of dysfunction represents a key mechanism by which tumour cells evade recognition and elimination by the immune system. Limited knowledge about the intracellular mediators of DC dysfunction restricts success of therapies aimed at reactivating a DC-driven anti-tumour immune response. Using a cell type-specific murine knock-out model, we have identified MAPK-activated protein kinase 2 (MK2) as a major guardian of a suppressive DC phenotype in the melanoma tumour microenvironment. MK2 deletion in CD11c+ cells led to an expansion of stimulatory CD103+ DCs, mounting a potent CD8+ T cell response that resulted in elimination of highly aggressive B16-F10 tumours upon toll-like receptor (TLR) activation in the presence of tumour antigen. Moreover, tumour infiltration by suppressive myeloid cells was strongly diminished. These insights into the regulation of DC functionality reveal MK2 as a targetable pathway for DC-centred immunomodulatory cancer therapies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Imunidade Celular , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Melanoma Experimental/imunologia , Proteínas Serina-Treonina Quinases/deficiência , Microambiente Tumoral , Animais , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Células Dendríticas/enzimologia , Células Dendríticas/patologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Melanoma Experimental/enzimologia , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
5.
J Immunol ; 195(6): 2560-70, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26246144

RESUMO

The PI3K signaling cascade in APCs has been recognized as an essential pathway to initiate, maintain, and resolve immune responses. In this study, we demonstrate that a cell type-specific loss of the PI3K antagonist phosphatase and tensin homolog (PTEN) in myeloid cells renders APCs toward a regulatory phenotype. APCs deficient for PTEN exhibit reduced activation of p38 MAPK and reduced expression of T cell-polarizing cytokines. Furthermore, PTEN deficiency leads to upregulation of markers for alternative activation, such as Arginase 1, with concomitant downregulation of inducible NO synthase in APCs in vitro and in vivo. As a result, T cell polarization was dysfunctional in PTEN(-/-) APCs, in particular affecting the Th17 cell subset. Intriguingly, mice with cell type-specific deletions of PTEN-targeting APCs were protected from experimental autoimmune encephalomyelitis, which was accompanied by a pronounced reduction of IL-17- and IL-22-producing autoreactive T cells and reduced CNS influx of classically activated monocytes/macrophages. These observations support the notion that activation of the PI3K signaling cascade promotes regulatory APC properties and suppresses pathogenic T cell polarization, thereby reducing the clinical symptoms and pathology of experimental autoimmune encephalomyelitis.


Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , PTEN Fosfo-Hidrolase/genética , Células Th17/imunologia , Animais , Arginase/biossíntese , Autoimunidade/imunologia , Antígeno CD11c/biossíntese , Diferenciação Celular/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Interleucina-17/biossíntese , Interleucinas/biossíntese , Ativação Linfocitária , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/imunologia , Óxido Nítrico Sintase Tipo II/biossíntese , Fragmentos de Peptídeos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Interleucina 22
6.
J Immunol ; 195(2): 541-52, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26078274

RESUMO

Dendritic cell (DC)-mediated inflammation induced via TLRs is promoted by MAPK-activated protein kinase (MK)-2, a substrate of p38 MAPK. In this study we show an opposing role of MK2, by which it consolidates immune regulatory functions in DCs through modulation of p38, ERK1/2-MAPK, and STAT3 signaling. During primary TLR/p38 signaling, MK2 mediates the inhibition of p38 activation and positively cross-regulates ERK1/2 activity, leading to a reduction of IL-12 and IL-1α/ß secretion. Consequently, MK2 impairs secondary autocrine IL-1α signaling in DCs, which further decreases the IL-1α/p38 but increases the anti-inflammatory IL-10/STAT3 signaling route. Therefore, the blockade of MK2 activity enables human and murine DCs to strengthen proinflammatory effector mechanisms by promoting IL-1α-mediated Th1 effector functions in vitro. Furthermore, MK2-deficient DCs trigger Th1 differentiation and Ag-specific cytotoxicity in vivo. Finally, wild-type mice immunized with LPS in the presence of an MK2 inhibitor strongly accumulate Th1 cells in their lymph nodes. These observations correlate with a severe clinical course in DC-specific MK2 knockout mice compared with wild-type littermates upon induction of experimental autoimmune encephalitis. Our data suggest that MK2 exerts a profound anti-inflammatory effect that prevents DCs from prolonging excessive Th1 effector T cell functions and autoimmunity.


Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Células Th1/imunologia , Animais , Diferenciação Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica , Humanos , Imunização , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/patologia , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Transdução de Sinais , Células Th1/efeitos dos fármacos , Células Th1/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
7.
J Immunol ; 193(4): 1717-27, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25015834

RESUMO

The activation of innate immune cells triggers numerous intracellular signaling pathways, which require tight control to mount an adequate immune response. The PI3K signaling pathway is intricately involved in innate immunity, and its activation dampens the expression and release of proinflammatory cytokines in myeloid cells. These signaling processes are strictly regulated by the PI3K antagonist, the lipid phosphatase, PTEN, a known tumor suppressor. Importantly, PTEN is responsible for the elevated production of cytokines such as IL-6 in response to TLR agonists, and deletion of PTEN results in diminished inflammatory responses. However, the mechanisms by which PI3K negatively regulates TLR signaling are only partially resolved. We observed that Arginase I expression and secretion were markedly induced by PTEN deletion, suggesting PTEN(-/-) macrophages were alternatively activated. This was mediated by increased expression and activation of the transcription factors C/EBPß and STAT3. Genetic and pharmacologic experimental approaches in vitro, as well as in vivo autoimmunity models, provide convincing evidence that PI3K/PTEN-regulated extracellular Arginase I acts as a paracrine regulator of inflammation and immunity.


Assuntos
Arginase/metabolismo , Macrófagos/imunologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/imunologia , Imunidade Adaptativa , Animais , Arginase/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Genótipo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Células HEK293 , Humanos , Imunidade Inata , Inflamação/genética , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/enzimologia , Células Mieloides/imunologia , Inibidores de Fosfoinositídeo-3 Quinase , RNA Mensageiro/biossíntese , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese
8.
PLoS One ; 8(2): e54879, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23408948

RESUMO

The dendritic cell (DC) coordinates innate and adaptive immunity to fight infections and cancer. Our observations reveal that DCs exposed to the microbial danger signal lipopolysaccharide (LPS) in the presence of interferon-γ (IFN-γ) acquire a continuously changing activation/maturation phenotype. The DCs' initial mode of action is pro-inflammatory via up-regulation among others of the signaling molecule interleukin (IL) 12, which polarizes IFN-γ secreting type 1 helper T-cells (Th1). Within 24 hours the same DC switches from the pro- into an anti-inflammatory phenotype. This is mediated by autocrine IL-10 release and secretion of soluble IL-2 receptor alpha (sIL-2RA) molecules. T-cells, when contacted with DCs during their anti-inflammatory phase loose their proliferative capacity and develop regulatory T-cell (Treg) -like anti-inflammatory functions indicated by IL-10 secretion and elevated FoxP3 levels. Studying the kinetics of IL-12 and IL-10 expression from LPS/IFN-γ activated myeloid DCs on a single cell level confirmed these observations. When T-cells are separated from DCs within 24 hours, they are spared from the anti-inflammatory DC activity. We conclude that, in addition to differentiation of DCs into distinct subsets, the observed sequential functional phases of DC differentiation permit the fine-tuning of an immune response. A better understanding of time-kinetic DC features is required for optimally exploiting the therapeutic capacity of DCs in cancer immune therapy.


Assuntos
Diferenciação Celular/fisiologia , Células Dendríticas/citologia , Inflamação/fisiopatologia , Receptor 4 Toll-Like/fisiologia , Animais , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Camundongos
9.
J Immunother ; 33(1): 40-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19952957

RESUMO

In cancer patients undergoing immune therapy with lipopolysaccharides/interferon-gamma activated interleukin (IL)-12 secreting dendritic cells (DCs) we observed enhanced proliferative capacity of pyrophosphate-responsive peripheral blood (PB) gammadelta T-cells. This was not noted before as in other clinical trials DCs were used that were not enabled for IL-12 secretion and mice do not have a corresponding subset of PB gammadelta T-cells. In vitro examination of IL-12 DC/PB gammadelta T-cell interactions revealed a potential of PB gammadelta T-cells to negatively regulate the proliferative capacity of CD4 and CD8 T-cells. We further demonstrate that IL-12 is critical in the activation of PB gammadelta T-cells. In contrast, the regulatory activity of PB gammadelta T-cells in immune responses against strong recall antigens or alloantigens did not require activated DCs, but depended on pyrophosphate activation of PB gammadelta T-cells. Depletion of PB gammadelta T-cells abrogated the regulatory activity in IL-12 DC/peripheral blood mononuclear cell cocultures; adding back graded numbers of PB gammadelta T-cells restored it. Our observations revealed a potential PB gammadelta T cell-mediated negative regulatory feedback mechanism triggered by IL-12 DCs, which may critically impact on the design of DC cancer vaccines.


Assuntos
Células Dendríticas/transplante , Interleucina-12/imunologia , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Subpopulações de Linfócitos T/imunologia , Vacinas Anticâncer/imunologia , Ensaios Clínicos Fase I como Assunto , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Interleucina-12/metabolismo
10.
Blood ; 109(6): 2607-10, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17095626

RESUMO

The TEL/AML1 fusion gene results from the most frequent t(12;21)(p13;q22) translocation in childhood acute lymphoblastic leukemia (ALL). Its contribution to transformation is largely unknown, in particular with respect to survival and apoptosis. We therefore silenced TEL/AML1 expression in leukemic REH cells by RNA inhibition, which eventually led to programmed cell death. Microarray and 2D gel electrophoresis data demonstrated a differential regulation of heat-shock proteins (HSPs), among them HSP90, as well as of its client, survivin. Consistent with these findings, ectopic expression of TEL/AML1 in Ba/F3 cells increased protein levels of HSP90 and survivin and conferred resistance to apoptotic stimuli. Our data suggest that TEL/AML1 not only contributes to leukemogenesis by affecting an antiapoptotic network but also seems to be indispensable for maintaining the malignant phenotype. The functional relationship between TEL/AML1, HSP90, and survivin provides the rational for targeted therapy, be it the fusion gene or the latter 2 proteins.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Interferência de RNA , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Inibidoras de Apoptose , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA