Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 5(1): e1068493, 2016.
Article in English | MEDLINE | ID: mdl-26942088

ABSTRACT

Antitumor immunity can be enhanced by the coordinated release and delivery of antigens and immune-stimulating agents to antigen-presenting cells via biodegradable vaccine carriers. So far, encapsulation of TLR ligands and tumor-associated antigens augmented cytotoxic T cell (CTLs) responses. Here, we compared the efficacy of the invariant NKT (iNKT) cell agonist α-galactosylceramide (α-GalCer) and TLR ligands (R848 and poly I:C) as an adjuvant for the full length ovalbumin (OVA) in PLGA nanoparticles. We observed that OVA+α-GalCer nanoparticles (NP) are superior over OVA+TLR-L NP in generating and stimulating antigen-specific cytotoxic T lymphocytes without the need for CD4+ T cell help. Not only a 4-fold higher induction of antigen-specific T cells was observed, but also a more profound IFN-γ secretion was obtained by the addition α-GalCer. Surprisingly, we observed that mixtures of OVA containing NP with α-GalCer were ineffective, demonstrating that co-encapsulation of both α-GalCer and antigen within the same nanoparticle is essential for the observed T cell responses. Moreover, a single immunization with OVA+α-GalCer NP provided substantial protection from tumor formation and even delayed the growth of already established tumors, which coincided with a prominent and enhanced antigen-specific CD8+ T cell infiltration. The provided evidence on the advantage of antigen and α-GalCer coencapsulation should be considered in the design of future nanoparticle vaccines for therapeutic purposes.

2.
Eur J Immunol ; 45(10): 2798-809, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26255864

ABSTRACT

Novel approaches of dendritic cell (DC) based cancer immunotherapy aim at harnessing the unique attributes of different DC subsets. Classical monocyte-derived DC vaccines are currently being replaced by either applying primary DCs or specifically targeting antigens and adjuvants to these subsets in vivo. Appropriate DC activation in both strategies is essential for optimal effect. For this purpose TLR agonists are favorable adjuvant choices, with TLR7 triggering being essential for inducing strong Th1 responses. However, mouse CD8α(+) DCs, considered to be the major cross-presenting subset, lack TLR7 expression. Interestingly, this DC subset can respond to TLR7 ligand upon concurrent TLR3 triggering. Nevertheless, the mechanism underlying this synergy remains obscure. We now show that TLR3 ligation results in the production of IFN-α, which rapidly induces the expression of TLR7, resulting in synergistic activation. Moreover, we demonstrate that this mechanism conversely holds for plasmacytoid DCs that respond to TLR3 ligation when TLR7 pathway is mobilized. We further demonstrate that this mechanism of sharpening DC senses is also conserved in human BDCA1(+) DCs and plasmacytoid DCs. These findings have important implications for future clinical trials as it suggests that combinations of TLR ligands should be applied irrespective of initial TLR expression profiles on natural DC subsets for optimal stimulation.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Membrane Glycoproteins/agonists , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/agonists , Animals , Dendritic Cells/cytology , Humans , Membrane Glycoproteins/immunology , Mice , Th1 Cells/cytology , Th1 Cells/immunology , Toll-Like Receptor 7/immunology
3.
Cancer Immunol Immunother ; 64(5): 563-72, 2015 May.
Article in English | MEDLINE | ID: mdl-25687736

ABSTRACT

In around half of the patients with neuroblastoma (NBL), the primary tumor is located in one of the adrenal glands. We have previously reported on a transplantable TH-MYCN model of subcutaneous (SC) growing NBL in C57Bl/6 mice for immunological studies. In this report, we describe an orthotopic TH-MYCN transplantable model where the tumor cells were injected intra-adrenally (IA) by microsurgery. Strikingly, 9464D cells grew out much faster in IA tumors compared to the subcutis. Tumors were infiltrated by equal numbers of lymphocytes and myeloid cells. Within the myeloid cell population, however, tumor-infiltrating macrophages were more abundant in IA tumors compared to SC tumors and expressed lower levels of MHC class II, indicative of a more immunosuppressive phenotype. Using 9464D cells stably expressing firefly luciferase, enhanced IA tumor growth could be confirmed using bioluminescence. Collectively, these data show that the orthotopic IA localization of TH-MYCN cells impacts the NBL tumor microenvironment, resulting in a more stringent NBL model to study novel immunotherapeutic approaches for NBL.


Subject(s)
Adrenal Gland Neoplasms/pathology , Macrophages/immunology , Neoplasms, Experimental/pathology , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Tumor Microenvironment/immunology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/therapy , Adrenal Glands/cytology , Adrenal Glands/immunology , Adrenal Glands/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Luminescent Measurements , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Neuroblastoma/genetics , Neuroblastoma/therapy , Nuclear Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Subcutaneous Tissue/immunology , Subcutaneous Tissue/pathology
4.
Mol Immunol ; 50(1-2): 66-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22209087

ABSTRACT

Dendritic cells (DCs) are professional antigen presenting cells of the immune system that play a crucial role in initiating immune responses and maintaining self tolerance. Better understanding of the molecular basis of DC immunobiology is required to improve DC-based immunotherapies. We previously described the interaction of transcription factor LUMAN (also known as CREB3 or LZIP) with the DC-specific transmembrane protein DC-STAMP in DCs. Target genes of LUMAN and its role in DCs are currently unknown. In this study we set out to identify genes regulated by LUMAN in DCs using microarray analysis. Expression of a constitutively active form of LUMAN in mouse DC cell line D2SC/1 identified Apolipoprotein A4 (ApoA4) as its target gene. Subsequent validation experiments, bioinformatics-based promoter analysis, and silencing studies confirmed that ApoA4 is a true target gene of LUMAN in bone marrow-derived DCs (BMDCs).


Subject(s)
Apolipoproteins A/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Dendritic Cells/metabolism , Gene Expression Regulation , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Brefeldin A/pharmacology , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Female , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Protein Synthesis Inhibitors/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
5.
Blood ; 118(26): 6836-44, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-21967977

ABSTRACT

Effective vaccines consist of 2 components: immunodominant antigens and effective adjuvants. Whereas it has been demonstrated that targeted delivery of antigens to dendritic cells (DCs) improves vaccine efficacy, we report here that co-targeting of TLR ligands (TLRLs) to DCs strongly enhances adjuvanticity and immunity. We encapsulated ligands for intracellular TLRs within biodegradable nanoparticles coated with Abs recognizing DC-specific receptors. Targeted delivery of TLRLs to human DCs enhanced the maturation and production of immune stimulatory cytokines and the Ag-specific activation of naive CD8(+) T cells. In vivo studies demonstrated that nanoparticles carrying Ag induced cytotoxic T-lymphocyte responses at 100-fold lower adjuvant dose when TLRLs were co-encapsulated instead of administered in soluble form. Moreover, the efficacy of these targeted TLRLs reduced the serum cytokine storm and related toxicity that is associated with administration of soluble TLRLs. We conclude that the targeted delivery of adjuvants may improve the efficacy and safety of DC-based vaccines.


Subject(s)
Dendritic Cells/immunology , Ligands , Toll-Like Receptors/immunology , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Dendritic Cells/metabolism , Drug Delivery Systems/methods , Female , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Monocytes/immunology , Monocytes/metabolism , Nanoparticles/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Toll-Like Receptors/metabolism , Vaccines/administration & dosage
6.
J Immunol ; 187(3): 1281-8, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705625

ABSTRACT

Effective antitumor immunotherapy requires the identification of suitable target Ags. Interestingly, many of the tumor Ags used in clinical trials are present in preparations of secreted tumor vesicles (exosomes). In this study, we compared T cell responses elicited by murine MCA101 fibrosarcoma tumors expressing a model Ag at different localizations within the tumor cell in association with secreted vesicles (exosomes), as a nonsecreted cell-associated protein, or as secreted soluble protein. Remarkably, we demonstrated that only the tumor-secreting vesicle-bound Ag elicited a strong Ag-specific CD8(+) T cell response, CD4(+) T cell help, Ag-specific Abs, and a decrease in the percentage of immunosuppressive regulatory T cells in the tumor. Moreover, in a therapeutic tumor model of cryoablation, only in tumors secreting vesicle-bound Ag could Ag-specific CD8(+) T cells still be detected up to 16 d after therapy. We concluded that the localization of an Ag within the tumor codetermines whether a robust immunostimulatory response is elicited. In vivo, vesicle-bound Ag clearly skews toward a more immunogenic phenotype, whereas soluble or cell-associated Ag expression cannot prevent or even delay outgrowth and results in tumor tolerance. This may explain why particular immunotherapies based on these vesicle-bound tumor Ags are potentially successful. Therefore, we conclude that this study may have significant implications in the discovery of new tumor Ags suitable for immunotherapy and that their location should be taken into account to ensure a strong antitumor immune response.


Subject(s)
Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fibrosarcoma/immunology , Ovalbumin/metabolism , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Exosomes/genetics , Exosomes/immunology , Exosomes/metabolism , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/biosynthesis , Ovalbumin/genetics , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Transfection
7.
BMC Genomics ; 12: 330, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21708028

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play a fundamental role in the regulation of gene expression by translational repression or target mRNA degradation. Regulatory elements in miRNA promoters are less well studied, but may reveal a link between their expression and a specific cell type. RESULTS: To explore this link in myeloid cells, miRNA expression profiles were generated from monocytes and dendritic cells (DCs). Differences in miRNA expression among monocytes, DCs and their stimulated progeny were observed. Furthermore, putative promoter regions of miRNAs that are significantly up-regulated in DCs were screened for Transcription Factor Binding Sites (TFBSs) based on TFBS motif matching score, the degree to which those TFBSs are over-represented in the promoters of the up-regulated miRNAs, and the extent of conservation of the TFBSs in mammals. CONCLUSIONS: Analysis of evolutionarily conserved TFBSs in DC promoters revealed preferential clustering of sites within 500 bp upstream of the precursor miRNAs and that many mRNAs of cognate TFs of the conserved TFBSs were indeed expressed in the DCs. Taken together, our data provide evidence that selected miRNAs expressed in DCs have evolutionarily conserved TFBSs relevant to DC biology in their promoters.


Subject(s)
Dendritic Cells/metabolism , MicroRNAs/metabolism , Binding Sites , Cells, Cultured , Cluster Analysis , Dendritic Cells/cytology , Evolution, Molecular , Gene Expression Profiling , Humans , MicroRNAs/genetics , Monocytes/cytology , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transcription Factors/metabolism , Up-Regulation
8.
Mol Immunol ; 47(11-12): 1963-73, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20546900

ABSTRACT

Dendritic cells (DCs) are the professional antigen-presenting cells (APC) which efficiently prime the immune response or induce tolerance. We recently identified Dendritic Cell Specific TrAnsMembrane Protein (DC-STAMP), a novel 470 amino acid protein preferentially expressed by dendritic cells. Previously we demonstrated that DC-STAMP re-localizes towards the Golgi upon DC maturation. To identify proteins that interact with DC-STAMP, a yeast-2-hybrid analysis was performed. Here, we report a physically interacting partner of DC-STAMP in the endoplasmic reticulum (ER), called LUMAN (also known as CREB3 or LZIP). LUMAN was previously described as an ER-resident transcription factor with unknown function. It is activated in a process called regulated intramembrane proteolysis (RIP), which involves translocation to the Golgi and subsequent proteolytic cleavage. The proteolytically activated form of the protein then translocates to the nucleus. Our data indicate that DC-STAMP plays an important role in the modulation of LUMAN activation. Moreover, we demonstrate that LUMAN is endogenously expressed by DC and becomes activated by RIP upon DC maturation induced by various different stimuli. These data define LUMAN/DC-STAMP as a novel regulatory circuit in DC.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Dendritic Cells/physiology , Membrane Proteins/physiology , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cyclic AMP Response Element-Binding Protein/genetics , Humans , Membrane Proteins/genetics , Protein Transport , RNA, Messenger/analysis
9.
Blood ; 116(4): 564-74, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20424184

ABSTRACT

Currently dendritic cell (DC)-based vaccines are explored in clinical trials, predominantly in cancer patients. Murine studies showed that only maturation with Toll-like receptor (TLR) ligands generates mature DCs that produce interleukin-12 and promote optimal T-cell help. Unfortunately, the limited availability of clinical-grade TLR ligands significantly hampers the translation of these findings into DC-based vaccines. Therefore, we explored 15 commonly used preventive vaccines as a possible source of TLR ligands. We have identified a cocktail of the vaccines BCG-SSI, Influvac, and Typhim that contains TLR ligands and is capable of optimally maturing DCs. These DCs (vaccine DCs) showed high expression of CD80, CD86, and CD83 and secreted interleukin-12. Although vaccine DCs exhibited an impaired migratory capacity, this could be restored by addition of prostaglandin E(2) (PGE(2); vaccine PGE(2) DCs). Vaccine PGE(2) DCs are potent inducers of T-cell proliferation and induce Th1 polarization. In addition, vaccine PGE(2) DCs are potent inducers of tumor antigen-specific CD8(+) effector T cells. Finally, vaccine PGE(2)-induced DC maturation is compatible with different antigen-loading strategies, including RNA electroporation. These data thus identify a new clinical application for a mixture of commonly used preventive vaccines in the generation of Th1-inducing clinical-grade mature DCs.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/drug effects , Monocytes/drug effects , Toll-Like Receptors , Vaccines/pharmacology , BCG Vaccine/immunology , BCG Vaccine/pharmacology , Cell Differentiation/immunology , Cell Movement/drug effects , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Dinoprostone/pharmacology , Diphtheria-Tetanus-acellular Pertussis Vaccines/immunology , Diphtheria-Tetanus-acellular Pertussis Vaccines/pharmacology , Drug Evaluation, Preclinical , Humans , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Interleukin-12/metabolism , Ligands , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , Monocytes/physiology , Polysaccharides, Bacterial/immunology , Polysaccharides, Bacterial/pharmacology , Preventive Medicine , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Typhoid-Paratyphoid Vaccines/immunology , Typhoid-Paratyphoid Vaccines/pharmacology , Vaccines/immunology , Vaccines, Synthetic/metabolism , Vaccines, Synthetic/pharmacology
10.
J Innate Immun ; 2(4): 360-70, 2010.
Article in English | MEDLINE | ID: mdl-20375561

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells that provide a link between innate and adaptive immunity. Multiple DC subsets exist and their activation by microorganisms occurs through binding of conserved pathogen-derived structures to so-called pattern recognition receptors (PRRs). In this study we analyzed the expression of PRRs responding to viral RNA in human monocyte-derived DCs (moDCs) under steady-state or pro-inflammatory conditions. We found that mRNA and protein levels for most PRRs were increased under pro-inflammatory conditions, with the most pronounced increases in the RIG-like helicase (RLH) family. Additionally, freshly isolated human plasmacytoid DCs (pDCs) displayed significantly higher levels of TLR7, RIG-I, MDA5 and PKR as compared to myeloid DCs and moDCs. Finally, we demonstrate for the first time that cross-talk between TLR-matured or virus-stimulated pDCs and moDCs leads to a type I interferon-dependent antiviral state in moDCs. This antiviral state was characterized by enhanced RLH expression and protection against picornavirus infection. These findings might represent a novel mechanism by which pDCs can preserve the function and viability of myeloid DCs that are attracted to a site with ongoing infection, thereby optimizing the antiviral immune response.


Subject(s)
Cell Communication , Dendritic Cells , Picornaviridae Infections/immunology , RNA/metabolism , Receptors, Pattern Recognition/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/physiology , Humans , Monocytes/cytology , Monocytes/immunology , Picornaviridae/pathogenicity , RNA/genetics , Receptors, Pattern Recognition/genetics
11.
Cancer Immunol Immunother ; 58(7): 1109-15, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19018531

ABSTRACT

Tumor-derived peptides are used frequently as antigen (Ag) source in dendritic cell (DC) therapy in cancer patients. An alternative is to load DC with tumor-associated Ag (TAA)-encoding RNA. RNA-loading obviates prior knowledge of CTL and Th epitopes in the Ag. Multiple epitopes for many HLA alleles (both MHC class I and class II) are encoded by the RNA and loading is independent of the patient's HLA make-up. Herein, we determined the optimal conditions for mRNA-electroporation of monocyte-derived DC for clinical application in relation to different maturation cocktails. The data demonstrate that TAA carcinoembryonic antigen, gp100 and tyrosinase are expressed already 30 min after electroporation with the encoding mRNA. Moreover, gp100-specific CTL are activated by gp100 mRNA-electroporated DC. Importantly, we show here that the presence of polyinosinic-polycytidylic acid [poly(I:C)] in the maturation cocktail prevents effective protein expression of the electroporated mRNA as well as subsequent CTL recognition. This effect of poly(I:C) correlates with the induction of IFN-induced genes and innate anti-viral effector molecules in DC. Together these data show that electroporation of mature DC with TAA-encoding mRNA is attractive for use in DC vaccination protocols in cancer patients, but protein expression should be tested for each maturation cocktail.


Subject(s)
Antigen Presentation/drug effects , Antigens, Neoplasm/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interferon Inducers/pharmacology , Poly I-C/pharmacology , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Dendritic Cells/transplantation , Electroporation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/immunology , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , gp100 Melanoma Antigen
12.
Mol Immunol ; 46(4): 505-15, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18952287

ABSTRACT

Dendritic cell-specific transmembrane protein (DC-STAMP) has been first identified as an EST in a cDNA library of human monocyte-derived dendritic cells (DC). DC-STAMP is a multimembrane spanning protein that has been implicated in skewing haematopoietic differentiation of bone marrow cells towards the myeloid lineage, and in cell fusion during osteoclastogenesis and giant cell formation. To gain molecular insight in how DC-STAMP exerts its function, DC-STAMP interacting proteins were identified in a yeast-2-hybrid analysis. Herein, we report that amplified in osteosarcoma 9 (OS9) physically interacts with DC-STAMP, and that both proteins colocalize in the endoplasmic reticulum in various cell lines, including immature DC. OS9 has previously been implicated in ER-to-Golgi transport and transcription factor turnover. Interestingly, we now demonstrate that toll-like receptor (TLR)-induced maturation of DC leads to the translocation of DC-STAMP from the ER to the Golgi while OS9 localization is unaffected. Applying TLR-expressing CHO cells we could confirm ER-to-Golgi translocation of DC-STAMP following TLR stimulation and demonstrated that the DC-STAMP/OS9 interaction is involved in this process. Collectively, the data indicate that OS9 is critically involved in the modulation of ER-to-Golgi transport of DC-STAMP in response to TLR triggering, suggesting a novel role for OS9 in myeloid differentiation and cell fusion.


Subject(s)
Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , CHO Cells , Cricetinae , Cricetulus , Dendritic Cells/cytology , Dendritic Cells/immunology , Endoplasmic Reticulum/metabolism , Humans , Lectins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Mutant Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Protein Transport/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Sequence Deletion/genetics , Sequence Deletion/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...